Open Access

Anti-Semaphorin-7A single chain antibody demonstrates beneficial effects on pulmonary inflammation during acute lung injury

Retraction in: /10.3892/etm.2023.12165

  • Authors:
    • Xiao Chen
    • Hailing Wang
    • Kui Jia
    • Hao Wang
    • Tao Ren
  • View Affiliations

  • Published online on: January 8, 2018     https://doi.org/10.3892/etm.2018.5724
  • Pages: 2356-2364
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pulmonary inflammation is a primary characteristic of lung injury initiated by the accession of immune cells into the alveolar space. Neutrophil migration serves an important role in pulmonary inflammation mediated by the migration of neutrophils into hypoxic tissue sites. The elimination of pulmonary inflammation is directly associated with rehabilitation in patients with lung injury. Anti‑inflammatory treatment is essential following lung injury and ultimately determines patient outcomes. Semaphorin‑7A (SEMA‑7A) is a member of the Semaphorin family that influences the migration of neutrophils into hypoxic tissue sites, thus promoting inflammation. However, understanding of the role of SEMA‑7A serves during lung injury is limited and the immunological function of SEMA‑7A during the migration of neutrophils into acute injury sites remains unknown. The present study investigated SEMA‑7A expression and constructed a single chain antibody for SEMA‑7A (Anti‑SEMA‑7A) to study its therapeutic efficacy against pulmonary inflammation in a mouse model of acute injury sites. The data indicated that the expression of SEMA‑7A was upregulated due to induction by pro‑inflammatory cytokines and demonstrated that Anti‑SEMA‑7A inhibited SEMA‑7A expression in vitro and in vivo. The current study also indicated that the production of pro‑inflammatory cytokines induced by SEMA‑7A in endothelial and epithelial cells enhanced pulmonary inflammation. Anti‑SEMA‑7A suppressed the transendothelial migration of neutrophils mediated by SEMA‑7A. Anti‑SEMA‑7A treatment neutralized SEMA‑7A expression and reduced signs of pulmonary inflammation, leading to the elimination of pulmonary inflammation in rat with acute lung injury. The current study identified Anti‑SEMA‑7A as a potential agent to interfere with the inflammatory pathway during acute lung injury, which may be the basis for anti-inflammatory strategies to treat lung injuries in the future.

Introduction

Acute lung injury is caused by a variety of direct and indirect injuries of alveolar epithelial and capillary endothelial cells that lead to diffuse pulmonary interstitial, alveolar edema and acute hypoxic respiratory insufficiency (1,2). Acute lung injury is the most common respiratory disease in clinical settings (3). Acute lung injury can be induced in a number of ways, including via stress, trauma, sepsis, seawater aspiration and burns. It frequently leads to respiration distress syndrome, and may result in the development of life-threatening conditions (4). The most severe form of acute lung injury is respiration distress syndrome, which is a leading cause of morbidity and mortality in critical patients with acute lung injury (5,6). The most commonly used treatments for acute lung injury are major surgery and prolonged mechanical ventilation, in addition to the use of anti-inflammatory treatments when inflammation develops in response to pneumonia (7,8). It has been demonstrated that acute lung injury is associated with refractory lung disease and the mortality rates of patients with advanced infectious lung injuries are high (9).

The mechanism of acute lung injury is well known and research indicates that lung injury is aggravated by the migration of neutrophils from the vascular compartment into the alveolar space (10). Pneumonia often occurs in conjunction with acute lung injury as the process of self-propagating inflammation and develops within the alveolar space (11). Pulmonary inflammation is associated with deterioration during acute lung injury. In addition, previous studies have demonstrated that the severity of associated pulmonary inflammation is associated with the extent of alveolar inflammation that occurs, which is a significant factor influencing the outcome of patients with acute lung injury (10,12). Furthermore, it has been indicated that neutrophil infiltration promotes the development of inflammation in the alveolar space, which is regulated by the chemokine system (13). It has also been suggested that the efficacy of the neuronal guidance protein signaling pathway is due to the control and orchestration of acute inflammation by neutrophil migration (11).

Semaphorin7A (SEMA-7A) is a multifunctional membrane-anchored protein, which serves a role in regulating pulmonary fibrosis. SEMA-7A participates in immune and inflammatory responses during acute lung injury (14). It is also a member of the semaphorin family that induces the expression of cytokines in macrophages and monocytes via a glycophosphatidylinositol linkage and serves a significant role during the effector phase of the inflammatory immune response (15). A previous study indicated that SEMA-7A aggravated pulmonary inflammation by inducing pro-inflammatory cytokine production in epithelial and endothelial cells, thus enhancing the condition of pulmonary inflammation and production of cytokines during lung injury (16). However, understanding of the role SEMA-7A serves during lung injury is limited and not well understood regarding the immunological function of SEMA-7A in inducing the migration of neutrophils into acute injury sites.

