AM1241

Effects of cannabinoid receptor 2 synthetic agonist, AM1241, on bleomycin induced pulmonary fibrosis

Ali Parlar , Seyfullah Oktay Arslan , Onder Yumrutas , Ebru Elibol , Alper Yalcin , Fatih Uckardes , Hasan Aydin , Muhammed Fatih Dogan , Elif Kayhan Kustepe & Mehmet Kaya Ozer

To cite this article: Ali Parlar , Seyfullah Oktay Arslan , Onder Yumrutas , Ebru Elibol , Alper Yalcin , Fatih Uckardes , Hasan Aydin , Muhammed Fatih Dogan , Elif Kayhan Kustepe & Mehmet Kaya Ozer (2020): Effects of cannabinoid receptor 2 synthetic agonist, AM1241, on bleomycin induced pulmonary fibrosis, Biotechnic & Histochemistry, DOI: 10.1080/10520295.2020.1758343
To link to this article: https://doi.org/10.1080/10520295.2020.1758343

Full Terms & Conditions of access and use can be found at https://www.tandfonline.com/action/journalInformation?journalCode=ibih20

BIOTECHNIC & HISTOCHEMISTRY
https://doi.org/10.1080/10520295.2020.1758343

Effects of cannabinoid receptor 2 synthetic agonist, AM1241, on bleomycin induced pulmonary fibrosis
Ali Parlar a, Seyfullah Oktay Arslanb, Onder Yumrutas c, Ebru Elibold, Alper Yalcind, Fatih Uckardese,
Hasan Aydinf, Muhammed Fatih Doganb, Elif Kayhan Kustepe d, and Mehmet Kaya Ozer a
aFaculty of Medicine, Department of Pharmacology, University of Adıyaman, Adıyaman, Turkey; bPharmacology Department, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, Turkey; cFaculty of Medicine, Department of Medical Biology, University of Adıyaman, Adıyaman, Turkey; dFaculty of Medicine, Department of Histology and Embryology, University of Adıyaman, Adıyaman, Turkey; eFaculty of Medicine, Department of Biostatistics and Medical Informatics, University of Adıyaman, Adıyaman, Turkey; fFaculty of Pharmacy, Department of Pharmaceutical Toxicology, University of Adıyaman, Adıyaman, Turkey

KEYWORDS
AM1241; bleomycin; cannabinoid receptor 2; CB2 receptor; CB2 agonist; lung; pulmonary fibrosis; rats

Fibrosis is characterized by rapid accumulation of extracellular matrix, which damages lung, liver, kidney, heart and skin (de Jesus et al. 2011; Heinzmann-Filho et al. 2013; Richter et al. 2015). Bleomycin (BLM) is a chemotherapeutic agent used to treat many types of cancers (Della Latta et al. 2015). Pulmonary fibrosis is a common side effect of BLM treatment (Arslan et al. 2002). Earlier reports indicate that metabolic homeostasis is disrupted in tissues of patients treated with BLM as well as oxidative stress associated with inflammation, which together contribute to fibrosis (Richter et al. 2015).
Cannabinoid compounds have been used medically for centuries (Mackie 2008) to regulate immune responses, e.g. leukocyte activation and cytokine release (Klein 2005), and inflammatory processes in neurological diseases (Klein 2005). Cannabinoids affect the endocannabinoid system by binding to two types of receptors: cannabinoid (CB) 1 receptors and CB2 receptors. Activation of these two types of receptors reduces liver fibrosis (Dai et al. 2017) and CB2 receptors exhibit anti-fibrotic effects in fibrosis induced by BLM (Akhmetshina et al. 2009). CB2 receptors were

activated in a BLM induced pulmonary fibrosis animal model (Arslan et al. 2002). Also, by activation of endocannabinoids or synthetic agonists, CB2 receptors provided protection against lung tissue damage in BLM treated mice (García-Martín et al. 2018). In addition, activation of CB2 receptors prevented hydrocephalus fibrillation, and JWH-133, an agonist of CB2, provided protection against ventricular fibrosis by suppressing tumor necrosis factor beta (TNF-β) (Tan et al. 2017). JWH-133 reduced skin and lung fibrosis by inhibiting proliferation of fibroblasts (Servettaz et al. 2010).
AM1241 is a selective CB2 receptor agonist. Activation of CB2 receptors by AM1241 delayed the progression of amyotrophic lateral sclerosis (Kim et al. 2006), and pharmacological activation of CB2 receptors by AM1241 decreased pentylenetetrazole induced seizures in epileptic rats (de Carvalho et al. 2016). The effect of activation of CB2 receptors by the AM1241 agonist on lung fibrosis is not clear. We investigated the effects of CB2 receptor activation by AM1241 in a BLM induced lung fibrosis rat model.

Materials and methods
Chemicals
Bleomycin sulfate was purchased from Onko Inc. (Istanbul, Turkey). Dimethylsulfoxide (DMSO), trichloroacetic acid, disodium hydrogen phosphate, dithiobisnitrobenzene thiobarbituric acid, AM1241 and AM630 were purchased from Sigma-Aldrich (Sigma Chemical Co., St. Louis, MO).

Sample size calculation
A pilot study was conducted to determine the proper sample size. A power analysis calculated using the G*Power (version 3.1.9.2) program indicated that at least 30 animals were required to achieve a power of
0.85 and an effect size of 0.75 at a significance level of
0.05. Therefore, we used 30 animals for our study.

