Skip to main content
Erschienen in: Respiratory Research 1/2019

Open Access 01.12.2019 | Research

Progenitor cell mobilisation and recruitment in pulmonary arteries in chronic obstructive pulmonary disease

verfasst von: Olga Tura-Ceide, Sandra Pizarro, Jéssica García-Lucio, Josep Ramírez, Laureano Molins, Isabel Blanco, Yolanda Torralba, Marta Sitges, Cristina Bonjoch, Victor I. Peinado, Joan Albert Barberà

Erschienen in: Respiratory Research | Ausgabe 1/2019

Abstract

Background

Pulmonary vascular abnormalities are a characteristic feature of chronic obstructive pulmonary disease (COPD). Cigarette smoking is the most important risk factor for COPD. It is believed that its constant exposure triggers endothelial cell damage and vascular remodelling. Under pathological conditions, progenitor cells (PCs) are mobilized from the bone marrow and recruited to sites of vascular injury. The aim of the study was to investigate whether in COPD the number of circulating PCs is related to the presence of bone marrow-derived cells in pulmonary arteries and the association of these phenomena to both systemic and pulmonary endothelial dysfunction.

Methods

Thirty-nine subjects, 25 with COPD, undergoing pulmonary resection because of a localized carcinoma, were included. The number of circulating PCs was assessed by flow cytometry using a triple combination of antibodies against CD45, CD133 and CD34. Infiltrating CD45+ cells were identified by immunohistochemistry in pulmonary arteries. Endothelial function in systemic and pulmonary arteries was measured by flow-mediated dilation and adenosine diphosphate-induced vasodilation, respectively.

Results

COPD patients had reduced numbers of circulating PCs (p < 0.05) and increased numbers of CD45+ cells (< 0.05) in the pulmonary arterial wall than non-COPD subjects, being both findings inversely correlated (r = − 0.35, p < 0.05). In pulmonary arteries, the number of CD45+ cells correlated with the severity of vascular remodelling (r = 0.4, p = 0.01) and the endothelium-dependent vasodilation (r = − 0.3, p = 0.05). Systemic endothelial function was unrelated to the number of circulating PCs and changes in pulmonary vessels.

Conclusion

In COPD, the decrease of circulating PCs is associated with their recruitment in pulmonary arteries, which in turn is associated with endothelial dysfunction and vessel remodelling, suggesting a mechanistic link between these phenomena. Our findings are consistent with the notion of an imbalance between endothelial damage and repair capacity in the pathogenesis of pulmonary vascular abnormalities in COPD.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12931-019-1024-z) contains supplementary material, which is available to authorized users.
Abkürzungen
ADP
Adenosine diphosphate
BNP
Brain natriuretic peptide
COPD
Chronic obstructive pulmonary disease
CS
Cigarette smoke
DLco
Lung diffusing capacity for carbon monoxide
FEV1
Forced expiratory volume in 1 s
FMD
Flow mediated dilation
FVC
Forced vital capacity
PaCO2
Partial pressure or arterial carbon dioxide
PaO2
Partial pressure of arterial oxygen
PC
Progenitor cells
RV
Residual volume
TLC
Total lung capacity

Background

Chronic obstructive pulmonary disease (COPD) is a highly prevalent, non-curable, life-threatening disease, characterised by irreversible changes in lung structures [13]. Pulmonary vascular abnormalities, as shown by alterations in vessel structure, abnormal cell growth, endothelial dysfunction and resistance to apoptosis, are characteristic features in COPD [4, 5]. An altered pulmonary vascular function in COPD patients may predispose to pulmonary hypertension, which is associated with adverse outcomes [6]. Growing evidence suggests that endothelial cell damage found in pulmonary vessels of COPD patients is an initial and important triggering factor that promotes pulmonary vascular remodelling [5]. It has been shown that COPD patients present endothelial dysfunction in both pulmonary and systemic arteries at early disease stages [79]. This endothelial damage may result from an imbalance between vascular injury and the body’s repair capacity [10].
Bone marrow-derived progenitor cells (PCs) are a population of rare, pre-differentiated adult stem cells that circulate in the blood with the ability to proliferate and differentiate into mature endothelial cells [11]. Although, the contribution of PCs in vascular remodelling and repair is unclear, it is believed that under pathological conditions, PCs are mobilized from the bone marrow and recruited to sites of vascular injury to maintain vascular homeostasis [7]. PCs are believed to be essential in maintenance of the endothelium’s integrity and restoration of normal function, replacing terminally differentiated cells lost as a consequence of physiological cell turnover or tissue damage [12]. Conversely, it has also been suggested that in COPD, PCs mobilization and recruitment may contribute to COPD pathogenesis. PCs intrinsic dysfunctional activity, mainly due to cigarette smoke (CS) exposure, promotes inflammation, pulmonary vessel remodelling and pulmonary hypertension [13].
Reduced number and function of circulating PCs has been established as an independent prognostic factor associated with endothelial dysfunction, high cardiovascular risk and increased mortality [1416]. In COPD, few studies to date have investigated the number of circulating PCs [10, 12, 17]. We and others have previously shown reduced levels of circulating PCs in COPD patients, as compared to controls [10, 12, 17]. To date, two different mechanisms have been suggested to explain the reduced number of circulating PCs seen in COPD, an impairment of the bone marrow to generate enough circulating PC numbers and/or an increased PC recruitment in pulmonary vessels in response to tissue injury. Accordingly, in this study we aimed to quantify in the same individuals both circulating PCs and bone marrow-derived cells localized in pulmonary arteries, as well as their association to pulmonary artery remodelling, and systemic and pulmonary artery functionality.

