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

Open Access 01.12.2019 | Research

Reverse-D-4F improves endothelial progenitor cell function and attenuates LPS-induced acute lung injury

verfasst von: Nana Yang, Hua Tian, Enxin Zhan, Lei Zhai, Peng Jiao, Shutong Yao, Guohua Lu, Qingjie Mu, Juan Wang, Aihua Zhao, Yadong Zhou, Shucun Qin

Erschienen in: Respiratory Research | Ausgabe 1/2019

Abstract

Background

Patients with acute lung injury (ALI) have increased levels of pro-inflammatory mediators, which impair endothelial progenitor cell (EPC) function. Increasing the number of EPC and alleviating EPC dysfunction induced by pro-inflammatory mediators play important roles in suppressing ALI development. Because the high density lipoprotein reverse-D-4F (Rev-D4F) improves EPC function, we hypothesized that it might repair lipopolysaccharide (LPS)-induced lung damage by improving EPC numbers and function in an LPS-induced ALI mouse model.

Methods

LPS was used to induce ALI in mice, and then the mice received intraperitoneal injections of Rev-D4F. Immunohistochemical staining, flow cytometry, MTT, transwell, and western blotting were used to assess the effect of Rev-D4F on repairment of lung impairment, and improvement of EPC numbers and function, as well as the signaling pathways involved.

Results

Rev-D4F inhibits LPS-induced pulmonary edema and decreases plasma levels of the pro-inflammatory mediators TNF-α and ET-1 in ALI mice. Rev-D4F inhibited infiltration of red and white blood cells into the interstitial space, reduced lung injury-induced inflammation, and restored injured pulmonary capillary endothelial cells. In addition, Rev-D4F increased numbers of circulating EPC, stimulated EPC differentiation, and improved EPC function impaired by LPS. Rev-D4F also acted via a PI3-kinase-dependent mechanism to restore levels of phospho-AKT, eNOS, and phospho-eNOS suppressed by LPS.

Conclusions

These findings indicate that Rev-D4F has an important vasculoprotective role in ALI by improving the EPC numbers and functions, and Rev-D4F reverses LPS-induced EPC dysfuncion partially through PI3K/AKT/eNOS signaling pathway.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12931-019-1099-6) contains supplementary material, which is available to authorized users.
Nana Yang, Hua Tian and Enxin Zhan contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALI
Acute lung injury
ARDS
Acute respiratory distress syndrome
EPC
Endothelial progenitor cell
HDL
High density lipoprotein
LPS
Lipopolysaccharide
PCEC
Pulmonary capillary endothelial cells

Background

Acute lung injury (ALI) is a common clinical syndrome characterized by high mortality [1]. ALI pathogenesis can be caused by biological and abiotic pathogens. Biological pathogens include bacteria, viruses, fungi, atypical pathogens, and malignant tumors, while abiotic pathogens include acidic substances, drugs, toxic gas inhalation, and mechanical ventilation-related injuries [2]. Initially, endotoxins and other harmful substances in the blood circulation injure pulmonary capillary endothelial cells (PCEC), causing an increase in permeability in endothelial cell monolayers, as well as endothelial cell shrinkage and death. Approximately two hours post-injury, pulmonary interstitial edema can occur, and alveolar edema occurs approximately 12–24 h post-injury [3].
Endothelial cell dysfunction and inflammation play important roles in ALI initiation and development [3]. In ALI, an improved patient survival correlates with a higher colony count of endothelial progenitor cell (EPC) [4]. EPC engraftment maintains the integrity of pulmonary alveolar-capillary barrier, reestablishes endothelial function in vessels, and ameliorates the inflammatory state in LPS-induced ALI in rat models [5]. Therefore, EPC contributes to the repair of endothelial impairment and prevention of ALI development. However, patients with ALI have increased levels of pro-inflammatory mediators (e.g., ox-LDL, IL-6, TNF-α), which impair EPC function, including proliferation, migration, and tube formation. These impaired EPC are unable to restore the damaged endothelial system in patients with ALI [6]. Therefore, increasing the number of EPC and alleviating EPC dysfunction induced by pro-inflammatory mediators may suppress the development of ALI.
High-density lipoprotein (HDL) regulates EPC mobilization from bone marrow, prevents EPC apoptosis, and promotes differentiation of EPC into endothelial cells [7]. Apolipoprotein A-I (apoA-I), an important functional component of HDL, has 243 amino acids, and improves survival in septic rats [8]. Because the lipid-binding properties of apoA-I are largely related to its class A amphipathic helices, designed mimetic peptides, which contain four phenylalanine residues on the hydrophobic face (4F; F being the biochemical symbol for phenylalanine) and mimic the class A amphipathic helices, have no sequence homology with apoA-I [9]. The 4F residues have anti-inflammatory and antioxidant activities, and improve survival in septic rats [8]. Kruger et al. have shown that the mimetic peptide D-4F can prevent the loss of certain EPC functions by increasing endothelial nitric oxide synthase (eNOS) in EPC [10]. Our previous studies revealed that reverse D-4F (Rev-D4F) acts via a PI3K/AKT/eNOS-dependent mechanism to significantly improve EPC proliferation, migration, and tube formation [11]. Thus, HDL, apoA-I, and its mimetic peptides are new drugs for the treatment of lung injury [12]. However, the precise mechanisms of the protective effects of HDL and HDL mimetic peptides Rev-D4F on EPC damage induced by LPS have not been entirely clarified. In this study, we investigated the effect of Rev-D4F on EPC numbers and function in LPS-induced ALI mice.

