Next Article in Journal
The Tumor Necrosis Factor-α Level in Platelet-Rich Plasma Might Be Associated with Treatment Outcome in Patients with Interstitial Cystitis/Bladder Pain Syndrome or Recurrent Urinary Tract Infection
Next Article in Special Issue
Chemical Compositions of Scutellaria baicalensis Georgi. (Huangqin) Extracts and Their Effects on ACE2 Binding of SARS-CoV-2 Spike Protein, ACE2 Activity, and Free Radicals
Previous Article in Journal
Teleost Eye Is the Portal of IHNV Entry and Contributes to a Robust Mucosal Immune Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Loliolide in Sargassum horneri Alleviates Ultrafine Urban Particulate Matter (PM 0.1)-Induced Inflammation in Human RPE Cells

1
AceBiome Inc., Seoul 06164, Republic of Korea
2
R&D Center, AceBiome Inc., Daejeon 34013, Republic of Korea
3
Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheonju-si 28116, Republic of Korea
4
Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheonju-si 28116, Republic of Korea
5
Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(1), 162; https://doi.org/10.3390/ijms25010162
Submission received: 10 November 2023 / Revised: 18 December 2023 / Accepted: 20 December 2023 / Published: 21 December 2023

Abstract

:
Owing to increasing air pollution due to industrial development, fine dust has been associated with threatening public health. In particular, ultrafine urban particulate matter (uf-UP, PM 0.1) can easily enter our bodies, causing inflammation-related diseases. Therefore, in the present study, we evaluated the effects of hydrothermal extracts of Sargassum horneri and its bioactive compound, loliolide, on uf-UP-induced inflammation as a potential treatment strategy for retinal disorders. Human retinal pigment epithelial cells (ARPE-19) stimulated with TNF-α or uf-UPs were treated with S. horneri extract and loliolide. S. horneri extracts exhibited anti-inflammatory effects on uf-UP-induced inflammation without cell toxicity through downregulating the mRNA expression of MCP-1, IL-8, IL-6, and TNF-α. UPLC-QTOF/MS analysis confirmed that the hydrothermal extract of S. horneri contained loliolide, which has anti-inflammatory effects. Loliolide effectively reduced the mRNA expression and production of proinflammatory chemokines (IL-8) and cytokines (IL-1β and IL-6) by downregulating the MAPK/NF-ĸB signaling pathway on TNF-α-stimulated inflammatory ARPE-19 cells. These effects were further confirmed in inflammatory ARPE-19 cells after stimulation with uf-UPs. Collectively, these results suggested the application of S. horneri as a functional ingredient for treating ocular disorders caused by particular matters.

1. Introduction

Environmental pollution has become increasingly severe worldwide, and many studies have reported that exposure to particulate matter (PM) harms human health. According to their aerodynamic diameters, PMs are divided into fine and ultrafine urban particulate matter (uf-UP), ranging from nanometers (nm) to microns (μm) [1]. Briefly, uf-UPs enter the body through the respiratory tract and are then translocated to all organs. Therefore, exposure to uf-UPs is closely correlated with respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), neurodegenerative diseases, as well as cardiovascular, metabolic, cancer, and emotional disorders [2,3,4]. In addition, PMs, including uf-UPs, cause eye diseases, such as allergic conjunctivitis, increased intraocular pressure, glaucoma, age-related macular degeneration (AMD), and dry eye syndrome [5,6,7,8].
Sargassum horneri (S. horneri) is a well-known brown alga commonly inhabiting the coast of Japan and Korea that is reported to exhibit various biological activities such as anti-inflammatory, anti-apoptotic and anti-allergic effects [9,10,11,12]. According to many studies, the main components of S. horneri mainly consist of polyphenols and carbohydrates [13,14]. S. horneri contains bioactive compounds, such as fucoidan and loliolide, with promising bioactivities, such as antioxidant and anti-inflammatory functions [15]. In particular, loliolide, the phenolic compound known as one of the active ingredients of S. horneri, is reported to have a variety of functions, such as cell protection, anti-cancer, anti-aging, and anti-apoptosis activities, including antioxidant and anti-inflammatory functions [16,17,18]. Furthermore, many studies have suggested that S. horneri is a potential inhibitor of PM-induced inflammatory responses, oxidative stress, and cell death [19,20]. We previously reported that fucoidan exhibits anti-inflammatory effects by suppressing the inflammatory response of TNF-α-stimulated ARPE-19 cells [14]. In particular, TNF-α, which is one of the key regulators of the PM-induced inflammatory response, is not only associated with dry eye syndrome and corneal epithelium inflammation but is also known to cause allergic immune responses that lead to the development of ocular allergy [8,21]. Nevertheless, the effects of uf-UPs on ocular disease remain relatively unknown compared with those of PMs.
In this study, we investigated the anti-inflammatory effects of S. horneri on uf-UP-stimulated ARPE-19 cells. We also evaluated whether loliolide reduces inflammation in a TNF-α-induced cellular model. We expect the present study will improve our knowledge of S. horneri and its functional compound, loliolide, which can relieve the uf-UP-induced inflammation of the retina.

2. Results

2.1. Cellular Changes in uf-UP-Exposed ARPE-19 Cells

We performed a cell viability assay to determine the toxicity of uf-UPs in APRE-19 cells. As shown in Figure 1A,B, no significant cytotoxicity or morphological changes were observed in cells exposed to concentrations of uf-UPs up to 10 μg/mL.
To determine whether uf-UP exposure induces an inflammatory response in ARPE-19 cells, we examined the levels of mRNA expression of inflammation-related genes using qRT-PCR. We observed that the mRNA levels of MCP-1 and TNF-α were significantly increased in ARPE-19 cells exposed to 10 μg/mL uf-UPs, while the mRNA expression of IL-6 was increased in cells treated with various concentrations of uf-UPs (Figure 1C). Therefore, we used 10 μg/mL uf-UPs for further experiments.

