You are on page 1of 14

Blood Cells, Molecules and Diseases 86 (2021) 102487

Contents lists available at ScienceDirect

Blood Cells, Molecules and Diseases


journal homepage: www.elsevier.com/locate/bcmd

CD133+CD34+ cells can give rise to EPCs: A comparative rabbit and human T
study
Hitesh Chopra, Yuanyuan Han, Chengfei Zhang, Edmond Ho Nang Pow

Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China

ARTICLE INFO

Editor: Mohandas Narla

1. Introduction morphology, whereas transformed EPCs (tEPCs) for the spindle-shaped


cells instead of eEPCs and endeavor to explain the reason behind it
Blood has been the center stage for assessing the pathophysiological based on our findings and previous studies.
functions of the human body for many decades. The reporting of EPCs In translational research, EPCs were isolated and characterized from
has triggered a mammoth investigation by clinical researchers globally various animals such as rats [20], rhesus monkeys [21], broiler
[1]. Encouraging results in human and experimental trials expanded the chickens [22], baboons [23], and pigs [24]. Although in 35% of the
role of EPCs in physiological and pathophysiological angiogenesis for musculoskeletal research [25] rabbits were used, rEPCs were only iso­
therapeutic use in various diseases such as diabetes [2], cancer [3], and lated and partially characterized as a prerequisite for their use in the
cardiovascular disorders [4]. Concomitant with their potential for experiments [26,27]. Studies comparing EPCs of rabbits with the
vascularization, EPCs have also been used extensively in tissue en­ human were lacking.
gineering. Tissue-engineered grafts pre-vascularized with EPCs can Therefore, to our knowledge, this is the first study to characterize
minimize cell death and improve integration to enhance graft survival rabbit EPCs in the full spectrum. Further, we hypothesized that the
[5]. rEPCs were similar to hEPCs regarding the proliferative potential,
Classically EPCs have been isolated from MNCs present in the per­ functional characterization, and the expression of surface markers.
ipheral blood using various isolation methods [6]. However, the phy­ Hence, rEPCs could provide an excellent research model for transla­
logenetic origin of these EPCs have shrouded a laudable controversy. tional research in medicine. Additionally, we also believe this is the first
Asahara et al. described “Blood-island” like morphologic pattern in the study to report that EPCs might co-exist as a subset within MNCs. Our
emergence of these EPCs with central round cells and peripheral spindle findings will be beneficial to researchers and the scientific community.
cells [1]. These spindle-shaped cells were later referred to as either
circulating endothelial cells (CACs) [7] or early EPCs (eEPCs) [8]. 2. Material and methods
Further studies revealed two distinct populations of EPCs based on the
timing of emergence in the MNC culture, eEPCs and late EPCs (lEPCs) 2.1. Blood sampling
[8,9]. In yet other studies, these lEPCs were also referred to as out­
growth endothelial cells OECs [10] or endothelial colony-forming cells 2.1.1. Rabbit blood sampling
(ECFCs) [11] having a characteristic cobblestone morphology. Al­ The committee on the use of live animals in teaching and research
though both eEPCs and late lEPCs share some standard features like the (CULATR: 3558-15) of The University of Hong Kong (HKU) approved
expression of CD31, CD34, DiIacLDL (1,1′-dioctadecyl-3,3,3′,3′-tetra­ 25 healthy New Zealand white male rabbits (≈9-month-old and
methyindocarbocyanide-labeled low-density lipoprotein) uptake, and weighing ≈ 4 kg) for the study. Under topical anesthesia, 30–100 ml of
UEA-1 lectin (Ulex europaeus agglutinin-1) binding, they also have blood was extracted from the marginal ear vein of the rabbit [28,29],
distinct attributes concerning their phenotype, morphology, pro­ followed by heparinization with heparin sodium (150 IU/ml; Leo
liferative potential, tubulogenic potential in vitro and mechanism of pharma, Denmark). The first 5 ml of collected blood contaminated with
neovascularization in vivo [7–19]. To avoid confusion in the present mature endothelial cells (ECs) was discarded. After selection, sample
study, we will use the term EPCs for the cells with cobblestone collection, and initial processing in the Laboratory animal unit (LAU)


Corresponding author at: 3/F, The Prince Philip Dental Hospital, 34 Hospital Road, Sai Ying Pun, Hong Kong.
E-mail addresses: u3002631@connect.hku.hk (H. Chopra), u3006886@connect.hku.hk (Y. Han), zhangcf@hku.hk (C. Zhang), ehnpow@hku.hk (E.H.N. Pow).

https://doi.org/10.1016/j.bcmd.2020.102487
Received 28 July 2020; Received in revised form 17 August 2020; Accepted 18 August 2020
Available online 01 September 2020
1079-9796/ © 2020 Elsevier Inc. All rights reserved.
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