The present study investigated SEMA-7A expression and constructed an antibody for SEMA-7A (Anti-SEMA-7A). The therapeutic efficacy of pulmonary inflammation during a mouse model of acute injury sites was also assessed. In addition, the production of pro-inflammatory cytokines induced by SEMA-7A in endothelial and epithelial cells was analyzed in vivo. Data indicated that Anti-SEMA-7A inhibited the transendothelial migration of neutrophils mediated by SEMA-7A inactivation. Furthermore, Anti-SEMA-7A significantly alleviated the degree of pulmonary inflammation, leading to the elimination of pulmonary inflammation in rat with acute lung injury.

Materials and methods

Cell culture and reagents

Human endothelial HMEC-1 cells (Cell Center of Nanyang Medicine College, Nanyang, China) were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.). Alveolar cells were isolated from experimental rats and cultured in DMEM medium supplemented with 10% FBS, 100 U/ml penicillin and 100 µg/ml streptomycin. All cells were maintained at 37°C, 5% CO2 and saturated humidity.

Cells treatments

Human endothelial HMEC-1 cells (1×105) were cultured in six-well plates for 12 h at 37°C. Cell were then incubated with LPS (5 mg/ml, Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) or PBS for 4 h at 37°C.

Enzyme-linked immunosorbent assay (ELISA)

SEMA-7A concentration levels were analyzed by an ELISA kit according to the manufacturer's protocol (cat no. SEB448Mu; Wuhan Uscn Business Co., Ltd., Wuhan, China). Cleaved Sema-7A from lysed lung cells membranes from rats was evaluated. The results were recorded at 450 nm using an ELISA microplate reader.

Construction expression and purification of Anti-SEMA-7A

Anti-SEMA-7A was screened using a filtering method described in previous studies (17,18). The Fc fragment and Anti-SEMA-7A are linked by a 32-amino-acid interlinker. Anti-SEMA-7A was expressed in E. coli Rossetta by transforming a recombinant pET27b-Anti-SEMA-7A plasmid (cat no. K30001, Invitrogen; Thermo Fisher Scientific, Inc.). E. coli Rossetta cells (Sigma-Aldrich; Merck KGaA) were cultured in LB medium (Sigma-Aldrich; Merck KGaA), harvested and subsequently disrupted and dissolved in phosphate-buffered saline (PBS) as described previously (19). The supernatant was subjected to affinity chromatography (20) (Columns: APPSEV; GE AKTA Pure System; GE Healthcare Life Sciences, Little Chalfont, UK) to purify the target protein. The purified protein was collected and gel filtration chromatography (cat no. 17014901; GE Healthcare Life Sciences) was used to further purify Anti-SEMA-7A, as described previously (21).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA was extracted from HMEC-1 and lung cells from experimental animals using an RNA Extract kit (cat no. 339390, Qiagen Sciences, Inc., Gaithersburg, MD, USA) according to the manufacturer's instructions. cDNA was synthesized using a RevertAid First Strand cDNA Synthesis kit (MBI Fermentas; Thermo Fisher Scientific, Inc., Pittsburgh, PA, USA). qPCR was performed using a 50 µl reaction mixture. All forward and reverse primers (Table I) were synthesized by Invitrogen (Thermo Fisher Scientific, Inc.). Expression of β-actin was used as an endogenous control (Invitrogen; Thermo Fisher Scientific, Inc.). Interleukin (IL)-1β, IL-17A, tumor necrosis factor-α (TNF-α), C-X-C motif chemokine receptor 2 (CXCR2), macrophage inflammatory protein 2 (MIP-2) and keratinocyte chemoattractant (KC) expression was analyzed with the SYBR-Green Master mix (Bio-Rad Laboratories, Inc., Hercules, CA, USA), according to the manufacturer's protocol. The reaction conditions were performed as follows: 40 cycles of 95°C for 10 min and 35 cycles of 95°C for 20 sec and 56.5°C for 1 min. Each experiment was repeated three times. Relative mRNA expression changes were calculated using the 2−ΔΔCq method and the results are expressed as the n-fold change compared with the control (22).

Table I.

Sequences of primers were used in this study.

Table I.

Sequences of primers were used in this study.

Primer sequences

GeneReverseForward
TNFα 5′-TCCAGACTTCCTTGAGACA-3′ 5′-GGCGATTACAGACACAACT-3′
IL-1β 5′-GGCTGCTTCCAAACCTTTGA-3′ 5′-GAAGACACGGATTCCATGGT-3′
IL-17A 5′-ATGCACAGCCACCGCGACTT-3′ 5′-CTTCATGACTGCCTCCAAGTAG-3′
SEMA-7A 5′-CTCCGCCCAGGGCCACCTAA-3′ 5′-ACATGGCCTTTCCAGACGGCG-3′
SEMA-7AR 5′-AGACCATACCTGCCGAATGTAG'-3′ 5′-GAGAGCTTCCTGTCCTGTAGAG-3′
CXCR2 5′-AACATGGAGAGTGACAGCTTTG-3′ 5′-TCACATGGGGCGGCATCC-3′
MIP-2 5′-AAGTTTGCCTTGACCCTGAA-3′ 5′-AGGCACATCAGGTACGATCC-3′
KC 5′-AGAACATCCAGAGCTTGAAGGTGTT-3′ 5′-GGACACCTTTTAGCATCTTTTGGACA-3′
β-actin 5′-CGGAGTCAACGGATTTGGTC-3′ 5′-AGCCTTCTCCATGGTCGTGA-3′

[i] SEMA-7A, Semaphorin-7A; SEMA-7AR, SEMA-7A target receptor IL, interleukin; TNF-α, tumor necrosis factor-α.