Methods

Subjects

Thirty-nine subjects, aged between 48 and 70 years, undergoing pulmonary resection because of a localized lung carcinoma were enrolled in the study. Patients were divided into two groups: (a) a non-COPD group (n = 14) with normal pulmonary function, and (b) a COPD patient group (n = 25), defined by a forced expiratory volume in the first second (FEV1)-to-forced vital capacity (FVC) ratio lower than 0.7. Patients with COPD were on regular bronchodilator treatment and some of them received inhaled corticosteroids. Before surgery, all patients underwent standard evaluation by means of medical history, clinical examination and pulmonary function tests (forced spirometry, lung volumes, carbon monoxide diffusing capacity (DLco), and arterial blood gas analysis), as previously described [18]. Plasma levels of endothelin-1, nitrites/nitrates, angiopoietin-2 and brain natriuretic peptide (BNP) were also measured. The study was conducted in accordance with the Declaration of Helsinki, approved by the institutional Committee on Human Research and all subjects gave written informed consent.

Circulating CD45+CD34+CD133+progenitor cells

The number of circulating progenitor cells was evaluated by flow cytometry using antibodies against CD45 (pan-leukocyte marker), CD133 (sub-population of haematopoietic stem cells) and CD34 (mature and progenitor endothelial cells) as previously described [10, 17, 19]. In brief, mononuclear cells (MNCs) were isolated by Ficoll density gradient separation, washed once with phosphate buffered saline (PBS) supplemented with 2% foetal calf serum (FCS) and resuspended at 2 × 106 cells (control tube) and at 4 × 106 cells (sample tube). Peripheral blood mononuclear cells were stained and analysed for phenotypic expression of surface markers using pre-conjugated anti-human monoclonal antibodies: anti-CD45-FITC, anti-CD34-PECy7 and anti-CD133-PE. The fluorescence minus one technique [10, 17, 20] was employed to provide negative controls. After 45 min of incubation, cells were washed, resuspended into 500ul of PBS + 2%FCS and proceeded to flow-cytometric analysis. Eighty thousand events were gated in the lymphocyte region as preciously described [10, 17].

Endothelial function in systemic and pulmonary arteries

Systemic endothelial function assessment

Endothelial function was assessed by high resolution ultrasound as the change in brachial artery diameter in response to reactive hyperaemia (flow-mediated dilation), as previously described [21]. Endothelium-independent, nitroglycerine-mediated dilation was also measured.

Endothelial pulmonary artery function assessment

Endothelial function of pulmonary artery segments was evaluated in vitro as previously described [4, 22]. Arterial segments with an external diameter of approximately 2 mm were carefully dissected free of visible fat and connective tissue and cut into 3 mm long rings. All rings were submaximally pre-contracted with norepinephrine (NE; 10− 7 to 10− 6 M) to obtain a stable plateau of tension. Rings were tested using cumulative concentrations of adenosine diphosphate (ADP) (10− 10 to 10− 4 M), an endothelium-dependent vasodilator. Relaxation of each pulmonary artery ring was determined by measuring the reduction in tone in response to cumulative doses of the vasodilating agent and expressed as the percent reduction from the value recorded after pre-contraction with NE. Maximal relaxation was the greatest reduction in tone in response to the vasodilator.

Number of CD45+ cells in pulmonary arteries

Cryostat sections of PBS-4% paraformaldehyde-fixed artery rings were immunostained using the avidin-biotin complex/horseradish peroxidase method (Vector Laboratories). We used a mouse monoclonal antibody against the pan-leukocyte marker CD45 (diluted 1/750) (Novocastra Laboratories) and a progenitor cell marker CD133 (diluted 1/5) (Miltenyi Biotec) for immunolocalization of bone marrow-derived cells. Negative control experiments were conducted omitting the primary antibody. The number of positive cells infiltrating the arterial wall were counted and expressed as cell number per square millimetre of intimal surface as previously described [22].

Morphometric studies

Pulmonary muscular arteries were analysed in formalin-fixed paraffin-embedded lung tissue sections processed with elastic orcein stain. All arteries with an external diameter < 1 mm and with complete elastic laminas were evaluated using a specific software package (Leica-Qwin) as previously described [23]. Briefly, the external diameter was measured as the widest distance between external elastic laminas, perpendicular to the greatest longitudinal axis of each artery. Arterial wall thickness of each pulmonary artery was calculated as the area of the arterial wall divided by the internal perimeter and expressed as percentage of radius. All arteries with an index of narrowing (smaller diameter divided by larger diameter) lower than 1/3 were rejected.

Data analysis

Data are expressed as mean ± SD. Comparisons between groups were performed using Mann-Whitney U-test. Spearman rank correlation was used to assess correlations between variables. A p value < 0.05 was considered statistically significant.