Methods

Animals

Male C57 mice (6 weeks old) were obtained from the Vital River Company (Beijing, China) and randomly divided into four groups with six mice in each group: the control group (PBS), Rev-D4F group (Rev-D4F 10 mg/Kg and PBS), LPS group (PBS and LPS 10 mg/Kg), and Rev-D4F treatment group (Rev-D4F 10 mg/Kg and LPS 10 mg/Kg). Either vehicle or Rev-D4F (10 mg/kg) was administered by intraperitoneal injection.
All animal procedures complied with the Guide for the Care and Use of Laboratory Animals published by Weifang Medical University, and the protocol was approved by the Committee on the Ethics of Animal Experiments of Weifang Medical University. Mice were anesthetized by intraperitoneal injection of 400 mg/kg chloral hydrate.

Rev-D4F synthesis

The Reverse D4F mimetic peptide synthesis method was used to obtain inverse chirality with D-amino acids only and reverse order of D4F (Ac-DWFKAFYDKVAEKFKEAF-NH2) via a practical approach of Fmoc solid-phase peptide synthesis by the Scilight-Peptide INC (Beijing, BJ, China). The structure and purity (98%) were determined by GC/MS.

Analysis of wet: dry weight ratio

The left lung was taken, and lung surface liquid was absorbed with an absorbent paper. Weight of the left lung was considered the wet weight. The lung was dried at 60 °C for 48 h, and then reweighed to obtain the dry weight. The degree of lung edema was evaluated by calculating the wet: dry weight ratio.

Measurement of serum cytokines

ELISA kits (Blue Gene, Shanghai, China) were used to measure TNF-α and ET-1 levels in mice plasma according to the manufacturer’s instructions.

Histopathologic evaluations and Immunohistochemical staining

The right lobes of the lungs were fixed in 10% formalin (Sigma-Aldrich, St Louis, MO, USA), embedded in paraffin, cut to 6 μm sections, and then stained with Hematoxylin-eosin (Sigma-Aldrich, St Louis, MO, USA). Immunohistochemistry (IHC) was performed with specific antibodies against Sca-1 (Abcam, Cambridge, UK), VCAM-1, ICAM-1, eNOS, iNOS, or vWF (Santa Cruz Biotechnology, Santa Cruz, CA, USA), and the samples were then counterstained with Hematoxylin.
Red and white blood cells in the lung parenchyma were counted. Red and white blood cell counts, as well as quantitative alveolar wall thickness measurements were performed based on the segmented morphology in 10 randomly selected fields in each section (n = 6/group) using high power fields (× 200).

Flow cytometric analysis of EPC

Red blood cell-lysing buffer (BD Biosciences, San Jose, CA, USA) was used to lyse peripheral blood erythrocytes. Then, other cells were labeled with the FITC-conjugated antibodies against CD34, PE-conjugated antibodies against FLK-1, and APC-conjugated antibodies against CD133 (BD Biosciences, San Jose, CA, USA). A gating strategy and isotype-identical antibodies were used to exclude debris and nonspecific fluorescent signals.

EPC isolation

In vitro isolation of mice bone marrow-derived EPC were performed as described previously [11]. The mononuclear cells (MNCs) were obtained from the femurs of 4-week-old male C57 mice by density gradient centrifugation (Sigma-Aldrich, St Louis, MO, USA), and plated on fibronectin-coated 6 well plates in EGM-2MV (Endothelial cell basal medium-2, plus FBS, VEGF, R-IGF-1, rhEGF, rhFGF-B, GA-1000, hydrocortisone and ascorbic acid) (Lonza, Basel, Switzerland). After 3 days, non-adherent cells were removed, and fresh medium was applied every 3 days.

Characterization of EPC

MNCs from mice bone marrow were cultured for 10 days, incubated with 2.5 mg/L DiI-ac-LDL (Thermo Fisher Scientific Inc., Waltham, MA, USA) for 2 h at 37 °C, and 10 mg/L FITC-UEA (Sigma-Aldrich, St Louis, MO, USA) for 1 h at 37 °C, and then fixed for 5 min with 1–2% paraformaldehyde (Sigma-Aldrich, St Louis, MO, USA). Double-positive cells and total cell numbers were counted in five random fields under a fluorescence microscope (× 100). The positive cells (%) represent the numbers of double-positive cells compared with the total number of cells.

EPC treatment

MNCs obtained from mice bone marrow were induced for 21 days by EGM-2MV and differentiated into late outgrowth EPC. Before experiment, the cell medium was changed to basic medium (M199 + 5% FBS), and cells were separated into 5 groups and treated as follows: Control (M199 + 5% FBS), Rev-D4F (M199 + 5% FBS + 50 μg/ml reverse D-4F), LPS (M199 + 5% FBS + 30 μg/ml LPS), LPS + Rev-D4F (M199 + 5% FBS + 50 μg/ml reverse D-4F + 30 μg/ml LPS), LPS + Rev-D4F + LY294002 (M199 + 5% FBS + 50 μg/ml reverse D-4F + 30 μg/ml LPS + 30 μM LY294002). After pretreatment with the PI3-kinase inhibitor LY294002 (30 μM) (Sigma-Aldrich, St Louis, MO, USA) for 2 h, EPC were incubated with Rev-D4F (50 μg/ml) for 6 h, and then treated with LPS for 36 h to induce EPC damage without removal of LY294002 and Rev-D4F.