2.2. Effect of S. horneri on uf-UP-Induced Inflammation Response in ARPE-19 Cells

To investigate whether S. horneri exerts anti-inflammatory effects on uf-UP-treated ARPE-19 cells, we analyzed the levels of mRNA expression of chemokines and cytokines. We found that exposure to uf-UPs significantly upregulated the mRNA levels of the MCP-1 and IL-8 chemokines, whereas pretreatment with S. horneri significantly downregulated their mRNA levels in a dose-dependent manner (Figure 2A). Although we detected that the mRNA level of IL-1β showed a decreasing trend after S. horneri pretreatment of cells under uf-UP-induced inflammatory conditions, it was not statistically significant. Of note, we determined that the mRNA expression of IL-6 and TNF-α was significantly increased following exposure to uf-UPs. Conversely, we observed that S. horneri pretreatment significantly and dose-dependently reduced the upregulated expression of the above-mentioned genes (Figure 2B). These results suggested that S. horneri exerts anti-inflammatory effects on uf-UP-induced ARPE-19 cells.

2.3. UPLC-QTOF/MS Analysis

Based on the aforementioned results and a previous study [15], we considered loliolide to be the major component responsible for the anti-inflammatory effect of S. horneri. We, therefore, performed content analyses by chromatographically analyzing aqueous extracts of S. horneri using UPLC-QTOF/MS (Figure 3A). As shown in Figure 3B, the major peak observed at 4.5 min in the mass chromatogram of the S. horneri water extract corresponded to a protonated ion at m/z 197.1169 (Calcd for C11H17O3, 197.1178) in the positive mode. Based on optical rotation and 1H and 13C NMR spectral data, we identified this peak as corresponding to (–)-loliolide, a monoterpene lactone that was previously isolated from marine natural products such as brown algae and marine microalgae [22]. Based on the content analysis of S. horneri, we further investigated the in vitro anti-inflammatory properties of loliolide, which was presumed to be a major bioactive component of S. horneri.

2.4. Effect of Loliolide on Cell Viability in ARPE-19 Cells

To determine whether loliolide was cytotoxic to ARPE-19 cells, we measured cell viability using the MTT assay. We found that high concentrations of loliolide did not significantly affect the viability of ARPE-19 cells (Figure 4A) nor change their morphology (Figure 4B). Therefore, we used loliolide at a concentration below 400 μM to further evaluate its anti-inflammatory effect as a bioactive compound contained in S. horneri.

2.5. Effect of Loliolide on Inflammation in TNF-α-Stimulated ARPE-19 Cells

We next evaluated the anti-inflammatory effect of loliolide using a TNF-α-induced inflammatory cell model, which could activate the NF-ĸb signaling pathway and mimic aspects of PM-induced RPE inflammation. As shown in Figure 5A, the mRNA levels of chemokine IL-8 were significantly decreased, whereas those of MCP-1 was not changed after loliolide pretreatment of TNF-α-induced ARPE-19 cells. Regarding the mRNA expression of cytokine genes, we detected that the levels of IL-1β and IL-6 were significantly decreased after loliolide pretreatment of TNF-α-induced ARPE-19 cells (Figure 5B).
To further investigate the anti-inflammatory effects of loliolide, we measured the protein levels of chemokines and cytokines using ELISA. We accordingly found a significant upregulation in the production of IL-8 and IL-6 cytokines in TNF-α-induced ARPE-19 cells, which was conversely significantly and dose-dependently reduced by loliolide pretreatment (Figure 6A,B). To better understand the loliolide-induced anti-inflammatory response, we next performed Western blot analyses of proteins related to MAPK and NF-κB signaling, which are critical interconnected inflammatory pathways. We observed that loliolide pretreatment significantly attenuated the phosphorylation of JNK, p38, and NF-κB in TNF-α-treated ARPE-19 cells, whereas it did not affect ERK phosphorylation (Figure 6C,D). We then used the uf-UP-treated ARPE-19 model to extend and validate our previous findings to a TNF-α-induced ARPE-19 inflammation model. According to qRT-PCR results, the upregulated expression of MCP-1, IL-8, and IL-6 after treatment with uf-UPs was significantly decreased by loliolide pretreatment. (Supplementary Figure S4). Based on these results, we suggested that loliolide exhibits anti-inflammatory effects by inhibiting the activation of the NF-κB/MAPK signaling pathway in ARPE-19 cells under inflammatory conditions.