(HKU, Hong Kong), the samples were transferred to the centralized cultured, expanded, and evaluated for their characteristic morphology
research laboratory (Faculty of Dentistry, HKU, Hong Kong) in a cold by microscopic examination. The EPCs which maintained their mor­
storage box for subsequent experimentation. phology over subsequent passaging were used for analysis and com­
parison.
2.1.2. Human blood sampling
The study was approved by the Institutional review board (IRB: UW
18-222) of HKU. Following written informed consent, 30 ml of blood 2.6. Growth curve
was withdrawn by venepuncture technique from 10 randomly selected
adults of the Faculty of dentistry (HKU, Hong Kong). The exclusion The proliferative potential of EPCs was determined by the CCK-8
criteria included subjects with any prevailing medical conditions and assay (Sigma-Aldrich). In brief, rCD34+CD133+EPCs and
smoking habits. Also, 5 ml of first-pass blood was discarded to prevent hCD34+CD133+EPCs (r/h CD34+CD133+EPCs) at P5 passage were
contamination by the mature endothelial cells. seeded in 96-well plates at a density of 1000 cells/well and cultured in
medium X at 37 °C in a humidified atmosphere containing 5% CO2. The
2.2. Isolation of mononuclear cells (MNCs) Optical density (OD) was measured every two days for two weeks. On
the day of evaluation, medium X was replaced by a phenol red-free
Rabbit mononuclear cells (MNCs) were isolated by density gradient culture medium containing 5% FBS, 1% P/S (working medium), and
centrifugation using dual Histopaque (Histopaque 11191 & Histopaque 10 μl of CCK-8. The plates were measured for absorbance at 460 nm
18031, Sigma-Aldrich, Shanghai, China) according to the manufacturer (OD) by a microplate reader after incubating for 2 h at 37 °C. The ex­
instructions [30]. Briefly, undiluted blood was carefully layered on periment was performed in triplicate, with each set having three wells.
Histopaque 11191 and Histopaque 1803 [Ratio: 1(Undiluted blood): The negative control consisted of the working medium and 10 μl of the
0.5(Histopaque 11191) + 0.5(Histopaque 1803)]. The layered column CCK-8 solution. The OD of both experimental groups at each time point
was centrifuged at 700 ×g for 30 min. Subsequently, the buffy coat was calculated by subtracting the mean of negative control values from
containing rabbit MNCs at the interface was carefully withdrawn. the values of the experimental group at that time point. The population
On the other hand, Ficoll-Paque Premium (GE Healthcare doubling time (PDT) in the logarithmic growth phase of both r/h
Biosciences, PA, USA) was used according to the manufacturer's in­ CD34+CD133+EPCs was also calculated.
structions to isolate MNCs from human blood. Briefly, the blood was
first diluted with PBS (1,1 ratio). The diluted blood (4 ml) was then
carefully layered over Ficoll-Paque Premium (3 ml), followed by cen­ 2.7. Flow cytometry
trifugation at 400 ×g for 40 min. These MNCs (buffy coat) at the in­
terface were then carefully withdrawn and processed further. After obtaining the homogeneous CD34+CD133+/CD34+VEGFR-
2+ EPCs by FACs, the percentage of cell surface markers, such as CD34,
2.3. Culturing of rabbit and human MNCs CD133, CD31, VEGFR-2, CD45, and CD14 were evaluated by expanding
the respective cell population. 1.5–3.0 × 106 cells and 0.5–1.0 × 106
The MNCs from the rabbit and human PB were washed twice with cells were used for sorting and analysis, respectively. The cells were
PBS at 100 × g to remove separating media and platelets. These MNCs simultaneously blocked (0.1% BSA) and incubated with the antibody
were then plated at three different densities of 0.5 × 106 cells, 1 × 106 (Table 1) for 45 min in an icebox with gentle shaking. After washing
cells/cm2 and 5 × 106 cells/cm2 (n = 3) in the custom coated six-well twice with cold PBS for 5 min, the sample was further incubated with
plates (2.5 μg/cm2, human plasma fibronectin, Gibco; ThermoFisher the secondary or conjugated antibody for 45 min and washed. For
Scientific, MA, USA) [1] in the medium X [EGM-2 or EGM-2MV; Lonza, isolation, dual antibody CD34/CD133 or CD34/VEGFR-2 were used,
Basel, Switzerland]. The MNCs were cultured according to the deple­ whereas, for quantitative analysis of surface markers, the single stem
tion technique or the re-plating technique in either EGM-2 or EGM-2MV cell marker was used. Regarding the control groups, unstained cells
supplemented with an additional 8%, and 5% fetal bovine serum (FBS) served as the negative control, whereas cells with isotype of the cor­
respectively [6]. The first medium was changed after four days in the responding antibody served as an isotype control. The isotype control
depletion technique, whereas in the re-plating procedure, it was after was also incubated for 30–45 min followed by washing thrice for 5 min
48 h. Further, the fundamental difference was that nonadherent cells in PBS. All samples were strained with a 70 μm filter to obtain singlets.
were removed in depletion technique with the first medium change, BD SORP (BD Biosciences) and BD LSR Fortessa (BD Biosciences) were
whereas nonadherent cells were re-plated after 48 h in the re-plating used for sorting and analysis, respectively, using the markers described
technique. After the first medium change, the cells were carefully above. Minimum 1% of cells were collected after sorting, whereas a
monitored under a microscope every day for 14 days. acLDL uptake and minimum of 20,000 events were recorded for analysis. The data were
lectin binding were used to test the preliminary efficacy of the protocols analyzed by FlowJo Version 10.0 (FlowJo, LLC, Ash- land, OR, USA).
or the type of medium in the cells with an endothelial phenotype that
were later subjected to FACS for EPC isolation.
2.8. Immunofluorescence (IF)
2.4. Isolation of homogenous EPCs by FACS
r/h CD34+CD133+EPCs at P4-P6 were seeded in 6-well plates at a
After testing appropriate culture conditions, second and subsequent density of 15,000 cells/well where three wells were used as treatment
media was changed accordingly. At 70–80% confluency, EPCs were group while the other three wells were used as an isotype control group.
passaged to obtain a sufficient number for double sorting The experiment was performed in triplicate. At 60–70% confluency
(CD34+CD133+ or CD34+VEGFR-2+) with FACS. The protocol was cells were washed twice with PBS for 2 min and fixed in cold 4% PFA
described later in this section, along with the analysis of surface mar­ for 30 min. The cells were then blocked with 1% bovine serum albumin
kers. (BSA) (Sigma-Aldrich) for 1 h, followed by overnight incubation with
antibodies. For secondary antibody (CD34), two additional steps were
2.5. Culturing and expansion of sorted CD34+CD133+ or CD34+VEGFR- performed, first washing the primary antibody twice for 5 min and
2+EPCs secondly, incubating with the secondary antibody for another 45 min.
The cells were then counterstained with DAPI (Sigma-Aldrich) and
Both sorted CD34+CD133+ or CD34+ VEGFR-2+ EPCs were analyzed with a fluorescent microscope.

2
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Table 1
Antibodies and isotype control.
Stem cell marker Catalog number Host React. Conjn Fluorochrome Clone Isotype control

CD 34 GTX28158 ⁎
Rat R, H Unconjugated – MEC 14.7 –
46-4817-80† Mouse Rat Secondary antibody PerCP eFlour710 r2a-1B2 46-4724-82†
CD 133 17-1338-41† Mouse ×, H Conjugated APC TMP 4 17-4714-81†
CD 31 AB958‡ Mouse R, H Unconjugated – JC/70A –
555,988§ Goat Mouse Secondary antibody FITC – GTX350151
VEGRF-2 FAB357C|| Mouse ×, H Conjugated PerCP Clone 89106 IC002C||
CD 45 GTX74646⁎ Mouse R, H Conjugated FITC MEM-55 GTX35015⁎
CD 14 Ab186689‡ Mouse R, H Conjugated PE Tuk 4 Ab91363‡

R = rabbit, H = human.
× - not present for rabbit.

Genetex.

Ebioscience.

Abcam.
§
BD Biosciences.
||
R&D.