EPC viability and proliferation

EPC viability was determined by MTT assay. After different treatments, cells were incubated with 20 μl 5 mg/ml MTT (Sigma-Aldrich, St Louis, MO, USA) for 4 h at 37 °C. The supernatant was aspirated, and 150 μL of dimethyl sulfoxide (DMSO, Sigma-Aldrich, St Louis, MO, USA) was added to each well and shaken for 10 min. OD values were measured at 490 nm.
EPC proliferation was evaluated by flow cytometry analysis. After treatments, cells were digested with 0.25% trypsin (Sigma-Aldrich, St Louis, MO, USA), fixed with 75% ethanol, and stained with FITC-conjugated antibodies against Ki67 (BD Biosciences, San Jose, CA, USA). A gating strategy and isotype-identical antibodies were used to exclude debris and nonspecific fluorescent signals.

EPC migration

EPC migration was measured using an 8 μm pore 24-well Cell Migration Assay kit (BD Biosciences, San Jose, CA, USA). After treatments, cells were digested with 0.25% trypsin, and 1.2 × 104 cells in M199 were placed in the upper chamber. EGM-2MV medium was placed in the lower compartment. After 24 h incubation at 37 °C, the upper cells were removed with a cotton wool swab, and the transwell filters were fixed and stained with DAPI (Sigma-Aldrich, St Louis, MO, USA). The migratory cells were counted in five random microscopic fields (× 100).

EPC adhesion

Mice bone marrow-derived MNCs were cultured on fibronectin-coated 6 well plates at 106 cells per cm2. After 3 days, non-adherent cells were removed, and adherent cells were cultured for additional 4 days. Then, we randomly selected five fields of view and counted the number of adherent cells under a microscope (× 100).

Tube formation in vitro

After different treatments, cells were digested with 0.25% trypsin and 1.0 × 104 cells in M199 plus 10% FBS were placed on matrigel (BD Biosciences, San Jose, CA, USA) in a 96 wells plate. After 18 h incubation, the average total length of complete tubes formed by cells was measured under a microscope (× 40) in five random microscopic fields using computer software, Image-Pro Plus.

Western blot analysis

Total protein was extracted with RIPA lysis buffer and quantified using the BCA method. In total, 30 μg of protein was electrophoresed on a 10% denaturing polyacrylamide gel, and transferred onto PVDF membranes. After blocking with 5% dried skimmed milk, the membranes were incubated with antibodies against β-actin (1: 20000, Sigma-Aldrich, St Louis, MO, USA), AKT (1: 2000, Cell Signaling Technologies, Beverly, MA, USA), phosphor-AKT (1: 1000, Cell Signaling Technologies, Beverly, MA, USA), eNOS (1: 500, Santa Cruz Biotechnology, Santa Cruz, CA, USA), phosphor-eNOS (1: 1000, Cell Signaling Technologies, Beverly, MA, USA), CD133 (1: 500, Abcam, Cambridge, UK), FLK-1 (1: 500,Abcam, Cambridge, UK) or Sca-1 (1: 500, Abcam, Cambridge, UK) for 3 h, and subsequently incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG antibody (1: 3000, Santa Cruz Biotechnology, Santa Cruz, CA, USA). The signals were detected using the Phototope-HRP Western Detection Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA).

Immunofluorescence staining

MNCs were cultured and differentiated into relatively mature endothelial cells. The lung tissues were used to make frozen sections. Next, cells and frozen sections were blocked with goat serum at room temperature for 30 min, and then incubated with anti-mouse CD31 (Santa Cruz Biotechnology, Santa Cruz, CA, USA), vWF (Santa Cruz Biotechnology, Santa Cruz, CA, USA), CD133 (Abcam, Cambridge, UK) or FLK-1 (Abcam, Cambridge, UK) antibodies for 1 h at 37 °C. Finally, cells and tissues were incubated with secondary antibodies conjugated with Cy3 or FITC, and staining was assessed under fluorescence microscopy (× 200).

Statistical analysis

All data are presented as mean ± SD. Variables between groups were assessed by one-way ANOVA using the software program SPSS11.5. Values of P<0.05 were considered significant.

Results

Rev-D4F suppresses LPS-induced lung injury in ALI mice

Compared to control group, LPS induced pulmonary edema, which was significantly inhibited by Rev-D4F in ALI mice (Fig. 1a). The pro-inflammatory cytokine TNF-α and the vasoconstrictor endothelin-1 (ET-1) contribute to ALI pathogenesis. Accordingly, LPS increased ET-1 and TNF-α levels in ALI mice plasma, which were inhibited by Rev-D4F (Fig. 1b, c).
In order to assess pulmonary injury, hematoxylin-eosin was used to stain lung tissues. The alveolar walls were thickened, and the interstitial space was infiltrated with red and white blood cells in the LPS-induced group (Fig. 1d-g). The majority of white blood cells were observed in the septa, and fewer red and white blood cells were observed in the alveolar spaces. Importantly, Rev-D4F reversed the LPS-induced changes (Fig. 1d-g).

Rev-D4F reduces inflammation and restores injured lung PCEC in ALI mice

ICAM-1, VCAM-1, and iNOS are considered inflammation markers of endothelial injury [13], while eNOS and vWF are important surface markers of endothelial cells [14]. Compared with the control group, expression of ICAM-1, VCAM-1, and iNOS in lung tissue was significantly increased in the LPS treatment group. Notably, the levels of ICAM-1, VCAM-1, and iNOS were inhibited by Rev-D4F (Fig. 2).
The function of PCECs is often impaired in endotoxin-induced ALI, resulting in increased permeability of PCECs, and pulmonary interstitial and alveolar edema [3]. Importantly, Rev-D4F restored the levels of endothelial cell surface markers, eNOS and vWF, compared with the LPS-treated group (Fig. 2).