3. Discussion

The pathophysiological progression of retinal disease is closely linked to significant risk factors such as inflammation, which may affect vision and lead to vision loss. Increased ocular inflammation is associated with AMD, diabetic retinopathy (DR), and dry eye [23,24,25]. Because it is difficult to confirm or predict the progress of ocular disease, prevention by means of avoiding contact with toxic substances is important. Due to rapid industrial development, the use of carbon-containing fuels and traffic exhaust has increased, leading to increased PM emissions. Recent studies have reported that PMs can act as potent inducers of inflammatory responses in diverse tissues, including the eyes [26,27,28,29]. PMs generated reactive oxygen species (ROS) and activated stress kinase, such as JNK, whereas treatment of 2-isopropylmalic acid (2-IPMA) alleviated oxidative stress inflammatory response in RPE cells [30]. Sim et al. also showed that PMs enhance inflammation by upregulating the ROS-triggered apoptotic pathways via increases the ratio of Bax/Bcl-2, while sulforaphane treatment reversed these pro-apoptotic changes in ARPE-19 cells [31]. Despite evidence supporting the association between PM-mediated RPE inflammation and candidate for anti-inflammatory potential, it has not been thoroughly investigated, especially regarding the anti-inflammatory properties of natural marine ingredients. In the present study, we investigated the anti-inflammatory efficacy of S. horneri and loliolide, which is a bioactive compound of S. horneri, in uf-UP- and TNF-α-induced retinal cell inflammation.
PMs are classified according to their particle diameters, such as PM 10 (diameter less than 10 μm), PM 2.5 (diameter 2.5 to 10 μm), and PM 0.1 (also known as uf-UPs, diameter less than 0.1 μm), and are considered harmful factors affecting health as they can induce various pathologies, including cardiovascular toxicity, pulmonary diseases, hematological aberration, cutaneous inflammation, and metabolic syndrome [32,33,34,35,36]. Although uf-UPs are less well known than PM 2.5 and PM 10, recent studies have shown that uf-UPs pose a more serious danger to human health because they can be translocated into the bloodstream, spread to other organs, and have thus been positively associated with COPD, cardiovascular diseases, congestive heart failure, preterm birth, asthma, and acute myocardial infarction [37]. Therefore, effective prevention and treatment are required to reduce the health risks of exposure to uf-UPs.
Marine brown algae are an important food source and functional ingredients that are rich in bioactive compounds such as phlorotannins, fucoxanthin, alginic acid, fucoidans, and laminarin [38,39]. S. horneri extracts and its bioactive compounds, including loliolide, have demonstrated beneficial physiological activities such as antioxidant, antisenescence, and anti-inflammatory effects against external stress [40,41,42]. Despite the identification of various beneficial activities of loliolide, no studies have explored its effect on ocular inflammatory diseases, especially uf-UP-induced disorders.
The retinal pigment epithelium (RPE) eliminates external agents through its phagocytic function, while also transporting large amounts of nutrients to the eye [43,44]. In addition, the RPE regulates the activity of immune cells and maintains ocular immune privilege [45]. Owing to these interactions and internal or external stimuli, the RPE is relatively sensitive to stress, especially inflammation. Therefore, the inhibition of inflammatory responses that are upregulated by uf-UPs, one of the various external stimuli, is necessary to reduce the complications associated with long-term chronic inflammation, including ocular disease. In the current study, we found that uf-UPs stimulated inflammation in ARPE-19 cells by enhancing the mRNA expression of proinflammatory chemokines and cytokines, whereas pretreatment with S. horneri extract significantly attenuated the uf-UP-induced inflammation. In addition, mRNA expression analysis showed that the production of MCP-1, IL-8, IL-6, and TNF-α was reduced after pretreatment of uf-UPs-treated cells with S. horneri extracts. Based on previous studies and content analysis using UPLC-QTOF/MS, we confirmed that S. horneri contains loliolide (Figure 3). Therefore, we investigated whether loliolide improves the inflammatory response in ARPE-19 cells. Our results indicated that loliolide treatment decreased the mRNA expression of IL-6 and IL-8 in inflammatory ARPE-19 cells induced by uf-UPs and TNF-α treatment, whereas the effect on MCP1 and IL-1β was only partial (Figure 5 and Supplementary Figure S4). Additionally, in our previous study, we confirmed that fucoidan has an anti-inflammatory effect against inflammation induced by TNF-α in ARPE-19 cells. Compared to these results, loliolide reduced chemokine secretion more than fucoidan [14].

4. Materials and Methods

4.1. Chemicals and Reagents

All chemicals and reagents were obtained from Sigma-Aldrich (St. Louis, MO, USA) unless stated otherwise. Thiazolyl blue tetrazolium bromide (MTT) powder was purchased from Tokyo Chemical Industry Co., Ltd. (Tokyo, Japan). TNF-α was purchased from PeproTech (Rocky Hill, NJ, USA). Loliolide was purchased from ChemFaces (Wuhan, China). The TRIzol reagent and bicinchoninic acid (BCA) protein assay kits were purchased from Thermo Fisher Scientific (Waltham, MA, USA). The MAPK Family Antibody Sampler Kit (cat. no. 9926) and phospho-MAPK Family Antibody Sampler Kit (cat. no. 9910) were purchased from Cell Signaling Technology (Danvers, MA, USA).

4.2. Preparation of S. horneri Powder

S. horneri was collected from the South Sea of Korea, and S. horneri powder preparation was performed as previously described [14]. Briefly, water extraction was performed at 100 °C for 3 h. The extract was further filtered (50 μm) and concentrated using a rotary vacuum evaporator (50 ± 10 °C). After concentration, freeze-drying was performed for approximately 72 h until the moisture content was less than 5%. The obtained powder was analyzed in a UPLC-QTOF/MS instrument (Waters, Milford, MA, USA) and used for further experiments.

4.3. Preparation of uf-UPs

uf-UPs were prepared using a previously reported method [46] with slight modifications. Briefly, 2 g of UPs (NIST1648A, Sigma, St. Louis, MO, USA) were added to 10 mL phosphate-buffered saline (PBS), vortexed for 1 min, sonicated for 45 min using an ultrasonic bath (Power Sonic 510, Hwashin Technology, Seoul, Republic of Korea), and filtered through a 0.22 μm syringe filter (Sartorius, Goettingen, Germany). These UPs were collected from St. Louis, MO, USA, and all constituents are described in the Certificate of Analysis (https://tsapps.nist.gov/srmext/certificates/1648a.pdf, accessed on 20 February 2023.

4.4. Cell Culture

The human RPE cell line ARPE-19 was grown in a 1:1 mixture of Dulbecco’s modified Eagle’s medium/Ham’s F-12 Nutrient Mixture (Welgene, Daegu, Republic of Korea) supplemented with 10% fetal bovine serum (HyClone™, Logan, UT, USA) and antibiotic/antimycotic solution (HyClone™) at 37 °C and in an atmosphere containing 5% CO2.

4.5. Cell Viability Assay

Cell viability was measured using the MTT assay. ARPE-19 cells were seeded into 12-well plates at a density of 1 × 105 cells/well and incubated for 24 h. After incubation, uf-UPs (0.1, 1, and 10 μg/mL) or loliolide (25, 50, 100, 200, and 400 μM) was added at the indicated concentrations, and cells were further incubated for 24 h at 37 °C in humidified air with 5% CO2. The concentration of S. horneri was determined based on previous studies [14]. A stock solution of MTT (5 mg/mL in PBS) was added to each well at a final concentration of 0.5 mg/mL according to the manufacturer’s protocol. After incubation for 2 h at 37 °C, formazan was solubilized in dimethyl sulfoxide (DMSO). Cell viability was measured at 590 nm using a Multiskan™ SkyHigh microplate reader (Thermo Fisher Scientific). The percentage of treated cells exhibiting cytotoxicity was determined relative to that of the control group.