Animals and experimental groups
Our study was approved by the local ethics committee (decision no. 2015/4). All animal experiments were performed in accordance with the care and use guidelines of Dollvet institution.
We used 8-week-old 180 − 200 g female Wistar rats obtained from the Experimental Research Center of Kahramanmaras Sutcu Imam University. To induce lung fibrosis, a solution of 5 mg/kg BLM dissolved in
0.1 ml saline was instilled into the trachea following a vertical midline incision of the neck. The dose of BLM was based on an earlier report (Cao et al. 2014). Two series, A and B, consisting of 30 rats each were created. The animals in series A were sacrificed 7 days after drug treatment and those in series B were sacrificed 28 days after the drug treatment. For each series, the animals were divided into five groups of six: control, 0.1 ml saline placed in trachea; BLM, 0.1 ml BLM placed in trachea; BLMA, CB2 agonist (AM1241,
0.2 ml) injected intraperitoneally (i.p.) before BLM was placed in the trachea; BLMA + A, CB2 antagonist (AM630, 0.2 ml) and CB2 agonist (AM1241, 0.2 ml) were injected i.p. at 30 min intervals once before BLM was placed in the trachea; BLM + vehicle, 20% DMSO was injected i.p. before BLM was placed in the trachea. Both the CB2 agonist (AM1241) and CB2 antagonist (AM630) dissolved in 20% (v/v) DMSO were injected i.p. at doses of 3 mg/kg and 1 mg/kg, respectively. The doses of the CB2 agonist and antagonist compounds were those reported in the
literature (de Carvalho et al. 2016)

Animal weights and lung indexes
The animals were observed daily and their body weights were recorded on days 0, 3, 6, 12, 15, 21, 24 and 28 after the animals in series A and B were sacrificed on the days 7 and 28, respectively. Both lungs were removed in toto including the trachea and weighed at the end of the experiment to determine the lung:body weight ratio, i.e. lung weight divided by body weight (lung index) for both series A and B.

Collection of broncho-alveolar lavage fluid (BALF)
At 7 or 28 days after BLM was given, the rats were anesthetized with 100 mg/kg, ketamine i.p. and sacrificed with overdose of anesthesia. The thorax was opened by a median incision, and the trachea was cannulated using a plastic catheter attached to a 10 ml syringe. BALF was obtained by instilling 5 ml sterile saline into the lungs five times and gently massaging the lungs. After each 5 ml instillation, BALF was collected by withdrawal into the injector and collected in a centrifuge tube. The BALF then was centrifuged at 300 x g for 10 min at 4 ºC to obtain the supernatant for biochemical analysis.

Preparation of lung tissue
The lungs were divided into five pieces and washed in cold saline. One sample was placed in 10% formaldehyde for subsequent histopathological examination. The other samples were frozen quickly in liquid nitrogen before storing at −20 ºC for later determination of levels of hydroxyproline, collagen type 1, total protein, glutathione (GSH) and malondialdehyde (MDA). The frozen lung samples were thawed and homogenized in isotonic saline before the experiments.

Total protein, hydroxyproline, and collagen type 1
A BCA protein assay kit (Thermo Fisher Scientific, Paisley, UK) was used to determine the amount of hydroxyproline/g protein and MDA/g protein; total proteins in BALF and tissue were expressed as mg/ml and mg/g, respectively. A hydroxyproline assay kit (Ab222941; Abcam) and rat COL1 kit (collagen type
I) (E-EL-R0233; Elabscience, Houston, TX) were used to measure hydroxyproline and collagen type 1 levels, respectively. The hydroxyproline content and collagen type 1 levels were expressed as µg/mg protein and µg/ mg wet lung tissue, respectively.

Interleukin-6 (IL-6) and TNF-α levels
IL-6 and TNF-α levels in BALF were measured using an enzyme-linked immunosorbent assay according to the manufacturer’s instructions (ab100772 and ab100785, respectively; Abcam). The IL-6 and TNF-α levels in BALF were expressed as pg/ml. The lower limit of detection of IL- 6 and TNF-α was 8 pg/ml and 4 pg/ml, respectively.

MDA levels
For MDA measurement, 750 μl of the supernatant were mixed 1:1 with 0.67% thiobarbituric acid and placed in a water bath for 15 min. The absorbance then was measured spectrophotometrically at 535 nm. The results were expressed as mmol/l/g protein.

GSH levels
Lung tissues were homogenized in 10 ml 10% trichloroacetic acid, followed by centrifugation at 3,900 x g at 4 ºC for 15 min. as described earlier (Parlar et al. 2018). Then, 0.5 ml of the supernatant was removed and mixed with 2 ml 0.3 M disodium hydrogen phosphate. After addition of 0.2 ml dithiobisnitrobenzene dissolved in 1% sodium citrate, the mixture was thoroughly vortexed. Finally, absorption of aliquots was measured at 412 nm. The results were expressed as nmol/g tissue.

Histopathology
Lung samples were fixed in neutral buffered 10% formalin for 7 days. After fixation, samples were dehydrated through ascending alcohols, cleared with xylene and embedding in paraffin. Sections were cut at 7 µm with a manual microtome (RM 2125; Leica Instruments, Nussloch, Germany). Sections were mounted on slides, deparaffinized, rehydrated, then stained using a Masson trichrome kit (Bio-Optica, Milan, Italy). Sections were photographed using a light microscope (Axiocam ERc5 s; Carl Zeiss, Gottingen, Germany) with digital camera attachment for histopathologic evaluation at 40 x.

