Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-29T12:22:17.133Z Has data issue: false hasContentIssue false

Microplastics: A threat for developing and repairing organs?

Published online by Cambridge University Press:  20 October 2023

Lars T. Hofstede
Affiliation:
Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
Gwenda F. Vasse
Affiliation:
Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
Barbro N. Melgert*
Affiliation:
Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
*
Corresponding author: Barbro N. Melgert; Email: b.n.melgert@rug.nl
Rights & Permissions [Opens in a new window]

Abstract

Plastic production has greatly increased in the past decades and has become central to modern human life. Realization is dawning that plastics break down into smaller pieces resulting in micro- or nanoplastics (MNP) that can enter humans directly via the environment. Indeed, MNP have been detected in every part of the human body, including the placenta, which is concerning for development. Early developmental stages are crucial for proper growth and genome programming. Environmental disruptors in MNP can have detrimental effects during this critical window as well and can increase the risk of developing disease and dysfunction. In addition, MNP may impact situations in which developmental pathways are reactivated after birth such as during organ repair. Currently, there is no overview of how MNP can impair (human) development and repair. Therefore, we provide an extensive overview of available evidence on MNP impacting developmental and regenerative processes in various organs in humans and rodent models. In addition, we have included the impact of some additives that can leach from these MNP. We conclude that MNP and their additives can have modulating effects on developing and regenerating organs.

Topics structure

Topic(s)

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NC
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial licence (http://creativecommons.org/licenses/by-nc/4.0), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original article is properly cited. The written permission of Cambridge University Press must be obtained prior to any commercial use.
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Impact statement

Plastics have become central to modern human life and have led to the new environmental problem of microplastics. Humans are unavoidably exposed to these microplastics through air, food and water and therefore microplastics have been detected in many different compartments of the body. This exposure could have implications for the function of cells and organs in our bodies, including how they develop and repair themselves when damaged. However, there is currently no comprehensive overview of the effects of microplastics on organ development and processes related to organ repair. Therefore, this review aims to provide an extensive overview of available evidence present in the public domain describing how microplastics and additives leaching from plastics can affect developing and repairing organs. The available studies suggest that we do not have the luxury to be complacent about microplastic pollution anymore, clear effects on development and repair have been found. It is therefore imperative that action is taken to reduce plastic use and prevent further contamination of the environment and ourselves with microplastics.

Introduction

The global production of plastic has surged dramatically from 1.5 million tons in 1950 to over 390 million tons in 2021 (Plastics Europe, 2021). The most widely used polymers include polyethylene, polypropylene, polystyrene, polyvinylchloride and polyamide (Peñalver et al., Reference Peñalver, Arroyo-Manzanares, López-García and Hernández-Córdoba2020). Their popularity arises from their versatility, durability, ease of use and cost-effectiveness (Wijesekara et al., Reference Wijesekara, Bolan, Bradney, Obadamudalige, Seshadri, Kunhikrishnan, Dharmarajan, Ok, Rinklebe, Kirkham and Vithanage2018). Nevertheless, their lack of biodegradability results in them persisting in the environment, causing considerable pollution (Bahl et al., Reference Bahl, Dolma, Jyot Singh and Sehgal2021).

Over time, environmental factors cause plastics to break down into smaller fragments (Gijsman and Dozeman, Reference Gijsman and Dozeman1996; Andrady, Reference Andrady2011; Min et al., Reference Min, Cuiffi and Mathers2020; Rodriguez et al., Reference Rodriguez, Mansoor, Ayoub, Colin and Benzerga2020). When these fragments are reduced to sizes smaller than 5 mm, they are classified as either microplastics (5 mm–1 μm) or nanoplastics (<1 μm) (Hartmann et al., Reference Hartmann, Hüffer, Thompson, Hassellöv, Verschoor, Daugaard, Rist, Karlsson, Brennholt, Cole, Herrling, Hess, Ivleva, Lusher and Wagner2019). These micro- and nanoplastics (MNP) are further categorized, based on their mode of environmental release, into primary MNP specifically produced to be small and secondary MNP originating from degradation of larger plastic waste (Figure 1) (Gijsman and Dozeman, Reference Gijsman and Dozeman1996; Andrady, Reference Andrady2011; Min et al., Reference Min, Cuiffi and Mathers2020; Rodriguez et al., Reference Rodriguez, Mansoor, Ayoub, Colin and Benzerga2020; Allen et al., Reference Allen, Allen, Abbasi, Baker, Bergmann, Brahney, Butler, Duce, Eckhardt, Evangeliou, Jickells, Kanakidou, Kershaw, Laj, Levermore, Li, Liss, Liu, Mahowald, Masque, Materić, Mayes, McGinnity, Osvath, Prather, Prospero, Revell, Sander, Shim, Slade, Stein, Tarasova and Wright2022). As a result, MNP are highly heterogeneous, varying in size, shape and polymer composition (Koelmans et al., Reference Koelmans, Redondo-Hasselerharm, Nor, De Ruijter, Mintenig and Kooi2022).

Figure 1. Microplastics exposure routes. An overview of different microplastic exposure routes. Primary sources include clothes and cosmetics, whereas secondary sources include larger pieces of plastic. Microbeads from cosmetics, microfibers from clothes and smaller plastic particles derived from plastic degradation can enter humans directly via food and/or drinks or via the natural environment. When pregnant women are exposed, a developing fetus can be exposed too. Image created with BioRender.com.

High amounts of MNP have been isolated from all environmental compartments ranging from water to soil and air (Figure 1) (Gasperi et al., Reference Gasperi, Wright, Dris, Collard, Mandin, Guerrouache, Langlois, Kelly and Tassin2018; Li et al., Reference Li, Liu and Paul Chen2018; Zhou et al., Reference Zhou, Wang, Zou, Jia, Zhou and Li2020). In the latter case, MNP have been found both in indoor and outdoor air (Evangeliou et al., Reference Evangeliou, Grythe, Klimont, Heyes, Eckhardt, Lopez-Aparicio and Stohl2020; Zhang et al., Reference Zhang, Wang and Kannan2020; Jenner et al., Reference Jenner, Sadofsky, Danopoulos and Rotchell2021). The levels of MNP indoors are higher compared to outdoors and this is worrying because we spend 90% of our time indoors (Gaston et al., Reference Gaston, Woo, Steele, Sukumaran and Anderson2020; Amato-Lourenço et al., Reference Amato-Lourenço, dos Santos Galvão, Wiebeck, Carvalho-Oliveira and Mauad2022). In addition, MNP are added on purpose to cosmetics and have been identified in our drinking water and food (Guerranti et al., Reference Guerranti, Martellini, Perra, Scopetani and Cincinelli2019; Koelmans et al., Reference Koelmans, Mohamed Nor, Hermsen, Kooi, Mintenig and De France2019; Jin et al., Reference Jin, Wang, Ren, Wang and Shan2021; Dronjak et al., Reference Dronjak, Exposito, Rovira, Florencio, Emiliano, Corzo, Schuhmacher, Valero and Sierra2022; Shi et al., Reference Shi, Dong, Shi, Yin, He, An, Tang, Hou, Chong, Chen, Qin and Lin2022). Consequently, humans are unavoidably exposed to MNP, that can enter the body via ingestion, inhalation and possibly dermal contact. Indeed, MNP have been detected in different compartments of the human body such as the blood, colon, liver, testes and lungs (Amato-Lourenço et al., Reference Amato-Lourenço, Carvalho-Oliveira, Júnior, dos Santos Galvão, Ando and Mauad2021; Ibrahim et al., Reference Ibrahim, Tuan Anuar, Azmi, Wan Mohd Khalik, Lehata, Hamzah, Ismail, Ma, Dzulkarnaen, Zakaria, Mustaffa, Tuan Sharif and Lee2021; Horvatits et al., Reference Horvatits, Tamminga, Liu, Sebode, Carambia, Fischer, Püschel, Huber and Fischer2022; Jenner et al., Reference Jenner, Rotchell, Bennett, Cowen, Tentzeris and Sadofsky2022; Leslie et al., Reference Leslie, van Velzen, Brandsma, Vethaak, Garcia-Vallejo and Lamoree2022; Zhao et al., Reference Zhao, Zhu, Weng, Jin, Cao, Jiang and Zhang2023). Notably, MNP were found in human placental tissue too and this may especially be of concern for a fetus (Ragusa et al., Reference Ragusa, Svelato, Santacroce, Catalano, Notarstefano, Carnevali, Papa, Rongioletti, Baiocco, Draghi, D’Amore, Rinaldo, Matta and Giorgini2021; Amereh et al., Reference Amereh, Amjadi, Mohseni-Bandpei, Isazadeh, Mehrabi, Eslami, Naeiji and Rafiee2022; Zhu et al., Reference Zhu, Zhu, Zuo, Xu, Qian and An2023). However, to date, there is no overview of effects of MNP on organ development and processes related to development such as regeneration and repair. Therefore, we here provide a comprehensive overview of current state-of-the-art knowledge regarding effects of MNP on developmental processes, including embryonic to childhood development and reactivation of developmental processes during regeneration in humans and rodent models.