4.6. Instruments and Analytical Conditions

The aqueous extract of S. horneri was chromatographed using a C18 column (Waters BEH C18, 1.7 μm, 2.1 mm × 100 mm) on an ultraperformance liquid chromatography system (UPLC, ACQUITY, Waters) equipped with a Micromass QTOF Premier mass spectrometer (Waters). A solvent system consisting of 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B) was applied at a flow rate of 0.4 mL/min with the gradient method being as follows: 0.0–1.0 min, 10% B; 1.0–4.0 min, 20% B; 4.0–6.0 min, 40% B; 6.0–8.0 min, 85% B; 8.0–12.0 min, 100% B; 12.0–13.4 min, 100% B; 13.4–13.5 min, 10% B, 13.5–15.0 min, 10% B. The temperature of the column oven was kept at 35 °C, and the injection volume was 2 μL. The parameters applied for mass spectrometric analysis were the same as those used in the analysis method described by Jang et al. [47]. NMR spectroscopic data were recorded using a JEOL ECZ500R spectrometer (JEOL, Tokyo, Japan). Optical rotation was measured using a P-2000 polarimeter (JASCO, Tokyo, Japan). Reference chemical (–)-loliolide (CAS No. 5989-02-6) was supplied by the Korea Plant Extract Bank of the Korea Research Institute of Bioscience and Biotechnology (KRIBB, Daejeon, Republic of Korea). The NMR and MS spectroscopic data for (–)-loliolide are provided in the Supplementary data (Figures S1–S3).

4.7. RNA Preparation and Quantitative Real-Time PCR

Total RNA was extracted using the TRIzol® reagent (Thermo Fisher Scientific), according to the manufacturer’s protocol. cDNA was synthesized using a RevertAid RT Reverse Transcription Kit (Thermo Fisher Scientific). Quantitative real-time PCR (real-time qPCR) was performed using the 2× GreenStar™ qPCR Master Mix (Bioneer, Daejeon, Korea), according to the manufacturer’s instructions. A total of 18S rRNA and GAPDH were used as reference genes for the normalization of all samples, and the levels of gene expression were calculated using a previously reported method [48]. All primers used are listed in Table S1.

4.8. Measurement of Cytokine and Chemokine Production

The amount of secreted cytokines in cell culture supernatants was determined using enzyme-linked immunosorbent assays (ELISA). The content of IL-6 (cat. no. ab178013) and IL-8 (cat. no. ab214030) in the supernatant was measured using relevant ELISA kits according to the manufacturer’s instructions (Abcam, Cambridge, UK).

4.9. Western Blot Analysis

To extract total protein, ARPE-19 cells were washed with cold PBS and lysed using a radioimmunoprecipitation assay (RIPA) buffer containing a protease and phosphatase inhibitor cocktail. The concentration of extracted proteins was measured using a BCA protein assay kit (Thermo Fisher Scientific) according to the manufacturer’s protocol. Samples of extracted total protein (20 μg/mL) were separated on 8% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gels and transferred to polyvinylidene difluoride (PVDF) membranes. Membranes were blocked with 3% BSA or 5% skim milk for 1 h at room temperature. Membranes were then incubated with primary antibodies at 4 °C overnight. Primary antibodies included antibodies against p-JNK, JNK, p-p38, p-38, p-ERK, ERK, p-NF-κB, and NF-κB. After incubation, membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h at 25 °C. Protein bands were detected using SuperSignal™ West Pico PLUS Chemiluminescent Substrate (Thermo Fisher Scientific) and photographed under an iBright™ CL750 Imaging System (Thermo Fisher Scientific). GAPDH was used as the protein-loading control.

4.10. Statistical Analysis

All experiments were performed independently at least three times, and data were analyzed using GraphPad Prism 7.0 (San Diego, CA, USA). Data were expressed as the mean ± standard error of the mean. Statistical analyses were performed using Student’s t-test (for two-group comparisons) or one-way analysis of variance (for multiple-group comparisons). For all comparisons, a p-value < 0.05 was considered statistically significant.

5. Conclusions

In summary, this study suggested that S. horneri extract reduces the levels of proinflammatory markers in uf-UP-stimulated ARPE-19 cells. In a TNF-α-induced inflammation model, S. horneri extracts significantly alleviated proinflammatory phenotypes as indicated by the levels of mRNA expression and production of chemokines and cytokines via downregulating the MAPK/NF-ĸB signaling pathway. Likewise, loliolide treatment showed significant anti-inflammatory effects on uf-UP- and TNF-α-stimulated inflammation in ARPE-19 cells. Therefore, the present study demonstrated that S. horneri extract can be a good functional ingredient for alleviating retinal inflammatory diseases caused by external stimuli.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25010162/s1.

Author Contributions

E.J.L., S.L. and W.Y.: conceptualization, methodology, investigation, data curation, visualization, formal analysis, writing—original draft, and preparation. H.-J.J.: validation and formal analysis, writing—original draft, and preparation. W.Y. wrote the manuscript—reviewing, editing, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Korea Institute of Marine Science and Technology Promotion (KIMST) funded by the Ministry of Oceans and Fisheries (20210376).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

We thank Sei-Ryang Oh (Natural Product Research Center, Korea Research Institute of Bioscience and Biotechnology, Republic of Korea) for scientific advices and Junghyun Kim (Jeonbuk National University, Jeonju, Republic of Korea) for providing the human RPE cell line, ARPE-19.

