Exposure to Polypropylene Microplastics via Oral Ingestion Induces Colonic Apoptosis and Intestinal Barrier Damage through Oxidative Stress and Inflammation in Mice
Abstract
:1. Introduction
2. Materials and Methods
2.1. Characterization of PP Particles and Suspension Preparation
2.2. Animals and Experimental Design
2.3. Histopathological Examination and Electron Microscopy Analysis
2.4. Alcian Blue/Periodic Acid–Schiff (AB-PAS) Staining
2.5. Immunohistochemical Analysis
2.6. Detection of Oxidative Stress Markers
2.7. Enzyme-Linked Immunosorbent Assay (ELISA)
2.8. Apoptosis Detection in the Colon
2.9. Western Blot Analysis
2.10. Statistical Analysis
3. Results
3.1. Polypropylene Microplastic Exposure Changes Colonic Histopathology and Ultrastructure
3.2. Polypropylene Microplastic Exposure Induces Colonic Oxidative Stress and Inflammation
3.3. Polypropylene Microplastic Exposure Activates the TLR4/NF-κB Signaling Pathway
3.4. Polypropylene Microplastic Exposure Destroys the Intestinal Mucosal Barrier in Mice
3.5. Polypropylene Microplastic Exposure Promotes Apoptosis of Colonic Cells
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Geyer, R.; Jambeck, J.R.; Law, K.L. Production, use, and fate of all plastics ever made. Sci. Adv. 2017, 3, e1700782. [Google Scholar] [CrossRef] [PubMed]
- Zarus, G.M.; Muianga, C.; Hunter, C.M.; Pappas, R.S. A review of data for quantifying human exposures to micro and nanoplastics and potential health risks. Sci. Total Environ. 2021, 756, 144010. [Google Scholar] [CrossRef] [PubMed]
- Aragaw, T.A. Surgical face masks as a potential source for microplastic pollution in the COVID-19 scenario. Mar. Pollut. Bull. 2020, 159, 111517. [Google Scholar] [CrossRef] [PubMed]
- Fadare, O.O.; Okoffo, E.D. COVID-19 face masks: A potential source of microplastic fibers in the environment. Sci. Total Environ. 2020, 737, 140279. [Google Scholar] [CrossRef]
- Saadat, S.; Rawtani, D.; Hussain, C.M. Environmental perspective of COVID-19. Sci. Total Environ. 2020, 728, 138870. [Google Scholar] [CrossRef]
- Thompson, R.C.; Olsen, Y.; Mitchell, R.P.; Davis, A.; Rowland, S.J.; John, A.W.G.; McGonigle, D.; Russell, A.E. Lost at sea: Where is all the plastic? Science 2004, 304, 838. [Google Scholar] [CrossRef]
- Courtney, A.; Baker, J.; Bamford, H. (Eds.) Proceedings of the International ReseArch. Workshop on the Occurrence, Effects, and Fate of Microplastic Marine Debris, September 9–11, 2008, University of Washington Tacoma, Tacoma, WA, USA; NOAA Technical Memorandum NOS-OR&R-30; NOAA: Silver Spring, MD, USA, 2009. [Google Scholar]
- EFSA Panel on Contaminants in the Food Chain (CONTAM). Presence of microplastics and nanoplastics in food, with particular focus on seafood. EFSA J. 2016, 14, e04501. [Google Scholar] [CrossRef]
- He, D.; Chen, X.; Zhao, W.; Zhu, Z.; Qi, X.; Zhou, L.; Chen, W.; Wan, C.; Li, D.; Zou, X.; et al. Microplastics contamination in the surface water of the Yangtze River from upstream to estuary based on different sampling methods. Environ. Res. 2021, 196, 110908. [Google Scholar] [CrossRef]
- Lindeque, P.K.; Cole, M.; Coppock, R.L.; Lewis, C.N.; Miller, R.Z.; Watts, A.J.R.; Wilson-McNeal, A.; Wright, S.L.; Galloway, T.S. Are we underestimating microplastic abundance in the marine environment? A comparison of microplastic capture with nets of different mesh-size. Environ. Pollut. 2020, 265, 114721. [Google Scholar] [CrossRef]
- Cheng, Y.; Song, W.; Tian, H.; Zhang, K.; Li, B.; Du, Z.; Zhang, W.; Wang, J.; Wang, J.; Zhu, L. The Effects of High-Density Polyethylene and Polypropylene Microplastics on the Soil and Earthworm Metaphire Guillelmi Gut Microbiota. Chemosphere 2021, 267, 129219. [Google Scholar] [CrossRef]
- Zhu, B.K.; Fang, Y.M.; Zhu, D.; Christie, P.; Ke, X.; Zhu, Y.G.; Christie, X.K.; Zhu, Y.G. Exposure to Nanoplastics Disturbs the Gut Microbiome in the Soil Oligochaete Enchytraeus Crypticus. Environ. Pollut. 2018, 239, 408–415. [Google Scholar] [CrossRef] [PubMed]