Effect of rev-D4F on EPC numbers

EPC numbers positively correlate with the prognosis of patients with ALI [4]. Therefore, EPC mobilization contributes to the treatment of ALI. Compared with the control group, LPS significantly increased the numbers of CD34+, FLK-1+, CD133+, CD34+/FLK-1+, FLK-1+/CD133+, and CD34+/CD133+ cells in mice peripheral blood (Fig. 3a). Compared with the LPS treatment group, Rev-D4F inhibited the levels of CD34+, CD34/FLK-1+, and CD34+/CD133+ cells, while it increased the levels of FLK-1+, CD133+, and FLK-1+/CD133+ cells (Fig. 3a). The stem cells antigen-1 (Sca-1) has been used as a marker of EPC. Compared with control group, the number of Sca-1+ and FLK-1+/CD133+ cells was significantly increased in LPS-treated mice. Interestingly, Rev-D4F further increased the numbers of Sca-1+ and FLK-1+/CD133+ cells in lung tissues compared to LPS group (Fig. 3b-e). Compared with control group, the level of Sca-1 in lung tissues of LPS-treated mice increased significantly, while the level of FLK-1 and CD133 increased slightly (Fig. 3f-i).

Rev-D4F restores EPC function in ALI mice

Compared with the control group, more EPC were observed in the LPS treatment group. In contrast, EPC functions, such as differentiation, migration, and adherence were impaired by LPS (Fig. 4). However, the impaired adherence and migration of EPC in the LPS treatment group were reversed by Rev-D4F (Fig. 4). Because EPC take up LDL and bind lectin (Fig. 4), these activities were considered as differentiation markers of EPC. The double-positive cell numbers decreased in LPS treatment group compared with control group, which was improved by Rev-D4F (Fig. 4).

Characterization of EPC

After 14 days of culture, MNCs isolated from mouse bone marrow showed cobblestone-like morphology (Additional file 1: Figure S1). These induced MNCs took up LDL and bound lectin (Additional file 1: Figure S1). When cultured continuously, MNCs showed the characteristics of mature ECs, expressing CD31 and vWF as identified by immunofluorescence (Additional file 1: Figure S1). Therefore, these induced MNCs were characterized as differentiated EPC.

LPS impairs EPC function

Pro-inflammatory mediators, such as LPS or TNF-α, damage the function of vascular wall cells [15]. Thus, we investigated the effect of LPS (0–30 μg/ml; 36 h) on EPC proliferation. We found that LPS significantly reduced proliferation (Additional file 2: Figure S2) and inhibited the EPC cycle in G1/S phase (Additional file 2: Figure S2). We also found that LPS significantly reduced EPC migration and tube formation (Fig. 5).

Rev-D4F reverses LPS-induced impairment of EPC function

Previously, we found that Rev-D4F (25–100 μg/ml) could improve proliferation, migration, and tube formation of EPC [11]. To investigate whether Rev-D4F could reverse the LPS-induced impairment of EPC function, cells were pretreated with Rev-D4F for 6 h, incubated with LPS for 36 h, and their proliferation, migration, and tube formation in vitro were assessed. We found that Rev-D4F rescued the impairment of EPC function induced by LPS (Fig. 5).

LY294002 inhibits rev-D4F-mediated restoration of EPC function

To investigate the protective mechanism of Rev-D4F, EPC were pre-incubated (2 h) with LY294002, a PI3-kinase inhibitor, followed by 6 h incubation with Rev-D4F, and stimulation with LPS. As shown in Fig. 5, inhibition of PI3-kinase activity by LY294002 inhibited the Rev-D4F-mediated restoration of LPS-induced EPC dysfunction (Fig. 5).

Rev-D4F restores LPS-Iimpaired PI3K/AKT/eNOS pathway

The PI3K/AKT/eNOS pathway plays an important role in the biological functions of EPC, such as proliferation, migration, differentiation, and tube formation. Here we investigated the influence of Rev-D4F on the PI3K/AKT/eNOS signaling pathway. As shown in Fig. 6, LPS decreased the levels of phosphor-AKT, eNOS, and phosphor-eNOS. Pretreatment with Rev-D4F restored the levels of phosphor-AKT, eNOS, and phosphor-eNOS. Compared to cells treated with Rev-D4F, LY294002 decreased the phosphor-AKT and phosphor-eNOS protein levels (Fig. 6), indicating that the restorative effect of Rev-D4F on AKT and eNOS signaling in EPC is mediated by the PI3K pathway.