Western blot analysis

HMEC-1 cells and lung endothelial cells were lysed with radioimmunoprecipitation assay buffer containing 0.5% Triton X-100 and protease-inhibitor (cat no. FNN0071; Thermo Fisher Scientific). Protein concentration was measured using a BCA protein assay kit. Proteins (20 µg/lane) were analyzed using 12% SDS-PAGE assays followed by transfer onto polyvinylfluoride (PVDF) membranes. Protein was blocked with 5% skimmed milk for 1 h at 37°C. IL-1β (1:1,000; cat no. ab200478), IL-17A (1:1,000; cat no. ab79056), TNF-α (1:1,000; cat no. ab6671), CXCR2 (1:1,000; cat no. ab14935), MIP-2 (1:1,000; cat no. ab9950), KC (1:1,000; cat no. ab798362), SEMA-7A (1:1,000; cat no. ab23578), ETS domain-containing transcription factor (ERF; 1:1,000; cat no. ab199672), extracellular signal-regulated kinase (ERK; 1:1,000; cat no. ab54230), ERF mutant1-7 (M1-7; 1:1,000; cat no. ab61108) and fold superfamily/FRT-Kan-FRT (FSF/FKF; 1:1,000; cat no. ab200478, all Abcam, Cambridge, UK) were incubated with PVDF membranes for 12 h at 4°C. Subsequently, HRP-conjugated goat anti-rabbit IgG mAb (cat no. PV-6001; ZSGB-BIO, Beijing, China) was added for 24 h at 4°C. A Ventana Benchmark automated staining system was used for analyzing protein expression (Olympus BX51; Olympus, Tokyo, Japan). The density of the bands was analyzed by Quantity one software version 4.62 (Bio-Rad, Laboratories). All experiments were performed at least three times.

Immunohistochemistry

HMEC-1 cells and lung endothelial tissue from experimental rats were fixed using 10% formaldehyde for 2 h at 37°C and then embedded in paraffin. For HMEC-1 cells, rat anti-human SEMA-7A antibody (1:1,000, cat no. ab23578; Abcam) labeled with fluorescein isothiocyanate (green fluorescence) for 12 h at 4°C was used to analyze SEMA-7A expression prior to and following treatment with Anti-SEMA-7A (2 mg/ml), SEMA-7A (2 mg/ml) or PBS (2 mg/ml) for 2 h at 4°C. Lung tissues were cut into sections 4-µm thick. Antigen retrieval was performed in tissue sections and the sections were incubated with the following primary antibodies: SEMA-7A (1: 1,000; cat no. ab23578), M1-7 (1:1,000; cat no. ab61108) and FSF/FKF (1:1,000; cat no. ab200478; all Abcam) for 8 h at 4°C and correlative secondary antibodies: HRP-conjugated goat anti-rabbit IgG mAb (1:2,000; cat no. PV-6001; ZSGB-BIO, Beijing, China) were applied for specimens for 24 h at 4°C. The staining of the slides was performed with the avidin-biotin-peroxidase complex. A Ventana Benchmark automated staining system (Version 3.0, Ventana Medical Systems, Inc; Roche Diagnostics, Basel, Switzerland) was used to analyze SEMA-7A, ERF, ERK, M1-7 and FSF/FKF levels using confocal microscopy (Carl Zeiss LSM780, Carl Zeiss AG, Oberkochen, Germany).

Immunofluorescence

Paraffin-embedded tissue sections (4-µm thick) were fixed with 4% paraformaldehyde on slides for 2 h at 37°C. The sections were washed with PBS three times and then stained with the appropriate antibody: SEMA-7A (1:1,000; cat no. ab23578; Abcam) for 12 h at 4°C. Samples were incubated at 4°C for 12 h, washed with PBS, and then incubated with a specific fluorescence-conjugated IgG (1:1,000; cat no. O-6382, Invitrogen; Thermo Fisher Scientific, Inc.) for 1 h in a light-protected chamber at 37°C. Subsequently, sections were counterstained with Von Willebrand factor (2 µg/ml, Sigma-Aldrich; Merck KGaA) or DAPI (2 µg/ml, Sigma-Aldrich; Merck KGaA) at 37°C for 2 h and mounted. In addition, tissue sections (4-µm thick) were incubated with IL-1β (1:1,000; cat no. ab200478), IL-17A (1:1,000; cat no. ab79056), TNF-α (1:1,000; cat no. ab6671), CXCR2 (1:1,000; cat no. ab14935), MIP-2 (1:1,000; cat no. ab9950), KC (1:1,000; cat no. ab798362; all Abcam) at 4°C for 12 h. Immunofluorescence signals were detected using a laser scanning confocal microscope (Carl Zeiss AG, Oberkochen, Germany).