- Kelly, F.J.; Fussell, J.C. Toxicity of Airborne Particles-Established Evidence, Knowledge Gaps and Emerging Areas of Importance. Philos. Trans. A Math. Phys. Eng. Sci. 2020, 378, 20190322. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Zhao, Y.; Du, F.; Cai, H.; Wang, G.; Shi, H. Microplastic fallout in different indoor environments. Environ. Sci. Technol. 2020, 54, 6530–6539. [Google Scholar] [CrossRef] [PubMed]
- Sridharan, S.; Kumar, M.; Singh, L.; Bolan, N.S.; Saha, M. Microplastics as an emerging source of particulate air pollution: A critical review. J. Hazard. Mater. 2021, 418, 126245. [Google Scholar] [CrossRef] [PubMed]
- Kosuth, M.; Mason, S.A.; Wattenberg, E.V. Anthropogenic contamination of tap water, beer, and sea salt. PLoS ONE 2018, 13, e0194970. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Lin, T.; Chen, W. Occurrence and removal of microplastics in an advanced drinking water treatment plant (ADWTP). Sci. Total Environ. 2020, 700, 134520. [Google Scholar] [CrossRef] [PubMed]
- Tong, H.; Jiang, Q.; Hu, X.; Zhong, X. Occurrence and identification of microplastics in tap water from China. Chemosphere 2020, 252, 126493. [Google Scholar] [CrossRef] [PubMed]
- Koelmans, A.A.; Mohamed Nor, N.H.; Hermsen, E.; Kooi, M.; Mintenig, S.M.; De France, J. Microplastics in freshwaters and drinking water: Critical review and assessment of data quality. Water Res. 2019, 155, 410–422. [Google Scholar] [CrossRef] [PubMed]
- Oßmann, B.E. Microplastics in Drinking Water? Present State of Knowledge and Open Questions. Curr. Opin. Food Sci. 2021, 41, 44–51. [Google Scholar] [CrossRef]
- Liebezeit, G.; Liebezeit, E. Non-pollen particulates in honey and sugar. Food Addit. Contam. Part A Chem. Anal. Control Exp. Risk Assess 2013, 30, 2136–2140. [Google Scholar] [CrossRef]
- Liebezeit, G.; Liebezeit, E. Origin of synthetic particles in honeys. Pol. J. Food Nutr. Sci. 2015, 65, 143–147. [Google Scholar] [CrossRef]
- Zhu, J.; Zhang, Q.; Li, Y.; Tan, S.; Kang, Z.; Yu, X.; Lan, W.; Cai, L.; Wang, J.; Shi, H. Microplastic pollution in the Maowei Sea, a typical mariculture bay of China. Sci. Total Environ. 2019, 658, 62–68. [Google Scholar] [CrossRef] [PubMed]
- Yang, D.; Shi, H.; Li, L.; Li, J.; Jabeen, K.; Kolandhasamy, P. Microplastic pollution in table salts from China. Environ. Sci. Technol. 2015, 49, 13622–13627. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Kunz, A.; Shim, W.J.; Walther, B.A. Microplastic contamination of table salts from Taiwan, including a global review. Sci. Rep. 2019, 9, 10145. [Google Scholar] [CrossRef] [PubMed]
- Zolotova, N.; Kosyreva, A.; Dzhalilova, D.; Fokichev, N.; Makarova, O. Harmful effects of the microplastic pollution on animal health: A literature review. PeerJ 2022, 10, e13503. [Google Scholar] [CrossRef] [PubMed]
- Schwabl, P.; Köppel, S.; Königshofer, P.; Bucsics, T.; Trauner, M.; Reiberger, T.; Liebmann, B. Detection of various microplastics in human stool: A prospective case series. Ann. Intern. Med. 2019, 171, 453–457. [Google Scholar] [CrossRef]
- Zhang, N.; Li, Y.B.; He, H.R.; Zhang, J.F.; Ma, G.S. You are what you eat: Microplastics in the feces of young men living in Beijing. Sci. Total Environ. 2021, 767, 144345. [Google Scholar] [CrossRef]
- Zhang, J.; Wang, L.; Trasande, L.; Kannan, K. Occurrence of polyethylene terephthalate and polycarbonate microplastics in infant and adult feces. Environ. Sci. Technol. Lett. 2021, 8, 989–994. [Google Scholar] [CrossRef]
- Ragusa, A.; Svelato, A.; Santacroce, C.; Catalano, P.; Notarstefano, V.; Carnevali, O.; Papa, F.; Rongioletti, M.C.A.; Baiocco, F.; Draghi, S.; et al. Plasticenta: First evidence of microplastics in human placenta. Environ. Int. 2021, 146, 106274. [Google Scholar] [CrossRef]
- Braun, T.; Ehrlich, L.; Henrich, W.; Koeppel, S.; Lomako, I.; Schwabl, P.; Liebmann, B. Detection of microplastic in human placenta and meconium in a clinical setting. Pharmaceutics 2021, 13, 921. [Google Scholar] [CrossRef]
- Li, D.; Shi, Y.; Yang, L.; Xiao, L.; Kehoe, D.K.; Gun’ko, Y.K.; Boland, J.J.; Wang, J.J. Microplastic release from the degradation of polypropylene feeding bottles during infant formula preparation. Nat. Food 2020, 1, 746–754. [Google Scholar] [CrossRef]