Conflicts of Interest

E.J.L. and S.L. were employed by AceBiome Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Kwon, H.S.; Ryu, M.H.; Carlsten, C. Ultrafine particles: Unique physicochemical properties relevant to health and disease. Exp. Mol. Med. 2020, 52, 318–328. [Google Scholar] [CrossRef] [PubMed]
  2. Halonen, J.I.; Lanki, T.; Yli-Tuomi, T.; Kulmala, M.; Tiittanen, P.; Pekkanen, J. Urban air pollution, and asthma and COPD hospital emergency room visits. Thorax 2008, 63, 635–641. [Google Scholar] [CrossRef]
  3. Heusinkveld, H.J.; Wahle, T.; Campbell, A.; Westerink, R.H.S.; Tran, L.; Johnston, H.; Stone, V.; Cassee, F.R.; Schins, R.P.F. Neurodegenerative and neurological disorders by small inhaled particles. Neurotoxicology 2016, 56, 94–106. [Google Scholar] [CrossRef]
  4. Chu, C.; Zhang, H.; Cui, S.; Han, B.; Zhou, L.; Zhang, N.; Su, X.; Niu, Y.; Chen, W.; Chen, R.; et al. Ambient PM2.5 caused depressive-like responses through Nrf2/NLRP3 signaling pathway modulating inflammation. J. Hazard. Mater. 2019, 369, 180–190. [Google Scholar] [CrossRef] [PubMed]
  5. Mimura, T.; Ichinose, T.; Yamagami, S.; Fujishima, H.; Kamei, Y.; Goto, M.; Takada, S.; Matsubara, M. Airborne particulate matter (PM2.5) and the prevalence of allergic conjunctivitis in Japan. Sci. Total Environ. 2014, 487, 493–499. [Google Scholar] [CrossRef] [PubMed]
  6. Nwanaji-Enwerem, J.C.; Wang, W.; Nwanaji-Enwerem, O.; Vokonas, P.; Baccarelli, A.; Weisskopf, M.; Herndon, L.W.; Wiggs, J.L.; Park, S.K.; Schwartz, J. Association of Long-term Ambient Black Carbon Exposure and Oxidative Stress Allelic Variants with Intraocular Pressure in Older Men. JAMA Ophthalmol. 2019, 137, 129–137. [Google Scholar] [CrossRef] [PubMed]
  7. Chua, S.Y.L.; Khawaja, A.P.; Dick, A.D.; Morgan, J.; Dhillon, B.; Lotery, A.J.; Strouthidis, N.G.; Reisman, C.; Peto, T.; Khaw, P.T.; et al. Ambient Air Pollution Associations with Retinal Morphology in the UK Biobank. Investig. Ophthalmol. Vis. Sci. 2020, 61, 32. [Google Scholar] [CrossRef]
  8. Tan, G.; Li, J.; Yang, Q.; Wu, A.; Qu, D.Y.; Wang, Y.; Ye, L.; Bao, J.; Shao, Y. Air pollutant particulate matter 2.5 induces dry eye syndrome in mice. Sci. Rep. 2018, 8, 17828. [Google Scholar] [CrossRef]
  9. Komatsu, T.; Fukuda, M.; Mikami, A.; Mizuno, S.; Kantachumpoo, A.; Tanoue, H.; Kawamiya, M. Possible change in distribution of seaweed, Sargassum horneri, in northeast Asia under A2 scenario of global warming and consequent effect on some fish. Mar. Pollut. Bull 2014, 85, 317–324. [Google Scholar] [CrossRef]
  10. Jayawardena, T.U.; Kim, H.-S.; Sanjeewa, K.A.; Kim, S.-Y.; Rho, J.-R.; Jee, Y.; Ahn, G.; Jeon, Y.-J. Sargassum horneri and isolated 6-hydroxy-4,4,7a-trimethyl-5,6,7,7a-tetrahydrobenzofuran-2(4H)-one (HTT); LPS-induced inflammation attenuation via suppressing NF-κB, MAPK and oxidative stress through Nrf2/HO-1 pathways in RAW 264.7 macrophages. Algal. Res. 2019, 40, 101513. [Google Scholar] [CrossRef]
  11. Han, E.J.; Kim, S.Y.; Han, H.J.; Kim, H.S.; Kim, K.N.; Fernando, I.P.S.; Madusanka, D.M.D.; Dias, M.; Cheong, S.H.; Park, S.R.; et al. UVB protective effects of Sargassum horneri through the regulation of Nrf2 mediated antioxidant mechanism. Sci. Rep. 2021, 11, 9963. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, H.-S.; Han, E.J.; Fernando, I.P.S.; Sanjeewa, K.K.A.; Jayawardena, T.U.; Kim, H.-J.; Jee, Y.; Kang, S.-H.; Jang, J.-H.; Jang, J.-P. Anti-allergy effect of mojabanchromanol isolated from Sargassum horneri in bone marrow-derived cultured mast cells. Algal. Res. 2020, 48, 101898. [Google Scholar] [CrossRef]
  13. Sanjeewa, K.; Jayawardena, T.U.; Kim, H.-S.; Kim, S.-Y.; Ahn, G.; Kim, H.-J.; Fu, X.; Jee, Y.; Jeon, Y.-J. Ethanol extract separated from Sargassum horneri (Turner) abate LPS-induced inflammation in RAW 264.7 macrophages. Fish. Aquat. Sci. 2019, 22, 6. [Google Scholar] [CrossRef]
  14. Lee, S.; Lee, E.J.; Lee, G.M.; Yun, J.H.; Yoo, W. Inhibitory effect of fucoidan on TNF-alpha-induced inflammation in human retinal pigment epithelium cells. Front. Nutr. 2023, 10, 1162934. [Google Scholar] [CrossRef] [PubMed]
  15. Saraswati; Giriwono, P.E.; Iskandriati, D.; Tan, C.P.; Andarwulan, N. Sargassum Seaweed as a Source of Anti-Inflammatory Substances and the Potential Insight of the Tropical Species: A Review. Mar. Drugs 2019, 17, 590. [Google Scholar] [CrossRef] [PubMed]
  16. Yang, X.; Kang, M.-C.; Lee, K.-W.; Kang, S.-M.; Lee, W.-W.; Jeon, Y.-J. Antioxidant activity and cell protective effect of loliolide isolated from Sargassum ringgoldianum subsp. coreanum. Algae 2011, 26, 201–208. [Google Scholar] [CrossRef]
  17. Cho, D.H.; Yun, J.H.; Heo, J.; Lee, I.K.; Lee, Y.J.; Bae, S.; Yun, B.S.; Kim, H.S. Identification of Loliolide with Anti-Aging Properties from Scenedesmus deserticola JD052. J. Microbiol. Biotechnol. 2023, 33, 1250–1256. [Google Scholar] [CrossRef]