Plastic additives

Not only the plastic particles themselves, but also plastic additives can influence biological processes. Properties of plastic polymers can be modified to achieve a desired material performance by the addition of chemical additives such as plasticizers, flame retardants, photo- and heat stabilizers, antioxidants and pigments (Fauser et al., Reference Fauser, Vorkamp and Strand2022). Notably, endocrine disruptors bisphenol A and phthalates are commonly used as additives in plastics and can therefore also leach from MNP (Liu et al., Reference Liu, Shi, Xie, Dionysiou and Zhao2019; Cao et al., Reference Cao, Lin, Zhang, Xu, Yan, Leung and Lam2022). In addition, MNP can still contain residual monomers that failed to polymerize, as well as unintentional byproducts of reactions during the manufacturing process (Lewandowski et al., Reference Lewandowski, Hayes and Beck2005; Klaeger et al., Reference Klaeger, Tagg, Otto, Bienmüller, Sartorius and Labrenz2019). MNP can also retain toxic organic compounds, like polycyclic aromatic hydrocarbons, polychlorinated biphenyls, pesticides and inorganic compounds such as heavy metals from the environment (Wu et al., Reference Wu, Cai, Jin and Tang2019; Mei et al., Reference Mei, Chen, Bao, Song, Li and Luo2020; Lu et al., Reference Lu, Zeng, Wei, Gao, Abdurahman, Wang and Liang2022). Most of these can leach from MNP into the surrounding environment, including inside the human body and may consequently cause damage or affect development.

Bisphenol A and phthalates are the most studied plastic additives and have been found in human tissues and blood. Both can also cross the placental barrier and reach a developing fetus (Tang et al., Reference Tang, Xu, Deng, Z-X and Yu2020; Mok et al., Reference Mok, Jeong, Park, Kim, Lee, Park, Kim, Choi and Moon2021; Warner et al., Reference Warner, Dettogni, Bagchi, Flaws and Graceli2021). Bisphenol A is thought to be the first synthetic estrogen produced and the majority is metabolized in the liver by UDP-glucuronosyltransferase enzymes (Hanioka et al., Reference Hanioka, Naito and Narimatsu2008). Bisphenol A and phthalates exhibit endocrine effects by modulating androgen receptor and estrogen receptors alpha and beta, thereby disturbing normal signaling (Rubin, Reference Rubin2011; Engel et al., Reference Engel, Buhrke, Imber, Jessel, Seidel, Völkel and Lampen2017). Notably, developing fetuses do not or only lowly express UDP-glucuronosyltransferase enzymes and are consequently exposed to higher bisphenol A concentrations (Hines, Reference Hines2008). However, the contribution of MNP to bisphenol A and phthalate exposure may be limited as humans are already exposed to substantial amounts of these chemicals by consuming food and beverages that are contaminated with leachate from reusable plastic bottles or food packages (Liu et al., Reference Liu, Shi, Xie, Dionysiou and Zhao2019; da Silva Costa et al., Reference da Silva Costa, Sainara Maia Fernandes, de Sousa Almeida, Tomé Oliveira, Carvalho Guedes, Julião Zocolo, Wagner de Sousa and do Nascimento2021; Sessa et al., Reference Sessa, Polito, Monda, Scarinci, Salerno, Carotenuto, Cibelli, Valenzano, Campanozzi, Mollica, Monda and Messina2021). Interestingly though, animals exposed to MNP showed higher levels of these endocrine disrupting chemicals compared to animals without or with less microplastic exposure (Fossi et al., Reference Fossi, Panti, Guerranti, Coppola, Giannetti, Marsili and Minutoli2012; Chen et al., Reference Chen, Yin, Jia, Schiwy, Legradi, Yang and Hollert2017; Barboza et al., Reference Barboza, Cunha, Monteiro, Fernandes and Guilhermino2020; Lu et al., Reference Lu, Chao, Mansor, Peng, Hsu, Yu, Chang and Fu2021). Recently, López-Vázquez et al. (Reference López-Vázquez, Rodil, Trujillo-Rodríguez, Quintana, Cela and Miró2022) showed that over 65% of bisphenol A or phthalates in MNP can become bioaccessible when exposed to physiologically relevant human digestive conditions. While precise data on the quantities of additives leaching from MNP remain unavailable, existing studies suggest that it does occur. Hence, the potential contribution of MNP to exposure levels of bisphenol A and phthalates warrants consideration. Relevant data concerning the impact of bisphenol A and phthalates on developmental and repair mechanisms will therefore also be addressed.

Microplastic exposure on various organs and tissues

Placental and fetal development

Human gestation starts when a sperm and egg cell fuse during fertilization to form a one-celled diploid totipotent zygote (Figure 2) (Clift and Schuh, Reference Clift and Schuh2013). This initiates a highly sensitive phase of development with the zygote maturing into a blastocyst containing an outer layer of trophoblasts and enclosing an inner cell mass, the precursors of the future fetus (Rossant and Tam, Reference Rossant and Tam2022). Trophoblasts will differentiate into components of the placenta, establishing a critical link between maternal and fetal tissues (Cindrova-Davies and Sferruzzi-Perri, Reference Cindrova-Davies and Sferruzzi-Perri2022). This stage of development is particularly vulnerable to environmental disturbances as it is susceptible to gene expression modifications in both the fetus and the placenta (Assou et al., Reference Assou, Boumela, Haouzi, Anahory, Dechaud, De Vos and Hamamah2011). Perturbations in gene expression can have deleterious consequences for the fetus, either directly or indirectly by impacting placental development, and such risks persist even after removal of the stressors (Nesan et al., Reference Nesan, Sewell and Kurrasch2018; Maitre et al., Reference Maitre, Bustamante, Hernández-Ferrer, Thiel, Lau, Siskos, Vives-Usano, Ruiz-Arenas, Pelegrí-Sisó, Robinson, Mason, Wright, Cadiou, Slama, Heude, Casas, Sunyer, Papadopoulou, Gutzkow, Andrusaityte, Grazuleviciene, Vafeiadi, Chatzi, Sakhi, Thomsen, Tamayo, Nieuwenhuijsen, Urquiza, Borràs, Sabidó, Quintela, Carracedo, Estivill, Coen, González, Keun and Vrijheid2022).