2.9. Functional characterization values of the blank group from treatment group (AFI, Actual fluores­
cence intensity) as well as the control group followed by dividing the
2.9.1. Tubulogenic assay AFI treatment group by AFI control group.
r/h CD34+CD133+EPCs at P5 were evaluated for their ability to
form tube formation by Matrigel (BD Biosciences) assay. Briefly, 200 μl 2.10. Statistical analysis
of thawed (1.5 ml, overnight at 4 °C) Matrigel was carefully added
(avoid any air bubbles) into six wells (n = 3) of prechilled 96 well plate The growth of r/h CD34+CD133+EPCs was evaluated by analyzing
and incubated at 37 °C for 30 min. 2 × 105 cells/ml single-cell sus­ the difference in the mean OD by 2-way repeated-measures ANOVA for
pension of EPCs was prepared in EGM-2MV. 100 μl of cell suspension testing the difference in mean growth between two groups at the same
was added to the gel gently without disturbing the gel surface. The time point as well as between different time points within the same
plate was incubated at 37 °C with 5% CO2 and left undisturbed for 1 h, group. Similarly, in eNOS assay, the difference in the mean relative
after which tube formation was examined every 5 h beginning at 1 h till fluorescence intensity was analyzed by 2-way repeated-measures
25 h under an inverted microscope at 4×. Eight randomly selected ANOVA for testing the difference in mean RFI between 2 groups (rEPCs
areas from both the figures of r/h CD34+CD133+EPCs were chosen and and hEPCs) at the same concentrations as well as between different
quantitatively analyzed. concentrations within the same group. The Bonferroni correction was
used for adjusting the pairwise comparison. The above tests were per­
2.9.2. acLDL uptake and lectin binding formed as the two-sided tests at the 0.05 significance level using IBM
Expanded r/h CD34+CD133+EPCs at P5 were assessed for acLDL SPSS Statistics 24 (IBM Corp. Armonk, NY, USA).
uptake [31] and lectin binding [32]. Briefly, first ECs at day 7 (P0) (for
qualitative confirmation of ECs in EGM-2 or EGM-2MV) or expanded r/ 3. Results
h CD34+CD133+ (P5) EPCs were incubated with DiI-ac-LDL (Mole­
cular Probes, Invitrogen, Carlsbad, CA, USA) at 2.4 μg/ml for 4 h in the 3.1. Rabbit and human blood sampling
medium at 37 °C. These EPCs were then washed three times with PBS
and fixed with 4% paraformaldehyde (PFA) for 30 min at room tem­ Both the rabbit and human blood were successfully withdrawn.
perature. Subsequently, the ECs were counterstained with 200 μl of However, extracting rabbit blood was technically challenging and re­
mouse anti-human UEA-1 antibody-conjugated with fluorescein iso­ quired extended time as compared to removing human blood. After
thiocyanate (FITC) (Sigma, St. Louis, USA) at 4 °C for 1 h and then centrifugation, the buffy coat of MNCs was carefully pipetted and
washed three times with PBS. The fluorescent images were captured transferred into sterile tubes. However, while withdrawing the MNCs, if
under a laser scanning confocal microscope (Olympus IX81, Japan). any blood contamination occurred, the pipettes were discarded im­
DAPI (4′,6-diamidino-2-phenylindole) at a concentration of 0.1 μg/ml mediately.
was used as a negative control.
3.2. Culturing of MNCs and emergence of EPCs
2.9.3. eNOS assay
Nitric oxide (NO) production by endothelial nitric oxide synthase We found that MNCs at 1.0 × 106 cells/cm2 yielded optimal results.
(eNOS) in r/h CD34+CD133+EPCs was assessed by using 4,5 diamino- No ECs were obtained at 0.5 × 106 cells, whereas at 5.0 × 106 cells/
fluorescein diacetate (DAF 2-DA) [33]. Briefly, EPCs were cultured cm2, cell clumping, and cell death occurred (Fig. 1A). In our pre­
overnight in a black clear-bottom 96-well plate without serum and liminary experiments, the Hills technique was found to be ineffective
growth factors. The next day, medium in 96-well plates was replaced by and was only successful in 20% of cases (1/5 blood samples) tested,
the reaction mixture of 200 μl containing DAF-2 DA, 0.1 mM L arginine, whereas the depletion technique yielded EPCs in 40% of cases (2/5
and reaction buffer with β NADPH (induction group) or without β blood samples). Both EGM-2 and EGM-2MV gave positive results, but
NADPH (positive control and blank respectively). After incubating for EPCs in EGM-2MV emerged earlier and higher in number as observed
2 h at room temperature, fluorescence was measured (Ex/Em 490/ microscopically (Fig. 1B). acLDL uptake and lectin binding confirmed
520 nm) using a fluorimeter (VersaFluor Fluorometer, BioRad). Ex­ the emergence of cells with the endothelial phenotype (Fig. 2). In the
periments were performed in quadruplicate in which rEPCs and hEPCs depletion technique, from day 5 to day 7, 28% (7/25) of rabbit blood
were seeded together with control, blank, and experimental groups samples (rBS) and 40% (4/10) of human blood samples (hBS) yielded
together in 96 well plates. Relative fluorescence intensity (RFI) for each either EPCs in two different patterns; biphasic EPCs (Fig. 3C, a mixture
group at different concentrations was calculated by subtracting the of spindle-shaped shaped cells and EPCs), and de novo EPCs (Fig. 3D,

3
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 1. Culturing of EPCs. A. Rabbit MNCs were seeded in 3 different densities with representative images taken on day 6 1) 0.5 × 106 cells/cm2 2) 1.0 × 106 cells/
cm2 3) 5.0 × 106 cells/cm2 (n = 3, 10×, Pixel size: 0.9 μm, Scale bar: 100 μm) B. Influence of culture medium. Emergence of rEPCs on day 6 after culturing
1.0 × 106 cells/cm2 in different mediums. Representative images (n = 3, 10×, Pixel size: 0.9 μm, Scale bar: 100 μm) in 1) EGM-2 medium 2) EGM-2MV medium.