Statistical analysis
Data are reported as means ± SD. A two-way analysis of variance was used and the Bonferroni post hoc test was used for multiple comparisons. Statistical significance was set at p ≤ 0.05.

Results
Body weight changes and lung indexes
The body weight of the animals in the saline control group increased with time, whereas those in the BLM group decreased compared to the saline control group (Figure 1a, Table 1). We found that the body weight of the animals in the saline group was higher than those in the BLM group on day 3 (p = 0.013) and days 6, 12, 15, 21 and 28 (p < 0.0001). We found that the body weight of the animals in the BLMA group was higher than those in the BLM group on days 12, 15, 21, 24 and 28 (p < 0.0001). Comparison of the body weights in the BLMA and BLMA + A groups show that the CB2 antagonist inhibited the effectiveness of the CB2 agonist, which increased the body weight compared to BLM group (p < 0.0001). We found an increased in lung index for the BLM group compared to the saline control group on days 7 and 28 (p = 0.0024 and p < 0.0001, respectively) (Figure 1b). The CB2 agonist reduced the lung index only in the 28 day series (4.60 ± 0.24 mg/g). Although the CB2 agonist decreased the lung index in the 7 day series (3.86 ± 0.32 mg/g), the difference was not statistically significant. The CB2 antagonist (5.73 ± 0.68 mg/g) reversed the effect of the CB2 agonist only in the 28 day series (p = 0.0063). Total protein level in BALF and lung tissue For the 7 day and 28 day series, the total protein levels in BALF (7 days, 0.80 ± 0.19 mg/ml; 28 days, 0.71 ± 0.18 mg/ml) and tissue (7 days, 1.01 ± 0.18 mg/g tissue; 28 days 0.69 ± 0.10 mg/g tissue) in the BLM group were higher than those in the saline control group (BALF, 0.39 ± 0.14 mg/ml, 0.20 ± 0.03 mg/ml, for the 7 and 28 day series, respectively; for tissue, 0.33 ± 0.13 mg/g tissue, 0.29 ± 0.11 mg/g tissue for the 7 and 28 day series, respectively (Figure 2a, b). For the 7 day and 28 day series, the total protein levels in BALF (7 days, 0.75 ± 0.12 mg/ml; 28 d, 0.63 ± 0.15 mg/ml) and tissue (7 days, 0.90 ± 0.18 mg/g tissue; 28 days 0.69 ± 0.11 mg/g tissue) in the BLMA + A group were higher than those for the BLMA group (in BALF, 0.50 ± 0.12 mg/ml, 0.38 ± 0.13 mg/ml for the 7 and 28 day series, respectively; in tissue, 0.59 ± 0.13 mg/g tissue, 0.33 ± 0.15 mg/g tissue for the 7 and 28 day series, respectively) (Table 2). In the BLM group, although we found increased total protein level in tissue in the 7 day series compared to the 28 day series, the difference was not statistically significant for the total protein level in BALF in the 7 day vs. the 28 day series (Figure 2a, b). Figure 1. Effects of the CB2 agonist on body weight on days 0, 3, 6, 12, 15, 21, 24 and 28 (a) and lung indexes (b). Data are means ± SD (n = 6). *p < 0.05, **p < 0.001, and ***p < 0.0001 vs. saline control group. ###p < 0.0001 vs. BLM group. λλλp < 0.0001 vs. 7 day series. Hydroxyproline content We found a significant difference in hydroxyproline content between the BLM group (7 days, 1.64 ± 0.36 µg/mg protein; 28 days, 2.75 ± 0.26 µg/mg protein) and the saline control group (7 days 0.93 ± 0.27 µg/mg protein; 28 days 1.01 ± 0.36 µg/mg protein) (Figure 2c, Table 2). We found that the hydroxyproline content in the BLM group was greater in the 28 day series compared to the 7 day series (p = 0.0002). The CB2 agonist reduced the hydroxyproline content only in the 28 day series (1.42 ± 0.39 µg/mg protein). Compared to the BLM group, the hydroxyproline content for the BLMA group in the 7-day series (1.38 ± 0.37 µg/mg protein) was reduced, but the reduction of hydroxyproline content was not found in 28-day series. Collagen type 1 level We found no significant difference in the collagen type 1 content among the groups in the 7 day series (Figure 2d). For the 28 day series, we found a significant difference in collagen type 1 content between the BLM group (119.48 ± 18.62 µg/g tissue) and saline control group (29.63 ± 4.66 µg/g tissue), and between the CB2 agonist group (60.02 ± 18.68 µg/ g tissue) and BLM group. We found a decrease in the collagen type 1 content in the BLM group in the 7-day series compared to the 28 day series (p < 0.0001). IL-6 and TNF-α levels The levels of IL-6 in BALF increased in the BLM group (150.03 ± 7.21 pg/ml) compared to the saline control group (90.20 ± 1 0.26 pg/ml) in the 7 day series (p = 0.0020). Treatment with the CB2 agonist (102.42 ± 9.28 pg/ml) decreased the level of IL-6 in BALF (p = 0.0235 compared to the BLM group; the CB2 antagonist reversed this effect (147.59 ± 15.02 pg/ml) (Figure 3a). In the 28 day series, we found a significant difference in the IL-6 level in the BLM group (194.43 ± 34.87 pg/ml) compared to the saline control group (77.97 ± 21.26 pg/ml) and CB2 agonist pretreatment group (126.59 ± 20.47 pg/ml) (p < 0.0001 and p = 0.0004, respectively). We also found an increase in the IL-6 levels of the 7 day series, compared to the 28 day series (p = 0.0449). The levels of TNF-α