Discussion

EPC were first isolated from peripheral blood using magnetic microbeads [15]. EPC contributes to physiological neovascularization, wound healing in vessels, tissue regeneration in ischemia [16], and repair of lung injury [17]. Our present findings demonstrate that Rev-D4F inhibits LPS-induced pulmonary edema, decreases plasma levels of TNF-αand ET-1, inhibits infiltration of red and white blood cells into the interstitial space, reduces injury-induced lung inflammation, and restores injured PCEC. Our previous study indicated that Rev-D4F improved EPC proliferation, migration, tube formation, and NO-releasing function, which was partially dependent on the PI3K/AKT/eNOS/NO pathway [11]. Our present results demonstrate that Rev-D4F improves the function of EPC impaired by LPS both in vivo and in vitro, and that Rev-D4F increases EPC numbers to repair injured PCEC.
The protective effects of apoA-I mimetic peptide 4F on vascular function in endotoxemic rats have been demonstrated. The anti-inflammatory effects of apolipoprotein A-I mimetic peptide (L-4F) in ARDS are due to the direct inhibition of endotoxin activity and increase of HDL antioxidant activity [1820]. Similarly, our results demonstrate that another apoA-I mimetic peptide, Rev-D4F, repairs LPS-induced lung damage in mice. In addition, our results show that Rev-D4F increases numbers of EPC, both in peripheral blood and in lung tissues, and improves EPC function in LPS-treated mice.
Chronic inflammation decreases numbers of circulating EPC and suppresses their activity, resulting in suppressed restoration of vessel injury and continuous inflammation [21]. Reduced numbers of circulating EPC are associated with hypercholesterolemia [22], inflammation [23], obesity [24], and diabetes [25]. HDL, active in anti-inflammation and anti-oxidation, increases numbers of circulating EPC and promotes re-endothelialization in wound healing via the PI3K/Akt/cyclin D1 signaling pathway. In addition, HDL slows down EPC senescence by increasing NO and promoting telomerase activity via the PI3K/Akt signaling pathway [26]. ApoA-I, the major structural apolipoprotein of HDL, increases numbers of circulating EPC, promotes endothelial regeneration, and attenuates neointima formation in a murine model of transplant arteriosclerosis [27]. ApoA-I mimetic peptide D-4F, mimicking ApoA-I class A amphipathic helices, promotes EPC proliferation, migration, and adhesion [28]. Our previous study has indicated that reverse D-4F, a novel ApoA-I mimetic peptide, improves EPC proliferation, migration, tube formation, and NO-releasing function, partially through the PI3K/AKT/eNOS/NO pathway [11]. Few studies have investigated whether an ApoA-I mimetic peptide D-4F, especially reverse D-4F, exerts protective effects against EPC injury induced by LPS, and little is known about the potential mechanism.
In this study, we have found that LPS significantly impairs EPC functions, such as proliferation, migration, and tube formation, and Rev-D4F inhibits the LPS-induced EPC dysfunction. Patruno et al. have found that LPS reduces the phosphorylation levels of AKT in H9c2 cells, and that AKT is an important determinant of eNOS phosphorylation at Ser1177 [29]. PI3K/Akt activation plays an essential role in HDL-induced EPC proliferation, migration, and angiogenesis [30]. Our western blot results show that LPS reduces levels of eNOS, Ser1177-phosphor-eNOS, and phosphor-AKT in EPC, and that Rev-D4F restores the LPS-attenuated levels of these proteins. In addition, our results demonstrate that the PI3K/AKT inhibitor LY294002 inhibits the Rev-D4F-restorative effects on phosphor-AKT and phosphor-eNOS levels, indicating that the Rev-D4F-mediated restoration of EPC function in LPS-treated cells is mediated by PI3K/AKT /eNOS signaling pathway.

Conclusions

Our results demonstrate that Rev-D4F repairs LPS-induced lung damage and reverses the LPS-induced EPC dysfunction. Although the mechanism of improving EPC function by Rev-D4F is not well understood, our results support the hypothesis that the restorative function of Rev-D4F is mediated by PI3K/AKT/eNOS signaling. Together, our data suggest that by improving the EPC function and numbers, Rev-D4F has an important vasculoprotective role in lung inflammatory injury.

Ethics approval

All animal procedures complied with the Guide for the Care and Use of Laboratory Animals published by Weifang Medical University, and the protocol was approved by the Committee on the Ethics of Animal Experiments of Weifang Medical University.
All list authors consent to the submission and all data are used with the consent of the person generating the data.

Competing interests

The authors declare that they have no competing interests.
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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat RRubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ, Hudson LD. Incidence and outcomes of acute lung injury. N Engl J Med. 2005;353:1685–93.CrossRef RRubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ, Hudson LD. Incidence and outcomes of acute lung injury. N Engl J Med. 2005;353:1685–93.CrossRef
2.
Zurück zum Zitat Matthay MA, Zemans RL. The acute respiratory distress syndrome: pathogenesis and treatment. Annu Rev Pathol. 2011;6:147–63.CrossRef Matthay MA, Zemans RL. The acute respiratory distress syndrome: pathogenesis and treatment. Annu Rev Pathol. 2011;6:147–63.CrossRef
3.
Zurück zum Zitat Lee WL, Slutsky AS. Sepsis and endothelial permeability. N Engl J Med. 2010;363:689–91.CrossRef Lee WL, Slutsky AS. Sepsis and endothelial permeability. N Engl J Med. 2010;363:689–91.CrossRef
4.
Zurück zum Zitat Burnham EL, Taylor WR, Quyyumi AA, Rojas M, Brigham KL, Moss M. Increased circulating endothelial progenitor cells are associated with survival in acute lung injury. Am J Respir Crit Care Med. 2005;172:854–60.CrossRef Burnham EL, Taylor WR, Quyyumi AA, Rojas M, Brigham KL, Moss M. Increased circulating endothelial progenitor cells are associated with survival in acute lung injury. Am J Respir Crit Care Med. 2005;172:854–60.CrossRef
5.
Zurück zum Zitat Mao M, Wang SN, Lv XJ, Wang Y, Xu JC. Intravenous delivery of bone marrow-derived endothelial progenitor cells improves survival and attenuates lipopolysaccharide-induced lung injury in rats. Shock. 2010;34:196–204.CrossRef Mao M, Wang SN, Lv XJ, Wang Y, Xu JC. Intravenous delivery of bone marrow-derived endothelial progenitor cells improves survival and attenuates lipopolysaccharide-induced lung injury in rats. Shock. 2010;34:196–204.CrossRef
6.
Zurück zum Zitat Patschan SA, Patschan D, Temme J, Korsten P, Wessels JT, Koziolek M, Henze E, Müller GA. Endothelial progenitor cells (EPC) in sepsis with acute renal dysfunction (ARD). Crit Care. 2011;15:R94.CrossRef Patschan SA, Patschan D, Temme J, Korsten P, Wessels JT, Koziolek M, Henze E, Müller GA. Endothelial progenitor cells (EPC) in sepsis with acute renal dysfunction (ARD). Crit Care. 2011;15:R94.CrossRef
7.
Zurück zum Zitat Pellegatta F, Bragheri M, Grigore L, Raselli S, Maggi FM, Brambilla C, Reduzzi A, Pirillo A, Norata GD, Catapano AL. In vitro isolation of circulating endothelial progenitor cells is related to the high density lipoprotein plasma levels. Int J Mol Med. 2006;17:203–8.PubMed Pellegatta F, Bragheri M, Grigore L, Raselli S, Maggi FM, Brambilla C, Reduzzi A, Pirillo A, Norata GD, Catapano AL. In vitro isolation of circulating endothelial progenitor cells is related to the high density lipoprotein plasma levels. Int J Mol Med. 2006;17:203–8.PubMed
8.
Zurück zum Zitat Zhang Z, Datta G, Zhang Y, Miller AP, Mochon P, Chen YF, Chatham J, Anantharamaiah GM, White CR. Apolipoprotein A-I mimetic peptide treatment inhibits inflammatory responses and improves survival inseptic rats. Am J Physiol Heart Circ Physiol. 2009;297:H866–73.CrossRef Zhang Z, Datta G, Zhang Y, Miller AP, Mochon P, Chen YF, Chatham J, Anantharamaiah GM, White CR. Apolipoprotein A-I mimetic peptide treatment inhibits inflammatory responses and improves survival inseptic rats. Am J Physiol Heart Circ Physiol. 2009;297:H866–73.CrossRef
9.
Zurück zum Zitat Navab M, Anantharamaiah GM, Reddy ST, Hama S, Hough G, Grijalva VR, Yu N, Ansell BJ, Datta G, Garber DW, Fogelman AM. Apolipoprotein A-I mimetic peptides. Arterioscler Thromb Vasc Biol. 2005;25:1325–31.CrossRef Navab M, Anantharamaiah GM, Reddy ST, Hama S, Hough G, Grijalva VR, Yu N, Ansell BJ, Datta G, Garber DW, Fogelman AM. Apolipoprotein A-I mimetic peptides. Arterioscler Thromb Vasc Biol. 2005;25:1325–31.CrossRef
10.
Zurück zum Zitat Kruger AL, Peterson S, Turkseven S, Kaminski PM, Zhang FF, Quan S, Wolin MS, Abraham NG. D-4F induces heme oxygenase-1 and extracellular superoxide dismutase, decreases endothelial cell sloughing, and improves vascular reactivity in rat model of diabetes. Circulation. 2005;111:3126–34.CrossRef Kruger AL, Peterson S, Turkseven S, Kaminski PM, Zhang FF, Quan S, Wolin MS, Abraham NG. D-4F induces heme oxygenase-1 and extracellular superoxide dismutase, decreases endothelial cell sloughing, and improves vascular reactivity in rat model of diabetes. Circulation. 2005;111:3126–34.CrossRef
11.
Zurück zum Zitat Yang N, Yao S, Wang M, Jiao P, Zhang Y, Qin S. Apolipoprotein A-I mimetic peptide reverse D-4F improves the biological functions of mouse bone marrow-derived late EPCs via PI3K/AKT/eNOS pathway. Mol Cell Biochem. 2013;377:229–36.CrossRef Yang N, Yao S, Wang M, Jiao P, Zhang Y, Qin S. Apolipoprotein A-I mimetic peptide reverse D-4F improves the biological functions of mouse bone marrow-derived late EPCs via PI3K/AKT/eNOS pathway. Mol Cell Biochem. 2013;377:229–36.CrossRef
12.