Results

General patient’s characteristics

The COPD and non-COPD groups were well matched with respect to age and body mass index. Both groups showed a high percentage of male subjects. Five patients in the non-COPD group had never smoked. Approximately, half of the patients in each group were current smokers. All COPD patients were current or ex-smokers (Table 1). COPD patients had moderate-to-severe airflow limitation, moderately reduced DLco and mild hypoxemia. There were no differences in the Framingham risk score between groups. The number of leukocytes, monocytes, lymphocytes and neutrophils were similar in both groups. COPD patients presented lower platelet counts than non-COPD subjects (Table 1). Levels of endothelin-1, angiopoietin-2, C-reactive protein, fibrinogen, nitrites/nitrates, VEGF, IL-6, BNP and VEGR2 did not differ between groups (Table 1).
Table 1
Clinical characteristics, lung function and laboratory measurements
 
Non-COPD
COPD
Age, years
57.3 ± 7.2
58.5 ± 5.8
Male sex, n (%)
9 (64.2%)
24 (96%)
Body mass index (Kg/m2)
24.6 ± 1.7
25.4 ± 2.8
Never smoked, %
5 (35.7%)
0 (0%)
Current smokers, %
6 (43%)
17 (68%)
Ex-smokers, %
3 (21%)
8 (32%)
Smoking history, pack-years
38.6 ± 22.4
60.1 ± 33.9
Expiratory carbon monoxide, ppm
1.5 ± 1.5
2.4 ± 2.2
FEV1, % predicted
91.7 ± 10.5
56.7 ± 21.5***
FEV1/FVC, %
76.2 ± 4.5
51.0 ± 14.7***
TLC, % predicted
90.9 ± 14.5
106.6 ± 17.3*
RV, % predicted
108.8 ± 20.8
164.4 ± 69.2**
DLCO, % predicted
82.5 ± 11.9
63.4 ± 20.1**
PaO2, mmHg
85.0 ± 12.8
75.3 ± 10.5 *
PaCO2, mmHg
37.3 ± 2.8
37.3 ± 3.8
Total cholesterol, mg/dL
196.0 ± 31.4
203.2 ± 39.5
Triglycerides, mg/dL
92.4 ± 25.0
111.2 ± 46.7
HDL, mg/dL
47.0 ± 12.3
48.6 ± 12.1
LDL mg/dL
129.4 ± 28.0
132.4 ± 34.0
Framingham risk score$
8.4 ± 4.8
10.9 ± 5.7
Glucose, mg/dL
97.1 ± 10.3
100.7 ± 20.5
Leukocyte count, × 109/L
7.7 ± 1.7
8.4 ± 1.5
Lymphocyte count, × 109/L
1.6 ± 0.5
1.8 ± 0.4
Monocyte count, × 109/L
0.4 ± 0.1
0.4 ± 0.1
Neutrophils, ×109/L
5.2 ± 1.5
5.7 ± 1.4
Red blood cells, × 1012/L
4.6 ± 0.4
4.6 ± 0.3
Haemoglobin, g/L
137.0 ± 13.2
146.5 ± 9.3*
Platelet count, × 109/L
309.5 ± 88.0
252.2 ± 61.0*
C-reactive protein, mg/L
1.1 ± 1.5
1.0 ± 2.1
Fibrinogen, mg/L
5.4 ± 1.9
4.0 ± 1.2
Nitrites/nitrates, nMol/mL
20.4 ± 7.0
22.4 ± 10.6
Endothelin-1, pmol/L
6.5 ± 4.3
5.8 ± 2.3
Angiopoietin-2, pg/mL
527.0 ± 99.6
433.3 ± 142.0
VEGF, pg/mL
49.8 ± 41.0
57.8 ± 78.7
VEGFR2, pg/mL
34.4 ± 10.4
53.7 ± 7.8
IL-6, pg/mL
5 ± 7.2
7.7 ± 16.0
BNP. pg/mL
15.9 ± 6.2
21.8 ± 18.2
Definition of abbreviations: COPD chronic obstructive pulmonary disease, DLco lung diffusing capacity for carbon monoxide, RV residual volume, FEV1 forced expiratory volume in 1 s, FVC forced vital capacity, TLC total lung capacity, PaCO2 partial pressure or arterial carbon dioxide, PaO2 partial pressure of arterial oxygen. Framingham risk score$ can range from −6 to 19, with higher scores indicating greater cardiovascular risk. Non-COPD (n = 14), COPD (n = 25), * p < 0.05, ** p < 0.01, *** p < 0.001, compared with non-COPD, Mann Whitney test. Values expressed as mean ± SD

COPD patients showed lower number of circulating CD45+CD34+CD133+cells and greater number of CD45+ cells in pulmonary arteries than non-COPD subjects

Circulating progenitor cells, defined as CD45+CD34+CD133+cells were significantly reduced in COPD patients compared with the non-COPD group (Fig. 1a). Conversely, COPD patients had a higher number of CD45+ cells in pulmonary arteries than non-COPD subjects (Fig. 1b). Representative histological pictures showed CD45+and CD133+ cells localized within the intima of pulmonary arteries (Additional file 1: Figure S1a-b). There was a significant inverse correlation between the number of circulating CD45+CD34+CD133+ cells and the number of CD45+ infiltrating the intima of the pulmonary arteries (r = − 0.35, p = 0.03) (Fig. 1c).

Endothelial function in systemic and pulmonary arteries

Systemic endothelial function assessment

COPD patients and non-COPD subjects showed similar values for flow mediated dilation (FMD) (1.8 ± 1.0 vs 1.6 ± 1.7). FMD was unrelated to the number of circulating CD45+CD34+CD133+ cells (r = − 0.2, p = 0.3) or to the presence of CD45+cells in the intima of pulmonary arteries (r = − 0.1, p = 0.4).