only EPCs) or a “Blood-island” like cell cluster morphology (Fig. 3B, substantiated the findings from FACS analysis. Both rEPCs and hEPCs
spindle-shaped cells at the periphery and central round cells). Spindle- were positive for CD34, CD31, and VEGFR-2, whereas negative for
shaped cells (Fig. 3A) occurred in the rest of the cases and died within CD133, CD45, and CD14 (Fig. 6).
1–2 weeks.
3.6. Functional characterization
3.3. Isolation, culturing and expansion of EPCs
3.6.1. Tubulogenic assay
Both rEPCs and hEPCs were successfully sorted with The tubulogenic assay revealed that r/h CD34+CD133+EPCS
CD34+CD133+/CD34+VEGFR-2+ using FACS. However, the percen­ formed a characteristic tubule on Matrigel. In general, the tubules in
tage of CD34+CD133+ rEPCs was one-third (3%) (Fig. 4A) as compared hEPCs were more uniform and hence could sustain for a comparatively
to the hEPCs (12%). On the other hand, the percentage of longer time as compared to rEPCs (Fig. 7A). However, quantitative
CD34+VEGFR-2+ EPCs was significantly higher in both rabbits analysis of selected areas revealed that rCD34+CD133+EPCS had a
(39.7%), and humans (43.4%) as compared to CD34+CD133+ sorted significantly thicker junctional area, tube thickness, and longer tubule
EPCs. length (P < 0.05) at 5 h as well as 8 h as compared to
rEPCs and hEPCs were successfully cultured and expanded. hCD34+CD133+EPCs (Fig. 7B).
However, after P7/P8, CD34+VEGFR-2+EPCs began to lose their
characteristic cobblestone appearance and proliferative potential, 3.6.2. acLDL uptake and lectin binding
whereas both of the properties were intact in CD34+CD133+EPCs. Both r/h CD34+CD133+EPCs demonstrated specific acLDL binding
Therefore, for subsequent analysis and comparison, and uptake of UEA-1 with no observable difference observed between
CD34+CD133+EPCs were used. It was also observed that rEPCs ap­ rEPCs or hEPCs (Fig. 8A).
peared to be larger than hEPCs (Fig. 4B).
3.6.3. eNOS assay
3.4. Growth curve A significantly increased NO production in both r/h
CD34+CD133+EPCs was observed with 0.5 μM of β NADPH. Further,
The growth curve (Fig. 4C, Table 2) revealed a statistically sig­ at different concentrations of β NADPH, it was found that the rEPCs
nificant difference in overall time points (P < 0.05) as wells as be­ released higher but statistically insignificant (P > 0.05) NO as com­
tween time points (P < 0.05) and groups (P < 0.05). Significant pared to hEPCs (Fig. 8B, Table 3).
growth was observed from day 2 to day 8 (P < 0.05). PDT for rEPCS
was 21.18 h, whereas, for hEPCs, it was 20.01 h. Overall, both rEPCs 4. Discussion
and hEPCs revealed a similar growth pattern.
As blood is the source of EPCs, the foremost question was how much
3.5. Flow cytometry and immunofluorescence blood could be withdrawn from an animal/human? A rule of thumb is
the 10%-10% rule. Accordingly, safe sampling volume is 10% of the
FACS analysis demonstrated similar expression for r/h total blood volume, which is estimated to be 10% of the animal's body
CD34+CD133+EPCs. Both rEPCs and hEPCs were strongly positive for weight. In other words, safe sampling volume is 1% of total body
CD34, CD31, and VEGFR-2. However, rEPCs had a higher percentage of weight [28]. Therefore, 30 ml of blood was withdrawn from the rabbit
these markers as compared to hEPCs (Fig. 5). CD45, a pan leukocytic of approximately 3 kg, whereas in humans, a standard 30 ml of blood
cell marker, and CD14, a monocytic lineage marker, were absent in was collected. It was also observed that the greater the amount of
both cell populations. Further, it was noteworthy that expanded blood, the higher were the chances of isolating EPCs because only
CD34+CD133+ lacks CD133+ expression (Fig. 5). IF results 0.05–0.2 EPCs/ml are present in the blood [34]. Various studies had

4
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

(caption on next page)

5
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 2. acLDL binding and UEA-1 uptake. The emergence of EPCs was confirmed by acLDL binding and UEA-1 uptake assay. Rabbit MNCs cultured in EGM-2MV
revealed a higher number of EPCs as compared to EGM-2. Representative images of staining of rEPCs with DAPI, UEA-1, acLDL, and merged from the three
experiments (n = 3, 4×, Pixel size: 2.22 μm, Scale bar: 500 μm).

also collected a similar amount of blood (30–100 ml) for isolating EPCs coated dishes/flasks as compared to the collagen-coated dishes [1].
[8,11,19,35]. Further, it was also found that ECFC colonies appeared earlier when
If EPCs are difficult to isolate, the next question was, how many cells seeded on fibronectin than on collagen [36].
should be seeded/cm2 of the culture ware? There were two essential Culture media is decisive for defining cell characteristics. In litera­
considerations; firstly, efficient MNC recovery, and secondly, appro­ ture, there is a discrepancy in the type of media used for the isolation of
priate seeding density. MNCs were isolated by various techniques, such EPCs from PBMNCs [8,19,37,38]. In our study, we found that EPCs
as; magnetic bead separation, FACS, and density gradient centrifuga­ emerged on day six in EGM-2MV as compared to day 9 in EGM-2.
tion. However, density gradient centrifugation with Ficoll-Paque is the Further, uptake of acLDL and UEA-1 uptake was higher in EGM-2MV as
most widely used technique. For isolation of hMNCs, we adhered to compared to EGM-2, indicating that they were more metabolically ac­
Ficoll-Paque Premium. However, for isolation rMNCs, we used a dual tive in EGM-2MV than EGM-2. Our results were in correspondence with
Histopaque technique because the density of rabbit MNCs is higher as another study in which EGM-2MV increased colony count, cell differ­
compared to humans, and that could give the highest and purest entiation, adhesion, tubulogenic potential, and NO production of EPCs
mononuclear cell recovery [30]. As the density of solution increases, cells, particularly EPCs [39].
the osmotic shock is reduced in Histopaque as compared to Ficoll- The EPCs have been traditionally classified into eEPCs and lEPCs
Paque, thereby reducing cell death and increasing cell recovery [30]. based on the appearance in the culture media and their phenotypic
We also found that only 1.0 × 106 MNCs/cm2 yielded EPCs. In expression (CD34, CD 45, CD14, CD31, VEGFR-2, and CD133). Both
another study, 1.5 × 106 cells/cm2 was used as the initial seeding early and late EPCs express CD34. CD31/PECAM-1 (platelet endothelial
density. However, researchers did not test the minimum cell con­ cell adhesion molecule) is strongly expressed in lEPC but weakly ex­
centration required for EPCs isolation [35]. Another important con­ pressed (focal expression) in eEPC [8,13,40]. On the other hand, Flk-1
sideration is whether the coating of culture ware can improve the ef­ (VEGFR-2 in mouse, or KDR human homolog of VEGFR-2), is expressed
ficiency of EPCs isolation? We used custom made fibronectin-coated weakly in eEPCs but strongly expressed in lEPCs [8,12–14,40]. Re­
dishes instead of collagen because morpho-differentiation and pro­ garding CD45 and CD14, these can be found only on eEPCs but not on
liferation of cells were found to be significantly higher in fibronectin- lEPCs [8,13,15]. CD133, a member of 5 transmembrane glycoproteins,

Fig. 3. Variations in the emergence of EPCs. Both rabbit (n = 25 rBS) and human MNCs (n = 10 hBS) cultured at 1.0 × 106 cells/cm2 of the 6-well plate in EGM-
2MV medium revealed 4 different types of appearance from day 5 - day 7. A. Spindle-shaped cells (eEPCs) (n = 18 rBS + 6 hBS) B. Blood-island like cell clusters
(eEPCs at the periphery and central round cells) (n = 3 rBS + 2 hBS) C. Biphasic EPCs (mixed eEPCs and lEPCs) (n = 2 rBS + 1 hBS) D. de novo lEPCs (ECFCs)
(n = 2 rBS + 1 hBS) (4×, Pixel size: 2.22 μm, Scale bar: 500 μm).