- Jemec Kokalj, A.; Dolar, A.; Drobne, D.; Marinšek, M.; Dolenec, M.; Škrlep, L.; Strmljan, G.; Mušič, B.; Škapin, A.S. Environmental Hazard. of polypropylene microplastics from disposable medical masks: Acute toxicity towards Daphnia magna and current knowledge on other polypropylene microplastics. Microplast. Nanoplast. 2022, 2, 1. [Google Scholar] [CrossRef]
- Jeyavani, J.; Sibiya, A.; Gopi, N.; Mahboob, S.; Riaz, M.N.; Vaseeharan, B. Dietary consumption of polypropylene microplastics alter the biochemical parameters and histological response in freshwater benthic mollusc Pomacea paludosa. Environ. Res. 2022, 212, 113370. [Google Scholar] [CrossRef] [PubMed]
- Piccardo, M.; Bertoli, M.; Pastorino, P.; Barceló, D.; Provenza, F.; Lesa, D.; Anselmi, S.; Elia, A.C.; Prearo, M.; Pizzul, E.; et al. Lethal and sublethal responses of Hydropsyche pellucidula (Insecta, Trichoptera) to commercial polypropylene microplastics after different preconditioning treatments. Toxics 2021, 9, 256. [Google Scholar] [CrossRef] [PubMed]
- Esterhuizen, M.; Kim, Y.J. Effects of polypropylene, polyvinyl chloride, polyethylene terephthalate, polyurethane, high-density polyethylene, and polystyrene microplastic on Nelumbo nucifera (Lotus) in water and sediment. Environ. Sci. Pollut. Res. Int. 2022, 29, 17580–17590. [Google Scholar] [CrossRef]
- Shi, R.; Liu, W.; Lian, Y.; Wang, Q.; Zeb, A.; Tang, J. Phytotoxicity of polystyrene, polyethylene and polypropylene microplastics on tomato (Lycopersicon esculentum L.). J. Environ. Manag. 2022, 317, 115441. [Google Scholar] [CrossRef]
- Zhao, Y.; Qiao, R.; Zhang, S.; Wang, G. Metabolomic profiling reveals the intestinal toxicity of different length of microplastic fibers on zebrafish (Danio rerio). J. Hazard. Mater. 2021, 403, 123663. [Google Scholar] [CrossRef]
- Sheng, C.; Zhang, S.; Zhang, Y. The influence of different polymer types of microplastics on adsorption, accumulation, and toxicity of triclosan in zebrafish. J. Hazard. Mater. 2021, 402, 123733. [Google Scholar] [CrossRef]
- Lei, L.; Wu, S.; Lu, S.; Liu, M.; Song, Y.; Fu, Z.; Shi, H.; Raley-Susman, K.M.; He, D. Microplastic particles cause intestinal damage and other adverse effects in zebrafish Danio rerio and nematode Caenorhabditis elegans. Sci. Total Environ. 2018, 619–620, 1–8. [Google Scholar] [CrossRef]
- Hwang, J.; Choi, D.; Han, S.; Choi, J.; Hong, J. An assessment of the toxicity of polypropylene microplastics in human derived cells. Sci. Total Environ. 2019, 684, 657–669. [Google Scholar] [CrossRef]
- Woo, J.H.; Seo, H.J.; Lee, J.Y.; Lee, I.; Jeon, K.; Kim, B.; Lee, K. Polypropylene microplastic exposure leads to lung inflammation through p38-mediated NF-κB pathway due to mitochondrial damage. Part. Fibre Toxicol. 2022, 20, 2. [Google Scholar] [CrossRef]
- Stock, V.; Laurisch, C.; Franke, J.; Dönmez, M.H.; Voss, L.; Böhmert, L.; Braeuning, A.; Sieg, H. Uptake and cellular effects of PE, PP, PET and PVC microplastic particles. Toxicol. Vitr. 2021, 70, 105021. [Google Scholar] [CrossRef]
- Lehner, R.; Wohlleben, W.; Septiadi, D.; Landsiedel, R.; Petri-Fink, A.; Rothen-Rutishauser, B. A novel 3D intestine barrier model to study the immune response upon exposure to microplastics. Arch. Toxicol. 2020, 94, 2463–2479. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Maruthupandy, M.; An, K.S.; Lee, K.H.; Jeon, S.; Kim, J.S.; Cho, W.S. Acute and subacute repeated oral toxicity study of fragmented microplastics in Sprague-Dawley rats. Ecotoxicol. Environ. Saf. 2021, 228, 112964. [Google Scholar] [CrossRef] [PubMed]
- Deng, Y.; Yan, Z.; Shen, R.; Wang, M.; Huang, Y.; Ren, H.; Zhang, Y.; Lemos, B. Microplastics release phthalate esters and cause aggravated adverse effects in the mouse gut. Environ. Int. 2020, 143, 105916. [Google Scholar] [CrossRef] [PubMed]
- Jin, Y.; Lu, L.; Tu, W.; Luo, T.; Fu, Z. Impacts of polystyrene microplastic on the gut barrier, microbiota and metabolism of mice. Sci. Total Environ. 2019, 649, 308–317. [Google Scholar] [CrossRef] [PubMed]
- Djouina, M.; Vignal, C.; Dehaut, A.; Caboche, S.; Hirt, N.; Waxin, C.; Himber, C.; Beury, D.; Hot, D.; Dubuquoy, L.; et al. Oral exposure to polyethylene microplastics alters gut morphology, immune response, and microbiota composition in mice. Environ. Res. 2022, 212, 113230. [Google Scholar] [CrossRef] [PubMed]