Concentration of serum albumin

Venous blood samples were collected from the antecubital vein. Serum was obtained by centrifuging at 8,000 × g for 10 min at 4°C. Serum albumin levels were measured using the bromocresol green dye-binding method on a Roche Modular DP analyzer (Roche Diagnostics, Basel, Switzerland).

Animal study

A total of 60 Specific pathogen-free male SD rats (eight-week old, weighing 280–320 g) were purchased from the Shanghai Laboratory Animal Center (Shanghai, China). All animals were housed in a temperature-controlled facility at 23±1°C and relative humidity of 50±5% with a 12-h light/dark cycle. The current study was approved by the Ethics Committee of The First Affiliated Hospital, Nanyang Medicine College. All rats were then returned to their cages and given food and water. Animals were anesthetized with sodium pentobarbital (40 mg/kg, Sigma-Aldrich; Merck KGaA) by intravenous injection for further analysis. Following anesthesia, the limbs of rats were affixed to a board and necks were shaved, followed by a 1-cm incision. The trachea was punctured with a 25-Ga needle and LPS (500 µg/kg; Sigma-Aldrich; Merck KGaA) was injected into the trachea. Treatments were initiated on day 3 after LPS injection. Rats were divided into two groups (n=20, each) and received treatment of Anti-SEMA-7A (10 mg/ml; Sigma-Aldrich) or the same volume of PBS. The treatments were performed once a day for a total of 10 days. The therapeutic outcome of acute lung injury was evaluated by inflammatory factors Il-1β, IL-17A and TNF-α according to a previous study (23). In addition, Anti-SEMA-7A (10 mg/ml, Sigma-Aldrich) or PBS (10 mg/ml, Sigma-Aldrich) were transported to lesions using a hybrid nanometer carrier (liposome), following a previously described protocol (24). Animals were sacrificed on day 15 for further analysis.

Statistical analysis

Statistical analysis was completed using SPSS 19.0 statistical software (IBM SPSS, Armonk, NY, USA) with the assistance of Microsoft Excel (Windows 2010, Microsoft Corporation, Redmond, WA, USA). Data are presented as the mean ± standard deviation of triplicate experiments. Differences among multiple groups were determined using the Student's t-test or one-way analysis of variance followed by a Tukey HSD test and P<0.05 was considered to indicate a statistically significant difference.

Results

Analysis of Semaphorin-7A expression during acute lung injury

On the basis of a previous study (25) demonstrating that SEMA7A regulates neutrophil trafficking during acute pulmonary inflammation, the present study assessed SEMA-7A expression in rats with lung injury induced by LPS (500 µg/kg) in vivo. Levels of SEMA-7A mRNA and protein expression were significantly higher in the pulmonary tissue of rats with lung injury (P<0.05; Fig. 1A and B). In addition, the results in Fig. 1C indicate an increase in SEMA-7A expression in pulmonary tissue in the LPS group compared with the control. Furthermore, SEMA-7A target receptor expression was assessed in pulmonary tissue in rats with lung injury by RT-qPCR (Fig. 1D). Analysis indicated that the expression of SEMA-7A target receptor (SEMA-7AR) was significantly increased in acute pulmonary inflammation in vivo in the LPS group compared with the control group (P<0.01). These findings suggest that SEMA-7A expression is upregulated during acute pulmonary inflammation.

Construction and characteristics of Anti-SEMA-7A in vitro

Based on the aforementioned report (26), the present study hypothesized that SEMA-7A may serve as a potential therapeutic target molecular for acute lung injury therapy in LPS-induced acute lung injury rats. Therefore, antibody therapy target for SEMA-7A may provide potential novel target therapy for acute lung injury. In the current study, a recombinant single chain antibody liked with Fc was constructed and presented a high affinity for SEMA-7A. Fig. 2A presents the structure of Anti-SEMA-7A, which contains a cell-penetrating peptide (CCP) and Fc fragment to enhance transmembrane ability and stability. Anti-SEMA-7A was expressed by E. coli and the yield was ~42 mg/l. The molecular weight of Anti-SEMA-7A-CPP was ~38 kDa, as determined by SDS-PAGE and ~48 kDa under constant denatured gel electrophoresis (Fig. 2B). In addition, ELISA and western blot analysis assays indicated that Anti-SEMA-7A specifically bound to SEMA-7A (Fig. 2C and D). The data indicated that Anti-SEMA-7A was purified successfully and possessed the potential to integrate into the corresponding molecules.