in BALF increased in the BLM group (7 day, 74.46 ± 7.15; 28 day, 46.33 ± 6.80 pg/ml, respectively) compared to the saline control group (7 day, 25.79 ± 3.26; 28 day, 26.47 ± 4.34 pg/ml) (7 day p < 0.0001; 28 day, p = 0.0029) (Figure 3b). Compared to the BLM group (74.46 ± 7.15 pg/ml), the BLMA group (49.67 ± 5.89 pg/ml) exhibited decreased TNF- α in the 7 day series only. We found no statistically significant difference in TNF-α levels between the BLMA group and BLM groups or between BLMA and BLMA + A groups in the 28-day series. In the 7 day series, the CB2 antagonist countered the CB2 agonist’s reducing effect on TNF-α level when the BLMA + A group (68.50 ± 7.04) compared to the BLMA group (49.67 ± 5.89 pg/ml). The TNF-α level in the 7 day series was greater than that for the 28 day series (p < 0.0001). MDA and GSH levels The MDA level increased in the BLM group (1.52 ± 0.32 mmol/g protein) compared to the saline control group (0.71 ± 0.21 mmol/g protein) in the 7 day series (p < 0.0001). Pretreatment with the CB2 agonist decreased the level of MDA (0.84 ± 0.16 mmol/g protein) (p < 0.0001); the CB2 antagonist (1.43 ± 0.25 mmol/g protein) reversed this effect (Figure 3c). For the 28 day series, we found a significant difference in the MDA level in the BLM group (1.15 ± 0.19 mmol/g protein) compared to the saline control group (0.69 ± 0.18 mmol/g protein) and the CB2 agonist pretreated group (0.75 ± 0.13 mmol/g protein). We found a significant difference in the GSH level in the lung tissue of the BLM group (7 day, 138.47 ± 46.19 nmol/g tissue; 28 day, 255.62 ± 42.65 nmol/g tissue) compared to the saline control group (7 day, 455.04 ± 28.01 nmol/g tissue; 28 day, 421.18 ± 51.50 nmol/g tissue) in both the 7 day (p < 0.0001) and 28 day series (p = 0.0016) (Figure 3d). The CB2 agonist increased the GSH level in the 7 day (395.09 ± 33.59 nmol/g tissue) and 28 day series (392.48 ± 53.30 nmol/g tissue). Pretreatment with the CB2 antagonist (7 day 236.26 ± 41.15 nmol/g tissue; 28 day 263.28 ± 73.08 nmol/g tissue, reversed the effect of the CB2 agonist in both the 7 day and 28 day series. Histopathology Histopathological sections from all study groups are shown in Figures 4 and 5. Pulmonary fibrosis was assessed by morphological examination of Masson’s trichrome stained lung sections. Lung sections of the 7 day series were evaluated using a light microscope. Normal alveolar structure was evident in the sections of the saline control group; no fibrosis was observed in this group. By contrast, in the BLM group, marked thickening of alveolar septa was observed in addition to collapse of alveolar spaces, loss of alveolar structure, areas of perivascular fibrosis and collagen fibers. Alveolar cell necrosis and destruction of alveolar walls were seen in the BLM and BLMA + A groups. Granulomas also were observed in the BLMA + A group. The histological structure in the BLMA and BLM + vehicle groups was similar to that of the saline control group (Figure 4). In the 28 day series, the general histological structure of the lung tissue was normal in the saline control group. Increased fibrosis, especially in peribronchial locations, as well as necrosis and inflammation area, was observed in the BLM group. Intratracheal instillation of BLM caused prominent accumulation of collagen and extracellular matrix, thickened Figure 2. Masson’s trichrome stained sections of rat lung with pulmonary fibrosis induced by BLM and CB2 in 7 day series. Scale bars = 250 μm. Br, bronchioles; Bv, blood vessel; ad, alveolar duct; C, collagen fiber; SM, smooth muscle; T, terminal bronchioles; R, respiratory bronchioles; ↔, enlarged perivascular and peribronchiolar areas; arrow, destroyed epithelium of a pulmonary artery and bronchioles; G, granulomas; √, areas of alveolar collapse. interalveolar septae and obstruction of alveolar spaces in the BLM group. These findings were exacerbated in the BLMA + A group. The histological structure for the BLMA + A and BLM + vehicle groups were similar to the BLM group. Our histopathological findings demonstrated that BLM induced pulmonary fibrosis was more extensive in the 28 day series after BLM instillation compared to the 7 day series (Figure 5). For the BLM group in the 7 day and 28 day series we found fibrotic lesions in peribronchiolar and perivascular areas. Therefore, normal alveolar architecture was disrupted in this group. An increase was observed in the infiltration of inflammatory cells in interstitial and alveolar spaces. Collagen synthesis was increased significantly in the BLM and BLMA + A groups in response to BLM instillation compared to the other groups. Increased collagen fibers and hypertrophic smooth muscle cells were particularly evident in perivascular and peribronchiolar areas. The groups with fibrosis exhibited severely damaged lungs, with infiltration of inflammatory Table 2. Lung indexes and total protein, hydroxyproline, collagen, oxidative stress and cytokine levels. Days Saline control BLM BLM + vehicle BLMA + A BLMA P1 P2 P3 Lung index (mg/g) 7 3.45 ± 0.10 4.68 ± 0.54 4.62 ± 0.41 4.32 ± 0.19 3.86 ± 0.32 0.0024 0.1299 > 0.999