6
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 4. Isolation and expansion of CD34+CD133+EPCs. Homogenous r/h CD34+CD133+EPCs were isolated from the primary culture of EPCs by double sorting with
CD34 and CD133 antibody. A. Representative scatter plots of r/h CD34+CD133+EPCs population (n = 3). B. Microscopic examination of expanded
rCD34+CD133+EPCs (60–70% confluency) and hCD34+CD133+EPCs (80–90% confluency) (n = 3, 4×, Pixel size: 2.22 μm). C. Growth curve of expanded
CD34+CD133+EPCs with statistically significant differences between human and rabbit group on day 2, 4, 6 and 8 (P < 0.05).

is expressed on both hematopoietic stem and progenitor cells (HSCs). It [6].


is a transitional marker found variably on circulating endothelial cells, In this study, we used two types of double sorting, CD34+CD133+
eEPCs, and lEPCs, but not on mature and differentiated endothelial cells and CD34+VEGFR2+ based on the above-stated reasons. We were able

7
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Table 2
Growth curve between expanded rCD34+CD133+EPCs and hCD34+CD133+EPCs.
Time Groups⁎ Mean ± standard deviation Pairwise comparison
(N = 9)
Between groups Within group†

p value rEPCs hEPCs

Day1 1 0.151 ± 0.012 p > 0.05 Day 1 < Day 2 < Day 4 < Day 6 < Day Day 1 < Day 2 < Day 4 < Day 6 < Day 8 < Day
2 0.160 ± 0.020 8 < Day 10 < Day 12 < Day 14 10 < Day12 < Day 14
Day2 1 0.609 ± 0.017 p < 0.001
2 0.721 ± 0.005
Day4 1 1.099 ± 0.017 p < 0.001
2 1.337 ± 0.014
Day6 1 1.700 ± 0.006 p < 0.001
2 1.957 ± 0.089
Day8 1 2.395 ± 0.018 p < 0.001
2 2.562 ± 0.011
Day10 1 2.994 ± 0.069 p > 0.05
2 2.964 ± 0.018
Day12 1 3.142 ± 0.014 p > 0.05
2 3.131 ± 0.018
Day14 1 3.232 ± 0.013 p > 0.05
2 3.222 ± 0.021


Group 1: rCD34+CD133+EPCs; Group 2: hCD34+CD133+EPCs.

Post Hoc Tukey HSD test for multiple comparisons with adjustment (statistically significant at p < 0.001).

to successfully isolate both CD34+CD133+ EPCs and CD34+VEGFR2+ physiological and pathophysiological processes in the human body
EPCs from the heterogenous MNC culture as well as able to expand [42]. DAF-2DA is a highly sensitive, specific, and a membrane-perme­
these EPCs. However, when the expanded r/hCD34+CD133+EPCs po­ able fluorescent probe where DAF-2 reacts rapidly and irreversibly in
pulations were further analyzed, CD133 was found to absent from both solution with NO and NO-derived reactive species in a concentration-
cell populations. The results confirmed that CD133 is an early marker, dependent manner to produce the highly fluorescent product triazolo­
and when sorted CD34+CD133+ were expanded, they transformed into fluorescein (DAF-2 T) [33]. We measured the RFI of
mature cells and lost their CD133 expression. The results were sup­ CD34+CD133+EPCs by using β NADPH as it is one of the co-factors in
ported by the study where the authors found that EPCs differentiated the pathways of NO production [43]. We found no significant difference
into mature cells having cobblestone appearance, which were CD34+ in the production of NO in either human or rabbit EPCs.
VEGRF-2+ but CD133− [16]. In yet another study, researchers were The other exciting finding in the present study, apart from the si­
able to generate EPCs from CD133+ cells [17]. In sharp contrast, some milarities between rEPCs and hEPCs, was the emergence of two distinct
researchers were neither able to detect VEGFR-2 transcripts in CD133+ colonies of EPCs. We found two other patterns apart from blood-island
cells nor generate EPCs from CD133+ cells. They hypothesized that like structures, biphasic EPCs, and de novo EPCs. In “biphasic EPCs”,
CD34+VEGFR-2+CD133− cells within the heterogenous CD34+ frac­ lEPCs as mentioned by various authors [8,9] did not need emerge after
tion might be the origin of the EPCs [15]. eEPCs. In fact, both eEPCs and lEPCs were simultaneously growing
The tubulogenic assay is a specific assay for endothelial cells. In the together, and because eEPCs have a short life span, they eventually die,
present study, characteristic tubules were formed on Matrigel. eEPCs allowing lEPCs to proliferate and increase in number. The corre­
failed to create a tube-like structure, whereas EPCs did, indicating an sponding presence of both eEPCs and lEPCs in our study, proved pre­
inferior endothelial function of eEPCs in vitro. EPCs expressed FLK-1, vious studies stipulating lEPCs arise from cells other than eEPCs
which might be responsible for VEGF mediated tube formation [8]. In [8,14,19]. On the other hand, “de novo EPCs” are termed so because
the current study, both rCD34+CD133+EPCs and these EPCs emerge without any significant spindle-shaped cells (eEPCs)
+ +
hCD34 CD133 EPCs did form a tubular network, as seen on micro­ in the culture. The said pattern is reminiscent of ECFCs described in
scopic examination, but the junctional area, tubule thickness, and various studies [11,34,44]. Therefore, the appearance of biphasic EPCs
length of tubules were larger in rEPCs as compared to hEPCs. It might and de novo EPCs suggested that some EPCs might coexist as a subset in
explain why rabbits are called as potential healers as compared to hu­ the MNC, albeit in very few numbers. In the rest of the cultures, spindle-
mans. shaped cells were observed, and even after culturing over an extended
The selectivity of endothelial cells can also be demonstrated by the period, these were not able to give cobblestone appearance, a hallmark
ability to bind with DiI acLDL [31] and UEA-1 uptake [32]. The distinct of EPCs. We concluded that these are either transformed EPCs or fi­
advantages of Dil-ac-LDL assay include its reproducibility, uniform la­ broblast-like cells, and the appearance of these cells was an indicator of
beling without any permeabilization or fixing, non-sensitivity to tryp­ failure to isolate EPCs.
sinization, and no effect on the growth rate of endothelial cells. On the To explain the above results, an overview of studies on EPCs and
other hand, lectin binds specifically to L-fructose residues on en­ monocytes is imperative. Initial studies revealed that eEPCs are a het­
dothelial cells [41]. Both types of EPCs bind with acLDL and uptake erogeneous population of CD14+ and CD14− and lEPCs originate from
UEA-1, but the level of binding and uptake in eEPCs was less as com­ CD14− cells [8,9,14]. A decade later, gene expression, microarray, and
pared to lEPCs suggesting higher metabolic activity in lEPCs [8]. Our proteomic analysis confirmed that eEPCs have a monocytic phenotype
results revealed the same that the uptake and binding intensity in the [40]. Further, eEPCs were found to be positive for CD16, where
primary culture of EPCs were significantly less than that in homogenous CD16+CD14− population generated more CFU-Hill colonies than
and sorted CD34+CD133+EPCs. CD16+CD14+. However, only these two populations were able to
The detection of the release of NO is another functional assay to produce eEPCs but not the CD16−CD14+ or CD16−CD14− [45]. Par­
characterize EPCs. NO is generated in vivo through the conversion of L- allelly, monocytes were classified into classical monocytes (CD14++,
arginine to L-citrulline by NO synthase (NOS). It mediates many CD16−), intermediate monocytes (CD14++, CD16+), and non-classical