Figure 2. Early human development. An overview of human development at different time points. First, a sperm cell fuses with an egg cell during fertilization to form a zygote and this time point is referred to as gestational day 0. The zygote develops further into a blastocyte, consisting of an inner cell mass (purple cells) and trophoblasts (pink cells) on day 5. The inner cell mass further differentiates into ectoderm (blue cells), mesoderm (red cells) and endoderm (yellow cells) on day 15 and is called a gastrula. The embryo will then further develop and is called a fetus after week 8. Image created with BioRender.com.

Research investigating the effects of MNP on fetal development has produced important insights. Amereh et al. (Reference Amereh, Amjadi, Mohseni-Bandpei, Isazadeh, Mehrabi, Eslami, Naeiji and Rafiee2022) delineated a potential dose–response relationship in humans between placental plastics and reduced fetal growth, suggesting possible interference in nutrient exchange in the placenta. This is supported by several in vitro studies showing that microplastic exposure is toxic for a variety of human placental cell lines (Lee et al., Reference Lee, Amarakoon, C-I, Choi, Smolensky and Lee2021; Dusza et al., Reference Dusza, Katrukha, Nijmeijer, Akhmanova, Vethaak, Walker and Legler2022; Shen et al., Reference Shen, Li, Guo and Chen2022; Ragusa et al., Reference Ragusa, Matta, Cristiano, Matassa, Battaglione, Svelato, De Luca, D’Avino, Gulotta, Rongioletti, Catalano, Santacroce, Notarstefano, Carnevali, Giorgini, Vizza, Familiari and Nottola2022a; Dusza et al., Reference Dusza, van Boxel, van Duursen, Forsberg, Legler and Vähäkangas2023). In pregnant mice exposed to polystyrene microparticles, similar outcomes were found, demonstrating disruptions in placental metabolism (Chen et al., Reference Chen, Xiong, Jing, van Gestel, van Straalen, Roelofs, Sun and Qiu2022; Aghaei et al., Reference Aghaei, Mercer, Schneider, Sled, Macgowan, Baschat, Kingdom, Helm, Simpson, Simpson, Jobst and Cahill2022a; Aghaei et al., Reference Aghaei, Sled, Kingdom, Baschat, Helm, Jobst and Cahill2022b). Moreover, pregnant rats or mice exposed to polystyrene/polyethylene MNP showed detectable spread of particles throughout mothers and pups within 24 h, alongside a variety of other effects like lower fetal body weights, less vascularization of the placenta and higher expression of genes involved in cholesterol/lipid metabolism, the complement system and the coagulation cascade (Fournier et al., Reference Fournier, D’Errico, Adler, Kollontzi, Goedken, Fabris, Yurkow and Stapleton2020; Park et al., Reference Park, Han, Park, Seong, Lee, Kim, Son, H-Y and Lee2020; Huang et al., Reference Huang, Zhang, Lin, Liu, Sun, Liu, Yuan, Xiang, Kuang, Yang and Zhang2022; Aghaei et al., Reference Aghaei, Mercer, Schneider, Sled, Macgowan, Baschat, Kingdom, Helm, Simpson, Simpson, Jobst and Cahill2022a, Reference Aghaei, Sled, Kingdom, Baschat, Helm, Jobst and Cahillb; Chen et al., Reference Chen, Xiong, Jing, van Gestel, van Straalen, Roelofs, Sun and Qiu2023). These findings suggest that MNP exposure may disrupt both placental function and fetal development (Figure 3), thereby potentially leading to detrimental consequences for embryonic development.

Figure 3. Effects of microplastics on various organs and tissues. Overview of effects of microplastics exposure on various organs and tissues of a developing fetus. Microplastics have detrimental effects on development of the placenta, central nervous system, liver, intestines, lungs, reproductive system and stem cells. Image created with BioRender.com.

With respect to developmental aberrations and endocrine disrupting chemicals like bisphenol A and phthalates, many more studies have been published. These have been elegantly reviewed by Rolfo et al. (Reference Rolfo, Nuzzo, De Amicis, Moretti, Bertoli and Leone2020) among others. In short, numerous studies, including in vitro studies, animal models and population-based studies, compellingly suggest that endocrine disrupting chemicals can adversely affect fetal and placental health. These disruptors potentially interfere with the developing embryonic epigenome, thereby predisposing individuals to disease in adulthood. Furthermore, endocrine-disrupting chemicals may trigger or contribute to serious pregnancy-related conditions such as preeclampsia, fetal growth restriction and gestational diabetes. Therefore, studies into effects of MNP should always consider additives contributing or being responsible for any effect that is found.

Reproductive system development

Reproductive structures already begin to form in the embryonic stage (Figure 2) (Pask, Reference Pask, Wilhelm and Bernard2016). The urogenital system is bipotential and undifferentiated until week 6 of gestation and can still develop into female and male primary sexual organs (Makiyan, Reference Makiyan2016; Garcia-Alonso et al., Reference Garcia-Alonso, Lorenzi, Mazzeo, Alves-Lopes, Roberts, Sancho-Serra, Engelbert, Marečková, Gruhn, Botting, Li, Crespo, Van Dongen, Kiselev, Prigmore, Herbert, Moffett, Chédotal, Bayraktar, Surani, Haniffa and Vento-Tormo2022). The presence of the sex-determining region Y gene on the Y chromosome is the main determining factor in development of the testes (Koopman et al., Reference Koopman, Gubbay, Vivian, Goodfellow and Lovell-Badge1991). The testes subsequently secrete anti-Müllerian hormone and the androgen testosterone, which induce differentiation toward male sex organs (Josso et al., Reference Josso, Lamarre, Picard, Berta, Davies, Morichon, Peschanski and Jeny1993; Nassar and Leslie, Reference Nassar and Leslie2023). In contrast, when an embryo lacks the Y chromosome, and thus the sex-determining region Y, there is no formation of the testes. Therefore, anti-Müllerian hormone and testosterone are not produced and this eventually leads toward development of female sex organs (Healey, Reference Healey, Mann, Blair and Garden2012; Cunha et al., Reference Cunha, Robboy, Kurita, Isaacson, Shen, Cao and Baskin2018).

Effects on reproductive system development were studied for two types of MNP, polystyrene and polyethylene. Maternal exposure to polystyrene nanoplastics resulted in lower testicular weights and altered morphology with lower sperm count in male offspring, while polyethylene microplastics reduced oocyte maturation and fertility in female offspring (Huang et al., Reference Huang, Zhang, Lin, Liu, Sun, Liu, Yuan, Xiang, Kuang, Yang and Zhang2022; Zhang et al., Reference Zhang, Wang, Zhao, Zhao, Yu, Yao, Zhao, Yu, Liu and Su2023). In both studies, oxidative stress was shown to be associated with the effects found. Further information on the effects of MNP on development of the male or female reproductive system is unfortunately limited.

An abundance of data exists concerning the impact of various plastic additives. For instance, bisphenol A is implicated in causing developmental abnormalities, as evidenced in animal models. These adverse effects seem to be particularly pronounced in the female reproductive organs. Different rodent studies found that fetal bisphenol A exposure resulted in abnormal follicle development (Susiarjo et al., Reference Susiarjo, Hassold, Freeman and Hunt2007; Rodríguez et al., Reference Rodríguez, Santambrosio, Santamaría, Muñoz-de-Toro and Luque2010; Karavan and Pepling, Reference Karavan and Pepling2012). Human oogenesis only takes place during embryonic development of the ovaries, meaning that the number of oocytes is established at birth and abnormalities during development will therefore impact fertility later in life (Feher, Reference Feher and Feher2012). Furthermore, Hunt et al. (Reference Hunt, Lawson, Gieske, Murdoch, Smith, Marre, Hassold and Vandevoort2012) treated pregnant rhesus monkeys with bisphenol A and found that second trimester fetuses had more oocytes with an abnormal number of chromosomes and abnormal morphology. Male offspring of pregnant rats exposed to bisphenol A during pregnancy and beyond had lower sperm counts and motility and less expression of steroid receptors in the testes. Worryingly this resulted in less fertility in these animals and their offspring up to the F3 generation (Salian et al., Reference Salian, Doshi and Vanage2009). Various other studies have confirmed that prenatal bisphenol A exposure can lead to sperm with abnormal morphology and to a lower sperm count and function (Vilela et al., Reference Vilela, Hartmann, Silva, Cardoso, Corcini, Varela-Junior, Martinez and Colares2014; Hass et al., Reference Hass, Christiansen, Boberg, Rasmussen, Mandrup and Axelstad2016; Rahman et al., Reference Rahman, Kwon, Karmakar, Yoon, Ryu and Pang2017). Moreover, bisphenol A exposure was also associated with a lower anogenital distance in humans (Mammadov et al., Reference Mammadov, Uncu and Dalkan2018; Sun et al., Reference Sun, Li, Liang, Miao, Song, Wang, Zhou and Yuan2018). This anogenital distance is a biomarker of fetal androgen exposure. A short distance in males is associated with genital malformations and reproductive disorders later in life (Schwartz et al., Reference Schwartz, Christiansen, Vinggaard, Axelstad, Hass and Svingen2019). Moreover, the anogenital distance is linked to adult testicular function, which is defined by sperm and testosterone production (Foresta et al., Reference Foresta, Valente, Di Nisio, Cacco, Magagna, Cosci, Presciutti and Garolla2018; Priskorn et al., Reference Priskorn, Petersen, Jørgensen, Kyhl, Andersen, Main, Andersson, Skakkebaek and Jensen2018, Reference Priskorn, Bang, Nordkap, Krause, Mendiola, Jensen, Juul, Skakkebaek, Swan and Jørgensen2019). Notably, prenatal exposure to phthalates has also been linked to a smaller anogenital distance in human infants (Swan et al., Reference Swan, Main, Liu, Stewart, Kruse, Calafat, Mao, Redmon, Ternand, Sullivan and Teague2005; Bornehag et al., Reference Bornehag, Carlstedt, Jönsson, Lindh, Jensen, Bodin, Jonsson, Janson and Swan2015). Various animal studies confirmed these human findings in offspring of rats or mice who were treated with different concentrations of phthalates during pregnancy (Ma et al., Reference Ma, Yin, Han, Ding, Zhang, Han and Li2017; Hsu et al., Reference Hsu, Jhong, Huang, Lee, Chen and Guo2021). The offspring of phthalate-exposed animals also showed signs of histological damage in the testes and apoptosis of cells in the seminiferous tubule, which is responsible for sperm production (Ma et al., Reference Ma, Yin, Han, Ding, Zhang, Han and Li2017). The female reproductive system was also affected by phthalate exposure. Mice that were prenatally exposed to phthalates had fewer healthy follicles and this effect was found up to the F3 generation also having lower total follicle numbers (Brehm et al., Reference Brehm, Rattan, Gao and Flaws2018; Repouskou et al., Reference Repouskou, Panagiotidou, Panagopoulou, Bisting, Tuck, Sjödin, Lindberg, Bozas, Rüegg, Gennings, Bornehag, Damdimopoulou, Stamatakis and Kitraki2019).

In summary, though the evidence from animal models is limited, it suggests that MNP could negatively affect the development of the reproductive system in fetuses, potentially leading to infertility or other reproductive issues (Figure 3). There is more robust and compelling evidence for the adverse effects of additives, indicating that MNP can harm developing reproductive organs through these substances. Therefore, studies investigating how much of the additive presence in humans is derived from microplastics exposure will be invaluable.

Central nervous system development

One of the first steps in the development of the nervous system begins in the third week of gestation and is the differentiation of ectoderm into neuroectoderm (Pleasure et al., Reference Pleasure, Pleasure, Pleasure, Polin, Abman, Rowitch, Benitz and Fox2017). Neuroectoderm cells are neural stem cells that are capable of self-renewal and can differentiate into neurons (Sansom et al., Reference Sansom, Griffiths, Faedo, Kleinjan, Ruan, Smith, Van Heyningen, Rubenstein and Livesey2009; Zhang et al., Reference Zhang, Huang, Chen, Pankratz, Xi, Li, Yang, Lavaute, Li, Ayala, Bondarenko, Du, Jin, Golos and Zhang2010; Thakurela et al., Reference Thakurela, Tiwari, Schick, Garding, Ivanek, Berninger and Tiwari2016). These processes can be modeled in vitro with embryonic or pluripotent stem cells and can then be used to study effects of MNP. Neurospheres generated from human embryonic stem cells were exposed to polyethylene nanoplastics and particles were found to penetrate deep into these neurospheres, causing oxidative stress with higher levels of malondialdehyde. Importantly, the expression of genes that play crucial roles in embryonic neural development was lower indicating that polyethylene nanoplastics can impair neural development (Hoelting et al., Reference Hoelting, Scheinhardt, Bondarenko, Schildknecht, Kapitza, Tanavde, Tan, Lee, Mecking, Leist and Kadereit2013). Another study used human-induced pluripotent stem cells and differentiated them into human forebrain cortical spheroids. Exposing these cortical spheroids to polystyrene MNP resulted in lower cell viability and expression of mature neuronal markers indicating that polystyrene MNP can also affect neural differentiation (Hua et al., Reference Hua, Kiran, Li and Sang2022).

Others have investigated the effects of microplastic exposure in mouse models. Yang et al. treated pregnant mice with polystyrene MNP. Polystyrene nanoplastics were observed throughout the fetus including the brain, especially in the thalamus. Excessive production of reactive oxygen species, more apoptosis, less proliferation, less gamma-aminobutyric acid synthesis and less expression of mature neuronal genes were found compared to unexposed fetuses. Moreover, mice showed more anxiety-like behavior in several tests after nanoplastic treatment. Together these findings indicate that polystyrene nanoparticle exposure during gestation can inhibit fetal brain development, which may result in anxiety (Yang et al., Reference Yang, Zhu, Zhou, Pan, Nan, Yin, Lei, Ma, Zhu, Chen, Han, Ding and Ding2022).

In addition to in utero exposure, maternally ingested polystyrene nanoplastics were also found to reach the brain via breast milk (Jeong et al., Reference Jeong, Baek, Koo, Park, Ryu, Kim, Zhang, Chung, Dogan, Choi, Um, Kim, Lee, Jeong, Shin, Lee, N-S and Lee2022). Milk-exposed pups showed less proliferation and lower expression of mature neuronal genes in the hippocampus, which indicates impairment of neuronal development. This reduced brain development resulted in neuronal dysfunction and cognitive deficit, which was dependent on estrogen receptor alpha and was more severe in exposed females.

Finally, bisphenol A and phthalates also have detrimental effects on neural development. Various studies found that these endocrine disrupting chemicals can diminish neural differentiation, impair neurotransmission pathways and diminish myelination (Zhou et al., Reference Zhou, Chen, Feng, Zhou, Li and Chen2015; Grohs et al., Reference Grohs, Reynolds, Liu, Martin, Pollock, Lebel and Dewey2019; Tiwari et al., Reference Tiwari, Agarwal, Chauhan, Mishra and Chaturvedi2019; Lucaccioni et al., Reference Lucaccioni, Trevisani, Passini, Righi, Plessi, Predieri and Iughetti2021). Moreover, exposure to bisphenol A and phthalates during gestation is linked to behavioral problems (particularly in girls), lower nonverbal IQ scores, anxiety and depression-like behavior (Zhou et al., Reference Zhou, Chen, Feng, Zhou, Li and Chen2015; Ejaredar et al., Reference Ejaredar, Lee, Roberts, Sauve and Dewey2017; Daniel et al., Reference Daniel, Balalian, Insel, Liu, Whyatt, Calafat, Rauh, Perera, Hoepner, Herbstman and Factor-Litvak2020; Van Den Dries et al., Reference Van Den Dries, Guxens, Spaan, Ferguson, Philips, Santos, Jaddoe, Ghassabian, Trasande, Tiemeier and Pronk2020; Guilbert et al., Reference Guilbert, Rolland, Pin, Thomsen, Sakhi, Sabaredzovic, Slama, Guichardet and Philippat2021; Rolland et al., Reference Rolland, Lyon-Caen, Thomsen, Sakhi, Sabaredzovic, Bayat, Slama, Méary and Philippat2023).

Together these studies indicate that MNP and/or their additives can directly or indirectly impair neural development. Whether this is happening in humans will depend on the number of particles that can reach the human brain and this information is sadly lacking (Figure 3). Moving forward in this field, large longitudinal studies following brain development in children in combination with analysis of MNP exposure will be necessary. Having the possibility to image MNP presence in brain tissue would be an enormous step forward, but for now is still science fiction.

Intestinal development and regeneration

Intestines develop from endoderm forming a hollow cylinder surrounded by cells of the mesoderm resulting in a primitive intestinal tube, whereas the ectoderm forms the enteric nervous system during week three of gestation (Spence et al., Reference Spence, Lauf and Shroyer2011). Then the foregut develops into esophagus, lung, stomach, liver and pancreas and the midgut and hindgut into the small and large intestines (Sheaffer and Kaestner, Reference Sheaffer and Kaestner2012; Chin et al., Reference Chin, Hill, Aurora and Spence2017; McCracken and Wells, Reference McCracken and Wells2017; Zhang et al., Reference Zhang, Jiang, Kim, Lin, Liu, Lan and Que2017). Signaling pathways controlling intestinal stem cell self-renewal include Wnt and Notch (Van Camp et al., Reference Van Camp, Beckers, Zegers and Van Hul2014; Demitrack and Samuelson, Reference Demitrack and Samuelson2016).

Unfortunately, no studies investigated effects of microplastics or additives on intestinal development, but some did use organoids. Intestinal organoids are an excellent way to study intestinal repair mechanisms and reactivated developmental pathways. Intestinal mouse organoids exposed to polystyrene MNP had higher expression of Notch pathway genes, other intestinal stem cell markers and proliferation markers compared to unexposed organoids. In contrast, the expression of endothelial and goblet cell markers was lower, indicating that polystyrene MNP can stimulate stemness but impair cell differentiation by overstimulation of Notch signaling (Xie et al., Reference Xie, Zhang, Li, Liu, Chen and Yu2023).

Effects of MNP on microbiome development were also investigated and this is of relevance because the microbiome can influence the function of the gut. Environmental stimuli in their turn can influence development of the microbiome, especially up until the age of 5 (Stiemsma and Turvey, Reference Stiemsma and Turvey2017; Roswall et al., Reference Roswall, Olsson, Kovatcheva-Datchary, Nilsson, Tremaroli, Simon, Kiilerich, Akrami, Krämer, Uhlén, Gummesson, Kristiansen, Dahlgren and Bäckhed2021; Wernroth et al., Reference Wernroth, Peura, Hedman, Hetty, Vicenzi, Kennedy, Fall, Svennblad, Andolf, Pershagen, Theorell-Haglöw, Nguyen, Sayols-Baixeras, Dekkers, Bertilsson, Almqvist, Dicksved and Fall2022). Infants can be exposed to high levels of MNP through bottle feeding, since MNP are found in various milk products and can be released from feeding bottles (Li et al., Reference Li, Shi, Yang, Xiao, Kehoe, Gun’Ko, Boland and Wang2020; Da Costa Filho et al., Reference Da Costa Filho, Andrey, Eriksen, Peixoto, Carreres, Ambühl, Descarrega, Dubascoux, Zbinden, Panchaud and Poitevin2021). In addition, infants can ingest MNP through breastfeeding, as MNP have been found in human breastmilk (Ragusa et al., Reference Ragusa, Notarstefano, Svelato, Belloni, Gioacchini, Blondeel, Zucchelli, De Luca, D’Avino, Gulotta, Carnevali and Giorgini2022b; Liu et al., Reference Liu, Guo, Liu, Yang, Wang, Sun, Chen and Dong2023). Notably, MNP were found in both infant formula and in the feces of infants consuming these types of milk (Zhang et al., Reference Zhang, Wang, Trasande and Kannan2021; Liu et al., Reference Liu, Guo, Liu, Yang, Wang, Sun, Chen and Dong2023). Fournier et al. processed stool of infants in a novel fermentation system that simulates physicochemical and microbial conditions in the intestines of a toddler. Exposing the microbiome of infants to polyethylene MNP resulted in lower numbers of healthy microbes and more opportunistic pathogens. Consequently, an altered microbial metabolic activity was found including a changed volatile organic compounds profile with less butyrate production (Fournier et al., Reference Fournier, Ratel, Denis, Leveque, Ruiz, Mazal, Amiard, Edely, Bezirard, Gaultier, Lamas, Houdeau, Engel, Lagarde, Etienne-Mesmin, Mercier-Bonin and Blanquet-Diot2023). This is of interest because higher butyrate production is associated with protection against allergies and asthma (Roduit et al., Reference Roduit, Frei, Ferstl, Loeliger, Westermann, Rhyner, Schiavi, Barcik, Rodriguez-Perez, Wawrzyniak, Chassard, Lacroix, Schmausser-Hechfellner, Depner, von Mutius, Braun-Fahrländer, Karvonen, Kirjavainen, Pekkanen, Dalphin, Riedler, Akdis, Lauener and O’Mahony2019; Depner et al., Reference Depner, Taft, Kirjavainen, Kalanetra, Karvonen, Peschel, Schmausser-Hechfellner, Roduit, Frei, Lauener, Divaret-Chauveau, Dalphin, Riedler, Roponen, Kabesch, Renz, Pekkanen, Farquharson, Louis, Mills, von Mutius, Genuneit, Hyvärinen, Illi, Laurent, Pfefferle, Schaub, von Mutius and Ege2020). These findings indicate that polyethylene MNP could cause significant disturbances in the microbiota of infants (Fournier et al., Reference Fournier, Ratel, Denis, Leveque, Ruiz, Mazal, Amiard, Edely, Bezirard, Gaultier, Lamas, Houdeau, Engel, Lagarde, Etienne-Mesmin, Mercier-Bonin and Blanquet-Diot2023).

Despite the intestinal system’s pivotal role in MNP entry into the body, knowledge regarding their effects on intestinal development and repair remains surprisingly sparse. A limited number of existing studies suggest that the ingestion of MNP may hinder the differentiation of intestinal epithelial cells and cause microbiota dysbiosis (Figure 3). Given the vital nature of the intestinal system as a primary entry point for MNP, it is imperative that this area receives a significantly greater level of research focus than it currently does.

Liver development

While the intestines originate from the midgut and hindgut, the liver develops from the more distal end of the foregut in the third week of gestation (Sheaffer and Kaestner, Reference Sheaffer and Kaestner2012; Chin et al., Reference Chin, Hill, Aurora and Spence2017). Endoderm-derived cells develop further in hepatoblasts, that differentiate with hepatocyte growth factor and Wnt signaling into hepatocytes or cholangiocytes (Shin and Monga, Reference Shin and Monga2013; Giancotti et al., Reference Giancotti, Monti, Nevi, Safarikia, D’Ambrosio, Brunelli, Pajno, Corno, Di Donato, Musella, Chiappetta, Bosco, Benedetti Panici, Alvaro and Cardinale2019).

With respect to effects of MNP on liver development, offspring of pregnant mice exposed to polystyrene MNP had higher liver weights in comparison to control mice. Moreover, expression of hepatic genes involved in fatty acid metabolism was lower. However, the observed results were sex-specific and dependent on the size of the polystyrene MNP. Whereas anabolic pathways were slower in males, females maintained the synthesis of lipids at the costs of amino acids. Eventually, these changes induced by maternal polystyrene microplastic exposure resulted in a higher risk of hepatic lipid accumulation and the development of metabolic disorders in offspring (Luo et al., Reference Luo, Zhang, Wang, Wang, Zhou, Shen, Zhao, Fu and Jin2019a, Reference Luo, Wang, Pan, Jin, Fu and Jin2019b). Interestingly, no significant change in liver weight in offspring was found after maternal exposure to nanoplastic in a study of Huang et al. (Reference Huang, Zhang, Lin, Liu, Sun, Liu, Yuan, Xiang, Kuang, Yang and Zhang2022). Therefore, the development of a higher liver weight is probably size-dependent since it was only found after maternal exposure of polystyrene microplastic of 5 μm (Luo et al., Reference Luo, Wang, Pan, Jin, Fu and Jin2019b). Interestingly, exposure of pregnant mice to polystyrene nanoplastics did induce higher levels of malondialdehyde and pro-inflammatory cytokines in liver tissue of their offspring indicating oxidative stress induction. Moreover, metabolomics revealed that levels of metabolites involved in carbohydrate metabolism were lower. Therefore, these results show that maternal exposure to polystyrene nanoplastics can trigger hepatic oxidative stress and inflammation resulting in a disrupted carbohydrate metabolism in their offspring (Huang et al., Reference Huang, Zhang, Lin, Liu, Sun, Liu, Yuan, Xiang, Kuang, Yang and Zhang2022).

In addition to studies on the direct effects of MNP on liver development, some examined the effects of additives. Different studies showed that prenatal bisphenol A and phthalate exposure can impair liver development, resulting in metabolic disorders including changes to glucose and lipid metabolism (Maranghi et al., Reference Maranghi, Lorenzetti, Tassinari, Moracci, Tassinari, Marcoccia, Di Virgilio, Eusepi, Romeo, Magrelli, Salvatore, Tosto, Viganotti, Antoccia, Di Masi, Azzalin, Tanzarella, Macino, Taruscio and Mantovani2010; Strakovsky et al., Reference Strakovsky, Wang, Engeseth, Flaws, Helferich, Pan and Lezmi2015; DeBenedictis et al., Reference DeBenedictis, Guan and Yang2016; Sol et al., Reference Sol, Santos, Duijts, Asimakopoulos, Martinez-Moral, Kannan, Jaddoe and Trasande2020; Long et al., Reference Long, Fan, Wu, Liu, Wu, Liu, Chen, Su, Cheng, Xu, Su, Cao, Zhang, Hai and Wang2021).

Although the evidence is not abundant, the available studies suggest that maternal exposure to MNP and additives leaching from plastics can detrimentally affect liver development, resulting in metabolic disorders (Figure 3). Again, given the proximity of the liver to a main port of MNP entry, more focus on this organ is warranted.

Lung development and regeneration

Another organ that develops from the primitive foregut is the lung. First, two independent outpouchings of the more proximal end of the foregut arise and these two lung buds develop into a tree-like system of airways ending in respiratory units called alveoli (Warburton et al., Reference Warburton, Bellusci, De Langhe, Del Moral, Fleury, Mailleux, Tefft, Unbekandt, Wang and Shi2005; Schittny, Reference Schittny2017). Eventually, airways consist of pseudostratified epithelial cells with basal cells, ciliated cells and secretory cells such as goblet and club cells, while alveoli are lined with alveolar epithelial cell types I and II (Desai et al., Reference Desai, Brownfield and Krasnow2014; Li et al., Reference Li, He, Wei, Cho and Liu2015). Some of these epithelial cells have progenitor functions that reactivate developmental pathways when repair of damaged structures is needed (Levardon et al., Reference Levardon, Yonker, Hurley and Mou2018; Olajuyin et al., Reference Olajuyin, Zhang and Ji2019; Davis and Wypych, Reference Davis and Wypych2021). Like for the intestine, pathways important in regulation of lung development and repair include Notch and Wnt (Kiyokawa and Morimoto, Reference Kiyokawa and Morimoto2020; Aros et al., Reference Aros, Pantoja and Gomperts2021).

We recently generated human and mouse lung organoids that self-assemble from isolated primary lung epithelial cells. These developing mouse and human lung organoids were exposed to polyester or polyamide 6,6 microfibers for 14 days. Both polyester and polyamide 6,6 microfiber exposure resulted in lower numbers and sizes of mouse and human organoids with the effects of polyamide 6,6 being most profound. Interestingly, this effect of polyamide 6,6 was mediated by compounds leaching from the microfibers. The observed effects of the leachate did not affect stemness of epithelial progenitors, or fully developed epithelial cells, but specifically inhibited differentiation of progenitor cells into airway epithelial cell types. Therefore, these results indicate that exposure to plastic microfibers and compounds leaching from them may affect developing lungs by impairing epithelial differentiation (Dijk et al., Reference Dijk, Song, Eck, Wu, Bos, Boom, Kooter, Spierings, Wardenaar, Cole, Salvati, Gosens and Melgert2021).

Winkler et al. (Reference Winkler, Cherubini, Rusconi, Santo, Madaschi, Pistoni, Moschetti, Sarnicola, Crosti, Rosso, Tremolada, Lazzari and Bacchetta2022) examined the effects of polyester microfiber exposure on developing human airway organoids in more detail. In this study, organoid growth was not affected by exposure to polyester microfibers and exposure did not induce oxidative stress or cell activation. However, lower levels of club cell markers were found, suggesting that exposure to polyester microfibers can inhibit generation of this epithelial cell type from progenitors.

Collectively, these two studies demonstrate that MNP and compounds leaching from MNP can have significant effects on differentiation of lung epithelial cells (Figure 3).

Regeneration in other tissues

Fetal development is highly dependent on specific pathways that are subsequently downregulated after birth. Interestingly, these pathways are reactivated when tissue damage occurs and regeneration is required. This includes, for instance, regeneration of damaged skeletal muscles after exercise, which is mainly governed by normally quiescent satellite cells (Relaix and Zammit, Reference Relaix and Zammit2012). After reactivation, these stem cells proliferate and differentiate into myogenic cells to repair damaged myofibers (Yin et al., Reference Yin, Price and Rudnicki2013; Zhang et al., Reference Zhang, Zhang, Gu, Lan, Liu, Wang, Su, Ge, Wang, Yu, Liu, Li, Li, Zhao, Yu, Wang, Li and Meng2018). When mice were exposed to polystyrene MNP for 30 days and skeletal muscle injury was induced on day 25, polystyrene microplastic exposure resulted in lower relative muscle weights, less and smaller myofibers, lower gene and protein expression of myogenic differentiation markers, more lipid deposition and more expression of adipogenic markers after injury compared to control mice (Shengchen et al., Reference Shengchen, Jing, Yujie, Yue and Shiwen2021). In another study, pregnant mice were exposed to polystyrene nanoplastics, which resulted in an abnormal morphology of skeletal muscles and in lower expression of genes involved in muscle development in their offspring (Chen et al., Reference Chen, Xiong, Jing, van Gestel, van Straalen, Roelofs, Sun and Qiu2023).

Other cells capable of regeneration are mesenchymal stromal cells (Pittenger et al., Reference Pittenger, Discher, Péault, Phinney, Hare and Caplan2019). These multipotent cells can differentiate into osteocytes, chondrocytes and adipocytes (Uccelli et al., Reference Uccelli, Moretta and Pistoia2008). Najahi et al. exposed human bone marrow and adipose mesenchymal stromal cells to polyethylene terephthalate MNP. Less proliferation and self-renewal and more senescence, reactive oxygen species and DNA damage was found in both types of mesenchymal stromal cells after exposure. Polyethylene terephthalate exposure also induced higher expression of early adipose markers in adipose mesenchymal stromal cells and expression of early chondrocyte markers in bone marrow mesenchymal stromal cells compared to untreated mesenchymal stromal cells. These findings indicate that polyethylene terephthalate MNP can induce stress and impair differentiation of mesenchymal stromal cells into more mature cells (Najahi et al., Reference Najahi, Alessio, Squillaro, Conti, Ferrante, Di Bernardo, Galderisi, Messaoudi, Minucci and Banni2022).

In a study conducted by Im et al., the exposure of human bone marrow mesenchymal stromal cells to polystyrene nanoplastics led to a decrease in reactive oxygen species levels and suppressed expression of markers for osteocytes, chondrocytes and neurons. Intriguingly, genes associated with adipogenesis were again markedly elevated, suggesting a shift in differentiation toward adipocytes (Im et al., Reference Im, Kim, Jo, Yoo, Kim, Park, Hyeon, G-R and Bhang2022). Therefore, MNP exposure appears to slow down muscle repair and promote adipogenic differentiation of mesenchymal stromal cells (Figure 3).

Perspectives and conclusion

The presence of MNP in our environment and our food and drinks is evident and exposure to significant levels appears unavoidable (Gasperi et al., Reference Gasperi, Wright, Dris, Collard, Mandin, Guerrouache, Langlois, Kelly and Tassin2018; Li et al., Reference Li, Liu and Paul Chen2018; Koelmans et al., Reference Koelmans, Mohamed Nor, Hermsen, Kooi, Mintenig and De France2019; Evangeliou et al., Reference Evangeliou, Grythe, Klimont, Heyes, Eckhardt, Lopez-Aparicio and Stohl2020; Zhang et al., Reference Zhang, Wang and Kannan2020; Zhou et al., Reference Zhou, Wang, Zou, Jia, Zhou and Li2020; Jenner et al., Reference Jenner, Sadofsky, Danopoulos and Rotchell2021; Jin et al., Reference Jin, Wang, Ren, Wang and Shan2021; Dronjak et al., Reference Dronjak, Exposito, Rovira, Florencio, Emiliano, Corzo, Schuhmacher, Valero and Sierra2022; Shi et al., Reference Shi, Dong, Shi, Yin, He, An, Tang, Hou, Chong, Chen, Qin and Lin2022). Consequently, MNP have been found throughout the entire human body including the placenta, which is of great concern for development (Ibrahim et al., Reference Ibrahim, Tuan Anuar, Azmi, Wan Mohd Khalik, Lehata, Hamzah, Ismail, Ma, Dzulkarnaen, Zakaria, Mustaffa, Tuan Sharif and Lee2021; Ragusa et al., Reference Ragusa, Svelato, Santacroce, Catalano, Notarstefano, Carnevali, Papa, Rongioletti, Baiocco, Draghi, D’Amore, Rinaldo, Matta and Giorgini2021; Horvatits et al., Reference Horvatits, Tamminga, Liu, Sebode, Carambia, Fischer, Püschel, Huber and Fischer2022; Jenner et al., Reference Jenner, Rotchell, Bennett, Cowen, Tentzeris and Sadofsky2022; Leslie et al., Reference Leslie, van Velzen, Brandsma, Vethaak, Garcia-Vallejo and Lamoree2022; Zhu et al., Reference Zhu, Zhu, Zuo, Xu, Qian and An2023). However, the effects of microplastic exposure on (human) development or repair mechanisms are not well known and the absence of evidence of risk cannot be translated into evidence for the absence of risk (Leslie and Depledge, Reference Leslie and Depledge2020; Gouin et al., Reference Gouin, Cunliffe, De France, Fawell, Jarvis, Koelmans, Marsden, Testai, Asami, Bevan, Carrier, Cotruvo, Eckhardt and Ong2021; Wardman et al., Reference Wardman, Koelmans, Whyte and Pahl2021).

Most studies have predominantly explored the impacts of MNP composed of polystyrene or polyethylene terephthalate, typically focusing on a singular size category or specific additive types such as bisphenol A or phthalates. Yet MNPs are diverse in their polymer composition, size and additives. As such, there is a pressing need for broader research into the impacts of environmentally relevant MNPs, incorporating varying types, sizes and their leachates.

The design and methodological approaches of these investigations are crucial. This includes the selection of the cell types and the experimental setup, which should closely emulate our constant exposure to probably low to moderate MNP levels. Regrettably, most current experimental models do not reflect these conditions, as they often focus on short-term, high-level exposure, resulting in immediate toxicological outcomes such as cell death, oxidative stress or cytokine release (Weis and Palmquist, Reference Weis and Palmquist2021). The study of continuous, low-dose exposure, relevant exposure routes and developmental or repair processes are rarely prioritized and neither are studies into mechanisms, even though they are highly pertinent to real-life exposure scenarios. The frequent use of cancer cell lines and culture conditions that do not represent in vivo conditions, such as serum-free cultures, further confounds the ability to predict the biological impact of MNP exposure accurately.

Organoids derived from primary cells could help overcome some of these limitations by mimicking complex organ systems. Nonetheless, they also have their own constraints, including atypical physiology, irrelevant exposure pathways and lack of interorgan communication. Thus, the use of animal models remains essential to study the complex human physiological interactions. Care should be taken to use these models in a way that represents common human exposure with credible exposure levels and modes and to focus on mechanisms behind any effects found in these systems.