Zurück zum Zitat Gordon EM, Figueroa DM, Barochia AV, Yao X, Levine SJ. High-density lipoproteins and Ap-olipoprotein A-I: potential new players in the prevention and treatment of lung disease. Front Pharmacol. 2016;7:323.CrossRef Gordon EM, Figueroa DM, Barochia AV, Yao X, Levine SJ. High-density lipoproteins and Ap-olipoprotein A-I: potential new players in the prevention and treatment of lung disease. Front Pharmacol. 2016;7:323.CrossRef
13.
Zurück zum Zitat Liao CH, Lin FY, Wu YN, Chiang HS. Androgens inhibit tumor necrosis factor-α-induced cell adhesion and promote tube formation of human coronary artery endothelial cells. Steroids. 2012;77:756–64.CrossRef Liao CH, Lin FY, Wu YN, Chiang HS. Androgens inhibit tumor necrosis factor-α-induced cell adhesion and promote tube formation of human coronary artery endothelial cells. Steroids. 2012;77:756–64.CrossRef
14.
Zurück zum Zitat Arya D, Chang S, DiMuzio P, Carpenter J, Tulenko TN. Sphingosine-1-phosphate promotes the differentiation of adipose-derived stem cells into endothelial nitric oxide synthase (eNOS) expressing endothelial-like cells. J Biomed Sci. 2014;21:55.CrossRef Arya D, Chang S, DiMuzio P, Carpenter J, Tulenko TN. Sphingosine-1-phosphate promotes the differentiation of adipose-derived stem cells into endothelial nitric oxide synthase (eNOS) expressing endothelial-like cells. J Biomed Sci. 2014;21:55.CrossRef
15.
Zurück zum Zitat Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–7.CrossRef Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–7.CrossRef
16.
Zurück zum Zitat George AL, Bangalore-Prakash P, Rajoria S, Suriano R, Shanmugam A, Mittelman A, Tiwari RK. Endothelial progenitor cell biology in disease and tissue regeneration. J Hematol Oncol. 2011;4:24.CrossRef George AL, Bangalore-Prakash P, Rajoria S, Suriano R, Shanmugam A, Mittelman A, Tiwari RK. Endothelial progenitor cell biology in disease and tissue regeneration. J Hematol Oncol. 2011;4:24.CrossRef
17.
Zurück zum Zitat Fan H, Goodwin AJ, Chang E, Zingarelli B, Borg K, Guan S, Halushka PV, Cook JA. Endothelial progenitor cells and a stromal cell-derived factor-1α analogue synergistically improve survival in sepsis. Am J Respir Crit Care Med. 2014;189:1509–19.CrossRef Fan H, Goodwin AJ, Chang E, Zingarelli B, Borg K, Guan S, Halushka PV, Cook JA. Endothelial progenitor cells and a stromal cell-derived factor-1α analogue synergistically improve survival in sepsis. Am J Respir Crit Care Med. 2014;189:1509–19.CrossRef
18.
Zurück zum Zitat Dai L, Datta G, Zhang Z, Gupta H, Patel R, Honavar J, Modi S, Wyss JM, Palgunachari M, Anantharamaiah GM, White CR. The apolipoprotein A-I mimetic peptide 4F prevents defects in vascular function in endotoxemic rats. J Lipid Res. 2010;51:2695–705.CrossRef Dai L, Datta G, Zhang Z, Gupta H, Patel R, Honavar J, Modi S, Wyss JM, Palgunachari M, Anantharamaiah GM, White CR. The apolipoprotein A-I mimetic peptide 4F prevents defects in vascular function in endotoxemic rats. J Lipid Res. 2010;51:2695–705.CrossRef
19.
Zurück zum Zitat Zhang Z, Datta G, Zhang Y, Miller AP, Mochon P, Chen YF, Chatham J, Anantharamaiah GM, White CR. Apolipoprotein A-I mimetic peptide treatment inhibits inflammatory responses and improves survival in septic rats. Am J Physiol Heart Circ Physiol. 2009;297:H866–73.CrossRef Zhang Z, Datta G, Zhang Y, Miller AP, Mochon P, Chen YF, Chatham J, Anantharamaiah GM, White CR. Apolipoprotein A-I mimetic peptide treatment inhibits inflammatory responses and improves survival in septic rats. Am J Physiol Heart Circ Physiol. 2009;297:H866–73.CrossRef
20.
Zurück zum Zitat OF S, Xu X, Gaggar A, Grizzle WE, Mishra VK, Honavar J, Litovsky SH, Palgunachari MN, White CR, Anantharamaiah GM, Gupta H. Anti-inflammatory mechanisms of apolipoprotein A-I mimetic peptide in acute respiratory distress syndrome secondary to sepsis. PLoS One. 2013;8:e64486.CrossRef OF S, Xu X, Gaggar A, Grizzle WE, Mishra VK, Honavar J, Litovsky SH, Palgunachari MN, White CR, Anantharamaiah GM, Gupta H. Anti-inflammatory mechanisms of apolipoprotein A-I mimetic peptide in acute respiratory distress syndrome secondary to sepsis. PLoS One. 2013;8:e64486.CrossRef
21.
Zurück zum Zitat Spinelli FR, Metere A, Barbati C, Pierdominici M, Iannuccelli C, Lucchino B, Ciciarello F, Agati L, Valesini G, Di Franco M. Effect of therapeutic inhibition of TNF on circulating endothelial progenitor cells in patients with rheumatoid arthritis. Mediat Inflamm. 2013;2013:537539.CrossRef Spinelli FR, Metere A, Barbati C, Pierdominici M, Iannuccelli C, Lucchino B, Ciciarello F, Agati L, Valesini G, Di Franco M. Effect of therapeutic inhibition of TNF on circulating endothelial progenitor cells in patients with rheumatoid arthritis. Mediat Inflamm. 2013;2013:537539.CrossRef
22.
Zurück zum Zitat Pirro M, Bagaglia F, Paoletti L, Razzi R, Mannarino MR. Hypercholesterolemia-associated endothelial progenitor cell dysfunction. Ther Adv Cardiovasc Dis. 2008;2:329–39.CrossRef Pirro M, Bagaglia F, Paoletti L, Razzi R, Mannarino MR. Hypercholesterolemia-associated endothelial progenitor cell dysfunction. Ther Adv Cardiovasc Dis. 2008;2:329–39.CrossRef