- Cui, Y.; Wang, Q.; Chang, R.; Zhou, X.; Xu, C. Intestinal barrier function-non-alcoholic fatty liver disease interactions and possible role of gut microbiota. J. Agric. Food Chem. 2019, 67, 2754–2762. [Google Scholar] [CrossRef]
- Huang, L.; He, F.; Wu, B. Mechanism of effects of nickel or nickel compounds on intestinal mucosal barrier. Chemosphere 2022, 305, 135429. [Google Scholar] [CrossRef] [PubMed]
- Johansson, M.E.; Ambort, D.; Pelaseyed, T.; Schütte, A.; Gustafsson, J.K.; Ermund, A.; Subramani, D.B.; Holmén-Larsson, J.M.; Thomsson, K.A.; Bergström, J.H.; et al. Composition and functional role of the mucus layers in the intestine. Cell Mol. Life Sci. 2011, 68, 3635–3641. [Google Scholar] [CrossRef]
- Schroeder, B.O. Fight them or feed them: How the intestinal mucus layer manages the gut microbiota. Gastroenterol. Rep. 2019, 7, 3–12. [Google Scholar] [CrossRef] [PubMed]
- Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Catalioto, R.M.; Maggi, C.A.; Giuliani, S. Intestinal epithelial barrier dysfunction in disease and possible therapeutical interventions. Curr. Med. Chem. 2011, 18, 398–426. [Google Scholar] [CrossRef] [PubMed]
- Liang, B.; Zhong, Y.; Huang, Y.; Lin, X.; Liu, J.; Lin, L.; Hu, M.; Jiang, J.; Dai, M.; Wang, B.; et al. Underestimated health risks: Polystyrene micro- and nanoplastics jointly induce intestinal barrier dysfunction by ROS-mediated epithelial cell apoptosis. Part. Fibre Toxicol. 2021, 18, 20. [Google Scholar] [CrossRef]
- Wang, K.; Qiu, L.; Zhu, J.; Sun, Q.; Qu, W.; Yu, Y.; Zhao, Z.; Yu, Y.; Shao, G. Environmental contaminant BPA causes intestinal damage by disrupting cellular repair and injury homeostasis in vivo and in vitro. Biomed. Pharmacother. 2021, 137, 111270. [Google Scholar] [CrossRef]
- Stock, V.; Fahrenson, C.; Thuenemann, A.; Dönmez, M.H.; Voss, L.; Böhmert, L.; Braeuning, A.; Lampen, A.; Sieg, H. Impact of artificial digestion on the sizes and shapes of microplastic particles. Food Chem. Toxicol. 2020, 135, 111010. [Google Scholar] [CrossRef]
- Schneider, C.A.; Rasband, W.S.; Eliceiri, K.W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 2012, 9, 671–675. [Google Scholar] [CrossRef] [PubMed]
- Senathirajah, K.; Attwood, S.; Bhagwat, G.; Carbery, M.; Wilson, S.; Palanisami, T. Estimation of the mass of microplastics ingested—A pivotal first step towards human health risk assessment. J. Hazard. Mater. 2021, 404, 124004. [Google Scholar] [CrossRef]
- Stock, V.; Böhmert, L.; Lisicki, E.; Block, R.; Cara-Carmona, J.; Pack, L.K.; Selb, R.; Lichtenstein, D.; Voss, L.; Henderson, C.J.; et al. Uptake and effects of orally ingested polystyrene microplastic particles in vitro and in vivo. Arch. Toxicol. 2019, 93, 1817–1833. [Google Scholar] [CrossRef]
- Mu, Y.; Sun, J.; Li, Z.; Zhang, W.; Liu, Z.; Li, C.; Peng, C.; Cui, G.; Shao, H.; Du, Z. Activation of pyroptosis and ferroptosis is involved in the hepatotoxicity induced by polystyrene microplastics in mice. Chemosphere 2022, 291, 132944. [Google Scholar] [CrossRef]
- Bao, W.; Liu, M.; Meng, J.; Liu, S.; Wang, S.; Jia, R.; Wang, Y.; Ma, G.; Wei, W.; Tian, Z. MOFs-based nanoagent enables dual mitochondrial damage in synergistic antitumor therapy via oxidative stress and calcium overload. Nat. Commun. 2021, 12, 6399. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.; Wei, P.; Chen, X.; Zhang, Z.; Jin, Z.; Liu, J.; Liu, L. Less is more: Biological effects of NiSe(2)/rGO nanocomposites with low dose provide new insight for risk assessment. J. Hazard. Mater. 2021, 415, 125605. [Google Scholar] [CrossRef] [PubMed]
- Yun, S.H.; Moon, Y.S.; Sohn, S.H.; Jang, I.S. Effects of cyclic heat stress or vitamin C supplementation during cyclic heat stress on HSP70, inflammatory cytokines, and the antioxidant defense system in Sprague Dawley rats. Exp. Anim. 2012, 61, 543–553. [Google Scholar] [CrossRef] [PubMed]
- Impellizzeri, D.; Cordaro, M.; Campolo, M.; Gugliandolo, E.; Esposito, E.; Benedetto, F.; Cuzzocrea, S.; Navarra, M. Anti-inflammatory and antioxidant effects of flavonoid-rich fraction of bergamot juice (BJe) in a mouse model of intestinal ischemia/reperfusion injury. Front. Pharmacol. 2016, 7, 203. [Google Scholar] [CrossRef] [PubMed]