Endothelial pulmonary artery function assessment

Endothelium-dependent dilation of isolated pulmonary arteries induced by ADP and distensibility were reduced in COPD patients compared to non-COPD subjects (Table 2). There was no significant correlation between systemic FMD and pulmonary endothelial function or between systemic FMD and distensibility (Table 2). The number of CD45+ infiltrating the artery wall of pulmonary arteries was inversely correlated with endothelial function (r = − 0.3, p = 0.05) and distensibility (r = − 0.4, p = 0.04), whereas the number of circulating CD45+CD34+CD133+ cells did not.
Table 2
Endothelial function and distensibility of pulmonary arteries
 
Non-COPD
COPD
ADP-induced vasodilation, % change from maximal contraction
−88.2 ± 26.0
−83.5 ± 22.6
Distensibility, vol/mmHg
2.0 ± 0.8
1.4 ± 0.5*
Definition of abbreviations: ADP adenosine diphosphate. Non-COPD (n = 11), COPD (n = 23), Mann Whitney test. Values expressed as mean ± SD. * p < 0.05

Morphological evaluation

Morphometric measurements of pulmonary arteries are shown in Table 3. COPD patients had thicker artery walls (Fig. 2a), reduced lumen area (Fig. 2b), thicker muscular layer and a trend to thicker intima than non-COPD subjects (Fig. 2c).
Table 3
Morphometric measurements on pulmonary arteries
Variables
Non-COPD
COPD
Mean wall thickness (μm)
44.1 ± 14.1
56.6 ± 15.8*
Measured external diameter (μm)
210.7 ± 61.0
252.3 ± 120.5
Calculated external diameter, um
538.4 ± 147.9
531.1 ± 117.2
Index of narrowing, %
73.5 ± 3.9
72.8 ± 8.1
Wall thickness, % measured diameter
34.0 ± 9.6
41.0 ± 16.5
Intimal area, % total area
22.8 ± 8.9
24.7 ± 7.2
Muscular area, % total area
32.8 ± 8.9
43.4 ± 5.9***
Lumen area, % total area
42.3 ± 12.8
31.8 ± 7.7**
Definition of abbreviations: COPD chronic obstructive pulmonary disease; Non-COPD (n = 9), COPD (n = 23), * p < 0.05, ** p < 0.01, *** p < 0.001 compared with non-COPD, Mann Whitney test. Values are expressed as mean ± SD
Whereas the number of CD45+ localized in the intima of the pulmonary arteries inversely correlated with artery lumen area, the number of circulating CD45+CD34+CD133+ cells did not (Fig. 3a, b).

Relationships among progenitor cells, pulmonary vascular remodelling, pulmonary function and CS exposure

In COPD patients, the number of circulating PCs or bone marrow-derived cells localized in pulmonary arteries was unrelated to the severity of airflow obstruction or DLco values (Fig. 4a, b). Yet, COPD patients with DLco values below the median value had greater pulmonary vascular remodelling, as shown by lower lumen area, than those with DLco values above the median value (Fig. 4c). Non-COPD controls were separated in non-smokers and smokers and compared to COPD patients (Additional file 2: Table S1). Smokers and COPD subjects showed a higher percentage of male subjects than non-smoker controls. In most parameters analyzed, non-COPD smokers presented an intermediate phenotype between non-smoker controls and COPD subjects (FEV1, % predicted; FEV1/FVC, %; DLCO, % predicted; PaO2, mmHg; CD45+CD34+CD133+ cells; mean wall thickness (um); lumen area, % total area and muscular area, % total area) (Additional file 2: Table S1).