Inhibition of pro-inflammatory cytokines through Anti-SEMA-7A in vitro

To confirm the efficacy of Anti-SEMA-7A against acute lung injury, the present study investigated whether administration with Anti-SEMA-7A decreases SEMA-7A expression in vitro. Endothelial HMEC-1 cells were exposed to LPS for 24 h and then treated with Anti-SEMA-7A or SEMA-7A. Following this, levels of the pro-inflammatory cytokines IL-1β, IL-17A and TNF-α were analyzed. IL-1β, IL-17A and TNF-α expression significantly decreased following Anti-SEMA-7A treatment (P<0.01; Fig. 3A-C). The expression of SEMA-7A was also significantly downregulated in HMEC-1 cells exposed to Anti-SEMA-7A, as determined by RT-qPCR (P<0.01; Fig. 3D). In addition, a markedly increased inhibition of SEMA-7A expression compared with non-stimulated HMEC-1 cells was identified (Fig. 3E). Furthermore, Anti-SEMA-7A exhibited a markedly suppressive effect on SEMA-7AR expression (Fig. 3F). These findings reveal that Anti-SEMA-7A neutralized SEMA-7A activity and downregulated IL-1β, IL-17A TNF-α and SEMA-7AR expression in HMEC-1 cells exposed to LPS, indicating that anti-SEMA-7A may neutralize SEMA-7A in acute lung injury.

Anti-SEMA-7A suppresses the transmigration of neutrophils in endothelial and epithelial cells

On the basis of the in vitro efficacy of Anti-SEMA-7A, the present study further assessed the role of Anti-SEMA-7A in the LPS-induced acute lung injury rat model. Rats with acute lung injury received Anti-SEMA-7A treatment a total of 7 times. The concentration level of serum albumin was used to determine the therapeutic effects of Anti-SEMA-7A. The results of the present study demonstrated that the number of neutrophils was markedly reduced, along with the expression of Claudin within the pulmonary tissue of Anti-SEMA-7A-treated rats compared with controls. The results in Fig. 4A indicate that the pathogenic condition of rats with acute lung injury was relieved following treatment with Anti-SEMA-7A. Lung sections were prepared for analysis of SEMA-7A and inflammatory factor expression. It was demonstrated that SEMA-7A expression decreased following Anti-SEMA-7A treatment in lung, which did not occur in the lung tissue of rats treated with PBS (Fig. 4B). Results demonstrated that levels of IL-1B, IL-17A and TNF-a are downregulated following Anti-SEMA-7A treatment, compared with controls (Fig. 4C). Furthermore, expression of the chemokines of CXCR2, MIP-2 and KC decreased within the alveolar space of Anti-SEMA-7A-treated rat (Fig. 4D). These findings suggest that Anti-SEMA-7A exhibits beneficial effects on rats with acute lung injury by neutralizing SEMA-7A expression.

Anti-SEMA-7A suppresses ERF activity for EMT signaling pathway

To investigate the potential mechanism of Anti-SEMA-7A in acute lung injury, the activity of ERF as a Ras/ERK mediator in EMT progression was investigated in alveolar cells. Alveolar epithelial cells were isolated from experimental rat that had undergone treatment with Anti-SEMA-7A or PBS. Levels of ERF, ERK, M1-7 and FSF/FKF expression were analyzed in alveolar cells by immunofluorescence. The results indicate that levels of ERF, ERK, M1-7 and FSF/FKF expression were downregulated following treatment with Anti-SEMA-7A (Fig. 5). These findings indicate that Anti-SEMA-7A obstructed the ERF-induced EMT process through the Ras/ERK pathway in alveolar cells.

Discussion

Acute lung injury is a serious acute respiratory disease that presents as the most common type of acute respiratory failure with high mortality and morbidity in critically ill patients (27). Acute lung injury frequently leads directly or indirectly to alveolar epithelial cell injury and capillary endothelial cell damage, which subsequently results in diffuse pulmonary interstitial, alveolar edema and acute hypoxic respiratory insufficiency (28). Pathological physiological characteristics of acute lung injury are evident decreases in the vital capacity, lung compliance and ventilation/blood flow ratio (29). The clinical characteristics of acute lung injury are progressive hypoxemia and respiratory distress, diagnosed by imaging to identify homogenous exudative lesions (30). Acute lung injury may develop into acute respiratory distress syndrome at severe stages (oxygenation index <200), which may become a clinical problem for patient treatments (31). In the present study, a rat model of acute lung injury induced by LPS was established by inhalation to investigate the primary mechanism and progression of this disease.

Pneumonia is the single most common feature for patients with acute lung injury (32). Inflammatory factors contribute to pneumonia and aggravate the condition of patients with acute lung injury (33). Despite pneumonia being identified as the most common risk factor for patients with acute lung injury, there is limited understanding of the molecular mechanism of inflammatory factors in patients with acute lung injury and pneumonia (34). Therefore, investigating the role of inflammatory factors in the progress of pneumonia within the alveolar space in patients with acute lung injury is essential.

SEMA-7A is part of the Semaphorins family of soluble and membrane-anchor proteins originally considered to be involved in the development of axonal signal factors in the nervous system. Previous studies have indicated that SEMA-7A influences polymorphonuclear leukocyte migration during hypoxia and demonstrated that SEMA-7A regulated pulmonary fibrosis, which participates in immune and inflammatory responses during acute lung injury (14,35). Furthermore, it has been reported that overexpression of SEMA-7A promotes the migration of transendothelial neutrophils into the alveoli, resulting in inflammation and stimulating changes in the morphology of lung cells in patients with acute lung injury (14). The most serious threat for patients with acute lung injury is that pneumonia may lead to lung failure. In addition, a previous study has suggested that SEMA-7A may initiate inflammation during acute lung injury induced by different factors, including hypoxia-inducible factor and β1 integrin (36). The etiology of lung injury is frequently associated with the process of inflammation within the lung, which leads to fatal infections deep in the lungs as a sequela of the primary process (37). SEMA-7A expression is associated with the inflammatory process within the lung, indicating that SEMA-7A may be a potential molecular target (38).