- Hou, J.; Lei, Z.; Cui, L.; Hou, Y.; Yang, L.; An, R.; Wang, Q.; Li, S.; Zhang, H.; Zhang, L. Polystyrene microplastics lead to pyroptosis and apoptosis of ovarian granulosa cells via NLRP3/Caspase-1 signaling pathway in rats. Ecotoxicol. Environ. Saf. 2021, 212, 112012. [Google Scholar] [CrossRef]
- Yin, K.; Lu, H.; Zhang, Y.; Hou, L.; Meng, X.; Li, J.; Zhao, H.; Xing, M. Secondary brain injury after polystyrene microplastic-induced intracerebral hemorrhage is associated with inflammation and pyroptosis. Chem. Biol. Interact. 2022, 367, 110180. [Google Scholar] [CrossRef]
- Hu, M.; Palić, D. Micro- and nano-plastics activation of oxidative and inflammatory adverse outcome pathways. Redox Biol. 2020, 37, 101620. [Google Scholar] [CrossRef]
- Kale, G.; Naren, A.P.; Sheth, P.; Rao, R.K. Tyrosine phosphorylation of occludin attenuates its interactions with ZO-1, ZO-2, and ZO-3. Biochem. Biophys. Res. Commun. 2003, 302, 324–329. [Google Scholar] [CrossRef]
- Salim, S.Y.; Söderholm, J.D. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 362–381. [Google Scholar] [CrossRef]
- Ulluwishewa, D.; Anderson, R.C.; McNabb, W.C.; Moughan, P.J.; Wells, J.M.; Roy, N.C. Regulation of tight junction permeability by intestinal bacteria and dietary components. J. Nutr. 2011, 141, 769–776. [Google Scholar] [CrossRef]
- Suzuki, T.; Tanabe, S.; Hara, H. Kaempferol enhances intestinal barrier function through the cytoskeletal association and expression of tight junction proteins in Caco-2 cells. J. Nutr. 2011, 141, 87–94. [Google Scholar] [CrossRef] [PubMed]
- Furuse, M.; Hirase, T.; Itoh, M.; Nagafuchi, A.; Yonemura, S.; Tsukita, S.; Tsukita, S. Occludin: A novel integral membrane protein localizing at tight junctions. J. Cell Biol. 1993, 123, 1777–1788. [Google Scholar] [CrossRef] [PubMed]
- Lynn, K.S.; Peterson, R.J.; Koval, M. Ruffles and spikes: Control of tight junction morphology and permeability by claudins. Biochim. Biophys. Acta Biomembr. 2020, 1862, 183339. [Google Scholar] [CrossRef] [PubMed]
- Griffiths, V.; Al Assaf, N.; Khan, R. Review of claudin proteins as potential biomarkers for necrotizing enterocolitis. Ir. J. Med. Sci. 2021, 190, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Zhang, H.S.; Fong, G.H.; Xi, Q.L.; Wu, G.H.; Bai, C.G.; Ling, Z.Q.; Fan, L.; Xu, Y.M.; Qin, Y.Q.; et al. PHD3 stabilizes the tight junction protein occludin and protects intestinal epithelial barrier function. J. Biol. Chem. 2015, 290, 20580–20589. [Google Scholar] [CrossRef] [PubMed]
- Camilleri, M. Leaky gut: Mechanisms, measurement and clinical implications in humans. Gut 2019, 68, 1516–1526. [Google Scholar] [CrossRef]
- Rao, R.K.; Basuroy, S.; Rao, V.U.; Karnaky, K.J., Jr.; Gupta, A. Tyrosine phosphorylation and dissociation of occludin-ZO-1 and E-cadherin-beta-catenin complexes from the cytoskeleton by oxidative stress. Biochem. J. 2002, 368, 471–481. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.Y.; Oh, T.W.; Do, H.J.; Yang, J.H.; Yang, I.J.; Jeon, Y.H.; Go, Y.H.; Ahn, S.C.; Ma, J.Y.; Park, K.I. Acer palmatum thumb. Ethanol extract alleviates interleukin-6-induced barrier dysfunction and dextran sodium sulfate-induced colitis by improving intestinal barrier function and reducing inflammation. J. Immunol. Res. 2018, 2018, 5718396. [Google Scholar] [CrossRef]
- Zhou, H.Y.; Zhu, H.; Yao, X.M.; Qian, J.P.; Yang, J.; Pan, X.D.; Chen, X.D. Metformin regulates tight junction of intestinal epithelial cells via MLCK-MLC signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 5239–5246. [Google Scholar] [CrossRef] [PubMed]
- Marchiando, A.M.; Shen, L.; Graham, W.V.; Weber, C.R.; Schwarz, B.T.; Austin, J.R., 2nd; Raleigh, D.R.; Guan, Y.; Watson, A.J.; Montrose, M.H.; et al. Caveolin-1-dependent occludin endocytosis is required for TNF-induced tight junction regulation in vivo. J. Cell Biol. 2010, 189, 111–126. [Google Scholar] [CrossRef] [PubMed]
- Capaldo, C.T.; Nusrat, A. Cytokine regulation of tight junctions. Biochim. Biophys. Acta 2009, 1788, 864–871. [Google Scholar] [CrossRef] [PubMed]