Discussion

The present study shows that in COPD the decrease of circulating PCs is associated with the presence of bone marrow-derived cells in pulmonary arteries and the number of CD45+ cells infiltrating the intima of pulmonary arteries directly correlated with pulmonary vascular remodelling.
Circulating CD45+CD34+CD133+ PCs were significantly reduced in COPD patients compared with the non-COPD group, confirming recent findings published by our group and others [10, 12, 17]. Reduced number of circulating PCs has been established as a prognostic risk factor associated with endothelial dysfunction and increased cardiovascular risk [14]. PCs residing in the bone marrow under homeostatic conditions can be mobilized into the circulation in response to tissue damage and are believed to be essential in tissue regeneration replacing terminally differentiated cells lost because of physiological cell turnover or tissue damage [10].
The role of PCs in COPD and tissue regeneration is not fully understood, and different mechanisms have been suggested to explain the reduced number of circulating PCs seen in COPD patients. One potential explanation is an increased recruitment of these cells to sites of tissue injury, namely in the pulmonary vasculature. Tissue repair and regeneration following injury is thought to involve resident cell proliferation as well as a selective recruitment of circulating progenitor cell populations through complex signalling cascades [24]. Our group has previously shown an increased number of CD133+ progenitor cells localized in the wall of pulmonary arteries of COPD patients [22]. Similarly, in this study a significant number of CD133+ progenitor cells localized within the pulmonary artery of COPD patients suggesting their involvement in pulmonary vessel repair mechanisms.
In the present study, we extend these previous findings and show that COPD patients present a greater number of CD45+ cells in pulmonary arteries than non-COPD subjects. Interestingly, the number of CD45+ cells within the pulmonary artery wall was inversely correlated with the number of circulating CD45+CD34+CD133+ cells, patients with lower number of circulating PCs had a higher number of CD45+ cells in the vessel wall. Although this correlation does not establish a cause and effect relationship, it is plausible that in COPD, progenitor cell mobilization and homing in pulmonary arteries may occur in response to vascular damage, causing the depletion of the pool of circulating PCs.
Exposure to CS is the primary cause of COPD and plays a key role in PC dysfunction [25]. Dysfunctional PCs show excessive apoptosis and are unable to respond to external injuries to mediate proper lung tissue healing [26]. Previous results from our group showed in an experimental animal model, that short-term exposure to CS induced PCs dysfunction, affecting pulmonary homing and proliferation [27]. In vitro, CS altered the rate of proliferation, senescence, differentiation and migration capacity of PCs [27]. In COPD patients, dysfunctional PCs may be unable to support the normal repair of the pulmonary vasculature promoting neointima formation, vessel remodelling and disease progression. In line with these results, our group has previously shown that bone marrow derived CD133+ cells had the capacity to migrate from the vessel lumen into the intima and differentiate into smooth muscle cells, exerting remodelling effects on injured vessels [28].
In the present study, COPD patients displayed thicker pulmonary artery walls and reduced lumens than non-COPD subjects. Interestingly, the increased number of CD45+ cells localized in the pulmonary artery wall was associated with a reduction in the arterial lumen. This is in agreement with previous observations from our group showing that structural alterations in pulmonary arteries occur at early stages in COPD and that the number of CD133+ cells attached to the endothelium was greater in COPD patients than in control subjects [23]. Overall, the current and previous findings suggest a potential causative role of progenitor cell recruitment in the pathogenesis of pulmonary vascular remodelling in COPD.
COPD patients and non-COPD subjects showed similar values of systemic endothelial function, though both groups had lower values than those previously observed in healthy non-smokers [17]. This indicates that both COPD patients and non-COPD subjects in this study presented systemic endothelial dysfunction. We have previously reported significant differences in FMD values between control non-smokers and both COPD patients and smokers without COPD [17]. Accordingly, the reduced value of FMD shown herein in the non-COPD group is likely due to the fact that most of the non-COPD subjects were heavy smokers. When the non-COPD group was divided in non-smokers and smokers a marked increase of FMD was seen in the non-smoker group compared to the smoker control group. We also observed that FMD values were unrelated to the number of circulating PCs or CD45+ cells localized in the pulmonary artery wall. This agrees with previous findings where no significant correlation was found between the number of circulating PCs and FMD values between COPD and non-COPD subjects [17].
Endothelium-dependent dilation and distensibility values measured in isolated pulmonary arteries were reduced in COPD patients compared to non-COPD subjects and correlated with the number of infiltrating CD45+ cells in pulmonary arteries. This is consistent with the notion that impairment of the endothelial function in pulmonary arteries may promote the mobilization and homing of bone marrow-derived PCs. Systemic endothelial dysfunction did not correlate with pulmonary endothelial dysfunction. We consider that endothelial dysfunction in systemic arteries is more likely due to chronic CS exposure rather than a systemic effect of COPD. On the other hand, pulmonary endothelial dysfunction appears to be more directly related to progenitor cell homing in response to injury and vessel remodelling. Overall, these findings suggest that CS might exert direct effects on endothelial function in both systemic and pulmonary arteries, irrespective of the number of recruited PCs in remodelled pulmonary arteries.
DLco values were not associated with variations in the number of circulating PCs or bone marrow-derived cells recruited in pulmonary vessels. Yet, lower DLco values were associated with greater remodelling of pulmonary arteries. Neither the severity of airflow obstruction nor the values of PaO2, were related to changes in PCs or pulmonary vascular remodelling. Overall, our findings are in line with the notion that DLco is a marker of pulmonary vascular integrity. Finally, non-COPD controls were separated in non-smokers and smokers and compared to COPD patients. In most parameters analyzed, non-COPD smokers presented an intermediate phenotype between non-smoker controls and COPD subjects indicating the key role of CS exposure in lung and endothelial dysfunction.
The main strength of this study was the simultaneous measurement of markers of vascular integrity and function in both systemic and pulmonary arteries, using lung tissue samples, in the same individual. Nevertheless, our study has some limitations. First, the reduced number of subjects, which is inherent to the need to evaluate patients undergoing lung resection in whom the lung neoplasm was localized and did not produce changes in lung parenchyma or respiratory function. Second, the CD45 maker used in tissue assessment is not specific to progenitor cells and could also indicate recruitment of other cell populations. Further studies using double staining for both CD133 and CD45 immunofluorescence are required to prove that these migrated cells in the intima of the pulmonary arteries are derived from circulating PCs. Third, in this study we could not be certain that the number of CD45+ cells present in the pulmonary walls and the reduced artery lumen area, were directly associated to the reduced number of circulating CD45+CD34+CD133+ cells in these patients. However, the direct correlation between the number of circulating CD45+CD34+CD133+ cells and the number of CD45+ infiltrates in pulmonary vessels and the presence of CD133+ cells, an intrinsic marker of progenitor cells, observed in the vessel wall might infer that in COPD patients, circulating CD45+CD34+CD133+ cells are recruited in response to injury to the pulmonary vessel wall, causing a reduction in circulating PCs numbers.
Interestingly, recent data suggested the existence of a “vasculogenic zone” in the wall of human blood vessels, which might serve as a reservoir for PCs capable to differentiate into mature endothelial cells [29]. Accordingly, the presence of PCs recruited as a result of an injury response could not solely be explained by the homing of bone-marrow derived circulating PCs as tissue resident PCs could also play an important role. Finally, some of the results are represented as correlations and therefore only describe an association, they do not prove a cause and effect relationship.