In the current study, a chimeric antibody containing cell-penetrating peptide and Fc target was produced for SEMA-7A and the function of Anti-SEMA-7A in an acute lung injury rat model was investigated. The present study indicated that SEMA-7A expression was dysfunctional in the acute lung injury model in vitro and in vivo, consistent with results of a previous study (39). In addition, the current study indicates that levels of IL-1β, IL-17A and TNF-α expression were upregulated in rats with LPS-induced acute lung injury. By contrast, levels of the chemokines CXCR2, MIP-2 and KC were decreased within the alveolar space of Anti-SEMA-7A-treated rat. These inflammatory factors and chemokines may stimulate neutrophil migration into the alveolar space and Anti-SEMA-7A decreased their levels of expression levels, thus inducing a remission effect in acute lung injury (40).

In addition, the data demonstrated SEMA-7A not only induced inflammation but also promoted the neutrophils migration into the alveolar space in vivo. This finding further explains the interaction between SEMA-7A expression and the upregulation of inflammatory factors, which is in accordance with a previous study (41). Despite SEMA-7A inducing the production of cytokines and chemokines in the injury sites of the lung, SEMA-7A also regulated the ERF-induced inhibition of EMT in Ras-dependent mouse mammary epithelial cells (42). Therefore, the current study suggests that Anti-SEMA-7A inhibits SEMA-7A expression and led to ERF-induced inhibition of EMT in Ras-dependent epithelial cells in vitro and in vivo (43). The hypothesis of the present study was confirmed by the data and results, as it suggested that SEMA-7A promoted EMT progress, leading to neutrophil migration into the alveolar space in vivo. These results may provide a novel target signaling pathway for inhibiting neutrophil migration in patients with acute lung injury.

In conclusion, the current study aimed to determine the therapeutic effects of Anti-SEMA-7A in acute lung injury rats. To the best of our knowledge, this is the first study to use Anti-SEMA-7A targeting of SEMA-7A to analyze the efficacy for rats in a model of lung injury. The results of the current study emphasise the importance of SEMA-7A for the control of the acute inflammatory response and treatment of acute lung injury. The interference of Anti-SEMA-7A with ERF-induced inhibition of EMT in Ras-dependent pathway in epithelial cells suggests its potential association with acute inflammation in lungs, suggesting that it may be developed for clinical applications in the future.

References

1 

Iwata K, Doi A, Ohji G, Oka H, Oba Y, Takimoto K, Igarashi W, Gremillion DH and Shimada T: Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): A systematic review and meta-analysis. Intern Med. 49:2423–2432. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Schmickl CN, Mastrobuoni S, Filippidis FT, Shah S, Radic J, Murad MH, Toy P and Gajic O: Male-predominant plasma transfusion strategy for preventing transfusion-related acute lung injury: A systematic review. Crit Care Med. 43:205–225. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Kim J, Song J and Lee M: Combinational delivery of HMGB1 A box and heparin for acute lung injury. J Control Release. 213:e572015. View Article : Google Scholar : PubMed/NCBI

4 

Butt Y, Kurdowska A and Allen TC: Acute lung injury: A clinical and molecular review. Arch Pathol Lab Med. 140:345–350. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Hirano Y, Aziz M, Yang WL, Ochani M and Wang P: Neutralization of osteopontin ameliorates acute lung injury induced by intestinal ischemia-reperfusion. Shock. 46:431–438. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Braun RK, Koch JM, Hacker TA, Pegelow D, Kim J, Raval AN, Schmuck EG, Schwahn DJ, Hei DJ, Centanni JM, et al: Cardiopulmonary and histological characterization of an acute rat lung injury model demonstrating safety of mesenchymal stromal cell infusion. Cytotherapy. 18:536–545. 2016. View Article : Google Scholar : PubMed/NCBI

7 

McIntyre LA, Moher D, Fergusson DA, Sullivan KJ, Mei SH, Lalu M, Marshall J, Mcleod M, Griffin G, Grimshaw J, et al: Efficacy of mesenchymal stromal cell therapy for acute lung injury in preclinical animal models: A Systematic review. PLoS One. 11:e01471702016. View Article : Google Scholar : PubMed/NCBI

8 

Eves ND, Song Y, Piper A and Maher TM: Year in review 2012: Acute lung injury, interstitial lung diseases, sleep and physiology. Respirology. 18:555–564. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Cermáková Z, Simetka O and Kořístka M: Transfusion-related acute lung injury (TRALI)-review. Ceska Gynekol. 78:211–215. 2013.(In Czech). PubMed/NCBI