- Zolotarevsky, Y.; Hecht, G.; Koutsouris, A.; Gonzalez, D.E.; Quan, C.; Tom, J.; Mrsny, R.J.; Turner, J.R. A membrane-permeant peptide that inhibits MLC kinase restores barrier function in in vitro models of intestinal disease. Gastroenterology 2002, 123, 163–172. [Google Scholar] [CrossRef] [PubMed]
- Al-Sadi, R.M.; Ma, T.Y. IL-1beta causes an increase in intestinal epithelial tight junction permeability. J. Immunol. 2007, 178, 4641–4649. [Google Scholar] [CrossRef] [PubMed]
- Desai, T.R.; Leeper, N.J.; Hynes, K.L.; Gewertz, B.L. Interleukin-6 causes endothelial barrier dysfunction via the protein kinase C pathway. J. Surg. Res. 2002, 104, 118–123. [Google Scholar] [CrossRef] [PubMed]
- Madsen, K.L.; Lewis, S.A.; Tavernini, M.M.; Hibbard, J.; Fedorak, R.N. Interleukin 10 prevents cytokine-induced disruption of T84 monolayer barrier integrity and limits chloride secretion. Gastroenterology 1997, 113, 151–159. [Google Scholar] [CrossRef]
- Madsen, K.L.; Malfair, D.; Gray, D.; Doyle, J.S.; Jewell, L.D.; Fedorak, R.N. Interleukin-10 gene-deficient mice develop a primary intestinal permeability defect in response to enteric microflora. Inflamm. Bowel Dis. 1999, 5, 262–270. [Google Scholar] [CrossRef]
- Linden, S.K.; Sutton, P.; Karlsson, N.G.; Korolik, V.; McGuckin, M.A. Mucins in the mucosal barrier to infection. Mucosal Immunol. 2008, 1, 183–197. [Google Scholar] [CrossRef]
- Sheng, Y.H.; Davies, J.M.; Wang, R.; Wong, K.Y.; Giri, R.; Yang, Y.; Begun, J.; Florin, T.H.; Hasnain, S.Z.; McGuckin, M.A. MUC1-mediated macrophage activation promotes colitis-associated colorectal cancer via activating the interleukin-6/ signal transducer and activator of transcription 3 axis. Cell Mol. Gastroenterol. Hepatol. 2022, 14, 789–811. [Google Scholar] [CrossRef]
- Gendler, S.J. MUC1, the renaissance molecule. J. Mammary Gland Biol. Neoplasia 2001, 6, 339–353. [Google Scholar] [CrossRef]
- Lu, L.; Wan, Z.; Luo, T. Polystyrene microplastics induce gut microbiota dysbiosis and hepatic lipid metabolism disorder in mice. Sci. Total Environ. 2018, 631–632, 449–458. [Google Scholar] [CrossRef]
- Luo, T.; Wang, C.; Pan, Z.; Jin, C.; Fu, Z.; Jin, Y. Maternal polystyrene microplastic exposure during gestation and lactation altered metabolic homeostasis in the dams and their F1 and F2 offspring. Environ. Sci. Technol. 2019, 53, 10978–10992. [Google Scholar] [CrossRef] [PubMed]
- Brownlee, I.A.; Knight, J.; Dettmar, P.W.; Pearson, J.P. Action of reactive oxygen species on colonic mucus secretions. Free Radic. Biol. Med. 2007, 43, 800–808. [Google Scholar] [CrossRef] [PubMed]
- Leppkes, M.; Roulis, M.; Neurath, M.F.; Kollias, G.; Becker, C. Pleiotropic functions of TNF-α in the regulation of the intestinal epithelial response to inflammation. Int. Immunol. 2014, 26, 509–515. [Google Scholar] [CrossRef]
- Hasnain, S.Z.; Tauro, S.; Das, I.; Tong, H.; Chen, A.C.; Jeffery, P.L.; McDonald, V.; Florin, T.H.; McGuckin, M.A. IL-10 promotes production of intestinal mucus by suppressing protein misfolding and endoplasmic reticulum stress in goblet cells. Gastroenterology 2013, 144, 357–368.e9. [Google Scholar] [CrossRef]
- Murek, M.; Kopic, S.; Geibel, J. Evidence for intestinal chloride secretion. Exp. Physiol. 2010, 95, 471–478. [Google Scholar] [CrossRef]
- Muchekehu, R.W.; Quinton, P.M. A new role for bicarbonate secretion in cervico-uterine mucus release. J. Physiol. 2010, 588, 2329–2342. [Google Scholar] [CrossRef] [PubMed]
- Garcia, M.A.; Yang, N.; Quinton, P.M. Normal mouse intestinal mucus release requires cystic fibrosis transmembrane regulator-dependent bicarbonate secretion. J. Clin. Investig. 2009, 119, 2613–2622. [Google Scholar] [CrossRef] [PubMed]
- Gustafsson, J.K.; Lindén, S.K.; Alwan, A.H.; Scholte, B.J.; Hansson, G.C.; Sjövall, H. Carbachol-induced colonic mucus formation requires transport via NKCC1, K⁺ channels and CFTR. Pflug. Arch. 2015, 467, 1403–1415. [Google Scholar] [CrossRef]
- Li, C.; Krishnamurthy, P.C.; Penmatsa, H.; Marrs, K.L.; Wang, X.Q.; Zaccolo, M.; Jalink, K.; Li, M.; Nelson, D.J.; Schuetz, J.D.; et al. Spatiotemporal coupling of cAMP transporter to CFTR chloride channel function in the gut epithelia. Cell 2007, 131, 940–951. [Google Scholar] [CrossRef]