28 3.67 ± 0.42 6.15 ± 0.92 6.08 ± 0.65 5.73 ± 0.68 4.60 ± 0.24 < 0.0001 0.0001 0.0063 Total protein (mg/ml) 7 0.39 ± 0.14 0.80 ± 0.19 0.82 ± 0.13 0.75 ± 0.12 0.50 ± 0.12 < 0.0001 0.0042 0.0239 28 0.20 ± 0.03 0.71 ± 0.18 0.70 ± 0.14 0.63 ± 0.15 0.38 ± 0.13 < 0.0001 0.0013 0.0255 Total protein (mg/g tissue) 7 0.33 ± 0.13 1.01 ± 0.18 0.97 ± 0.22 0.90 ± 0.18 0.59 ± 0.13 < 0.0001 0.0030 0.0485 28 0.29 ± 0.11 0.69 ± 0.10 0.70 ± 0.19 0.69 ± 0.11 0.33 ± 0.15 0.0036 0.0113 0.0112 Hidroxyproline 7 0.93 ± 0.27 1.64 ± 0.36 1.49 ± 0.23 1.55 ± 0.32 1.38 ± 0.37 0.0344 > 0.999 > 0.999
(µg/mg protein)
28 1.01 ± 0.36 2.75 ± 0.26 2.78 ± 0.24 2.12 ± 0.23 1.42 ± 0.39 < 0.0001 < 0.0001 0.0437 Col 1 (µg/g tissue) 7 30.71 ± 9.37 35.29 ± 8.34 36.01 ± 2.34 33.63 ± 3.89 31.94 ± 4.83 > 0.999 > 0.999 > 0.999
28 29.63 ± 4.66 119.48 ± 18.62 117.74 ± 17.21 92.98 ± 18.97 60.02 ± 18.68 < 0.0001 < 0.0001 0.0030 IL-6 (pg/ml) 7 90.20 ± 10.26 150.03 ± 7.21 149.39 ± 15.71 147.59 ± 15.02 102.42 ± 9.28 0.0020 0.0235 0.0385 28 77.97 ± 21.26 194.43 ± 34.87 190.21 ± 35.64 174.68 ± 25.62 126.59 ± 20.47 < 0.0001 0.0004 0.0213 TNF-α (pg/ml) 7 25.79 ± 3.26 74.46 ± 7.15 75.15 ± 5.96 68.60 ± 7.04 49.67 ± 5.89 < 0.0001 0.0002 0.0052 28 26.47 ± 4.34 46.33 ± 6.80 46.83 ± 6.85 44.11 ± 4.82 33.83 ± 6.49 0.0029 0.2180 0.8116 MDA (mmol/g protein) 7 0.71 ± 0.21 1.52 ± 0.32 1.43 ± 0.31 1.43 ± 0.25 0.84 ± 0.16 < 0.0001 < 0.0001 0.0006 28 0.69 ± 0.18 1.15 ± 0.19 1.14 ± 0.19 1.09 ± 0.22 0.75 ± 0.13 0.0125 0.0449 0.1843 GSH (nmol/g tissue) 7 455.04 ± 28.01 138.47 ± 46.19 166.99 ± 48.83 236.26 ± 41.15 395.09 ± 33.59 < 0.0001 < 0.0001 0.0026 28 421.18 ± 51.50 255.62 ± 42.65 237.10 ± 73.42 263.28 ± 73.08 392.48 ± 53.30 0.0016 0.0131 0.0231 Data are means ± SD (n = 6). p1, p value of the saline control and BLM groups; p2, p value of the BLMA and BLM groups; p3, p value of the BLMA + A and BLMA groups. Figure 3. Masson’s trichrome stained sections of rat lung with pulmonary fibrosis induced by BLM and CB2 in 28 day series. Scale bars = 250 μm. Br, bronchioles; Bv, blood vessel; C, collagen fiber; SM, smooth muscle; N, alveolar wall destruction and necrosis of alveolar cells; ↔, enlarged perivascular and peribronchiolar areas; arrow, destroyed epithelium of a pulmonary artery and bronchioles; √, areas of alveolar collapse; *inflammation. cells followed by infiltration of the interstitium and fibroblast deposition and excessive collagen deposition. By contrast, these inflammatory and fibrotic changes were reduced significantly by CB2 agonist treatment. Our findings indicate that CB2 inhibited BLM induced pulmonary fibrosis. Normal alveolar structure without accompanying fibrosis was observed in the saline control group. Discussion Previous reports indicate that activation of CB2 receptors produce a beneficial effect on various diseases. For example, activation of CB2 receptors inhibited pain (Gao et al. 2018) and reduced fibrogenesis and inflammation during skin wound healing (Wang et al. 2016). CB2 selective agonists exhibited immunomodulatory activities (Manera et al. 2015). Treatment with the CB2 receptor agonist, AM1241, reduced the progression of amyotrophic lateral sclerosis Figure 4. Total protein levels in BALF (a), tissue (b), hydroxyproline (c) and collagen type 1 levels (d). Data are means ± SD (n = 6). *p < 0.05, **p < 0.001, ***p < 0.0001 vs. saline control group. #p < 0.05, ##p < 0.001 and ###p < 0.0001 vs. BLM group. &p < 0.05 vs. BLMA group; λp < 0.05, λλλp < 0.0001 for the BLM group of the 28 day series vs. BLM group of the 7 day series. (Kim et al. 2006), and reduced oxidative stress and inflammation in ischemic heart disease by activation of the PI3 K/Akt/Nrf2 pathway (Li et al. 2016). These investigators also reported activation of the CB2 receptor via AM1241 reduced myocardial fibrosis formation and that AM1241 ameliorated cardiac fibrosis by inhibiting NF2 via the TGF-β1/Smad3 pathway (Li et al. 2016). Liu et al. (2014) proposed that AM1241 reduced secretion of inflammatory cytokines including p38, MAP1, ERK1/2 and JNK1/2 in acute lung injury by CB2 activation. Although AM1241 significantly reduced fibrosis by activating the CB2 receptor, its effects on pulmonary fibrosis have not been reported. Ours is the first report that AM1241 provides protection against fibrosis in lung tissue. We observed biochemical, molecular and histopathological changes as a result of CB2 receptor agonist treatment in the 7 day and 28 day study groups exposed to BLM. Wang et al. (2016) reported that application of a CB2 agonist, GP1a, decreased TNF-α and IL-6 levels compared to those in an antagonist group (AM630). The weight of the rats in the BLM group decreased during the study. By contrast, the weight of the rats in the BLMA and BLMA + A groups decreased until day 7, then increased during the following days. Therefore, it appears that AM1241 induced activation of the CB2 receptor may have participated in the weight increase of the rats following the