10 

Dzierba AL, Abel EE, Buckley MS and Lat I: A review of inhaled nitric oxide and aerosolized epoprostenol in acute lung injury or acute respiratory distress syndrome. Pharmacotherapy. 34:279–290. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Vlaar AP and Juffermans NP: Transfusion-related acute lung injury: A clinical review. Lancet. 382:984–994. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Sud S, Sud M, Friedrich JO, Meade MO, Ferguson ND, Wunsch H and Adhikari NK: High frequency oscillation in patients with acute lung injury and acute respiratory distress syndrome (ARDS): Systematic review and meta-analysis. BMJ. 340:c23272010. View Article : Google Scholar : PubMed/NCBI

13 

Jaswal DS, Leung JM, Sun J, Cui X, Li Y, Kern S, Welsh J, Natanson C and Eichacker PQ: Tidal volume and plateau pressure use for acute lung injury from 2000 to present: A systematic literature review. Crit Care Med. 42:2278–2289. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Zhang M, Wang L, Dong M, Li Z and Jin F: Endothelial Semaphorin 7A promotes inflammation in seawater aspiration-induced acute lung injury. Int J Mol Sci. 15:19650–19661. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Holmes S, Downs AM, Fosberry A, Hayes PD, Michalovich D, Murdoch P, Moores K, Fox J, Deen K, Pettman G, et al: Sema7A is a potent monocyte stimulator. Scand J Immunol. 56:270–275. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Roth JM, Köhler D, Schneider M, Granja TF and Rosenberger P: Semaphorin 7A aggravates pulmonary inflammation during lung injury. PLoS One. 11:e01469302016. View Article : Google Scholar : PubMed/NCBI

17 

Wu H, Yao L, Chou L, Yang JH, Zhang YX, Li XL and Shan BE: Construction and functional analysis of an anti-human cervical carcinoma/anti-human CD3 single-chain bispecific antibody. Mol Med Rep. 14:804–810. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Singh PK, Agrawal R, Kamboj DV and Singh L: Construction of recombinant single chain variable fragment (ScFv) antibody against superantigen for immunodetection using antibody phage display technology. Methods Mol Biol. 1396:207–225. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Pasello M, Zamboni S, Mallano A, Flego M, Picci P, Cianfriglia M and Scotlandi K: Design and construction of a new human naive single-chain fragment variable antibody library, IORISS1. J Biotechnol. 224:1–11. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Bjarnadottir SG and Flengsrud R: Affinity chromatography, two-dimensional electrophoresis, adapted immunodepletion and mass spectrometry used for detection of porcine and piscine heparin-binding human plasma proteins. J Chromatogr B Analyt Technol Biomed Life Sci. 944:107–113. 2014. View Article : Google Scholar : PubMed/NCBI

21 

London AS, Mackay K, Lihon M, He Y and Alabi BR: Gel filtration chromatography as a method for removing bacterial endotoxin from antibody preparations. Biotechnol Prog. 30:1497–1501. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Rauch S, Johannes A, Zollhofer B and Muellenbach RM: Evaluating intra-abdominal pressures in a porcine model of acute lung injury by using a wireless motility capsule. Med Sci Monit. 18:BR163–BR166. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Yang L, Wu X, Liu F, Duan Y and Li S: Novel biodegradable polylactide/poly(ethylene glycol) mice lles prepared by direct dissolution method for controlled delivery of anticancer drugs. Pharm Res. 26:2332–2342. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Gutierrez-Franco A, Costa C, Eixarch H, Castillo M, Medina-Rodríguez EM, Bribián A, de Castro F, Montalban X and Espejo C: Differential expression of sema3A and sema7A in a murine model of multiple sclerosis: Implications for a therapeutic design. Clin Immunol. 163:22–33. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Allegra M, Zaragkoulias A, Vorgia E, Ioannou M, Litos G, Beug H and Mavrothalassitis G: Semaphorin-7a reverses the ERF-induced inhibition of EMT in Ras-dependent mouse mammary epithelial cells. Mol Biol Cell. 23:3873–3881. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Jeon CM, Shin IS, Shin NR, Hong JM, Kwon OK, Kim JH, Oh SR, Bach TT, Hai DV, Quang BH, et al: Clausena anisata-mediated protection against lipopolysaccharide-induced acute lung injury in mice. Int J Mol Med. 37:1091–1098. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Bianchi AM, Reboredo MM, Lucinda LM, Reis FF, Silva MV, Rabelo MA, Holanda MA, Oliveira JC, Lorente JÁ and Pinheiro Bdo V: The Effects of Prone position ventilation on experimental mild acute lung injury induced by intraperitoneal lipopolysaccharide injection in rats. Lung. 194:193–199. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Ariza-Prota M, Pando-Sandoval A and Garcia-Clemente M: Lung injury caused by all-trans-retinoic acid in the treatment of acute promyelocytic leukemia. Arch Bronconeumol. 52:441–442. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Piper A, Song Y, Eves ND and Maher TM: Year in review 2013: Acute lung injury, interstitial lung diseases, sleep and physiology. Respirology. 19:428–437. 2014. View Article : Google Scholar : PubMed/NCBI