- Lynch, S.V.; Goldfarb, K.C.; Wild, Y.K.; Kong, W.; De Lisle, R.C.; Brodie, E.L. Cystic fibrosis transmembrane conductance regulator knockout mice exhibit aberrant gastrointestinal microbiota. Gut Microbes 2013, 4, 41–47. [Google Scholar] [CrossRef]
- Wang, J.; Wang, W.; Wang, H.; Tuo, B. Physiological and pathological functions of SLC26A6. Front. Med. 2020, 7, 618256. [Google Scholar] [CrossRef] [PubMed]
- Zheng, J.; Ahmad, A.A.; Yang, Y.; Liang, Z.; Shen, W.; Feng, M.; Shen, J.; Lan, X.; Ding, X. Lactobacillus rhamnosus CY12 enhances intestinal barrier function by regulating tight junction protein expression, oxidative stress, and inflammation response in lipopolysaccharide-induced caco-2 cells. Int. J. Mol. Sci. 2022, 23, 11162. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Sun, S.; Wu, X.; Yang, S.; Wu, Y.; Zhao, J.; Zhang, H.; Chen, W. Intestinal environmental disorders associate with the tissue damages induced by perfluorooctane sulfonate exposure. Ecotoxicol. Environ. Saf. 2020, 197, 110590. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Weng, Y.; Shen, Q.; Zhao, Y.; Jin, Y. Microplastic: A potential threat to human and animal health by interfering with the intestinal barrier function and changing the intestinal microenvironment. Sci. Total Environ. 2021, 785, 147365. [Google Scholar] [CrossRef] [PubMed]
- Yao, H.; Hu, C.; Yin, L.; Tao, X.; Xu, L.; Qi, Y.; Han, X.; Xu, Y.; Zhao, Y.; Wang, C.; et al. Dioscin reduces lipopolysaccharide-induced inflammatory liver injury via regulating TLR4/MyD88 signal pathway. Int. Immunopharmacol. 2016, 36, 132–141. [Google Scholar] [CrossRef] [PubMed]
- Gao, K.; Dai, D.L.; Martinka, M.; Li, G. Prognostic significance of nuclear factor-kappaB p105/p50 in human melanoma and its role in cell migration. Cancer Res. 2006, 66, 8382–8388. [Google Scholar] [CrossRef]
- Ryseck, R.P.; Bull, P.; Takamiya, M.; Bours, V.; Siebenlist, U.; Dobrzanski, P.; Bravo, R. RelB, a new Rel family transcription activator that can interact with p50-NF-kappa B. Mol. Cell Biol. 1992, 12, 674–684. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Yang, F.; Ma, C.; Cao, W.; Yang, J.; Zhao, Z.; Tian, H.; Zhu, Z.; Zheng, H. Porcine epidemic diarrhea virus nsp14 inhibits NF-κB pathway activation by targeting the IKK complex and p65. Anim. Dis. 2021, 1, 24. [Google Scholar] [CrossRef]
- Matelski, L.; Morgan, R.K.; Grodzki, A.C.; Van de Water, J.; Lein, P.J. Effects of cytokines on nuclear factor-kappa B, cell viability, and synaptic connectivity in a human neuronal cell line. Mol. Psychiatry 2021, 26, 875–887. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.N.; Li, X.N.; Wang, Y.X.; Ma, X.Y.; Li, J.L. Disrupted microbiota-barrier-immune interaction in phthalates-mediated barrier defect in the duodenum. Chemosphere 2022, 308, 136275. [Google Scholar] [CrossRef]
- Hu, X.; Liu, S.; Zhu, J.; Ni, H. Dachengqi decoction alleviates acute lung injury and inhibits inflammatory cytokines production through TLR4/NF-κB signaling pathway in vivo and in vitro. J. Cell Biochem. 2019, 120, 8956–8964. [Google Scholar] [CrossRef]
- Chang, Y.; Yuan, L.; Liu, J.; Muhammad, I.; Cao, C.; Shi, C.; Zhang, Y.; Li, R.; Li, C.; Liu, F. Dihydromyricetin attenuates Escherichia coli lipopolysaccharide-induced ileum injury in chickens by inhibiting NLRP3 inflammasome and TLR4/NF-κB signalling pathway. Vet. Res. 2020, 51, 72. [Google Scholar] [CrossRef] [PubMed]
- Bai, Q.; Wang, Z.; Piao, Y.; Zhou, X.; Piao, Q.; Jiang, J.; Liu, H.; Piao, H.; Li, L.; Song, Y.; et al. Sesamin alleviates asthma airway inflammation by regulating mitophagy and mitochondrial apoptosis. J. Agric. Food Chem. 2022, 70, 4921–4933. [Google Scholar] [CrossRef] [PubMed]
- Li, W.J.; Zhang, L.; Wu, H.X.; Li, M.; Wang, T.; Zhang, W.B.; Du, Z.Y.; Zhang, M.L. Intestinal microbiota mediates gossypol-induced intestinal inflammation, oxidative stress, and apoptosis in fish. J. Agric. Food Chem. 2022, 70, 6688–6697. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Huang, J.; Yang, J.; Cai, J.; Liu, Q.; Zhang, X.; Bao, J.; Zhang, Z. Cadmium induces apoptosis and autophagy in swine small intestine by downregulating the PI3K/Akt pathway. Environ. Sci. Pollut. Res. Int. 2022, 29, 41207–41218. [Google Scholar] [CrossRef] [PubMed]