  18. Park, S.H.; Kim, D.S.; Kim, S.; Lorz, L.R.; Choi, E.; Lim, H.Y.; Hossain, M.A.; Jang, S.; Choi, Y.I.; Park, K.J.; et al. Loliolide Presents Antiapoptosis and Antiscratching Effects in Human Keratinocytes. Int. J. Mol. Sci. 2019, 20, 651. [Google Scholar] [CrossRef]
  19. Sanjeewa, K.K.A.; Jayawardena, T.U.; Kim, S.Y.; Lee, H.G.; Je, J.G.; Jee, Y.; Jeon, Y.J. Sargassum horneri (Turner) inhibit urban particulate matter-induced inflammation in MH-S lung macrophages via blocking TLRs mediated NF-kappaB and MAPK activation. J. Ethnopharmacol. 2020, 249, 112363. [Google Scholar] [CrossRef]
  20. Jee, Y.; Kim, A.; Herath, K.; Kim, H.J.; Mihindukulasooriya, S.; Ko, M.-O.; Jeon, Y.-J. Sargassum horneri inhibits particulate matter-induced apoptotic cell death in splenocytes. J. Immunol. 2019, 202, 187.31. [Google Scholar] [CrossRef]
  21. Lee, H.S.; Han, S.; Seo, J.W.; Jeon, K.J. Exposure to Traffic-Related Particulate Matter 2.5 Triggers Th2-Dominant Ocular Immune Response in a Murine Model. Int. J. Environ. Res. Public. Health 2020, 17, 2965. [Google Scholar] [CrossRef] [PubMed]
  22. Chi, H.; Qi, X.; Wang, X.; Wang, Y.; Han, X.; Wang, J.; Wang, H. Preparative separation and purification of loliolide and epiloliolide from Ascophyllum nodosum using amine-based microporous organic polymer for solid phase extraction coupled with macroporous resin and prep-HPLC. Anal. Methods 2021, 13, 1939–1944. [Google Scholar] [CrossRef] [PubMed]
  23. Tan, W.; Zou, J.; Yoshida, S.; Jiang, B.; Zhou, Y. The Role of Inflammation in Age-Related Macular Degeneration. Int. J. Biol. Sci. 2020, 16, 2989–3001. [Google Scholar] [CrossRef] [PubMed]
  24. Rubsam, A.; Parikh, S.; Fort, P.E. Role of Inflammation in Diabetic Retinopathy. Int. J. Mol. Sci. 2018, 19, 942. [Google Scholar] [CrossRef] [PubMed]
  25. Yamaguchi, T. Inflammatory Response in Dry Eye. Investig. Ophthalmol. Vis. Sci. 2018, 59, DES192–DES199. [Google Scholar] [CrossRef] [PubMed]
  26. Arias-Perez, R.D.; Taborda, N.A.; Gomez, D.M.; Narvaez, J.F.; Porras, J.; Hernandez, J.C. Inflammatory effects of particulate matter air pollution. Env. Sci. Pollut. Res. Int. 2020, 27, 42390–42404. [Google Scholar] [CrossRef] [PubMed]
  27. Wu, W.; Jin, Y.; Carlsten, C. Inflammatory health effects of indoor and outdoor particulate matter. J. Allergy. Clin. Immunol. 2018, 141, 833–844. [Google Scholar] [CrossRef] [PubMed]
  28. Yang, Q.; Li, K.; Li, D.; Zhang, Y.; Liu, X.; Wu, K. Effects of fine particulate matter on the ocular surface: An in vitro and in vivo study. Biomed. Pharmacother. 2019, 117, 109177. [Google Scholar] [CrossRef]
  29. Li, L.; Xing, C.; Zhou, J.; Niu, L.; Luo, B.; Song, M.; Niu, J.; Ruan, Y.; Sun, X.; Lei, Y. Airborne particulate matter (PM2.5) triggers ocular hypertension and glaucoma through pyroptosis. Part Fibre Toxicol. 2021, 18, 10. [Google Scholar] [CrossRef]
  30. Choi, J.Y.; Bae, J.E.; Kim, J.B.; Jo, D.S.; Park, N.Y.; Kim, Y.H.; Lee, H.J.; Kim, S.H.; Kim, S.H.; Jeon, H.B.; et al. 2-IPMA Ameliorates PM2.5-Induced Inflammation by Promoting Primary Ciliogenesis in RPE Cells. Molecules 2021, 26, 5409. [Google Scholar] [CrossRef]
  31. Sim, H.; Lee, W.; Choo, S.; Park, E.K.; Baek, M.C.; Lee, I.K.; Park, D.H.; Bae, J.S. Sulforaphane Alleviates Particulate Matter-Induced Oxidative Stress in Human Retinal Pigment Epithelial Cells. Front. Med. 2021, 8, 685032. [Google Scholar] [CrossRef] [PubMed]
  32. Du, Y.; Xu, X.; Chu, M.; Guo, Y.; Wang, J. Air particulate matter and cardiovascular disease: The epidemiological, biomedical and clinical evidence. J. Thorac. Dis. 2016, 8, E8–E19. [Google Scholar] [CrossRef] [PubMed]
  33. Guo, C.; Zhang, Z.; Lau, A.K.H.; Lin, C.Q.; Chuang, Y.C.; Chan, J.; Jiang, W.K.; Tam, T.; Yeoh, E.K.; Chan, T.C.; et al. Effect of long-term exposure to fine particulate matter on lung function decline and risk of chronic obstructive pulmonary disease in Taiwan: A longitudinal, cohort study. Lancet Planet. Health 2018, 2, e114–e125. [Google Scholar] [CrossRef] [PubMed]
  34. Fongsodsri, K.; Chamnanchanunt, S.; Desakorn, V.; Thanachartwet, V.; Sahassananda, D.; Rojnuckarin, P.; Umemura, T. Particulate Matter 2.5 and Hematological Disorders From Dust to Diseases: A Systematic Review of Available Evidence. Front. Med. 2021, 8, 692008. [Google Scholar] [CrossRef]
  35. Jin, S.P.; Li, Z.; Choi, E.K.; Lee, S.; Kim, Y.K.; Seo, E.Y.; Chung, J.H.; Cho, S. Urban particulate matter in air pollution penetrates into the barrier-disrupted skin and produces ROS-dependent cutaneous inflammatory response in vivo. J. Dermatol. Sci. 2018, 91, 175–183. [Google Scholar] [CrossRef]
  36. Lee, S.; Park, H.; Kim, S.; Lee, E.K.; Lee, J.; Hong, Y.S.; Ha, E. Fine particulate matter and incidence of metabolic syndrome in non-CVD patients: A nationwide population-based cohort study. Int. J. Hyg. Environ. Health 2019, 222, 533–540. [Google Scholar] [CrossRef]
  37. Abdillah, S.F.I.; Wang, Y.F. Ambient ultrafine particle (PM0.1): Sources, characteristics, measurements and exposure implications on human health. Environ. Res. 2023, 218, 115061. [Google Scholar] [CrossRef]