8
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 5. Flow cytometric analysis of r/h CD34+CD133+EPCs. The expression of CD34, CD133, CD31, VEGFR-2, CD45, and CD14 surface antigens by
rCD34+CD133+EPCs and hCD34+CD133+EPCs were quantitatively analyzed by flow cytometry. Histograms were representative of at least three separate ex­
periments, each with a minimum of 20,000 events.

9
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

(caption on next page)


10
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 6. Immunofluorescence assay r/h CD34+CD133+EPCs. Both r/h CD34+CD133+EPCs at P4-P6 were seeded in 6-well plates at a density of 15,000 cells/well and
qualitatively analyzed at 60–70% confluency. Representative images of both rEPCs and hEPCs were positive for CD34, CD31, and VEGFR-2, whereas negative for
CD133, CD45, and CD14 (n = 3, 60×, Pixel size: 0.25 μm, Scale bar: 100 μm).

monocytes (CD14−, CD16+) [46]. Additionally, it was also found out prospective use of rCD34+CD133+ in preclinical animal trials where
that some cells from intermediate monocytes expressed several surface the potential of rabbit periodontal ligament cells (rPDLCs) [49] with
markers associated with pro-angiogenesis, including endoglin (ENG), rCD34+CD133+ in neoosteogenesis and neovascularization in an irra­
TEK tyrosine kinase (Tie2, CD202b) and KDR (VEGFR-2) [47]. If we diated rabbit model will be evaluated, respectively.
correlate the two parallel themes of studies in the hematopoietic do­
main, early EPCs might probably arise from intermediate monocytes,
whereas lEPCs are true endothelial cells. This hypothesis can be sup­ CRediT authorship contribution statement
ported by various studies in which endothelial cells could transform the
phenotype and function of monocytes [18,48]. Hitesh Chopra: Conceptualization, Methodology, Project ad­
Hence, our study was the first to underscore different patterns in the ministration, Visualization, Investigation, Data curation, Writing -
origin of EPCs supported by hypothesis to explain the origin of tEPCs original draft. Yuanyuan Han: Visualization, Investigation, Writing -
(referred to as eEPCs/CACs) and EPCs (referred to as lEPCs/OECs/ review & editing. Chengfei Zhang: Conceptualization, Supervision,
ECFCs). Further, both CD34+CD133+ and VEGFR2+CD34+ cells can Funding acquisition, Validation, Writing - review & editing.
give rise to EPCs, but EPCs from CD34+CD133+ cells can be passaged Edmond Ho Nang Pow: Supervision, Funding acquisition,
to a greater extent as compared to EPCs from VEGFR2+CD34+ cells. Resources, Validation, Writing - review & editing.
Therefore, we compared rabbit and human CD34+CD133+ EPCs for the

Fig. 7. Tubulogenic assay. A. Time course of the tubulogenic assay. 100 μl of 2 × 104 r/h CD34+CD133+EPCs/well were seeded in 96-well microplate.
Representative images of tube formation in Matrigel at a 5 h interval up to 25 h (n = 3, 4×, Pixel size: 2.22 μm, Scale bar: 500 μm). B. Quantitative analysis of tubule
formation. Mean (μm ± SD) tube thickness, and tubule length, whereas, mean (μm2 ± SD) junctional area were analyzed in eight randomly selected areas at 5 h as
well as 10 h. The mean surface area of rEPCs was significantly higher than the mean surface area of hEPCs (P < 0.05) at both 5 h and 10 h. rCD34+CD133+ had a
significantly thicker junctional area, tubule thickness, and a longer tubule length (P < 0.05) as compared to hCD34+CD133+EPCs.

11
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

(caption on next page)


12
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

Fig. 8. Other functional characterization essays. A. acLDL binding and UEA-1 uptake. r/h CD34+CD133+EPCs cultured in EGM-2MV demonstrated characteristic
acLDL binding and UEA-1 uptake (DAPI was used as a negative control) (n = 3, 20×, Pixel size: 0.44 μm, Scale bar: 100 μm). B. eNOS assay. DAF 2-DA did not reveal
a significant difference (P > 0.05) in NO production by r/h CD34+CD133+EPCs after induction with β NADPH.

Table 3
eNOS assay.
β NADPH Groupsa Mean RFI ± standard deviation (N = 4) Pairwise comparison

Between groups Within groupb

P value rEPCs hEPCs

I (0.05 μM) 1 1.525 ± 0.033 P = 0.087 I=II < III=IV=V I=II < III=IV=V
2 1.451 ± 0.032
II (0.01 μM) 1 1.568 ± 0.024 P = 0.618
2 1.506 ± 0.023
III (0.5 μM) 1 1.987 ± 0.039 P = 0.377
2 1.844 ± 0.036
IV (1 μM) 1 2.019 ± 0.028 P = 0.746
2 1.902 ± 0.050
V (5 μM) 1 2.056 ± 0.017 P = 0.525
2 1.923 ± 0.050

a
Group 1: r CD34+CD133+EPCs; Group 2: h CD34+CD133+EPCs.
b
Post-Hoc Tukey HSD test for multiple comparisons with adjustment (statistically significant at p < 0.001).

Declaration of competing interest [8] J. Hur, C.H. Yoon, H.S. Kim, et al., Characterization of two types of endothelial
progenitor cells and their different contributions to neovasculogenesis, Arterioscler.
Thromb. Vasc. Biol. 24 (2004) 288–293, https://doi.org/10.1161/01.Atv.
The authors declare that they have no competing interests. 0000114236.77009.06.
[9] R. Gulati, D. Jevremovic, T.E. Peterson, et al., Diverse origin and function of cells
with endothelial phenotype obtained from adult human blood, Circ. Res. 93 (2003)
Acknowledgments 1023–1025, https://doi.org/10.1161/01.Res.0000105569.77539.21.
[10] Y. Lin, D.J. Weisdorf, A. Solovey, R.P. Hebbel, Origins of circulating endothelial
We are thankful to Dr. Mei Ki Hung for human blood sampling. We cells and endothelial outgrowth from blood, J. Clin. Invest. 105 (2000) 71–77,
https://doi.org/10.1172/JCI8071.
also appreciate Dr. Dan Zhao (Ph.D. student, Faculty of Dentistry, HKU) [11] D.A. Ingram, L.E. Mead, H. Tanaka, et al., Identification of a novel hierarchy of
and Prof. Jin (Clinical Professor, Periodontology) for helping us with endothelial progenitor cells using human peripheral and umbilical cord blood,
their appointed nurse in human blood collection. Blood 104 (2004) 2752–2760, https://doi.org/10.1182/blood-2004-04-1396.
[12] N. Mukai, T. Akahori, M. Komaki, et al., A comparison of the tube forming po­
tentials of early and late endothelial progenitor cells, Exp. Cell Res. 314 (2008)
Availability of data and materials 430–440, https://doi.org/10.1016/j.yexcr.2007.11.016.
[13] C.C. Cheng, S.J. Chang, Y.N. Chueh, et al., Distinct angiogenesis roles and surface
The complete datasets supporting this article are provided with the markers of early and late endothelial progenitor cells revealed by functional group
analyses, BMC Genomics 14 (2013) 182, https://doi.org/10.1186/1471-2164-14-
article. 182.
[14] C.H. Yoon, J. Hur, K.W. Park, et al., Synergistic neovascularization by mixed
Grant support transplantation of early endothelial progenitor cells and late outgrowth endothelial
cells: the role of angiogenic cytokines and matrix metalloproteinases, Circulation
112 (2005) 1618–1627, https://doi.org/10.1161/CIRCULATIONAHA.104.503433.
Chopra H was supported through a seed grant no. 201511159105 [15] F. Timmermans, F. Van Hauwermeiren, M. De Smedt, et al., Endothelial outgrowth
awarded to Pow EHN and Zhang CF by University Research Grants for cells are not derived from CD133(+) cells or CD45(+) hematopoietic precursors,
Arterioscler. Thromb. Vasc. Biol. 27 (2007) 1572–1579, https://doi.org/10.1161/
“3D cell sheets cultured from endothelial progenitor cells and period­ Atvbaha.107.144972.
ontal ligament stem cells (2016).” [16] M. Peichev, A.J. Naiyer, D. Pereira, et al., Expression of VEGFR-2 and AC133 by
circulating human CD34(+) cells identifies a population of functional endothelial
precursors, Blood 95 (2000) 952–958, https://doi.org/10.1182/blood.V95.3.952.
References
003k27_952_958.
[17] U.M. Gehling, S. Ergun, U. Schumacher, et al., In vitro differentiation of endothelial
[1] T. Asahara, T. Murohara, A. Sullivan, et al., Isolation of putative progenitor en­ cells from AC133-positive progenitor cells, Blood 95 (2000) 3106–3112, https://
dothelial cells for angiogenesis, Science 275 (1997) 964–967, https://doi.org/10. doi.org/10.1182/blood.V95.10.3106.
1126/science.275.5302.964. [18] S. Thomas-Ecker, A. Lindecke, W. Hatzmann, C. Kaltschmidt, K.S. Zanker,
[2] G.P. Fadini, S. Sartore, C. Agostini, A. Avogaro, Significance of endothelial pro­ T. Dittmar, Alteration in the gene expression pattern of primary monocytes after
genitor cells in subjects with diabetes, Diabetes Care 30 (2007) 1305–1313, https:// adhesion to endothelial cells, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 5539–5544,
doi.org/10.2337/dc06-2305. https://doi.org/10.1073/pnas.0700732104.
[3] P. Mancuso, F. Bertolini, Circulating endothelial cells as biomarkers in clinical [19] R.J. Medina, C.L. O’Neill, M. Sweeney, et al., Molecular analysis of endothelial
oncology, Microvasc. Res. 79 (2010) 224–228, https://doi.org/10.1016/j.mvr. progenitor cell (EPC) subtypes reveals two distinct cell populations with different
2010.02.007. identities, BMC Med. Genet. 3 (2010) 18, https://doi.org/10.1186/1755-8794-
[4] C. Dong, P.J. Goldschmidt-Clermont, Endothelial progenitor cells: a promising 3-18.
therapeutic alternative for cardiovascular disease, J. Interv. Cardiol. 20 (2007) [20] R.A. Thomas, D.C. Pietrzak, M.S. Scicchitano, H.C. Thomas, D.C. McFarland,
93–99, https://doi.org/10.1111/j.1540-8183.2007.00251.x. K.S. Frazier, Detection and characterization of circulating endothelial progenitor
[5] J. Rouwkema, N.C. Rivron, C.A. van Blitterswijk, Vascularization in tissue en­ cells in normal rat blood, J. Pharmacol. Toxicol. Methods 60 (2009) 263–274,
gineering, Trends Biotechnol. 26 (2008) 434–441, https://doi.org/10.1016/j. https://doi.org/10.1016/j.vascn.2009.06.002.
tibtech.2008.04.009. [21] W. Sun, L. Zheng, P. Han, Y.J. Kang, Isolation and characterization of endothelial
[6] H. Chopra, M.K. Hung, D.L. Kwong, C.F. Zhang, E.H.N. Pow, Insights into en­ progenitor cells from Rhesus monkeys, Regen Med Res 2 (2014) 5, https://doi.org/
dothelial progenitor cells: origin, classification, potentials, and prospects, Stem 10.1186/2050-490X-2-5.
Cells Int. 2018 (2018) 9847015, , https://doi.org/10.1155/2018/9847015. [22] S. Bi, X. Tan, S.Q. Ali, L. Wei, Isolation and characterization of peripheral blood-
[7] J. Rehman, J. Li, C.M. Orschell, K.L. March, Peripheral blood “endothelial pro­ derived endothelial progenitor cells from broiler chickens, Vet. J. 202 (2014)
genitor cells” are derived from monocyte/macrophages and secrete angiogenic 396–399, https://doi.org/10.1016/j.tvjl.2014.08.017.
growth factors, Circulation 107 (2003) 1164–1169, https://doi.org/10.1161/01. [23] M.T. Hinds, M. Ma, N. Tran, et al., Potential of baboon endothelial progenitor cells
CIR.0000058702.69484.A0. for tissue engineered vascular grafts, J. Biomed. Mater. Res. A 86 (2008) 804–812,

13
H. Chopra, et al. Blood Cells, Molecules and Diseases 86 (2021) 102487

https://doi.org/10.1002/jbm.a.31672. https://doi.org/10.1371/journal.pone.0066734.
[24] M. Avci-Adali, A. Nolte, P. Simon, G. Ziemer, H.P. Wendel, Porcine EPCs down­ [37] C. Kalka, H. Masuda, T. Takahashi, et al., Transplantation of ex vivo expanded
regulate stem cell markers and upregulate endothelial maturation markers during in endothelial progenitor cells for therapeutic neovascularization, Proc. Natl. Acad.
vitro cultivation, J. Tissue Eng. Regen. Med. 3 (2009) 512–520, https://doi.org/10. Sci. U. S. A. 97 (2000) 3422–3427, https://doi.org/10.1073/pnas.070046397.
1002/term.189. [38] X.L. Mai, Z.L. Ma, J.H. Sun, S.H. Ju, M. Ma, G.J. Teng, Assessments of proliferation
[25] J. Neyt, J.A. Buckwalter, N. Carroll, Use of animal models in musculoskeletal re­ capacity and viability of New Zealand rabbit peripheral blood endothelial pro­
search, Iowa Orthopaedic J. 18 (1998) 118. genitor cells labeled with superparamagnetic particles, Cell Transplant. 18 (2009)
[26] A.R. Amini, C.T. Laurencin, S.P. Nukavarapu, Differential analysis of peripheral 171–181, https://doi.org/10.3727/096368909788341306.
blood- and bone marrow-derived endothelial progenitor cells for enhanced vascu­ [39] X.M. Guan, M. Cheng, H. Li, et al., Biological properties of bone marrow-derived
larization in bone tissue engineering, J. Orthop. Res. 30 (2012) 1507–1515, https:// early and late endothelial progenitor cells in different culture media, Mol. Med.
doi.org/10.1002/jor.22097. Rep. 8 (2013) 1722–1728, https://doi.org/10.3892/mmr.2013.1718.
[27] C.F. Lam, Y.C. Liu, J.K. Hsu, et al., Autologous transplantation of endothelial pro­ [40] S.M. Cheng, S.J. Chang, T.N. Tsai, et al., Differential expression of distinct surface
genitor cells attenuates acute lung injury in rabbits, Anesthesiology 108 (2008) markers in early endothelial progenitor cells and monocyte-derived macrophages,
392–401, https://doi.org/10.1097/ALN.0b013e318164ca64. Gene Expr. 16 (2013) 15–24, https://doi.org/10.3727/
[28] M.W. McGuill, A.N. Rowan, Biological effects of blood loss: implications for sam­ 105221613x13776146743307.
pling volumes and techniques, ILAR J. 31 (1989) 5–20, https://doi.org/10.1093/ [41] M. Hormia, V.P. Lehto, I. Virtanen, Identification of UEA I-binding surface glyco­
ilar.31.4.5. proteins of cultured human endothelial cells, Cell Biol Int Rep 7 (1983) 467–475,
[29] S. Parasuraman, R. Raveendran, R. Kesavan, Blood sample collection in small la­ https://doi.org/10.1016/0309-1651(83)90136-4.
boratory animals, J. Pharmacol. Pharmacother. 1 (2010) 87–93, https://doi.org/10. [42] F. Murad, Discovery of some of the biological effects of nitric oxide and its role in
4103/0976-500X.72350. cell signaling, Biosci. Rep. 24 (2004) 452–474, https://doi.org/10.1007/s10540-
[30] D.L. Feldman, T.C. Mogelesky, Use of histopaque for isolating mononuclear-cells 005-2741-8.
from rabbit-blood, J. Immunol. Methods 102 (1987) 243–249, https://doi.org/10. [43] U. Forstermann, W.C. Sessa, Nitric oxide synthases: regulation and function, Euro
1016/0022-1759(87)90083-4. Heart J. 33 (2012) 829–837d, https://doi.org/10.1093/eurheartj/ehr304.
[31] J.C. Voyta, D.P. Via, C.E. Butterfield, B.R. Zetter, Identification and isolation of [44] M.C. Yoder, L.E. Mead, D. Prater, et al., Redefining endothelial progenitor cells via
endothelial-cells based on their increased uptake of acetylated-low density lipo­ clonal analysis and hematopoietic stem/progenitor cell principals, Blood 109
protein, J. Cell Biol. 99 (1984) 2034–2040, https://doi.org/10.1083/jcb.99.6.2034. (2007) 1801–1809, https://doi.org/10.1182/blood-2006-08-043471.
[32] H. Holthofer, I. Virtanen, A.L. Kariniemi, M. Hormia, E. Linder, A. Miettinen, Ulex [45] T. Kimura, H. Kohno, Y. Matsuoka, et al., CD16 antigen is a positive marker of
europaeus I lectin as a marker for vascular endothelium in human tissues, Lab. peripheral blood-derived early endothelial progenitor cells, Int. J. Hematol. 93
Investig. 47 (1982) 60–66. (2011) 123–125, https://doi.org/10.1007/s12185-010-0739-x.
[33] H. Kojima, N. Nakatsubo, K. Kikuchi, et al., Detection and imaging of nitric oxide [46] L. Ziegler-Heitbrock, P. Ancuta, S. Crowe, et al., Nomenclature of monocytes and
with novel fluorescent indicators: diaminofluoresceins, Anal. Chem. 70 (1998) dendritic cells in blood, Blood 116 (2010) e74–e80, https://doi.org/10.1182/
2446–2453, https://doi.org/10.1021/ac9801723. blood-2010-02-258558.
[34] D.N. Prater, J. Case, D.A. Ingram, M.C. Yoder, Working hypothesis to redefine en­ [47] A.M. Zawada, K.S. Rogacev, B. Rotter, et al., SuperSAGE evidence for CD14+
dothelial progenitor cells, Leukemia 21 (2007) 1141–1149, https://doi.org/10. +CD16+ monocytes as a third monocyte subset, Blood 118 (2011) e50–e61,
1038/sj.leu.2404676. https://doi.org/10.1182/blood-2011-01-326827.
[35] C. Bueno-Beti, S. Novella, M. Lazaro-Franco, et al., An affordable method to obtain [48] C. Tso, K.-A. Rye, P. Barter, Phenotypic and functional changes in blood monocytes
cultured endothelial cells from peripheral blood, J. Cell. Mol. Med. 17 (2013) following adherence to endothelium, PLoS One 7 (2012) e37091, https://doi.org/
1475–1483, https://doi.org/10.1111/jcmm.12133. 10.1371/journal.pone.0037091.
[36] E. Colombo, F. Calcaterra, M. Cappelletti, D. Mavilio, S. Della Bella, Comparison of [49] H. Chopra, C. Liao, C.F. Zhang, E.H.N. Pow, Lapine periodontal ligament stem cells
fibronectin and collagen in supporting the isolation and expansion of endothelial for musculoskeletal research in preclinical animal trials, J. Transl. Med. 16 (2018)
progenitor cells from human adult peripheral blood, PLoS One 8 (2013) e66734, , 174, https://doi.org/10.1186/s12967-018-1551-2.

14

You might also like