31 

McMullen SM, Meade M, Rose L, Burns K, Mehta S, Doyle R and Henzler D; Canadian Critical Care Trials Group (CCCTG), : Partial ventilatory support modalities in acute lung injury and acute respiratory distress syndrome-a systematic review. PLoS One. 7:e401902012. View Article : Google Scholar : PubMed/NCBI

32 

Osaka D, Shibata Y, Kanouchi K, Nishiwaki M, Kimura T, Kishi H, Abe S, Inoue S, Tokairin Y, Igarashi A, et al: Soluble endothelial selectin in acute lung injury complicated by severe pneumonia. Int J Med Sci. 8:302–308. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Seki H, Fukunaga K, Arita M, Arai H, Nakanishi H, Taguchi R, Miyasho T, Takamiya R, Asano K, Ishizaka A, et al: The anti-inflammatory and proresolving mediator resolvin E1 protects rat from bacterial pneumonia and acute lung injury. J Immunol. 184:836–843. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Nieuwenhuizen L, de Groot PG, Grutters JC and Biesma DH: A review of pulmonary coagulopathy in acute lung injury, acute respiratory distress syndrome and pneumonia. Eur J Haematol. 82:413–425. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Nakamura S, Yanagihara K, Izumikawa K, Seki M, Kakeya H, Yamamoto Y, Mukae H, Tashiro T and Kohno S: Efficacy of sivelestat for acute lung injury due to severe bacterial pneumonia with systemic inflammatory response syndrome. Nihon Kokyuki Gakkai Zasshi. 46:793–797. 2008.(In Japanese). PubMed/NCBI

36 

Czopik AK, Bynoe MS, Palm N, Raine CS and Medzhitov R: Semaphorin 7A is a negative regulator of T cell responses. Immunity. 24:591–600. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Garcia-Gonzalez MJ, Dominguez-Rodriguez A and Ferrer-Hita JJ: Unusual etiology of acute lung injury in a patient with acute myocardial infarction. Int J Cardiol. 117:e95–e97. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Delorme G, Saltel F, Bonnelye E, Jurdic P and Machuca-Gayet I: Expression and function of semaphorin 7A in bone cells. Biol Cell. 97:589–597. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Morote-Garcia JC, Napiwotzky D, Köhler D and Rosenberger P: Endothelial Semaphorin 7A promotes neutrophil migration during hypoxia. Proc Natl Acad Sci USA. 109:pp. 14146–14151. 2012; View Article : Google Scholar : PubMed/NCBI

40 

Zarbock A, Bishop J, Muller H, Schmolke M, Buschmann K, Van Aken H and Singbartl K: Chemokine homeostasis vs. chemokine presentation during severe acute lung injury: the other side of the Duffy antigen receptor for chemokines. Am J Physiol Lung Cell Mol Physiol. 298:L462–L471. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Bhatia M, Zemans RL and Jeyaseelan S: Role of chemokines in the pathogenesis of acute lung injury. Am J Respir Cell Mol Biol. 46:566–572. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Allegra M, Zaragkoulias A, Vorgia E, Ioannou M, Litos G, Beug H and Mavrothalassitis G: Semaphorin-7a reverses the ERF-induced inhibition of EMT in Ras-dependent mouse mammary epithelial cells. Mol Biol Cell. 23:3873–3881. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Kojicic M, Li G, Hanson AC, Lee KM, Thakur L, Vedre J, Ahmed A, Baddour LM, Ryu JH and Gajic O: Risk factors for the development of acute lung injury in patients with infectious pneumonia. Crit Care. 16:R462012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 15 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Wang H, Jia K, Wang H and Ren T: Anti-Semaphorin-7A single chain antibody demonstrates beneficial effects on pulmonary inflammation during acute lung injury Retraction in /10.3892/etm.2023.12165. Exp Ther Med 15: 2356-2364, 2018
APA
Chen, X., Wang, H., Jia, K., Wang, H., & Ren, T. (2018). Anti-Semaphorin-7A single chain antibody demonstrates beneficial effects on pulmonary inflammation during acute lung injury Retraction in /10.3892/etm.2023.12165. Experimental and Therapeutic Medicine, 15, 2356-2364. https://doi.org/10.3892/etm.2018.5724
MLA
Chen, X., Wang, H., Jia, K., Wang, H., Ren, T."Anti-Semaphorin-7A single chain antibody demonstrates beneficial effects on pulmonary inflammation during acute lung injury Retraction in /10.3892/etm.2023.12165". Experimental and Therapeutic Medicine 15.3 (2018): 2356-2364.
Chicago
Chen, X., Wang, H., Jia, K., Wang, H., Ren, T."Anti-Semaphorin-7A single chain antibody demonstrates beneficial effects on pulmonary inflammation during acute lung injury Retraction in /10.3892/etm.2023.12165". Experimental and Therapeutic Medicine 15, no. 3 (2018): 2356-2364. https://doi.org/10.3892/etm.2018.5724