  38. Li, Y.; Zheng, Y.; Zhang, Y.; Yang, Y.; Wang, P.; Imre, B.; Wong, A.C.Y.; Hsieh, Y.S.Y.; Wang, D. Brown Algae Carbohydrates: Structures, Pharmaceutical Properties, and Research Challenges. Mar. Drugs 2021, 19, 620. [Google Scholar] [CrossRef]
  39. Silva, A.; Cassani, L.; Grosso, C.; Garcia-Oliveira, P.; Morais, S.L.; Echave, J.; Carpena, M.; Xiao, J.; Barroso, M.F.; Simal-Gandara, J.; et al. Recent advances in biological properties of brown algae-derived compounds for nutraceutical applications. Crit. Rev. Food Sci. Nutr. 2022, 2022, 2115004. [Google Scholar] [CrossRef]
  40. Fernando, I.P.S.; Heo, S.J.; Dias, M.; Madusanka, D.M.D.; Han, E.J.; Kim, M.J.; Sanjeewa, K.K.A.; Lee, K.; Ahn, G. (-)-Loliolide Isolated from Sargassum horneri Abate UVB-Induced Oxidative Damage in Human Dermal Fibroblasts and Subside ECM Degradation. Mar. Drugs 2021, 19, 435. [Google Scholar] [CrossRef]
  41. Yang, H.H.; Hwangbo, K.; Zheng, M.S.; Cho, J.H.; Son, J.K.; Kim, H.Y.; Baek, S.H.; Choi, H.C.; Park, S.Y.; Kim, J.R. Inhibitory effects of (−)-loliolide on cellular senescence in human dermal fibroblasts. Arch. Pharm. Res. 2015, 38, 876–884. [Google Scholar] [CrossRef] [PubMed]
  42. Han, E.J.; Fernando, I.P.S.; Kim, H.S.; Lee, D.S.; Kim, A.; Je, J.G.; Seo, M.J.; Jee, Y.H.; Jeon, Y.J.; Kim, S.Y.; et al. (-)-Loliolide Isolated from Sargassum horneri Suppressed Oxidative Stress and Inflammation by Activating Nrf2/HO-1 Signaling in IFN-gamma/TNF-alpha-Stimulated HaCaT Keratinocytes. Antioxidants 2021, 10, 856. [Google Scholar] [CrossRef] [PubMed]
  43. Qin, S.; Rodrigues, G.A. Progress and perspectives on the role of RPE cell inflammatory responses in the development of age-related macular degeneration. J. Inflamm. Res. 2008, 1, 49–65. [Google Scholar] [CrossRef] [PubMed]
  44. Kwon, W.; Freeman, S.A. Phagocytosis by the Retinal Pigment Epithelium: Recognition, Resolution, Recycling. Front Immunol. 2020, 11, 604205. [Google Scholar] [CrossRef] [PubMed]
  45. Taylor, A.W.; Hsu, S.; Ng, T.F. The Role of Retinal Pigment Epithelial Cells in Regulation of Macrophages/Microglial Cells in Retinal Immunobiology. Front. Immunol. 2021, 12, 724601. [Google Scholar] [CrossRef]
  46. Kim, J.Y.; Kim, J.H.; Kim, Y.D.; Seo, J.H. High Vulnerability of Oligodendrocytes to Oxidative Stress Induced by Ultrafine Urban Particles. Antioxidants 2020, 10, 4. [Google Scholar] [CrossRef]
  47. Jang, H.-J.; Kim, W.J.; Lee, S.U.; Kim, M.-O.; Park, M.H.; Song, S.; Kim, D.-Y.; Lee, S.M.; Yuk, H.J.; Lee, D.Y. Optimization of chiisanoside and chiisanogenin isolation from Eleutherococcus sessiliflorus (Rupr. & Maxim.) leaves for industrial application: A pilot study. Ind. Crops Prod. 2022, 185, 115099. [Google Scholar]
  48. Lee, S.; Park, H.O.; Yoo, W. Anti-Melanogenic and Antioxidant Effects of Cell-Free Supernatant from Lactobacillus gasseri BNR17. Microorganisms 2022, 10, 788. [Google Scholar] [CrossRef]
Figure 1. Cell viability and inflammation in uf-UP-treated ARPE-19 cells. ARPE-19 cells were treated with uf-UPs (0.1, 1, and 10 μg/mL). (A) Cell viability and (B) cell morphology 24 h after treatment. (C) Levels of mRNA expression of chemokines and cytokines in uf-UP-stimulated ARPE-19 cells. ARPE-19 cells were treated with uf-UPs (0.1, 1, and 10 μg/mL) for 1 h. Values represent the mean ± SD of three independent experiments. * p < 0.05 and ** p < 0.01 compared with the control.
Figure 1. Cell viability and inflammation in uf-UP-treated ARPE-19 cells. ARPE-19 cells were treated with uf-UPs (0.1, 1, and 10 μg/mL). (A) Cell viability and (B) cell morphology 24 h after treatment. (C) Levels of mRNA expression of chemokines and cytokines in uf-UP-stimulated ARPE-19 cells. ARPE-19 cells were treated with uf-UPs (0.1, 1, and 10 μg/mL) for 1 h. Values represent the mean ± SD of three independent experiments. * p < 0.05 and ** p < 0.01 compared with the control.
Ijms 25 00162 g001
Figure 2. Effect of S. horneri on mRNA expression of inflammation-related genes in uf-UP-stimulated ARPE-19 cells. ARPE-19 cells were pretreated with S. horneri (10 and 100 μg/mL) for 1 h and then treated with 10 μg/mL uf-UPs for 1 h. Levels of mRNA expression of (A) chemokines MCP-1 and IL-8 and (B) cytokines IL-1β, IL-6, and TNF-α. Levels of mRNA expression were analyzed by RT-PCR. Values represent the mean ± SD of three independent experiments. # p < 0.05, ## p < 0.01, and ### p < 0.001 compared with the control. * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with the uf-UP-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Figure 2. Effect of S. horneri on mRNA expression of inflammation-related genes in uf-UP-stimulated ARPE-19 cells. ARPE-19 cells were pretreated with S. horneri (10 and 100 μg/mL) for 1 h and then treated with 10 μg/mL uf-UPs for 1 h. Levels of mRNA expression of (A) chemokines MCP-1 and IL-8 and (B) cytokines IL-1β, IL-6, and TNF-α. Levels of mRNA expression were analyzed by RT-PCR. Values represent the mean ± SD of three independent experiments. # p < 0.05, ## p < 0.01, and ### p < 0.001 compared with the control. * p < 0.05, ** p < 0.01, and *** p < 0.001 compared with the uf-UP-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Ijms 25 00162 g002