23.
Zurück zum Zitat Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY, Huang YC, Liao WL, Huang HC, Liu PL, Chen YH. Endothelial progenitor cell dysfunction in cardiovascular diseases: role of reactive oxygen species and inflammation. Biomed Res Int. 2013;2013:845037.PubMed Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY, Huang YC, Liao WL, Huang HC, Liu PL, Chen YH. Endothelial progenitor cell dysfunction in cardiovascular diseases: role of reactive oxygen species and inflammation. Biomed Res Int. 2013;2013:845037.PubMed
24.
Zurück zum Zitat Westerweel PE, Visseren FL, Hajer GR, Olijhoek JK, Hoefer IE, de Bree P, Rafii S, Doevendans PA, Verhaar MC. Endothelial progenitor cell levels in obese men with the metabolic syndrome and the effect of simvastatin monotherapy vs. simvastatin/ezetimibe combination therapy. Eur Heart J. 2008;29:2808–17.CrossRef Westerweel PE, Visseren FL, Hajer GR, Olijhoek JK, Hoefer IE, de Bree P, Rafii S, Doevendans PA, Verhaar MC. Endothelial progenitor cell levels in obese men with the metabolic syndrome and the effect of simvastatin monotherapy vs. simvastatin/ezetimibe combination therapy. Eur Heart J. 2008;29:2808–17.CrossRef
25.
Zurück zum Zitat Hamed S, Brenner B, Aharon A, Daoud D, Roguin A. Nitric oxide and superoxide dismutase modulate endothelial progenitor cell function in type 2 diabetes mellitus. Cardiovasc Diabetol. 2009;8:56.CrossRef Hamed S, Brenner B, Aharon A, Daoud D, Roguin A. Nitric oxide and superoxide dismutase modulate endothelial progenitor cell function in type 2 diabetes mellitus. Cardiovasc Diabetol. 2009;8:56.CrossRef
26.
Zurück zum Zitat Zhang Q, Yin H, Liu P, Zhang H, She M. Essential role of HDL on endothelial progenitor cell proliferation with PI3K/Akt/cyclin D1 as the signal pathway. Exp Biol Med (Maywood). 2010;235:1082–92.CrossRef Zhang Q, Yin H, Liu P, Zhang H, She M. Essential role of HDL on endothelial progenitor cell proliferation with PI3K/Akt/cyclin D1 as the signal pathway. Exp Biol Med (Maywood). 2010;235:1082–92.CrossRef
27.
Zurück zum Zitat Peterson SJ, Husney D, Kruger AL, Olszanecki R, Ricci F, Rodella LF, Stacchiotti A, Rezzani R, McClung JA, Aronow WS, Ikehara S, Abraham NG. Long-term treatment with the apolipoprotein A1 mimetic peptide increases antioxidants and vascular repair in type I diabetic rats. J Pharmacol Exp Ther. 2007;322:514–20.CrossRef Peterson SJ, Husney D, Kruger AL, Olszanecki R, Ricci F, Rodella LF, Stacchiotti A, Rezzani R, McClung JA, Aronow WS, Ikehara S, Abraham NG. Long-term treatment with the apolipoprotein A1 mimetic peptide increases antioxidants and vascular repair in type I diabetic rats. J Pharmacol Exp Ther. 2007;322:514–20.CrossRef
28.
Zurück zum Zitat Zhang Z, Qun J, Cao C, Wang J, Li W, Wu Y, Du L, Zhao P, Gong K. Apolipoprotein A-I mimetic peptide D-4F promotes human endothelial progenitor cell proliferation,migration, adhesion though eNOS/NO pathway. Mol Biol Rep. 2012;39:4445–54.CrossRef Zhang Z, Qun J, Cao C, Wang J, Li W, Wu Y, Du L, Zhao P, Gong K. Apolipoprotein A-I mimetic peptide D-4F promotes human endothelial progenitor cell proliferation,migration, adhesion though eNOS/NO pathway. Mol Biol Rep. 2012;39:4445–54.CrossRef
29.
Zurück zum Zitat Patruno A, Franceschelli S, Pesce M, Maccallini C, Fantacuzzi M, Speranza L, Ferrone A, De Lutiis MA, Ricciotti E, Amoroso R, Felaco M. Novel aminobenzyl-acetamidine derivative modulate the differential regulation of NOSs in LPS induced inflammatory response: role of PI3K/Akt pathway. Biochim Biophys Acta. 2012;1820:2095–104.CrossRef Patruno A, Franceschelli S, Pesce M, Maccallini C, Fantacuzzi M, Speranza L, Ferrone A, De Lutiis MA, Ricciotti E, Amoroso R, Felaco M. Novel aminobenzyl-acetamidine derivative modulate the differential regulation of NOSs in LPS induced inflammatory response: role of PI3K/Akt pathway. Biochim Biophys Acta. 2012;1820:2095–104.CrossRef
30.
Zurück zum Zitat Everaert BR, Van Craenenbroeck EM, Hoymans VY, Haine SE, Van Nassauw L, Conraads VM, Timmermans JP, Vrints CJ. Current perspective of pathophysiological and interventional effects on endothelial progenitor cell biology: focus on PI3K/AKT/eNOS pathway. Int J Cardiol. 2010;144:350–66.CrossRef Everaert BR, Van Craenenbroeck EM, Hoymans VY, Haine SE, Van Nassauw L, Conraads VM, Timmermans JP, Vrints CJ. Current perspective of pathophysiological and interventional effects on endothelial progenitor cell biology: focus on PI3K/AKT/eNOS pathway. Int J Cardiol. 2010;144:350–66.CrossRef
Metadaten
Titel
Reverse-D-4F improves endothelial progenitor cell function and attenuates LPS-induced acute lung injury
verfasst von
Nana Yang
Hua Tian
Enxin Zhan
Lei Zhai
Peng Jiao
Shutong Yao
Guohua Lu
Qingjie Mu
Juan Wang
Aihua Zhao
Yadong Zhou
Shucun Qin
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2019
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-019-1099-6

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

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

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