- Spierings, D.; McStay, G.; Saleh, M.; Bender, C.; Chipuk, J.; Maurer, U.; Green, D.R. Connected to death: The (unexpurgated) mitochondrial pathway of apoptosis. Science 2005, 310, 66–67. [Google Scholar] [CrossRef]
- Oberst, A.; Bender, C.; Green, D.R. Living with death: The evolution of the mitochondrial pathway of apoptosis in animals. Cell Death Differ. 2008, 15, 1139–1146. [Google Scholar] [CrossRef]
- Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef]
- Hou, L.; Wang, D.; Yin, K.; Zhang, Y.; Lu, H.; Guo, T.; Li, J.; Zhao, H.; Xing, M. Polystyrene microplastics induce apoptosis in chicken testis via crosstalk between NF-κB and Nrf2 pathways. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2022, 262, 109444. [Google Scholar] [CrossRef]
- Zhou, W.J.; Wang, S.; Hu, Z.; Zhou, Z.Y.; Song, C.J. Angelica sinensis polysaccharides promotes apoptosis in human breast cancer cells via CREB-regulated caspase-3 activation. Biochem. Biophys. Res. Commun. 2015, 467, 562–569. [Google Scholar] [CrossRef]
- Shou, Y.; Li, N.; Li, L.; Borowitz, J.L.; Isom, G.E. NF-kappaB-mediated up-regulation of Bcl-X(S) and Bax contributes to cytochrome c release in cyanide-induced apoptosis. J. Neurochem. 2002, 81, 842–852. [Google Scholar] [CrossRef] [PubMed]
- Gill, J.S.; Windebank, A.J. Ceramide initiates NFkappaB-mediated caspase activation in neuronal apoptosis. Neurobiol. Dis. 2000, 7, 448–461. [Google Scholar] [CrossRef] [PubMed]
- Visalli, G.; Facciolà, A.; Pruiti Ciarello, M.; De Marco, G.; Maisano, M.; Di Pietro, A. Acute and sub-chronic effects of microplastics (3 and 10 µm) on the Human Intestinal Cells Ht-29. Int. J. Environ. Res. Public Health 2021, 18, 5833. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Zhu, S.; Liu, Q.; Wei, J.; Jin, Y.; Wang, X.; Zhang, L. Polystyrene microplastics cause cardiac fibrosis by activating Wnt/β-catenin signaling pathway and promoting cardiomyocyte apoptosis in rats. Environ. Pollut. 2020, 265, 115025. [Google Scholar] [CrossRef]
- Liu, Z.; Huang, Y.; Jiao, Y.; Chen, Q.; Wu, D.; Yu, P.; Li, Y.; Cai, M.; Zhao, Y. Polystyrene nanoplastic induces ROS production and affects the MAPK-HIF-1/NFkB-mediated antioxidant system in Daphnia pulex. Aquat. Toxicol. 2020, 220, 105420. [Google Scholar] [CrossRef]
- Jeong, C.B.; Won, E.J.; Kang, H.M.; Lee, M.C.; Hwang, D.S.; Hwang, U.K.; Zhou, B.; Souissi, S.; Lee, S.J.; Lee, J.S. Microplastic size-dependent toxicity, oxidative stress induction, and p-JNK and p-p38 activation in the Monogonont Rotifer (Brachionus Koreanus). Environ. Sci. Technol. 2016, 50, 8849–8857. [Google Scholar] [CrossRef]
- Qiang, L.; Cheng, J. Exposure to polystyrene microplastics impairs gonads of zebrafish (Danio Rerio). Chemosphere 2021, 263, 128161. [Google Scholar] [CrossRef]
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. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jia, R.; Han, J.; Liu, X.; Li, K.; Lai, W.; Bian, L.; Yan, J.; Xi, Z. Exposure to Polypropylene Microplastics via Oral Ingestion Induces Colonic Apoptosis and Intestinal Barrier Damage through Oxidative Stress and Inflammation in Mice. Toxics 2023, 11, 127. https://doi.org/10.3390/toxics11020127
Jia R, Han J, Liu X, Li K, Lai W, Bian L, Yan J, Xi Z. Exposure to Polypropylene Microplastics via Oral Ingestion Induces Colonic Apoptosis and Intestinal Barrier Damage through Oxidative Stress and Inflammation in Mice. Toxics. 2023; 11(2):127. https://doi.org/10.3390/toxics11020127
Chicago/Turabian StyleJia, Rui, Jie Han, Xiaohua Liu, Kang Li, Wenqing Lai, Liping Bian, Jun Yan, and Zhuge Xi. 2023. "Exposure to Polypropylene Microplastics via Oral Ingestion Induces Colonic Apoptosis and Intestinal Barrier Damage through Oxidative Stress and Inflammation in Mice" Toxics 11, no. 2: 127. https://doi.org/10.3390/toxics11020127
APA StyleJia, R., Han, J., Liu, X., Li, K., Lai, W., Bian, L., Yan, J., & Xi, Z. (2023). Exposure to Polypropylene Microplastics via Oral Ingestion Induces Colonic Apoptosis and Intestinal Barrier Damage through Oxidative Stress and Inflammation in Mice. Toxics, 11(2), 127. https://doi.org/10.3390/toxics11020127