Figure 3. UPLC-QTOF/MS analysis. (A) Chemical structure of (–)-loliolide and (B) UPLC-QTOF/MS chromatogram of S. horneri aqueous extracts. Red arrow: BPI (Base Peak Intensity).
Figure 3. UPLC-QTOF/MS analysis. (A) Chemical structure of (–)-loliolide and (B) UPLC-QTOF/MS chromatogram of S. horneri aqueous extracts. Red arrow: BPI (Base Peak Intensity).
Ijms 25 00162 g003
Figure 4. Cell viability and morphology in loliolide-treated ARPE-19 cells. ARPE-19 cells were treated with various concentrations of loliolide. (A) Cell viability and (B) cell morphology 24 h after treatment. Values represent the mean ± SD of three independent experiments.
Figure 4. Cell viability and morphology in loliolide-treated ARPE-19 cells. ARPE-19 cells were treated with various concentrations of loliolide. (A) Cell viability and (B) cell morphology 24 h after treatment. Values represent the mean ± SD of three independent experiments.
Ijms 25 00162 g004
Figure 5. Effect of loliolide on mRNA expression of genes related to inflammation in TNF-α-treated ARPE-19 cells. ARPE-19 cells were pretreated with loliolide (50, 100, and 200 μg/mL) for 1 h and then treated with 50 ng/mL TNF-α for 4 h. Levels of mRNA expression of (A) chemokines MCP-1 and IL-8 and (B) cytokines IL-1β and IL-6. Levels of mRNA expression were analyzed by RT-PCR. Values represent the mean ± SD of three independent experiments. # p < 0.05 and ### p < 0.001 compared with the control. * p < 0.05 and ** p < 0.01 compared with the TNF-α-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Figure 5. Effect of loliolide on mRNA expression of genes related to inflammation in TNF-α-treated ARPE-19 cells. ARPE-19 cells were pretreated with loliolide (50, 100, and 200 μg/mL) for 1 h and then treated with 50 ng/mL TNF-α for 4 h. Levels of mRNA expression of (A) chemokines MCP-1 and IL-8 and (B) cytokines IL-1β and IL-6. Levels of mRNA expression were analyzed by RT-PCR. Values represent the mean ± SD of three independent experiments. # p < 0.05 and ### p < 0.001 compared with the control. * p < 0.05 and ** p < 0.01 compared with the TNF-α-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Ijms 25 00162 g005
Figure 6. Effect of loliolide on cytokine production and MAPK/NF-κB signaling in TNF-α-treated ARPE-19 cells. ARPE-19 cells were pretreated with loliolide (100, 200, and 400 μM) for 1 h and then treated with 50 ng/mL TNF-α for 24 h. (A) Content of chemokine IL-8 and (B) cytokine IL-6. Phosphorylation of proteins in the (C) MAPK and (D) NF-κB signaling pathways. ARPE-19 cells were pretreated with loliolide (200 and 400 μM) for 1 h and then stimulated with 50 ng/mL TNF-α for 30 min. GAPDH was used as a loading control. Values represent the mean ± SD of three independent experiments. # p < 0.05, ### p < 0.001 compared with the control. * p < 0.05, ** p < 0.01 and *** p < 0.001 compared with the TNF-α-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Figure 6. Effect of loliolide on cytokine production and MAPK/NF-κB signaling in TNF-α-treated ARPE-19 cells. ARPE-19 cells were pretreated with loliolide (100, 200, and 400 μM) for 1 h and then treated with 50 ng/mL TNF-α for 24 h. (A) Content of chemokine IL-8 and (B) cytokine IL-6. Phosphorylation of proteins in the (C) MAPK and (D) NF-κB signaling pathways. ARPE-19 cells were pretreated with loliolide (200 and 400 μM) for 1 h and then stimulated with 50 ng/mL TNF-α for 30 min. GAPDH was used as a loading control. Values represent the mean ± SD of three independent experiments. # p < 0.05, ### p < 0.001 compared with the control. * p < 0.05, ** p < 0.01 and *** p < 0.001 compared with the TNF-α-treated control, respectively. +; treated TNF-α, −; non-treated TNF-α or loliolide.
Ijms 25 00162 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lee, E.J.; Lee, S.; Jang, H.-J.; Yoo, W. Loliolide in Sargassum horneri Alleviates Ultrafine Urban Particulate Matter (PM 0.1)-Induced Inflammation in Human RPE Cells. Int. J. Mol. Sci. 2024, 25, 162. https://doi.org/10.3390/ijms25010162

AMA Style

Lee EJ, Lee S, Jang H-J, Yoo W. Loliolide in Sargassum horneri Alleviates Ultrafine Urban Particulate Matter (PM 0.1)-Induced Inflammation in Human RPE Cells. International Journal of Molecular Sciences. 2024; 25(1):162. https://doi.org/10.3390/ijms25010162

Chicago/Turabian Style

Lee, Eun Jeoung, Sol Lee, Hyun-Jae Jang, and Wonbeak Yoo. 2024. "Loliolide in Sargassum horneri Alleviates Ultrafine Urban Particulate Matter (PM 0.1)-Induced Inflammation in Human RPE Cells" International Journal of Molecular Sciences 25, no. 1: 162. https://doi.org/10.3390/ijms25010162

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop