Next Article in Journal
Short-Term Microplastics Exposure to the Common Mysid Shrimp, Americamysis bahia: Effects on Mortality and DNA Methylation
Previous Article in Journal
Uptake and Cellular Effects of Polymethylmethacrylate on Human Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progress in Intestinal Toxicity of Microplastics and Nanoplastics: Systematic Review of Preclinical Evidence

by
Madjid Djouina
,
Suzie Loison
and
Mathilde Body-Malapel
*
Université de Lille, Inserm, CHU Lille, U1286- INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microplastics 2024, 3(2), 217-233; https://doi.org/10.3390/microplastics3020013
Submission received: 10 January 2024 / Revised: 14 March 2024 / Accepted: 2 April 2024 / Published: 8 April 2024

Abstract

:
The tremendous plastic production and poor post-use management are current and future sources of environmental and human contamination due to their degradation products: microplastics and nanoplastics (MNPLs). Methodological developments have allowed MNPLs to be detected in an increasing variety of human foods, as well as in stool and colonic mucosa. It was suggested early that the direct contact between MNPLs and intestinal tissues could represent a potential risk for human health. In order to assess this, over the last 3 years, numerous studies have evaluated the impact of MNPL ingestion on intestinal homeostasis in rodents. This comprehensive review reports the preclinical studies published between January 2021 and January 2024, and analyzes their contributions as well as their shortcomings. It shows that evidence is accumulating of the intestinal toxicity of spherical MNPLs, which lead to pro-inflammatory, pro-oxidative, barrier-disruptive and dysbiotic effects. However, the available literature has addressed only a minor part of the potential health issues of MNPLs. Many parameters contributing to MNPL toxicity need to be better taken into account in future studies. Particular attention should be paid to improve the representativeness of MNPLs, as well as to better consider the susceptibility factors of MNPL toxicity, generated especially by an underlying pathology or pathological imprinting.

Graphical Abstract

1. Introduction

Plastic is a long-carbon-chain polymer that is massively integrated in our daily life. Plastic products are used in packaging, building and construction, automotives, electricals and electronics, agriculture and houseware [1]. During the last 70 years, plastic production has extensively expanded, reaching 390.7 metric tons of plastic production worldwide in 2021 [1]. The accumulation of post-consumer plastic waste in the environment coupled with mismanaged waste programs have led to plastic contamination in all environmental niches [2]. Overall, nearly 60–75% of plastic products are discarded to the environment or landfilled, and this colossal plastic burden will remain in the earth for centuries [3]. Indeed, in the environment, large plastics undergo continuous physical, chemical and biological degradation processes, generating a broad amount of microplastics. Microplastics (MPLs) refer to small plastics ranging in size from 1 µm to 5 mm [4]. They can be further degraded into plastic particles with a size range between 1 and 1000 nm, known as nanoplastics (NPLs) [5]. Microplastics and nanoplastics (MNPLs) have many shapes such as granules, fragments, microbeads, fibers and foams [6]. MNPLs spread through the air, land and sea. The mass of MPLs which accumulates in the oceans has led to contamination by MPLs in fish, zooplankton, shrimp, crab, clam, mussel and many other aquatic organisms [7]. Microplastics can also be absorbed and accumulated by plants, such as rice, wheat, lettuce and other crops. Plants can absorb MPLs through their roots and then transfer them to their aboveground parts [8,9]. Moreover, MPLs easily enter the human food chain as marine and terrestrial organisms ingest MPLs [10]. Overall, human food can be contaminated by plastic particles either through direct exposure to the environment or indirectly by the transfer of MPLs within the ecosystem [10]. Therefore, MPLs accumulate in the human food chain. They have been detected in water, tea, beer, white wine, energy drinks, soft drinks, fish, shellfish, salt, sugar, honey, milk, poultry meat, fruit and vegetables [11]. The most common detected MPLs in food are polyethylene terephthalate (PET), polyethylene (PE), polypropylene (PP), polystyrene (PS), polyvinyl chloride (PVC), polyamide (PA) and polycarbonate (PC) [11]. Still, there is a lack of data quantifying MNPLs in meat, cereals and many other food products, and the amount of MNPLs ingested by human through food is recognized to be underestimated [6]. Moreover, exposure to MPLs through dust also decisively contributes to MPL intake. The amount of MPLs ingested from exposure to indoor dust has been estimated to be 0.21, 0.23 and 0.6 MPL/kg bw/day for adults worldwide [12], in China [13] and in Iran [14], respectively. Infants and newborns are exposed to higher MPL concentrations, namely 3.04, 7.4 and 13.7 MPL/kg bw/day, respectively. Despite the knowledge gaps regarding human exposure to MNPLs, the total burden of human exposure to MNPLs has been recently estimated to be 2.93 × 1010 particle/year [6]. The contact between MPLs and the colonic epithelium has been confirmed by several studies describing the detection of MPLs in the stools of healthy adults [15,16,17,18,19]. MPLs have also been detected in the meconium and in infant stools [20,21]. MPLs are therefore present in the intestinal lumen, but could also penetrate the intestinal epithelium, as suggested by their detection in human colectomy specimens [22,23]. Indeed, MPL presence was reported in colons from healthy adults and from patients with colorectal carcinoma. Therefore, it is essential to determine the health impact generated by MPL exposure, particularly at the intestinal level. Numerous studies carried out mainly in aquatic organisms and to a lesser extent in rodents have since demonstrated the intestinal toxicity caused by the ingestion of MPLs. The available data were reviewed in 2020 [24,25]. Since then, knowledge of the effects of MNPLs has extended to mammals, thanks to numerous studies which have assessed the effect of ingestion of MPLs and NPLs in rodents. In comparison with previous studies, these new studies focus more closely on human physiology. They studied a wide variety of MPLs and NPLs; they addressed several types of polymers, presenting a wide variety of sizes, sometimes with different surface charges. They were carried out for varying durations, in healthy or pathological individuals. They strengthened the evidence of MNPL-induced gut toxicity, but also identified new intestinal adverse effects as well as novel pathophysiological mechanisms impaired by MNPL exposure. The aim of this work was to offer a comprehensive analysis of the recent literature in order to provide an up-to-date understanding of the research area of MNPL intestinal toxicity.

2. Methods

A systematic search for published articles and documents was conducted in databases such PubMed database (https://www-ncbi-nlm-nih-gov.proxy.insermbiblio.inist.fr, accessed on 29 February 2024), ScienceDirect (https://www-sciencedirect-com.proxy.insermbiblio.inist.fr, https://www-ncbi-nlm-nih-gov.proxy.insermbiblio.inist.fr, accessed on 29 February 2024), SpringerLink (https://link-springer-com.proxy.insermbiblio.inist.fr, https://www-ncbi-nlm-nih-gov.proxy.insermbiblio.inist.fr, accessed on 29 February 2024) and Google Scholar (https://scholar-google-com.proxy.insermbiblio.inist.fr, https://www-ncbi-nlm-nih-gov.proxy.insermbiblio.inist.fr, accessed on 29 February 2024), in the period from January 2021 to February 2024.
The search queries were “microplastic or nanoplastic” and “intestinal” and “mouse or rat or rodent” in titles/abstracts. All the relevant studies were retrieved by sorting the title and the abstract. Only studies aimed at studying the impact of MPLs and/or NPLs by ingestion in rodents were included. Publications including in vitro and in vivo studies were included but only in vivo data were reported. Studies reporting only data on the gut microbiota were included. Studies for which it was not possible to report a primary characterization of the MNPLs used, such as the type of polymer, were excluded.

3. Results and Discussion

The preclinical studies which assessed the intestinal toxicity of MNPLs in rodents are detailed in Table 1.
Most of the preclinical studies focused on PS. There are a plethora of studies showing gut toxicity of 5 µm PS MPLs, which is characterized by impaired gut barrier and mucus production, increased oxidative stress and cytokine levels, and gut microbiota dysbiosis [26,31,32,33,34,35,37,41,44,45,48]. Most studies reported MPL toxicity in the colon, but when the small intestine was analyzed, impairments to the duodenum, jejunum and ileum were also reported [28,48]. Two studies found that gut toxicity occurred from a concentration of 0.1 mg/kg bw/day for 5 µm PS MPLs and worsened the intensity of DSS-induced colitis [26,31]. Similar effects were even found at the lowest dosage of 0.02 mg/kg bw/day [34,37]. The dysbiotic effects of 5 µm PS MPLs were detected from 0.02 mg/kg bw/day in male C57BL/6 mice [32,44] and from 0.006 mg/kg/bw/day in female BALB/C mice [49]. These findings related to 5 µm PS MPL are consistent with the ones observed for other sizes of MPLs [40,43], which all showed strong evidence of the gut toxicity of PS MPLs.
Numerous publications have also reported the gut toxicity of PS NPLs, which is, as for PS MPLs, characterized by impaired gut barrier function and mucus production, increased oxidative stress and cytokine levels, and gut microbiota dysbiosis [38,39,42,46,50,53,54,56,57]. Li et al. reported intestinal mechanical and immune barrier dysfunction in mice exposed to 0.02 mg/kg bw/day of PS NPLs [50]. Accordingly, colon inflammation was observed after exposure to PS NPLs at a similar concentration by Teng et al. [53]. The 0.02 mg/kg bw/day concentration can therefore be considered so far as the lowest observed adverse effect level for both PS MPL and PS NPL intestinal toxicity. Compared with PS MPLs, ingestion of PS nano-sized NPLs induced similar gut disturbances, except in the studies of Xiao et al. and Schwarzfisher et al., who showed, respectively, slight and null gut toxicity following exposure to PS NPLs [29,30]. PS NPL ingestion impaired both the colon and the small intestine epithelium. PS MNPL toxicity can be associated with disturbances to gut immune response, involving macrophages, innate lymphoid cells and B and T lymphocytes [39,49,50]. Overall, PS MNPL intestinal toxicity appeared in most studies after an exposure duration of about 4 weeks, but was also described after 2 and 32 weeks of exposure. Among the studies with exposure to both MPLs and NPLs in the same experimental conditions, four studies supported a greater intestinal toxicity of MPLs compared to NPLs [27,47,51,52]. The other five studies showed that the intestinal toxicity of NPLs was of a similar intensity to that of MPLs, but that the features of toxic effects were dependent on the size of the MNPLs [28,40,43,49,55]. The complete study performed by Zhang et al., which included different PS MNPL sizes and concentrations and 2-week and 4-week exposure durations, argued in favor of a greater influence of exposure duration and MNPL size compared to MNPL concentration on the gut toxicity of MNPLs [49]. Furthermore, the influence of the surface charge of PS NPLs on adverse gut effects was assessed: two studies consistently observed a gradually increasing gut toxicity from pristine PS NPLs, to negatively charged carboxylated PS NPLs and then positively charged aminated PS NPLs [27,42], whereas similar levels of colon impairments were observed for pristine PS, PS-COOH and PS-NH2 NPLs by Teng et al. [53].
PS was the only polymer that could be administered in the drinking water of rodents. Other polymers required administration by gavage or through feed. Oral administration of PE, PVC, PP and PET MPLs also promoted gut microbiota dysbiosis and permeability defects, but these effects were observed at the highest exposure concentrations (minimum 0.2 mg/kg for PE [58,63], 22 and 100 mg/kg for PVC [64,65], 22 to 2272 mg/kg for PP [66] and 200 mg/kg for PET [67]). However, the dosage of 0.02 mg/kg bw/day has never been assessed for these polymers. Therefore, to date, it remains difficult to assess the relative gut toxicity of the different polymers. The best way to achieve this goal is to compare the polymers under the same experimental conditions, as shown by Xie et al., who concluded that the pro-inflammatory properties on the colon followed this trend: PS > PVC > PET > PE > PP [69].
It must be emphasized that apart from the consistently described pro-inflammatory, pro-oxidative, barrier-disruptive and dysbiotic effects, other adverse gut effects have been reported which deserve further investigation, such as the promotion of H. pylori infection [59], hyperproliferation and tumorigenesis [37,60,72], dyslipidemic effects [39] and alterations to microbial antibiotic resistance genes and virulence factors [40].
One important limitation is that most of the rodent studies have been performed with commercially available MNPLs. Therefore, they only reflect the toxicity of spherical MNPLs. They assessed only one of the many forms of MNPLs which are found in the environment and food [74]. Moreover, commercial MNPLs are devoid of additives and contaminants unlike MNPLs resulting from the degradation of industrial plastics. It is known that some plastic additives and contaminants have their own intestinal toxicity, such as bisphenols and phthalates [75,76]. Some plastic contaminants such as heavy metals and persistent organic pollutants could also present combined intestinal toxicity with MNPLs [77]. In addition to the known additives and contaminants of MNPLs, other unknown chemical products could additionally be released and alter gut homeostasis: this is the new avenue of research which was opened by Wang et al., who showed that human gut enzymes catalyzed the release of oligomer nanoparticles by polylactic acid. In mice, these oligomers induced small intestine and colon inflammation according to histological and molecular studies, associated with MMP12 inactivation [78]. An important limitation is therefore that the combined effect of MNPLs and their additives and contaminants is not taken into consideration in the studies using commercially available MNPL beads.
Furthermore, most of the studies did not consider either the mixture effects of various shapes and polymer types, or the weathering and aging of MNPLs. To date, only four studies have been performed on more realistic MNPLs [36,61,66,68]. They used ground or crushed MNPLs. Mouse exposure to pin-made PET MPLs did not induce colon barrier defects and inflammation. By contrast, it led to alterations in the gut immune transcriptome and metagenome [68]. Some adverse gut effects of spherical PET NPLs have also been shown at high concentrations in mice [67]. Ingestion of PET MPLs induced structural and functional alterations to duodenal myenteric neurons in pigs [79]. But the scarcity of studies precludes drawing conclusions on the toxicity of PET MPLs. Regarding the other polymers, studies on fragmented MPLs showed consistent evidence of pro-inflammatory effects induced in the small intestine for PS MPLs and in the colon for PE and PP MPLs. Feed contamination with crushed PS MPLs for 3 weeks induced small intestine epithelium damage and fecal dysbiosis [36]. Polydisperse, ground PE NPLs and MPLs led to overt colitis after only 1 week of exposure [61]. Colon oxidative stress, inflammation, barrier impairment and apoptosis were induced by oral administration of irregular ground PP MPLs [66]. These studies performed with more realistic MPLs tend to converge towards those performed with commercial microbeads. The types of damage observed appear similar, including pro-inflammatory, pro-oxidative, barrier-disruptive and dysbiotic effects. However, to date, the available data are too few and the protocols are too heterogeneous to determine whether real MPLs have increased or reduced toxicity compared to commercial microbeads.
Another important knowledge gap comes from the scarcity of research on the effects of mixtures of the various polymers. Except for the dysbiotic impact induced in rats by co-exposure to PP, PET, PS, rayon, PE, POM, PC, PA, PVC and PU [71], the cocktail effects induced by the ingestion of realistic mixtures of several polymers have been studied very little in mammals.
Another important issue is the individual susceptibility to MNPL toxicity. In addition to most of the studies carried out in healthy adult rodents, a few studies were performed in the presence of susceptibility factors. They suggested that the intestinal toxicity of MPLs could be worsened in undernourished patients as well as in patients with obesity [33,48]. Because MPLs are more likely to enter the intestinal epithelial cells in patients with leaky gut and PS MPLs exacerbated colitis in murine models [26,34], MPLs were proposed as emerging risk factors for inflammatory bowel disease [80]. Consistent with this hypothesis, Yan et al. showed that the fecal MPL concentration in IBD patients was significantly higher than that in healthy individuals. They also revealed a positive correlation between the fecal MPL concentration and the severity of IBD, suggesting that MPL exposure may be related to the disease’s development [18]. In IBD as in irritable bowel syndrome (IBS), the increased gut permeability may favor MNPLs crossing the gut barrier, inducing a vicious circle maintaining the barrier defects and subsequently the intestinal inflammation. MNPL exposure may also exacerbate IBD or IBS pathology by worsening dysbiosis, dysregulating the intestinal immune response or exerting direct inflammatory impacts and metabolic toxicity in intestinal epithelial cells, as supported by several in vitro studies on intestinal cell lines or organoids [81,82,83]. Of particular interest, a hyperproliferative effect induced by MPLs under basal conditions and a worsening of established colon cancer progress induced by NPL exposure have been reported, suggesting that MNPLs could favor the development of colorectal cancer [37,60,72]. These in vivo findings were reinforced by in vitro data showing that MNPLs may promote colorectal cancer by increasing the propensity for cell migration and the potential for pro-metastatic effects in human gastrointestinal cancer cells [84]. Lastly, it is worthy of note that gestational exposure to PS NPLs was reported to induce small intestine histological changes, oxidative damage and ferroptosis initiation in female and male offspring [38]. Extraintestinal health outcomes have been reported following in utero exposure to MNPLs in mice, further supporting the need to assess the transgenerational effect on MNPL ingestion on gut health [85,86,87].

4. Conclusions and Future Directions

During the last three years, it has been shown that in rodents, the ingestion of MNPLs impacts the main parameters of intestinal homeostasis, which are the barrier function, the immune response, the oxidative status and the balance of the intestinal microbiota. It is established that MNPL ingestion in healthy subjects disrupts intestinal functions, and critical studies showed that these disruptions could contribute to making individuals more susceptible to the development of inflammation, cancer or infections. However, despite these numerous advances, our knowledge of the intestinal impact of MNPLs remains limited. In order to address MNPLs in their whole physico-chemical complexity and to decipher their effects in various physiological and pathological states, more collaborative efforts must be developed involving experts from diverse fields (e.g., polymer chemistry, analytical chemistry, toxicology, pathophysiology). Future studies will need to better investigate the impact of realistic MNPLs by taking into consideration their diversity in terms of polymer type, shape and size. Above all, the presence in MNPLs of additives and contaminants that are likely to influence the overall toxicity of MNPLs must be better investigated. It is also essential to decipher the impact of MNPL ingestion in individuals weakened by an underlying pathology, particularly pathologies in which the intestinal barrier function is impaired, including inflammatory bowel diseases, irritable bowel syndrome and colorectal cancer [88], but also extraintestinal diseases such as metabolic and neurological diseases [89,90]. Finally, since pioneering studies have shown that gestational exposure to MNPLs could be hazardous for offspring, particular attention should be paid to the effects of MNPLs as determinants of health at early ages but also later in life.

Author Contributions

M.B.-M.: writing—original draft preparation, supervision, funding acquisition; M.D. and S.L.: writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by ANR-22-CE34-0002 and by the French Region Hauts-de-France.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Plastics—The Fast Facts 2023. Plastics Europe. Plast. Eur. Available online: https://plasticseurope.org/knowledge-hub/plastics-the-fast-facts-2023/ (accessed on 1 January 2024).
  2. Thacharodi, A.; Meenatchi, R.; Hassan, S.; Hussain, N.; Bhat, M.A.; Arockiaraj, J.; Ngo, H.H.; Le, Q.H.; Pugazhendhi, A. Microplastics in the Environment: A Critical Overview on Its Fate, Toxicity, Implications, Management, and Bioremediation Strategies. J. Environ. Manag. 2024, 349, 119433. [Google Scholar] [CrossRef]
  3. Chamas, A.; Moon, H.; Zheng, J.; Qiu, Y.; Tabassum, T.; Jang, J.H.; Abu-Omar, M.; Scott, S.L.; Suh, S. Degradation Rates of Plastics in the Environment. ACS Sustain. Chem. Eng. 2020, 8, 3494–3511. [Google Scholar] [CrossRef]
  4. Sources, Fate and Effects of Microplastics in the Marine Environment (Part 1). Available online: http://www.gesamp.org/publications/reports-and-studies-no-90 (accessed on 14 December 2023).
  5. Gigault, J.; ter Halle, A.; Baudrimont, M.; Pascal, P.-Y.; Gauffre, F.; Phi, T.-L.; El Hadri, H.; Grassl, B.; Reynaud, S. Current Opinion: What Is a Nanoplastic? Environ. Pollut. 2018, 235, 1030–1034. [Google Scholar] [CrossRef]
  6. Domenech, J.; Marcos, R. Pathways of Human Exposure to Microplastics, and Estimation of the Total Burden. Curr. Opin. Food Sci. 2021, 39, 144–151. [Google Scholar] [CrossRef]
  7. Kibria, G.; Nugegoda, D.; Haroon, A.K.Y. Microplastic Pollution and Contamination of Seafood (Including Fish, Sharks, Mussels, Oysters, Shrimps and Seaweeds): A Global Overview. In Microplastic Pollution: Environmental Occurrence and Treatment Technologies; Hashmi, M.Z., Ed.; Emerging Contaminants and Associated Treatment Technologies; Springer International Publishing: Cham, Switzerland, 2022; pp. 277–322. ISBN 978-3-030-89220-3. [Google Scholar]
  8. Liu, Y.; Guo, R.; Zhang, S.; Sun, Y.; Wang, F. Uptake and Translocation of Nano/Microplastics by Rice Seedlings: Evidence from a Hydroponic Experiment. J. Hazard. Mater. 2022, 421, 126700. [Google Scholar] [CrossRef]
  9. Hua, Z.; Zhang, T.; Luo, J.; Bai, H.; Ma, S.; Qiang, H.; Guo, X. Internalization, Physiological Responses and Molecular Mechanisms of Lettuce to Polystyrene Microplastics of Different Sizes: Validation of Simulated Soilless Culture. J. Hazard. Mater. 2024, 462, 132710. [Google Scholar] [CrossRef] [PubMed]
  10. Toussaint, B.; Raffael, B.; Angers-Loustau, A.; Gilliland, D.; Kestens, V.; Petrillo, M.; Rio-Echevarria, I.M.; Van den Eede, G. Review of Micro- and Nanoplastic Contamination in the Food Chain. Food Addit. Contam. Part Chem. Anal. Control Expo. Risk Assess. 2019, 36, 639–673. [Google Scholar] [CrossRef]
  11. Kadac-Czapska, K.; Knez, E.; Grembecka, M. Food and Human Safety: The Impact of Microplastics. Crit. Rev. Food Sci. Nutr. 2022, 0, 1–20. [Google Scholar] [CrossRef] [PubMed]
  12. Zuri, G.; Karanasiou, A.; Lacorte, S. Microplastics: Human Exposure Assessment through Air, Water, and Food. Environ. Int. 2023, 179, 108150. [Google Scholar] [CrossRef]
  13. Zhu, J.; Zhang, X.; Liao, K.; Wu, P.; Jin, H. Microplastics in Dust from Different Indoor Environments. Sci. Total Environ. 2022, 833, 155256. [Google Scholar] [CrossRef]
  14. Nematollahi, M.J.; Zarei, F.; Keshavarzi, B.; Zarei, M.; Moore, F.; Busquets, R.; Kelly, F.J. Microplastic Occurrence in Settled Indoor Dust in Schools. Sci. Total Environ. 2022, 807, 150984. [Google Scholar] [CrossRef]
  15. 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] [PubMed]
  16. Luqman, A.; Nugrahapraja, H.; Wahyuono, R.A.; Islami, I.; Haekal, M.H.; Fardiansyah, Y.; Putri, B.Q.; Amalludin, F.I.; Rofiqa, E.A.; Götz, F.; et al. Microplastic Contamination in Human Stools, Foods, and Drinking Water Associated with Indonesian Coastal Population. Environments 2021, 8, 138. [Google Scholar] [CrossRef]
  17. 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] [PubMed]
  18. Yan, Z.; Liu, Y.; Zhang, T.; Zhang, F.; Ren, H.; Zhang, Y. Analysis of Microplastics in Human Feces Reveals a Correlation between Fecal Microplastics and Inflammatory Bowel Disease Status. Environ. Sci. Technol. 2022, 56, 414–421. [Google Scholar] [CrossRef]
  19. 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]
  20. 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]
  21. Liu, S.; Liu, X.; Guo, J.; Yang, R.; Wang, H.; Sun, Y.; Chen, B.; Dong, R. The Association between Microplastics and Microbiota in Placentas and Meconium: The First Evidence in Humans. Environ. Sci. Technol. 2022, 57, 17774–17785. [Google Scholar] [CrossRef]
  22. Ibrahim, Y.S.; Anuar, S.T.; Azmi, A.A.; Khalik, W.M.A.W.M.; Lehata, S.; Hamzah, S.R.; Ismail, D.; Ma, Z.F.; Dzulkarnaen, A.; Zakaria, Z.; et al. Detection of Microplastics in Human Colectomy Specimens. JGH Open 2021, 5, 116–121. [Google Scholar] [CrossRef]
  23. Cetin, M.; Demirkaya Miloglu, F.; Kilic Baygutalp, N.; Ceylan, O.; Yildirim, S.; Eser, G.; Gul, H.İ. Higher Number of Microplastics in Tumoral Colon Tissues from Patients with Colorectal Adenocarcinoma. Environ. Chem. Lett. 2023, 21, 639–646. [Google Scholar] [CrossRef]
  24. Hirt, N.; Body-Malapel, M. Immunotoxicity and Intestinal Effects of Nano- and Microplastics: A Review of the Literature. Part. Fibre Toxicol. 2020, 17, 57. [Google Scholar] [CrossRef] [PubMed]
  25. Paul, M.B.; Stock, V.; Cara-Carmona, J.; Lisicki, E.; Shopova, S.; Fessard, V.; Braeuning, A.; Sieg, H.; Böhmert, L. Micro- and Nanoplastics—Current State of Knowledge with the Focus on Oral Uptake and Toxicity. Nanoscale Adv. 2020, 2, 4350–4367. [Google Scholar] [CrossRef] [PubMed]
  26. Zheng, H.; Wang, J.; Wei, X.; Chang, L.; Liu, S. Proinflammatory Properties and Lipid Disturbance of Polystyrene Microplastics in the Livers of Mice with Acute Colitis. Sci. Total Environ. 2021, 750, 143085. [Google Scholar] [CrossRef] [PubMed]
  27. Qiao, J.; Chen, R.; Wang, M.; Bai, R.; Cui, X.; Liu, Y.; Wu, C.; Chen, C. Perturbation of Gut Microbiota Plays an Important Role in Micro/Nanoplastics-Induced Gut Barrier Dysfunction. Nanoscale 2021, 13, 8806–8816. [Google Scholar] [CrossRef]
  28. 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] [PubMed]
  29. Schwarzfischer, M.; Niechcial, A.; Lee, S.S.; Sinnet, B.; Wawrzyniak, M.; Laimbacher, A.; Atrott, K.; Manzini, R.; Morsy, Y.; Häfliger, J.; et al. Ingested Nano- and Microsized Polystyrene Particles Surpass the Intestinal Barrier and Accumulate in the Body. NanoImpact 2022, 25, 100374. [Google Scholar] [CrossRef] [PubMed]
  30. Xiao, J.; Jiang, X.; Zhou, Y.; Sumayyah, G.; Zhou, L.; Tu, B.; Qin, Q.; Qiu, J.; Qin, X.; Zou, Z.; et al. Results of a 30-Day Safety Assessment in Young Mice Orally Exposed to Polystyrene Nanoparticles. Environ. Pollut. 2022, 292, 118184. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, S.; Li, H.; Wang, J.; Wu, B.; Guo, X. Polystyrene Microplastics Aggravate Inflammatory Damage in Mice with Intestinal Immune Imbalance. Sci. Total Environ. 2022, 833, 155198. [Google Scholar] [CrossRef] [PubMed]
  32. Wen, S.; Zhao, Y.; Liu, S.; Chen, Y.; Yuan, H.; Xu, H. Polystyrene Microplastics Exacerbated Liver Injury from Cyclophosphamide in Mice: Insight into Gut Microbiota. Sci. Total Environ. 2022, 840, 156668. [Google Scholar] [CrossRef]
  33. Huang, D.; Zhang, Y.; Long, J.; Yang, X.; Bao, L.; Yang, Z.; Wu, B.; Si, R.; Zhao, W.; Peng, C.; et al. Polystyrene Microplastic Exposure Induces Insulin Resistance in Mice via Dysbacteriosis and Pro-Inflammation. Sci. Total Environ. 2022, 838, 155937. [Google Scholar] [CrossRef]
  34. Luo, T.; Wang, D.; Zhao, Y.; Li, X.; Yang, G.; Jin, Y. Polystyrene Microplastics Exacerbate Experimental Colitis in Mice Tightly Associated with the Occurrence of Hepatic Inflammation. Sci. Total Environ. 2022, 844, 156884. [Google Scholar] [CrossRef]
  35. Chen, W.; Tu, P.; Ye, X.; Tang, Q.; Yu, T.; Zheng, X. Cyanidin-3-O-Glucoside Impacts Fecal Discharge of Polystyrene Microplastics in Mice: Potential Role of Microbiota-Derived Metabolites. Toxicol. Appl. Pharmacol. 2022, 453, 116212. [Google Scholar] [CrossRef]
  36. Deng, Y.; Chen, H.; Huang, Y.; Zhang, Y.; Ren, H.; Fang, M.; Wang, Q.; Chen, W.; Hale, R.C.; Galloway, T.S.; et al. Long-Term Exposure to Environmentally Relevant Doses of Large Polystyrene Microplastics Disturbs Lipid Homeostasis via Bowel Function Interference. Environ. Sci. Technol. 2022, 56, 15805–15817. [Google Scholar] [CrossRef] [PubMed]
  37. Xie, S.; Zhang, R.; Li, Z.; Liu, C.; Chen, Y.; Yu, Q. Microplastics Perturb Colonic Epithelial Homeostasis Associated with Intestinal Overproliferation, Exacerbating the Severity of Colitis. Environ. Res. 2023, 217, 114861. [Google Scholar] [CrossRef] [PubMed]
  38. Tang, J.; Bu, W.; Hu, W.; Zhao, Z.; Liu, L.; Luo, C.; Wang, R.; Fan, S.; Yu, S.; Wu, Q.; et al. Ferroptosis Is Involved in Sex-Specific Small Intestinal Toxicity in the Offspring of Adult Mice Exposed to Polystyrene Nanoplastics during Pregnancy. ACS Nano 2023, 17, 2440–2449. [Google Scholar] [CrossRef] [PubMed]
  39. Okamura, T.; Hamaguchi, M.; Hasegawa, Y.; Hashimoto, Y.; Majima, S.; Senmaru, T.; Ushigome, E.; Nakanishi, N.; Asano, M.; Yamazaki, M.; et al. Oral Exposure to Polystyrene Microplastics of Mice on a Normal or High-Fat Diet and Intestinal and Metabolic Outcomes. Environ. Health Perspect. 2023, 131, 027006. [Google Scholar] [CrossRef]
  40. Gao, B.; Shi, X.; Li, S.; Xu, W.; Gao, N.; Shan, J.; Shen, W. Size-Dependent Effects of Polystyrene Microplastics on Gut Metagenome and Antibiotic Resistance in C57BL/6 Mice. Ecotoxicol. Environ. Saf. 2023, 254, 114737. [Google Scholar] [CrossRef] [PubMed]
  41. Tu, P.; Xue, J.; Niu, H.; Tang, Q.; Mo, Z.; Zheng, X.; Wu, L.; Chen, Z.; Cai, Y.; Wang, X. Deciphering Gut Microbiome Responses upon Microplastic Exposure via Integrating Metagenomics and Activity-Based Metabolomics. Metabolites 2023, 13, 530. [Google Scholar] [CrossRef] [PubMed]
  42. Xu, D.; Ma, Y.; Peng, C.; Gan, Y.; Wang, Y.; Chen, Z.; Han, X.; Chen, Y. Differently Surface-Labeled Polystyrene Nanoplastics at an Environmentally Relevant Concentration Induced Crohn’s Ileitis-like Features via Triggering Intestinal Epithelial Cell Necroptosis. Environ. Int. 2023, 176, 107968. [Google Scholar] [CrossRef]
  43. Chen, X.; Xu, L.; Chen, Q.; Su, S.; Zhuang, J.; Qiao, D. Polystyrene Micro- and Nanoparticles Exposure Induced Anxiety-like Behaviors, Gut Microbiota Dysbiosis and Metabolism Disorder in Adult Mice. Ecotoxicol. Environ. Saf. 2023, 259, 115000. [Google Scholar] [CrossRef]
  44. Huang, H.; Wei, F.; Qiu, S.; Xing, B.; Hou, J. Polystyrene Microplastics Trigger Adiposity in Mice by Remodeling Gut Microbiota and Boosting Fatty Acid Synthesis. Sci. Total Environ. 2023, 890, 164297. [Google Scholar] [CrossRef] [PubMed]
  45. Zolotova, N.; Dzhalilova, D.; Tsvetkov, I.; Makarova, O. Influence of Microplastics on Morphological Manifestations of Experimental Acute Colitis. Toxics 2023, 11, 730. [Google Scholar] [CrossRef] [PubMed]
  46. Meng, X.; Ge, L.; Zhang, J.; Xue, J.; Gonzalez-Gil, G.; Vrouwenvelder, J.S.; Li, Z. Systemic Effects of Nanoplastics on Multi-Organ at the Environmentally Relevant Dose: The Insights in Physiological, Histological, and Oxidative Damages. Sci. Total Environ. 2023, 892, 164687. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, Z.; Weng, Y.; Shen, Q.; Zhao, Y.; Luo, T.; Xiao, Y.; Yang, G.; Jin, Y. Nano- and Micro-Polystyrene Plastics Interfered the Gut Barrier Function Mediated by Exosomal miRNAs in Rats. Environ. Pollut. 2023, 335, 122275. [Google Scholar] [CrossRef] [PubMed]
  48. Lv, W.; Shen, Y.; Xu, S.; Wu, B.; Zhang, Z.; Liu, S. Underestimated Health Risks: Dietary Restriction Magnify the Intestinal Barrier Dysfunction and Liver Injury in Mice Induced by Polystyrene Microplastics. Sci. Total Environ. 2023, 898, 165502. [Google Scholar] [CrossRef]
  49. Zhang, Z.; Xu, M.; Wang, L.; Gu, W.; Li, X.; Han, Z.; Fu, X.; Wang, X.; Li, X.; Su, Z. Continuous Oral Exposure to Micro- and Nanoplastics Induced Gut Microbiota Dysbiosis, Intestinal Barrier and Immune Dysfunction in Adult Mice. Environ. Int. 2023, 182, 108353. [Google Scholar] [CrossRef]
  50. Li, L.; Lv, X.; He, J.; Zhang, L.; Li, B.; Zhang, X.; Liu, S.; Zhang, Y. Chronic Exposure to Polystyrene Nanoplastics Induces Intestinal Mechanical and Immune Barrier Dysfunction in Mice. Ecotoxicol. Environ. Saf. 2024, 269, 115749. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, Z.; Chen, W.; Chan, H.; Peng, J.; Zhu, P.; Li, J.; Jiang, X.; Zhang, Z.; Wang, Y.; Tan, Z.; et al. Polystyrene Microplastics Induce Size-Dependent Multi-Organ Damage in Mice: Insights into Gut Microbiota and Fecal Metabolites. J. Hazard. Mater. 2023, 461, 132503. [Google Scholar] [CrossRef] [PubMed]
  52. Zeng, G.; Li, J.; Wang, Y.; Su, J.; Lu, Z.; Zhang, F.; Ding, W. Polystyrene Microplastic-Induced Oxidative Stress Triggers Intestinal Barrier Dysfunction via the NF-κB/NLRP3/IL-1β/MCLK Pathway. Environ. Pollut. 2024, 345, 123473. [Google Scholar] [CrossRef]
  53. Teng, M.; Zhao, X.; Zhou, L.; Yan, H.; Zhao, L.; Sun, J.; Li, Y.; Zhu, W.; Wu, F. An Integrated Analysis of the Fecal Metabolome and Metagenome Reveals the Distinct Effects of Differentially Charged Nanoplastics on the Gut Microbiota-Associated Metabolites in Mice. Sci. Total Environ. 2024, 906, 167287. [Google Scholar] [CrossRef]
  54. Zhang, Y.; Jia, Z.; Gao, X.; Zhao, J.; Zhang, H. Polystyrene Nanoparticles Induced Mammalian Intestine Damage Caused by Blockage of BNIP3/NIX-Mediated Mitophagy and Gut Microbiota Alteration. Sci. Total Environ. 2024, 907, 168064. [Google Scholar] [CrossRef]
  55. Zha, H.; Tang, R.; Li, S.; Zhuge, A.; Xia, J.; Lv, J.; Wang, S.; Wang, K.; Zhang, H.; Li, L. Effects of Partial Reduction of Polystyrene Micro-Nanoplastics on the Immunity, Gut Microbiota and Metabolome of Mice. Chemosphere 2024, 349, 140940. [Google Scholar] [CrossRef] [PubMed]
  56. Kim, D.H.; Lee, S.; Ahn, J.; Kim, J.H.; Lee, E.; Lee, I.; Byun, S. Transcriptomic and Metabolomic Analysis Unveils Nanoplastic-Induced Gut Barrier Dysfunction via STAT1/6 and ERK Pathways. Environ. Res. 2024, 249, 118437. [Google Scholar] [CrossRef]
  57. Lee, S.-H.; Lin, W.-Y.; Cheng, T.-J. Microbiota-Mediated Metabolic Perturbations in the Gut and Brain of Mice after Microplastic Exposure. Chemosphere 2024, 350, 141026. [Google Scholar] [CrossRef]
  58. Sun, H.; Chen, N.; Yang, X.; Xia, Y.; Wu, D. Effects Induced by Polyethylene Microplastics Oral Exposure on Colon Mucin Release, Inflammation, Gut Microflora Composition and Metabolism in Mice. Ecotoxicol. Environ. Saf. 2021, 220, 112340. [Google Scholar] [CrossRef] [PubMed]
  59. Tong, X.; Li, B.; Li, J.; Li, L.; Zhang, R.; Du, Y.; Zhang, Y. Polyethylene Microplastics Cooperate with Helicobacter Pylori to Promote Gastric Injury and Inflammation in Mice. Chemosphere 2022, 288, 132579. [Google Scholar] [CrossRef]
  60. 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]
  61. Yang, Q.; Dai, H.; Cheng, Y.; Wang, B.; Xu, J.; Zhang, Y.; Chen, Y.; Xu, F.; Ma, Q.; Lin, F.; et al. Oral Feeding of Nanoplastics Affects Brain Function of Mice by Inducing Macrophage IL-1 Signal in the Intestine. Cell Rep. 2023, 42, 112346. [Google Scholar] [CrossRef]
  62. Wang, J.; Tian, H.; Shi, Y.; Yang, Y.; Yu, F.; Cao, H.; Gao, L.; Liu, M. The Enhancement in Toxic Potency of Oxidized Functionalized Polyethylene-Microplastics in Mice Gut and Caco-2 Cells. Sci. Total Environ. 2023, 903, 166057. [Google Scholar] [CrossRef] [PubMed]
  63. Xu, R.; Cao, J.; Lv, H.; Geng, Y.; Guo, M. Polyethylene Microplastics Induced Gut Microbiota Dysbiosis Leading to Liver Injury via the TLR2/NF-κB/NLRP3 Pathway in Mice. Sci. Total Environ. 2024, 917, 170518. [Google Scholar] [CrossRef]
  64. Chen, X.; Zhuang, J.; Chen, Q.; Xu, L.; Yue, X.; Qiao, D. Polyvinyl Chloride Microplastics Induced Gut Barrier Dysfunction, Microbiota Dysbiosis and Metabolism Disorder in Adult Mice. Ecotoxicol. Environ. Saf. 2022, 241, 113809. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, X.; Zhuang, J.; Chen, Q.; Xu, L.; Yue, X.; Qiao, D. Chronic Exposure to Polyvinyl Chloride Microplastics Induces Liver Injury and Gut Microbiota Dysbiosis Based on the Integration of Liver Transcriptome Profiles and Full-Length 16S rRNA Sequencing Data. Sci. Total Environ. 2022, 839, 155984. [Google Scholar] [CrossRef] [PubMed]
  66. 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. [Google Scholar] [CrossRef] [PubMed]
  67. Lin, X.; Xie, H.; Zhang, Y.; Tian, X.; Cui, L.; Shi, N.; Wang, L.; Zhao, J.; An, L.; Wang, J.; et al. The Toxicity of Nano Polyethylene Terephthalate to Mice: Intestinal Obstruction, Growth Retardant, Gut Microbiota Dysbiosis and Lipid Metabolism Disorders. Food Chem. Toxicol. 2023, 172, 113585. [Google Scholar] [CrossRef] [PubMed]
  68. Harusato, A.; Seo, W.; Abo, H.; Nakanishi, Y.; Nishikawa, H.; Itoh, Y. Impact of Particulate Microplastics Generated from Polyethylene Terephthalate on Gut Pathology and Immune Microenvironments. iScience 2023, 26, 106474. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, L.; Chen, T.; Liu, J.; Hou, Y.; Tan, Q.; Zhang, X.; Li, Z.; Farooq, T.H.; Yan, W.; Li, Y. Intestinal Flora Variation Reflects the Short-Term Damage of Microplastic to the Intestinal Tract in Mice. Ecotoxicol. Environ. Saf. 2022, 246, 114194. [Google Scholar] [CrossRef] [PubMed]
  70. Toto, B.; Refosco, A.; O’Keeffe, M.; Barkhald, Ø.H.; Brønstad, A.; Lied, G.A.; Yadetie, F.; Goksøyr, A.; Kögel, T.; Dierkes, J. Intestinal Permeability and Gene Expression after Polyethylene and Polyamide Microplastic Ingestion in Wistar Rats. Toxicol. Lett. 2022, 370, 35–41. [Google Scholar] [CrossRef] [PubMed]
  71. Zhao, N.; Zhao, M.; Jin, H. Microplastic-Induced Gut Microbiota and Serum Metabolic Disruption in Sprague-Dawley Rats. Environ. Pollut. 2023, 320, 121071. [Google Scholar] [CrossRef]
  72. Yang, Q.; Dai, H.; Wang, B.; Xu, J.; Zhang, Y.; Chen, Y.; Ma, Q.; Xu, F.; Cheng, H.; Sun, D.; et al. Nanoplastics Shape Adaptive Anticancer Immunity in the Colon in Mice. Nano Lett. 2023, 23, 3516–3523. [Google Scholar] [CrossRef]
  73. Zhuang, J.; Chen, Q.; Xu, L.; Chen, X. Combined Exposure to Polyvinyl Chloride and Polystyrene Microplastics Induces Liver Injury and Perturbs Gut Microbial and Serum Metabolic Homeostasis in Mice. Ecotoxicol. Environ. Saf. 2023, 267, 115637. [Google Scholar] [CrossRef]
  74. Koelmans, A.A.; Redondo-Hasselerharm, P.E.; Nor, N.H.M.; de Ruijter, V.N.; Mintenig, S.M.; Kooi, M. Risk Assessment of Microplastic Particles. Nat. Rev. Mater. 2022, 7, 138–152. [Google Scholar] [CrossRef]
  75. Zhu, M.; Zeng, R.; Wu, D.; Li, Y.; Chen, T.; Wang, A. Research Progress of the Effects of Bisphenol Analogues on the Intestine and Its Underlying Mechanisms: A Review. Environ. Res. 2023, 243, 117891. [Google Scholar] [CrossRef] [PubMed]
  76. Goyal, S.P.; Saravanan, C. An Insight into the Critical Role of Gut Microbiota in Triggering the Phthalate-Induced Toxicity and Its Mitigation Using Probiotics. Sci. Total Environ. 2023, 904, 166889. [Google Scholar] [CrossRef] [PubMed]
  77. Hu, L.; Zhao, Y.; Xu, H. Trojan Horse in the Intestine: A Review on the Biotoxicity of Microplastics Combined Environmental Contaminants. J. Hazard. Mater. 2022, 439, 129652. [Google Scholar] [CrossRef]
  78. Wang, M.; Li, Q.; Shi, C.; Lv, J.; Xu, Y.; Yang, J.; Chua, S.L.; Jia, L.; Chen, H.; Liu, Q.; et al. Oligomer Nanoparticle Release from Polylactic Acid Plastics Catalysed by Gut Enzymes Triggers Acute Inflammation. Nat. Nanotechnol. 2023, 18, 403–411. [Google Scholar] [CrossRef] [PubMed]
  79. Gałęcka, I.; Szyryńska, N.; Całka, J. Influence of Polyethylene Terephthalate (PET) Microplastic on Selected Active Substances in the Intramural Neurons of the Porcine Duodenum. Part. Fibre Toxicol. 2024, 21, 5. [Google Scholar] [CrossRef] [PubMed]
  80. Chen, X.; Wang, S.; Mao, X.; Xiang, X.; Ye, S.; Chen, J.; Zhu, A.; Meng, Y.; Yang, X.; Peng, S.; et al. Adverse Health Effects of Emerging Contaminants on Inflammatory Bowel Disease. Front. Public Health 2023, 11, 1140786. [Google Scholar] [CrossRef] [PubMed]
  81. Chen, Y.; Williams, A.M.; Gordon, E.B.; Rudolph, S.E.; Longo, B.N.; Li, G.; Kaplan, D.L. Biological Effects of Polystyrene Micro- and Nano-Plastics on Human Intestinal Organoid-Derived Epithelial Tissue Models without and with M Cells. Nanomed. Nanotechnol. Biol. Med. 2023, 50, 102680. [Google Scholar] [CrossRef] [PubMed]
  82. Xuan, L.; Luo, J.; Qu, C.; Guo, P.; Yi, W.; Yang, J.; Yan, Y.; Guan, H.; Zhou, P.; Huang, R. Predictive Metabolomic Signatures for Safety Assessment of Three Plastic Nanoparticles Using Intestinal Organoids. Sci. Total Environ. 2024, 913, 169606. [Google Scholar] [CrossRef]
  83. Busch, M.; Bredeck, G.; Kämpfer, A.A.; Schins, R.P. Investigations of Acute Effects of Polystyrene and Polyvinyl Chloride Micro-and Nanoplastics in an Advanced in Vitro Triple Culture Model of the Healthy and Inflamed Intestine. Environ. Res. 2021, 193, 110536. [Google Scholar] [CrossRef]
  84. Brynzak-Schreiber, E.; Schögl, E.; Bapp, C.; Cseh, K.; Kopatz, V.; Jakupec, M.A.; Weber, A.; Lange, T.; Toca-Herrera, J.L.; del Favero, G.; et al. Microplastics Role in Cell Migration and Distribution during Cancer Cell Division. Chemosphere 2024, 353, 141463. [Google Scholar] [CrossRef] [PubMed]
  85. 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]
  86. Zhang, Y.; Wang, X.; Zhao, Y.; Zhao, J.; Yu, T.; Yao, Y.; Zhao, R.; Yu, R.; Liu, J.; Su, J. Reproductive Toxicity of Microplastics in Female Mice and Their Offspring from Induction of Oxidative Stress. Environ. Pollut. 2023, 327, 121482. [Google Scholar] [CrossRef] [PubMed]
  87. Huang, T.; Zhang, W.; Lin, T.; Liu, S.; Sun, Z.; Liu, F.; Yuan, Y.; Xiang, X.; Kuang, H.; Yang, B.; et al. Maternal Exposure to Polystyrene Nanoplastics during Gestation and Lactation Induces Hepatic and Testicular Toxicity in Male Mouse Offspring. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2022, 160, 112803. [Google Scholar] [CrossRef] [PubMed]
  88. Dixit, K.; Chaudhari, D.; Dhotre, D.; Shouche, Y.; Saroj, S. Restoration of Dysbiotic Human Gut Microbiome for Homeostasis. Life Sci. 2021, 278, 119622. [Google Scholar] [CrossRef]
  89. Fan, Y.; Pedersen, O. Gut Microbiota in Human Metabolic Health and Disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  90. Zhang, H.; Chen, Y.; Wang, Z.; Xie, G.; Liu, M.; Yuan, B.; Chai, H.; Wang, W.; Cheng, P. Implications of Gut Microbiota in Neurodegenerative Diseases. Front. Immunol. 2022, 13, 785644. [Google Scholar] [CrossRef]
Table 1. Overview of rodent studies of the intestinal toxicity of MNPLs.
Table 1. Overview of rodent studies of the intestinal toxicity of MNPLs.
Sex
Background
Specie
Polymer Type, Shape, Mean Aerodynamic Diameter and Other Specificities of MPLs/NPLsExposure Conditions
Concentration *
Duration
Administration Pathway
Main Alterations ObservedReference
Polystyrene (PS)
Male
C57BL/6
mice
Spherical
5 µm
PS
500 µg/L
so ≈0.11 mg/kg bw/day
4 weeks
Drinking water
Increased intestinal permeability in mice with acute DSS 1-induced colitis[26]
Male
C57BL/6
mice
Spherical
70 nm NPL and 5 µm MPL PS, pristine PS, negatively charged carboxylated PS (PS-COOH) and positively charged aminated PS (PS-NH2)
0.2 and 2 mg/kg bw/day
4 weeks
Gavage
Gut toxicity PS-NH2 > PS-COOH > pristine PS
Decreased expression of tight-junction proteins for PS-NH2 MPL and NPL
Dysbiosis:
MPL > NPL
Chemically modified MNPL > pristine MNPL
[27]
Male
C57BL/6
mice
Spherical
50, 500, 5000 nm individually and combined
PS
2,5, 50, 250, 500 mg/kg bw/day
4 weeks
Gavage
Numerous significant effects on duodenum, jejunum, ileum, colon: dose-dependent and size-dependent, including
Decreased mucus
Increased ROS 2 generation (DHE)
Increased apoptosis (TUNEL)
Increased caspase-3
Increased intestinal permeability (4 and 70 kDa dextran) and decreased Ecad levels
Combined toxicity of 50 and 500 nm NPLs
[28]
Female
C57BL/6 mice
Spherical
50 nm and 1 µm
PS
0.2 mg/mouse
so ≈9 mg/kg bw/day
12 and 23.7 weeks
Drinking water
No effects under basal, acute colitis or chronic colitis conditions[29]
Male
C57BL/6 mice
Spherical
47 nm
PS
0.2, 1 and 10 mg/kg bw/day
4.2 weeks
Gavage
Slight dysbiosis (impact on 3 to 8 taxa/group)
Slight intestinal damage (only Tff3 and Klf3 mRNA decrease at highest dose)
[30]
Male C57BL/6 miceSpherical
5 µm
PS
500 µg/L so
≈0.11 mg/kg bw/day
4 weeks
Drinking water
In colon:
Increased TNFα, IFNγ, IL1β, GPx
Decreased Claudin1 and Occludin1
Worsened DSS 1-induced colitis (colon length)
Decreased glycine and taurine production only under DSS 1-induced conditions
[31]
Male
C57BL/6 mice
Spherical
5 µm
PS
18 and 180 µg/kg bw/day
12.8 weeks
Drinking water
Dysbiosis[32]
Male ICR
mice
Spherical
5, 50, 100 and 200 µm PS
80 mg/kg bw/day, including 20 mg/kg of 5, 50, 100 and 200 µm MPL
10 weeks
Gavage
In normal-diet mice: induction of dysbiosis (including decreased Firmicutes/Bacteroides ratio)
In high-fat-fed (HFD) mice: modification of HFD-induced dysbiosis (including worsening of Enterobacteriaceae abundance increase)
[33]
C57BL/6J mice
(sex not provided)
Spherical
5 µm
PS
0.5 and 5 µg/mouse so
≈0.023 and 0.23 mg/kg bw/day
2 and 9 weeks
Gavage
DSS 1-induced acute and chronic colitis exacerbation[34]
Male C57BL/6 miceSpherical
5 µm
PS
0.1 mg/day so
≈4.5 mg/kg bw/day
6 weeks
Gavage
Alterations in gut microbiota and metabolites[35]
Male C57BL/6 miceCrushed
51 and 88 µm
PS
50 and 500 mg/kg bw/day
3 weeks
Feed
Decreased SI 3 mucus layer thickness
Increased SI 3 epithelial injury
Fecal dysbiosis
Host plasma lipid metabolism disturbance
[36]
Male C57BL/6 miceSpherical
5 µm
PS
100 µg/L
so ≈0.02 mg/kg bw/day
6 weeks
Drinking water
In colon:
Decreased goblet cell number
Increased crypt depth and density
Decreased Tff3 and Muc2 mRNA
Increased Il1β, Il6, Dll1, Dll4, Jag1, Notch1, Hes1, Lgr5, Bmi1, Olfm4 and c-Myc mRNA levels
Increased c-MYC and PCNA protein levels
Worsened DSS 1-induced colitis (body weight, colon length, histological score, serum LPS, colon Il1β and Il6)
[37]
Male and female
C57BL/6
mice
Spherical
71 nm
PS
50, 250 or 1250 µg/mouse/day
So ≈2.3, 11.5 or 57.5 mg/kg bw/day
7.52 × 1011, 3.76 × 1012, and 1.88 × 1013 particles/day
3 times per week during gestation
Oropharyngeal aspiration
In adult male and female offspring:
Duodenum, jejunum and ileum histomorphological alterations in female and male mice
Small intestine oxidative stress
Small intestine ferroptosis
[38]
Male
C57BL/6 mice
Spherical
0.45–0.53µm
PS COOH
1000 µg/L
so ≈0.22 mg/kg bw/day
4 weeks
Drinking water
On SI of high-fat-diet-fed mice:
Decreased goblet cells
Decreased mucus thickness
Decreased villus height and higher crypt depth
Increased innate lymphoid cells ILC1 and T bet+ ILC3 cells
Increased M1/M2 macrophage ratio
Decreased ILC3 cells
Decreased palmitic acid, acetic acid, propionic acid and butanoic acid
in feces
Increased inflammatory cytokines and lipid metabolism gene expression
[39]
Male
C57BL/6 mice
Spherical
0.05–0.1 µm
and 9–10 µm
PS
1 ppm, 1 mg/L
so ≈0.22 mg/kg bw/day
12 weeks
Drinking water
Size-dependent
Gut dysbiosis (bacteria and fungi)
Disturbed microbial metabolic pathways
Altered microbial antibiotic resistance genes and virulence factors
[40]
Male C57BL/6 miceSpherical
5 µm
PS
0.1 mg/mouse
so ≈4.5 mg/kg bw/day
6 weeks
Gavage
Gut dysbiosis and variations in predicted functional pathways
Modifications to metabolite profiles (bile acid metabolism enrichment)
Increased bile acids, decreased purine and pyrimidine nucleosides
Decreased fecal levels of acetic acid, propionic acid, butyric acid and
isobutyric acid (SCFA)
[41]
Male
BALB/c mice
Spherical
100 nm
PS,
PS-COOH
and PS-NH2
1 mg/mouse/day
so ≈45.5 mg/kg bw/day
4 weeks
Gavage
Decreased villus length
Increased crypt depth and lower ratio of villus length to crypt depth
Increased the secretion of mucus
in ileum
Increased IL-17a-positive cells
in the ileal lamina propria
Increased ileal apoptosis (TUNEL)
Increased necroptosis (RIPK3 and MLCK protein expression)
Increased mitophagy (PINK1, PARKIN, SQSTM1/p62, LC3B, TOMM20 protein expression)
[42]
Male C57BL/6 miceSpherical
100 nm NPL and 1 µm MPL
PS
0.5 mg/mouse
so ≈22.7 mg/kg bw/day
8.5 weeks
Gavage
Decreased colon mucus secretion
Increased gut permeability (FITC dextran)
Size-dependent dysbiosis
Altered serum metabolites
[43]
Male C57BL/6 miceSpherical
5 µm
PS
0.001, 0.1, 1 and 10 µg/mL
so ≈0.002, 0.02, 0.2 and 2 mg/kg/day
10 weeks
Drinking water
Dysbiosis
from the 0.1 µg/mL concentration
[44]
Male C57BL/6 miceSpherical
5 µm
PS
10 mg/L
so ≈2.3 mg/kg bw/day
6 weeks
Drinking water
In distal colon of healthy mice:
Increase in the number of endocrine cells
Increase in the content of highly
sulfated mucins in goblet cells
Increase in the number of cells in the lamina propria
Decrease in the volume fraction of
macrophages
Worsening of DSS 1-induced colitis:
greater prevalence of ulcers and inflammation; decrease in the content of neutral mucins in goblet cells
[45]
KN
Mice
(sex not provided)
Spherical
300 nm
PS
12 and 500 mg/kg bw/day
4 weeks
Gavage
Dose-dependent
Increased gut permeability
Increased ileum Muc2 expression
Decreased jejunum and ileum villus number and length
Decreased colon mucus thickness and goblet cells number
Ileum and jejunum oxidative stress
[46]
Male
Wistar
rat
Spherical
50 nm
and
5 µm
PS
0.1 and 1 mg/kg bw/day
4 weeks
Gavage
Higher for MPLs than NPLs:
Decreased colon mucus secretion and MUC2 expression
Decreased colon ZO-1 and occludin expression
For both MPLs and NPLs: decreased colon exosomal miR-126a expression
[47]
Male C57/BL/6
mice
Spherical
5 µm
PS
200 µg/L
so ≈0.04 mg/kg bw/day
5 weeks
Drinking water
With normal diet:
Increased serum Diamine oxidase (DAO), D-Lactate, D-Lactate dehydrogenase (D-LDH) and intestinal fatty acid-binding protein (IFABP)
Decreased ileum secretory IgA
Fecal dysbiosis
Worsened with dietary restriction:
Decreased ileum mucus secretion
Increased serum DAO, D-Lactate, IFABP and TNF-α levels
[48]
Female
BALB/C
mice
Spherical
20, 500, 5000 nm
PS
6, 60, 600 µg/kg bw/day
2 or 4 weeks
Gavage
Gut microbiota composition alterations
Variation in short-chain
fatty acid levels
Modifications to intestinal permeability (decreased after 2 weeks,
increased after 4 weeks)
Increased leukocytes in SI (4 weeks)
Decreased secretory IgA levels
Decreased CD4+ and CD8+ T cells
in MLN 4
[49]
Female C57BL/6 miceSpherical
50 nm
PS
0.1, 1 or 10 mg/L
so ≈0.02, 0.23 or 2.3 mg/kg bw/day
32 weeks
Drinking water
Increased caveolin and clathrin levels (endocytosis proteins)
Histological damage in jejunum, ileum, colon
Decreased Claudin-1, Occludin, ZO-1 levels
Increased ROS2 and MDA levels
Decreased SOD and GSH-Px levels
Increased B lymphocytes in MLN4
Decreased γδ+CD8+ and CD3+ CD8+ T cells in intestine
Increased intestinal mucosal IL1β, IL-6 and TNF-α.
[50]
Male
C57BL/6
mice
Spherical
0.5 and 5 µm
PS
0.5 mg/mouse/day
so ≈22.7 mg/kg bw/day
8 weeks
Gavage
Decreased colon length (5 µm only)
Increased colon IL6, TNFα, Il1β secretion (higher for 5 µm than 0.5 µm)
Dysbiosis and variation in fecal metabolome (higher for 5 µm than 0.5 µm)
[51]
Male
C57BL/6
mice
Spherical
0.2, 1 and 5 µm
PS
1 mg/kg bw/day
4 weeks
Gavage
In colon:
5 µm PS:
Oxidative stress
Increased inflammatory cytokines
Impaired tight junctions and mucins
Lower impairments for 1 µm
Fewest impairments for 0.2 µm
[52]
Male
ICR
mice
Spherical
44 nm
nonfunctionalized PS,
51 nm PS-NH2,
50 nm PS-COOH
80 µg/L
0.018 mg/kg bw/day
9 weeks
Drinking water
Similar for the 3 NPLs:
Increased colon histological score
Decreased colon Ifnγ, Il6, Il10, Tff3
Increased Tlr3 expression
NPL-specific variations in fecal microbiome and metabolome
[53]
Male
BALB/C
mice
Spherical
140 nm
PS
5 mg/kg bw/day
4 weeks
Gavage
Jejunum and colon mitophagy
(increased secretion of LC3B2/LC3B-1, BNIP3, NIX and p62)
Dysbiosis
[54]
Male
ICR mice
Spherical
99 nm and 5 µm
PS
200 or 500 µg/mouse
so ≈9 or 22.5 mg/kg bw/day
5 weeks
Gavage
Size- and concentration-dependent dysbiotic effects[55]
Male
C57BL/6 mice
Size and shape not provided
PS
NP
0.5 and 1.5 mg/mouse/day
so ≈22.7 and 68
mg/kg bw/day
4 or 6 weeks
Gavage
Activation of pro-inflammatory gene expression (RNA sequencing)
Activation JAK-STAT and
ERK1/2 signaling pathways
Depletion of taurine in colon
Increased gut permeability
(FITC-dextran and reduced
Tight-junction proteins)
DSS 1-induced colitis exacerbation
[56]
Male
C57BL/6
mice
Spherical
200 and 800 nm
Nile-red-labeled PS
109/mouse
3 times per week for 4 weeks
Gavage
Disruptions to cecal microbiome and metabolome[57]
Polyethylene (PE)
Female
ICR
mice
Spherical
1–10 µm
PE
0.002 and 0.2 µg/g bw/day
4 weeks
Gavage
At 0.2 µg dosage:
Decreased colon mucin
Decreased Il1β, ERK1, NF-κB, and increased Il10 and Il8 mRNA
Dysbiosis
[58]
Male
BALB/C
mice
Spherical
61 µm
PE
25 or 50 µg/mouse
so ≈1.25 or 2.5 mg/kg bw/day
3 times in a week
Gavage
Without H. pylori infection:
Gastric pathological damage
Increased gastric IL6 and TNF-α levels
With H. pylori infection:
Increased H. pylori gastric
colonization
Increased gastric injury
Increased gastric inflammation
(MPO, IL6, TNF-α)
[59]
Female C57BL/6 MiceSpherical
36 and 116 µm
PE
100 µg/g feed
so ≈16 mg/kg bw/day
6 weeks
Feed
Colon hyperproliferation
Increased colon mucus and Muc2 expression
Colon inflammation
Whole-gut immune population and
epithelial cell disturbances
Dysbiosis
[60]
BALB/c mice (sex not provided)Polydisperse, grinded
530 and 2300 nm
PE
10 mg/kg bw/day
in 0.5% CMC
1 week
Gavage
Overt colitis
Decreased colon length
Increased colon Il1β, Th2, Treg, Th17 cells
[61]
Male
C57BL/6
mice
Spherical
2.6 to 13 µm
LDPE
and
oxidized LDPE
5 mg/mouse/day
so ≈227 mg/kg bw/day
4 weeks
Gavage
Higher for oxidized LDPE than LDPE
Decreased duodenum length
Increased duodenum and colon crypt depth
Oxidative stress
Increase in Tnfα, Il1β, Il6 expression in duodenum and colon
Same extent for both LDPEs: dysbiosis
[62]
Male
C57BL/6
mice
Spherical
5 µm
PE
1 and 10 mg/L
so ≈0.22 and 2.2
mg/kg bw/day
3 weeks
Drinking water
Concentration-specific
dysbiosis
[63]
Polyvinylchloride (PVC)
Male C57BL/6 MiceSpherical
2 µm
PVC
100 mg/kg bw/day
8.5 weeks
Gavage
Increased intestinal permeability
Decreased mucus secretion
Decreased colon Muc1, Muc2, Muc3, Klf4, Retnlb mRNA expression
Gut dysbiosis
Modification to fecal metabolic profile
[64]
Male C57BL/6 MiceSpherical
2 µm
PVC
0.5 mg/mouse
So ≈22.7 mg/kg bw/day
8.5 weeks
Gavage
Gut dysbiosis[65]
Polypropylene (PP)
Male C57BL/6 MiceIrregular
Grinded
8 and 70 µm
PP
0.1, 1, 10 mg/mL
So ≈22, 227 and 2272 mg/kg bw/day
4 weeks
Gavage
Mild colon submucosa edema
Colon oxidative stress and inflammation
Disruption of intestinal barrier
TLR4/NF-κB signaling pathway
Colon apoptosis
[66]
Polyethylene terephthalate (PET)
Male and female KM miceSpherical
200 nm and 700 nm
PET
200 mg/kg bw/day
4 weeks
Gavage
LD50:
266 mg/kg bw for 200 nm-PET
and 523 mg/kg bw for 700 nm-PET
200 nm PET only:
Intestinal obstruction
Perturbations to gut microbiome and metabolome
[67]
Female
C57BL/6 mice
Pin made
1 µm
PET
3 × 104/mouse
Gavage
8 weeks
In colon:
No evidence of impaired histomorphology and mucus barrier
No low-grade inflammation
139 differentially expressed genes
In gut immune cells:
Oxidative phosphorylation and
reactive oxygen species pathways
enrichment
Dysbiosis
[68]
Several polymer types
Kunming mice
(sex not provided)
Spherical 150–130 µm
PE, PET, PP, PS and PVC
4 mg/mouse/day
So, ≈182 mg/kg/day
7 days
All: colon damage (PS > PVC > PET > PE > PP)
Polymer-specific oxidative stress
Polymer-specific dysbiosis
[69]
Male and female
Wistar rats
Spherical
15–20 µm
polyamide (PA)
40–48 µm PE
0.1%W/W (100 mg/kg) in feed
So, ≈227 mg/kg/day
5 weeks
Feed
Increased duodenum occludin and ZO1 expression[70]
Male
Sprague-Dawley rats
100 µm PP, PET, PS, Rayon, PE, polyoxymethylene (POM), polycarbonate (PC), PA, PVC, polyurethane (PU) mixture12 mg/kg bw/day
6 weeks
Gavage
Dysbiosis[71]
BALB/C mice (sex not provided)Spherical
500 nm
PE
500 nm
PS
10 mg/kg bw/day
1 week
Gavage
Both PE and PS NPL: In colon:
Decreased length
Pathological damage
Increased ROS 2 production
Increased proportion of pro-inflammatory macrophages, Th2, Th17 and Treg cells
PE NPL: promotes the development of CT26-luc cell tumor
[72]
Male
C57BL/6J mice
Spherical
2 µm PVC
1 µm PS
0.5 mg/mouse day
So ≈22.7 mg/kg bw/day
8.5 weeks
Gavage
PS-PVC co-exposure:
Decreased mucus secretion
Increased gut permeability
(FITC-dextran, serum LPS)
Increased colon Il1β, Il6, Tnfα mRNA
Decreased colon
Muc2, Muc3, Klf4, Retnlb, Meprin-β, Claudin5, Claudin4, Tjp1
Dysbiosis
[73]
1 DSS: dextran sodium sulfate. 2 ROS: reactive oxygen species. 3 SI: small intestine. 4 MLN: mesenteric lymph node. * A standardized method according to Zolotova et al. [45] (5 mL drinking water per day for a 22 g mouse) was used to convert concentrations in drinking water to mg/kg bw/day. A 22 g body weight was used to convert concentrations from mg/mouse to mg/kg bw/day.
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

Djouina, M.; Loison, S.; Body-Malapel, M. Recent Progress in Intestinal Toxicity of Microplastics and Nanoplastics: Systematic Review of Preclinical Evidence. Microplastics 2024, 3, 217-233. https://doi.org/10.3390/microplastics3020013

AMA Style

Djouina M, Loison S, Body-Malapel M. Recent Progress in Intestinal Toxicity of Microplastics and Nanoplastics: Systematic Review of Preclinical Evidence. Microplastics. 2024; 3(2):217-233. https://doi.org/10.3390/microplastics3020013

Chicago/Turabian Style

Djouina, Madjid, Suzie Loison, and Mathilde Body-Malapel. 2024. "Recent Progress in Intestinal Toxicity of Microplastics and Nanoplastics: Systematic Review of Preclinical Evidence" Microplastics 3, no. 2: 217-233. https://doi.org/10.3390/microplastics3020013

Article Metrics

Back to TopTop