application of BLM. BLM increased free radical production by creating a triple complex in the presence of oxygen and iron, and these radicals cut one or two strands of the DNA (Chen et al. 2008). Inflammation and fibroblast proliferation occurred in response to DNA damage and deterioration of the antioxidant-reactive oxygen species (ROS) balance caused by BLM (Guan et al. 2016). Pinkerton et al. (2017) reported that application of BLM increased fibrosis in lung tissue. It has been reported that collagen synthesis increased in parallel with increases in hydroxyproline levels, with increased tissue fibrosis (Chen et al. 2012; Kabel et al. 2017). We found that the hydroxyproline and collagen type 1 content increased in the lung tissues of the BLM group. In addition, histopathological examination revealed significant thickening of alveolar septa, collapse of alveolar spaces, loss of alveolar structure, perivascular fibrotic areas, increased number of fibroblast cells and Figure 5. Effects of the CB2 agonist on IL-6 (a), TNF-α (b), MDA (c) and GSH (d) levels. Data are means ± SD (n = 6). *p < 0.05, **p < 0.001 and ***p < 0.0001 vs. saline control group. #p < 0.05; ###p < 0.0001 vs. BLM group, &p < 0.05, &&p < 0.001, &&&p < 0.0001 vs. BLMA group. λp < 0.05, λλλp < 0.0001 for the BLM group of the 28 day series vs. BLM group of the 7 day series. collagen accumulation. Kabel et al. (2017) reported severe bleeding, emphysema, thickened alveolar septa and leukocytic infiltration of alveolar walls associated with increased fibrosis after application of BLM. Chen et al. (2012) reported low level interstitial inflammation and a large amount of peribronchial fibrosis in rat tissue following application of BLM. We found, however, that after application of AM1241 (BLMA group), hydroxyproline and collagen type 1 levels were decreased compared to the BLM group, and these findings were supported by histopathology. Earlier studies indicated that various substances affect BLM induced lung fibrosis. Arslan et al. (2002) reported that although hydroxyproline, collagen type 1 and total protein levels were increased in rats exposed to BLM, they decreased after the application of melatonin, which is an antioxidant. These investigators concluded that the level of antioxidant enzymes were increased in the groups to which melatonin was applied and that BLM induced pulmonary fibrosis was inhibited in the presence of the antioxidant. Chen et al. (2012) reported that Cordyceps sinensis, a fungus used in traditional Chinese medicine, protected against BLM induced pulmonary fibrosis. BLM is an agonist of CB2. The similar effects observed in the BLM treated groups in our study suggest that CB2 activation may participate in regulating ROS. Richter et al. (2015) reported increased cytokine levels in response to oxidative stress resulting from increased ROS and associated inflammation Fouad and Jresat (2011) reported elevated levels of TNF-α as a result of increased ROS. Molecular and biochemical events in BLM induced pulmonary fibrosis induced inflammation with subsequent release of cytokines, such as TNF-α and TNF-β (Khalil et al. 1993). TNF-α levels also increased during response to inflammation in lung tissue (Sime et al. 1998). We found that increased IL-6 and TNF-α levels in the BLM treated groups were decreased significantly after application of AM1241. Our findings indicate that CB2 receptor activation may participate in rapid regulation of inflammatory processes by decreasing the levels of cytokines (Klein 2005). We found no difference in MDA levels among the groups in the 7 day series. In the 28 day series, however, the MDA level increased for the BLM group compared to the saline control group, and the MDA level for the BLMA group decreased compared to the BLM group. Kabel et al. (2017) also reported that the MDA level in lung tissue increased significantly after application of BLM. GSH is a low molecular weight antioxidant, whose level is decreased by pulmonary fibrosis compared to normal tissues (Cheresh et al. 2013). These investigators reported that pulmonary fibrosis induced by BLM was resolved by increased in GSH levels. We found that the GSH level decreased significantly in the BLM treated groups, and the GSH level in the 7 day and 28 day series increased significantly in the BLMA group compared to the BLM group. Therefore, application of BLM decreased the antioxidant level and increased ROS levels of lung tissue and that the application of the CB2 agonist, AM1241, reversed these effects. CB2 receptor activation by the agonist, AM1241, exhibited an ameliorating effect on BLM induced pulmonary fibrosis by inhibiting inflammation related cytokines, such as IL-6 and TNF-α, and reduction of fibrosis by decreasing hydroxyproline and collagen type 1 levels. Further investigation is required to elucidate the possible contributions of other inflammatory cytokines to BLM induced pulmonary fibrosis. The mechanisms underlying changes in collagen structure at the molecular level also remain to be clarified.