Conclusion

This integrative study shows a reduction of circulating PCs in COPD, which is associated with the recruitment of bone marrow-derived cells into the patient’s pulmonary artery wall. This, in turn, is associated with endothelial dysfunction and vessel remodelling of pulmonary arteries. Overall, our results suggest the contribution of bone marrow-derived PCs in pulmonary vascular remodelling and are consistent with the notion of an imbalance between endothelial damage and repair capacity in the pathogenesis of pulmonary vascular abnormalities in COPD.

Acknowledgements

The authors would like to thank S. Poyatos, G. Argemí, D. Domínguez-Fandos and M. Díez for their skilful collaboration in the study.

Funding

This study was supported by grants from the Instituto de Salud Carlos III PI05/0244, PI12/00510, PI16/01147 and PIE15/00582; Fundació La Marató de TV3 (04310), SEPAR 24/2015. S. Pizarro was the recipient of a predoctoral research fellowship from Hospital Clínic, J. García-Lucio was the recipient of a predoctoral research fellowship (PFIS) from the Instituto de Salud Carlos III and O. Tura-Ceide was the recipient of Marie Curie Post-Doctoral Fellowship Award BIOTRACK: IDIBAPS and a Miguel Servet grant from the Instituto de Salud Carlos III (CP17/00114).. Cofinanciado por el Fondo Europeo de Desarrollo Regional (FEDER). UnioÂn Europea. “Una manera de hacer Europa”.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
The study was conducted in accordance with the Declaration of Helsinki, was approved by the Committee on Human Research of our institution and all subjects gave written informed consent.
All subjects gave written informed consent.

Competing interests

The authors declare no conflict of interest. The funding sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Decramer M, Janssens W, Miravitlles M. Chronic obstructive pulmonary disease. Lancet. 2012;379:1341–51.CrossRef Decramer M, Janssens W, Miravitlles M. Chronic obstructive pulmonary disease. Lancet. 2012;379:1341–51.CrossRef
2.
Zurück zum Zitat Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442.CrossRef Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442.CrossRef
3.
Zurück zum Zitat Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet (London, England). 2012;380:2095–128. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet (London, England). 2012;380:2095–128.
4.
Zurück zum Zitat Peinado VI, Barberà JA, Ramírez J, Gomez FP, Roca J, Jover L, Gimferrer JM, Rodríguez-Roisin R. Endothelial dysfunction in pulmonary arteries of patients with mild COPD. Am J Phys. 1998;274:L908–13. Peinado VI, Barberà JA, Ramírez J, Gomez FP, Roca J, Jover L, Gimferrer JM, Rodríguez-Roisin R. Endothelial dysfunction in pulmonary arteries of patients with mild COPD. Am J Phys. 1998;274:L908–13.
6.
Zurück zum Zitat Barberà JA, Blanco I. Gaining insights into pulmonary hypertension in respiratory diseases. Eur Respir J. 2015;46:1247–50.CrossRef Barberà JA, Blanco I. Gaining insights into pulmonary hypertension in respiratory diseases. Eur Respir J. 2015;46:1247–50.CrossRef
7.
Zurück zum Zitat Peinado VI, Pizarro S, Barberà JA. Pulmonary vascular involvement in COPD. Chest. 2008;134:808–14.CrossRef Peinado VI, Pizarro S, Barberà JA. Pulmonary vascular involvement in COPD. Chest. 2008;134:808–14.CrossRef
8.
Zurück zum Zitat Salter B, Sehmi R. The role of bone marrow-derived endothelial progenitor cells and angiogenic responses in chronic obstructive pulmonary disease. J Thorac Dis. 2017;9:2168–77.CrossRef Salter B, Sehmi R. The role of bone marrow-derived endothelial progenitor cells and angiogenic responses in chronic obstructive pulmonary disease. J Thorac Dis. 2017;9:2168–77.CrossRef
9.
Zurück zum Zitat Maclay JD, McAllister DA, Mills NL, Paterson FP, Ludlam CA, Drost EM, Newby DE, MacNee W. Vascular dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:513–20.CrossRef Maclay JD, McAllister DA, Mills NL, Paterson FP, Ludlam CA, Drost EM, Newby DE, MacNee W. Vascular dysfunction in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;180:513–20.CrossRef
10.
Zurück zum Zitat García-Lucio J, Peinado VI, de Jover L, Pozo R, Blanco I, Bonjoch C, Coll-Bonfill N, Paul T, Tura-Ceide O, Barberà JA. Imbalance between endothelial damage and repair capacity in chronic obstructive pulmonary disease. PLoS One. 2018;13:e0195724.CrossRef García-Lucio J, Peinado VI, de Jover L, Pozo R, Blanco I, Bonjoch C, Coll-Bonfill N, Paul T, Tura-Ceide O, Barberà JA. Imbalance between endothelial damage and repair capacity in chronic obstructive pulmonary disease. PLoS One. 2018;13:e0195724.CrossRef
11.
Zurück zum Zitat Naito H, Kidoya H, Sakimoto S, Wakabayashi T, Takakura N. Identification and characterization of a resident vascular stem/progenitor cell population in preexisting blood vessels. EMBO J. 2012;31:842–55.CrossRef Naito H, Kidoya H, Sakimoto S, Wakabayashi T, Takakura N. Identification and characterization of a resident vascular stem/progenitor cell population in preexisting blood vessels. EMBO J. 2012;31:842–55.CrossRef
12.
Zurück zum Zitat Liu X, Liu Y, Huang X, Lin G, Xie C. Endothelial progenitor cell dysfunction in acute exacerbation of chronic obstructive pulmonary disease. Mol Med Rep. 2017;16:5294–302.CrossRef Liu X, Liu Y, Huang X, Lin G, Xie C. Endothelial progenitor cell dysfunction in acute exacerbation of chronic obstructive pulmonary disease. Mol Med Rep. 2017;16:5294–302.CrossRef
13.
Zurück zum Zitat Koutroumpi M, Dimopoulos S, Psarra K, Kyprianou T, Nanas S. Circulating endothelial and progenitor cells: evidence from acute and long-term exercise effects. World J Cardiol. 2012;4:312–26.CrossRef Koutroumpi M, Dimopoulos S, Psarra K, Kyprianou T, Nanas S. Circulating endothelial and progenitor cells: evidence from acute and long-term exercise effects. World J Cardiol. 2012;4:312–26.CrossRef
14.
Zurück zum Zitat Schmidt-Lucke C, Rössig L, Fichtlscherer S, Vasa M, Britten M, Kämper U, Dimmeler S, Zeiher AM. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111:2981–7.CrossRef Schmidt-Lucke C, Rössig L, Fichtlscherer S, Vasa M, Britten M, Kämper U, Dimmeler S, Zeiher AM. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111:2981–7.CrossRef
15.
Zurück zum Zitat Brittan M, Hoogenboom MM, Padfield GJ, Tura O, Fujisawa T, Maclay JD, Macnee W, Mills NL. Endothelial progenitor cells in patients with chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2013;305:L964–9.CrossRef Brittan M, Hoogenboom MM, Padfield GJ, Tura O, Fujisawa T, Maclay JD, Macnee W, Mills NL. Endothelial progenitor cells in patients with chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2013;305:L964–9.CrossRef
16.
Zurück zum Zitat Janssen WJ, Yunt ZX, Muldrow A, Kearns MT, Kloepfer A, Barthel L, Bratton DL, Bowler RP, Henson PM. Circulating hematopoietic progenitor cells are decreased in COPD. COPD. 2014;11:277–89.PubMed Janssen WJ, Yunt ZX, Muldrow A, Kearns MT, Kloepfer A, Barthel L, Bratton DL, Bowler RP, Henson PM. Circulating hematopoietic progenitor cells are decreased in COPD. COPD. 2014;11:277–89.PubMed
17.
Zurück zum Zitat Pizarro S, García-Lucio J, Peinado VI, Tura-Ceide O, Díez M, Blanco I, Sitges M, et al. Circulating progenitor cells and vascular dysfunction in chronic obstructive pulmonary disease. PLoS One. 2014;9:e106163.CrossRef Pizarro S, García-Lucio J, Peinado VI, Tura-Ceide O, Díez M, Blanco I, Sitges M, et al. Circulating progenitor cells and vascular dysfunction in chronic obstructive pulmonary disease. PLoS One. 2014;9:e106163.CrossRef
18.