Disclosure statement
The authors declare no conflict of interest.

Funding
Our work was supported by the Scientific Research ProjectsManagement Unit of Adiyaman University under Grant [TIPFMAP/2015-0010].

ORCID
Ali Parlar http://orcid.org/0000-0002-4656-3402
Onder Yumrutas http://orcid.org/0000-0001-9657-8306 Elif Kayhan Kustepe http://orcid.org/0000-0002-0030-5862 Mehmet Kaya Ozer http://orcid.org/0000-0002-7961-4130

References
Akhmetshina A, Dees C, Busch N, Beer J, Sarter K, Zwerina J, Zimmer A, Distler O, Schett G, Distler JHW. 2009. The cannabinoid receptor CB2 exerts antifibrotic effects in experimental dermal fibrosis. Arthr Rheum. 60:1129–1136. Arslan SO, Zerin M, Vural H, Coskun A. 2002. The effect of melatonin on bleomycin-induced pulmonary fibrosis in rats. J Pineal Res. 32:21–25. doi:10.1034/j.1600-
079x.2002.10796.x.

Cao H, Zhou X, Zhang J, Huang X, Zhai Y, Zhang X, Chu L. 2014. Hydrogen sulfide protects against bleomycin- induced pulmonary fibrosis in rats by inhibiting NF-κB expression and regulating Th1/Th2 balance. Toxicol Lett. 224:387–394. doi:10.1016/j.toxlet.2013.11.008.
Chen J, Ghorai MK, Kenney G, Stubbe J. 2008. Mechanistic studies on bleomycin-mediated DNA damage: multiple binding modes can result in double-stranded DNA cleavage. Nucl Acids Res. 36:3781–3790. doi:10.1093/nar/gkn302.
Chen M, Cheung FWK, Chan MH, Hui PK, Ip S-P, Ling YH, Che C-T, Liu WK. 2012. Protective roles of Cordyceps on lung fibrosis in cellular and rat models. J Ethnopharmacol. 143:448–454. doi:10.1016/j.jep.2012.06.033.
Cheresh P, Kim S-J, Tulasiram S, Kamp DW. 2013. Oxidative stress and pulmonary fibrosis. Biochim Biophys Acta. 1832:1028–1040. doi:10.1016/j.bbadis.2012.11.021.
Dai E, Zhang L, Ye L, Wan S, Feng L, Qi Q, Yao F, Li Z. 2017. Hepatic expression of cannabinoid receptors CB1 and CB2 correlate with fibrogenesis in patients with chronic hepatitis B. Int J Infect Dis. 59:124–130. doi:10.1016/j. ijid.2017.03.008.
de Carvalho CR, Hoeller AA, Franco PLC, Martini APS, Soares FMS, Lin K, Prediger RD, Whalley BJ, Walz R. 2016. The cannabinoid CB2 receptor-specific agonist AM1241 increases pentylenetetrazole-induced seizure severity in Wistar rats. Epilep Res. 127:160–167. doi:10.1016/j.eplepsyres.2016.08.011.
de Jesus CR, I-Ching L, de Neiva TJC, de Vituri CL. 2011. Assessment of fibrosis and vascularization of bone marrow stroma of chronic myeloid Leukemia patients treated with imatinib mesylate and their relationship with the cytogenetic response. Braz J Pharm Sci. 47:313–322. doi:10.1590/S1984-82502011000200012.
Della Latta V, Cecchettini A, Del Ry S, Morales MA. 2015. Bleomycin in the setting of lung fibrosis induction: from biological mechanisms to counteractions. Pharmacol Res. 97:122–130. doi:10.1016/j.phrs.2015.04.012.
Fouad AA, Jresat I. 2011. Therapeutic potential of cannabidiol against ischemia/reperfusion liver injury in rats. Eur J Pharmacol. 670:216–223. doi:10.1016/j. ejphar.2011.08.048.
Gao F, Xiang H-C, Li H-P, Jia M, Pan X-L, Pan H-L LM. 2018. Electroacupuncture inhibits NLRP3 inflammasome activation through CB2 receptors in inflammatory pain. Brain Behav Immun. 67:91–100. doi:10.1016/j. bbi.2017.08.004.
García-Martín A, Garrido-Rodríguez M, Navarrete C, Del Río C, Bellido ML, Appendino G, Calzado MA, Muñoz E. 2018. EHP-101, an oral formulation of the cannabidiol aminoquinone VCE-004.8, alleviates bleomycin-induced skin and lung fibrosis. Biochem Pharmacol. 157:304–313. doi:10.1016/j.bcp.2018.07.047.
Guan R, Zhao X, Wang X, Song N, Guo Y, Yan X, Jiang L, Cheng W, Shen L. 2016. Emodin alleviates bleomycin-induced pulmonary fibrosis in rats. Toxicol Lett. 262:161–172. doi:10.1016/j.toxlet.2016.10.004.
Heinzmann-Filho JP, Vargas MHM, Piva TC, Vendrusculo FM, Pinto LA, Marostica PJC, Donadio MVF. 2013. Equações internacionais superestimam a força muscular ventilatória em crianças e adolescentes com fibrose cística. Fisioterapia e Pesquisa. 20:387–393. doi:10.1590/S1809-29502013000400014.

Kabel AM, Abd Elmaaboud MA, Atef A, Baali MH. 2017. Ameliorative potential of linagliptin and/or calcipotriol on bleomycin-induced lung fibrosis: in vivo and in vitro study. Env Toxicol Pharmacol. 50:216–226. doi:10.1016/j. etap.2017.02.008.
Khalil N, Whitman C, Zuo L, Danielpour D, Greenberg A. 1993. Regulation of alveolar macrophage transforming growth factor-beta secretion by corticosteroids in bleomycin-induced pulmonary inflammation in the rat. J Clin Invest. 92:1812–1818. doi:10.1172/JCI116771.
Kim K, Moore DH, Makriyannis A, Abood ME. 2006. AM1241, a cannabinoid CB2 receptor selective compound, delays disease progression in a mouse model of amyotrophic lateral sclerosis. Eur J Pharmacol. 542:100–105. doi:10.1016/j.ejphar.2006.05.025.
Klein TW. 2005. Cannabinoid-based drugs as anti-inflammatory therapeutics. Nat Rev Immunol. 5:400–411. doi:10.1038/nri1602.
Li X, Han D, Tian Z, Gao B, Fan M, Li C, Li X, Wang Y, Ma S, Cao F. 2016. Activation of cannabinoid receptor type II by AM1241 ameliorates myocardial fibrosis via Nrf2-mediated inhibition of TGF-β1/Smad3 pathway in myocardial infarction mice. Cell Physiol Biochem. 39:1521–1536. doi:10.1159/000447855.
Mackie K. 2008. Cannabinoid receptors: where they are and what they do. J Neuroendocrinol. 20 (Suppl. 1):10–14. doi:10.1111/j.1365-2826.2008.01671.x.
Manera C, Malfitano AM, Parkkari T, Lucchesi V, Carpi S, Fogli S, Bertini S, Laezza C, Ligresti A, Saccomanni G, Savinainen JR, Ciaglia E, Pisanti S, Gazzerro P, Di Marzo V, Nieri P, Macchia M, Bifulco M. 2015. New quinolone- and 1,8-naphthyridine-3-carboxamides as selective CB2 receptor agonists with anticancer and immuno-modulatory activity. Eur J Med Chem. 97:10–18. doi:10.1016/j.ejmech.2015.04.034. Parlar A, Arslan SO, Doğan MF, Çam SA, Yalçin A, Elibol E, Özer MK, Üçkarde F, Kara H. 2018. The exogenous

administration of CB2 specific agonist, GW405833, inhibits inflammation by reducing cytokine production and oxidative stress. Exp Ther Med. 16:4900–4908. doi:10.3892/etm.2018.6753.
Pinkerton JW, Kim RY, Robertson AAB, Hirota JA, Wood LG, Knight DA, Cooper MA, O’Neill LAJ, Horvat JC, Hansbro PM. 2017. Inflammasomes in the lung. Mol Immunol. 86:44–55. doi:10.1016/j.molimm.2017.01.014.
Richter K, Konzack A, Pihlajaniemi T, Heljasvaara R, Kietzmann T. 2015. Redox-fibrosis: impact of TGFβ1 on ROS generators, mediators and functional consequences. Redox Biol. 6:344–352. doi:10.1016/j.redox.2015.08.015.
Servettaz A, Kavian N, Nicco C, Deveaux V, Chéreau C, Wang A, Zimmer A, Lotersztajn S, Weill B, Batteux F. 2010. Targeting the cannabinoid pathway limits the development of fibrosis and autoimmunity in a mouse model of systemic sclerosis. Am J Pathol. 177:187–196. doi:10.2353/ajpath.2010.090763.
Sime PJ, Marr RA, Gauldie D, Xing Z, Hewlett BR, Graham FL, Gauldie J. 1998. Transfer of tumor necrosis factor-alpha to rat lung induces severe pulmonary inflammation and patchy interstitial fibrogenesis with induction of transforming growth factor-beta1 and myofibroblasts. Am J Pathol. 153:825–832. doi:10.1016/S0002-9440(10)65624-6.
Tan Q, Chen Q, Feng Z, Shi X, Tang J, Tao Y, Jiang B, Tan L, Feng H, Zhu G, Yang Y, Chen Z. 2017. Cannabinoid receptor 2 activation restricts fibrosis and alleviates hydrocephalus after intraventricular hemorrhage. Brain Res. 1654:24–33. doi:10.1016/j.brainres.2016.10.016.
Wang -L-L, Zhao R, Li J-Y, Li -S-S, Liu M, Wang M, Zhang M-Z, Dong -W-W, Jiang S-K, Zhang M, Tian Z-L, Liu C-S, Guan D-W. 2016. Pharmacological activation of cannabinoid 2 receptor attenuates inflammation, fibrogenesis, and promotes re-epithelialization during skin wound healing. Eur J Pharmacol. 786:128–136. doi:10.1016/j.ejphar.2016.06.006.