Zurück zum Zitat Barberà JA, Riverola A, Roca J, Ramírez J, Wagner PD, Ros D, Wiggs BR, Rodríguez-Roisin R. Pulmonary vascular abnormalities and ventilation-perfusion relationships in mild chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1994;149:423–9.CrossRef Barberà JA, Riverola A, Roca J, Ramírez J, Wagner PD, Ros D, Wiggs BR, Rodríguez-Roisin R. Pulmonary vascular abnormalities and ventilation-perfusion relationships in mild chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1994;149:423–9.CrossRef
19.
Zurück zum Zitat Diller GP, van Eijl S, Okonko DO, Howard LS, Ali O, Thum T, Wort SJ, et al. Circulating endothelial progenitor cells in patients with Eisenmenger syndrome and idiopathic pulmonary arterial hypertension. Circulation. 2008;117:3020–30.CrossRef Diller GP, van Eijl S, Okonko DO, Howard LS, Ali O, Thum T, Wort SJ, et al. Circulating endothelial progenitor cells in patients with Eisenmenger syndrome and idiopathic pulmonary arterial hypertension. Circulation. 2008;117:3020–30.CrossRef
20.
Zurück zum Zitat Tung YC, Zhang M, Lin CT, Kurabayashi K, Skerlos SJ. PDMS-based opto-fluidic micro flow cytometer with two-color, multi-angle fluorescence detection capability using PIN photodiodes. Sensors Actuators B Chem. 2004;98:356–67.CrossRef Tung YC, Zhang M, Lin CT, Kurabayashi K, Skerlos SJ. PDMS-based opto-fluidic micro flow cytometer with two-color, multi-angle fluorescence detection capability using PIN photodiodes. Sensors Actuators B Chem. 2004;98:356–67.CrossRef
21.
Zurück zum Zitat Corretti MC, Anderson TJ, Benjamin EJ, Celermajer D, Charbonneau F, Creager MA, Deanfield J, et al. Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: a report of the international brachial artery reactivity task force. J Am Coll Cardiol. 2002;39:257–65.CrossRef Corretti MC, Anderson TJ, Benjamin EJ, Celermajer D, Charbonneau F, Creager MA, Deanfield J, et al. Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: a report of the international brachial artery reactivity task force. J Am Coll Cardiol. 2002;39:257–65.CrossRef
22.
Zurück zum Zitat Peinado VI, Ramírez J, Roca J, Rodríguez-Roisin R, Barberà JA. Identification of vascular progenitor cells in pulmonary arteries of patients with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2006;34:257–63.CrossRef Peinado VI, Ramírez J, Roca J, Rodríguez-Roisin R, Barberà JA. Identification of vascular progenitor cells in pulmonary arteries of patients with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2006;34:257–63.CrossRef
23.
Zurück zum Zitat Boudreau N, Turley E, Rabinovitch M. Fibronectin, hyaluronan, and a hyaluronan binding protein contribute to increased ductus arteriosus smooth muscle cell migration. Dev Biol. 1991;143:235–47.CrossRef Boudreau N, Turley E, Rabinovitch M. Fibronectin, hyaluronan, and a hyaluronan binding protein contribute to increased ductus arteriosus smooth muscle cell migration. Dev Biol. 1991;143:235–47.CrossRef
24.
Zurück zum Zitat Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med. 2012;7:833–50.CrossRef Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med. 2012;7:833–50.CrossRef
25.
Zurück zum Zitat Kondo T, Hayashi M, Takeshita K, Numaguchi Y, Kobayashi K, Iino S, Inden Y, Toyoaki M. Smoking cessation rapidly increases circulating progenitor cells in peripheral blood in chronic smokers. Arterioscler Thromb Vasc Biol. 2004;24:1442–7.CrossRef Kondo T, Hayashi M, Takeshita K, Numaguchi Y, Kobayashi K, Iino S, Inden Y, Toyoaki M. Smoking cessation rapidly increases circulating progenitor cells in peripheral blood in chronic smokers. Arterioscler Thromb Vasc Biol. 2004;24:1442–7.CrossRef
26.
Zurück zum Zitat Heiss C, Amabile N, Lee AC, Real WM, Schick SF, Lao D, Wong ML, et al. Brief secondhand smoke exposure depresses endothelial progenitor cells activity and endothelial function: sustained vascular injury and blunted nitric oxide production. J Am Coll Cardiol. 2008;51:1760–71.CrossRef Heiss C, Amabile N, Lee AC, Real WM, Schick SF, Lao D, Wong ML, et al. Brief secondhand smoke exposure depresses endothelial progenitor cells activity and endothelial function: sustained vascular injury and blunted nitric oxide production. J Am Coll Cardiol. 2008;51:1760–71.CrossRef
27.
Zurück zum Zitat Tura-Ceide O, Lobo B, Paul T, Puig-Pey R, Coll-Bonfill N, García-Lucio J, Smolders V, Blanco I, Barberà JA, Peinado VI. Cigarette smoke challenges bone marrow mesenchymal stem cell capacities in Guinea pig. Respir Res. 2017;18:50.CrossRef Tura-Ceide O, Lobo B, Paul T, Puig-Pey R, Coll-Bonfill N, García-Lucio J, Smolders V, Blanco I, Barberà JA, Peinado VI. Cigarette smoke challenges bone marrow mesenchymal stem cell capacities in Guinea pig. Respir Res. 2017;18:50.CrossRef
28.
Zurück zum Zitat Díez M, Barberà JA, Ferrer E, Fernández-Lloris R, Pizarro S, Roca J, Peinado VI. Plasticity of CD133+ cells: role in pulmonary vascular remodeling. Cardiovasc Res. 2007;76:517–27.CrossRef Díez M, Barberà JA, Ferrer E, Fernández-Lloris R, Pizarro S, Roca J, Peinado VI. Plasticity of CD133+ cells: role in pulmonary vascular remodeling. Cardiovasc Res. 2007;76:517–27.CrossRef
29.
Zurück zum Zitat Zengin E, Chalajour F, Gehling UM, Ito WD, Treede H, Lauke H, Weil J, Reichenspurner H, Kilic N, Ergün S. Vascular wall resident progenitor cells: a source for postnatal vasculogenesis. Development. 2006;133:1543–51.CrossRef Zengin E, Chalajour F, Gehling UM, Ito WD, Treede H, Lauke H, Weil J, Reichenspurner H, Kilic N, Ergün S. Vascular wall resident progenitor cells: a source for postnatal vasculogenesis. Development. 2006;133:1543–51.CrossRef
Metadaten
Titel
Progenitor cell mobilisation and recruitment in pulmonary arteries in chronic obstructive pulmonary disease
verfasst von
Olga Tura-Ceide
Sandra Pizarro
Jéssica García-Lucio
Josep Ramírez
Laureano Molins
Isabel Blanco
Yolanda Torralba
Marta Sitges
Cristina Bonjoch
Victor I. Peinado
Joan Albert Barberà
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2019
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-019-1024-z

Weitere Artikel der Ausgabe 1/2019

Respiratory Research 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.