Another significant factor to keep in mind when evaluating literature is the presence of a positive publication bias that also affects the field of microplastics research. This bias tends to favor studies demonstrating harmful effects of microplastics on biological systems, which can inadvertently skew the overall understanding of their impact. As a result, studies that find no significant effects often face a higher hurdle in terms of getting published, which can potentially silence an essential aspect of the conversation. This imbalance can limit the diversity of our knowledge, creating a distorted picture of microplastics’ overall influence. It is crucial to address this bias to ensure a more comprehensive, balanced and accurate understanding of microplastics and their effects, or lack thereof, on biological systems. Ensuring the publication of all research outcomes, irrespective of their direction, will help to avoid overestimating the effects of microplastics and support the development of effective and proportionate response strategies.

In conclusion, we have provided an extensive overview of studies investigating effects of MNP on developmental and regenerative processes. MNP and additives commonly leaching from them can impair differentiation and/or promote aberrant differentiation. Therefore, MNP could have detrimental effects on a developing fetus and/or on repair processes in adult tissues of humans. The scant available data indicate that for all discussed organs, there is reason for concern (Vethaak and Legler, Reference Vethaak and Legler2021). By addressing these knowledge gaps in years to come, we may be able to protect ourselves from potential health risks induced by MNP.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/plc.2023.19.

Data availability statement

Data sharing not applicable – no new data generated.

Author contribution

L.T.H. and B.N.M. conceived the outline of the manuscript. L.T.H. collected published studies to include in this overview and drafted a first version of the manuscript. All the authors critically reviewed and revised the manuscript and approved the final version for publication.

Financial support

The authors thank ZonMw for their financial support with the ZonMw/Health Holland consortium grant MOMENTUM (458001101) led by Prof Dr. J. Legler and Prof Dr. D. Vethaak and awarded to them and Melgert among others.

Competing interest

The authors declare no competing interests exist.

References

Aghaei, Z, Mercer, GV, Schneider, CM, Sled, JG, Macgowan, CK, Baschat, AA, Kingdom, JC, Helm, PA, Simpson, AJ, Simpson, MJ, Jobst, KJ and Cahill, LS (2022a) Maternal exposure to polystyrene microplastics alters placental metabolism in mice. Metabolomics 19(1), 1. https://doi.org/10.1007/s11306-022-01967-8.CrossRefGoogle ScholarPubMed
Aghaei, Z, Sled, JG, Kingdom, JC, Baschat, AA, Helm, PA, Jobst, KJ and Cahill, LS (2022b) Maternal exposure to polystyrene micro- and nanoplastics causes fetal growth restriction in mice. Environmental Science & Technology Letters 9(5), 426430. https://doi.org/10.1021/acs.estlett.2c00186.CrossRefGoogle Scholar
Allen, D, Allen, S, Abbasi, S, Baker, A, Bergmann, M, Brahney, J, Butler, T, Duce, RA, Eckhardt, S, Evangeliou, N, Jickells, T, Kanakidou, M, Kershaw, P, Laj, P, Levermore, J, Li, D, Liss, P, Liu, K, Mahowald, N, Masque, P, Materić, D, Mayes, AG, McGinnity, P, Osvath, I, Prather, KA, Prospero, JM, Revell, LE, Sander, SG, Shim, WJ, Slade, J, Stein, A, Tarasova, O and Wright, S (2022) Microplastics and nanoplastics in the marine-atmosphere environment. Nature Reviews Earth & Environment 3(6), 393405. https://doi.org/10.1038/s43017-022-00292-x.CrossRefGoogle Scholar
Amato-Lourenço, LF, Carvalho-Oliveira, R, Júnior, GR, dos Santos Galvão, L, Ando, RA and Mauad, T (2021) Presence of airborne microplastics in human lung tissue. Journal of Hazardous Materials 416, 126124. https://doi.org/10.1016/j.jhazmat.2021.126124.CrossRefGoogle ScholarPubMed
Amato-Lourenço, LF, dos Santos Galvão, L, Wiebeck, H, Carvalho-Oliveira, R and Mauad, T (2022) Atmospheric microplastic fallout in outdoor and indoor environments in São Paulo megacity. Science of the Total Environment 821, 153450. https://doi.org/10.1016/j.scitotenv.2022.153450.CrossRefGoogle ScholarPubMed
Amereh, F, Amjadi, N, Mohseni-Bandpei, A, Isazadeh, S, Mehrabi, Y, Eslami, A, Naeiji, Z and Rafiee, M (2022) Placental plastics in young women from general population correlate with reduced foetal growth in IUGR pregnancies. Environmental Pollution 314, 120174. https://doi.org/10.1016/j.envpol.2022.120174.CrossRefGoogle ScholarPubMed
Andrady, AL (2011) Microplastics in the marine environment. Marine Pollution Bulletin 62(8), 15961605. https://doi.org/10.1016/j.marpolbul.2011.05.030.CrossRefGoogle ScholarPubMed
Aros, CJ, Pantoja, CJ and Gomperts, BN (2021) Wnt signaling in lung development, regeneration, and disease progression. Communications Biology 4(1), 601. https://doi.org/10.1038/s42003-021-02118-w.CrossRefGoogle ScholarPubMed
Assou, S, Boumela, I, Haouzi, D, Anahory, T, Dechaud, H, De Vos, J and Hamamah, S (2011) Dynamic changes in gene expression during human early embryo development: From fundamental aspects to clinical applications. Human Reproduction Update 17(2), 272290. https://doi.org/10.1093/humupd/dmq036.CrossRefGoogle ScholarPubMed
Bahl, S, Dolma, J, Jyot Singh, J and Sehgal, S (2021) Biodegradation of plastics: A state of the art review. Materials Today: Proceedings 39, 3134. https://doi.org/10.1016/j.matpr.2020.06.096.Google Scholar
Barboza, LGA, Cunha, SC, Monteiro, C, Fernandes, JO and Guilhermino, L (2020) Bisphenol A and its analogs in muscle and liver of fish from the north East Atlantic Ocean in relation to microplastic contamination. Exposure and risk to human consumers. Journal of Hazardous Materials 393, 122419. https://doi.org/10.1016/j.jhazmat.2020.122419.CrossRefGoogle ScholarPubMed
Bornehag, CG, Carlstedt, F, Jönsson, BA, Lindh, CH, Jensen, TK, Bodin, A, Jonsson, C, Janson, S and Swan, SH (2015) Prenatal phthalate exposures and anogenital distance in Swedish boys. Environmental Health Perspectives 123(1), 101107. https://doi.org/10.1289/ehp.1408163.CrossRefGoogle ScholarPubMed
Brehm, E, Rattan, S, Gao, L and Flaws, JA (2018) Prenatal exposure to di(2-ethylhexyl) phthalate causes long-term transgenerational effects on female reproduction in mice. Endocrinology 159(2), 795809. https://doi.org/10.1210/en.2017-03004.CrossRefGoogle ScholarPubMed
Cao, Y, Lin, H, Zhang, K, Xu, S, Yan, M, Leung, KMY and Lam, PKS (2022) Microplastics: A major source of phthalate esters in aquatic environments. Journal of Hazardous Materials 432, 128731. https://doi.org/10.1016/j.jhazmat.2022.128731.CrossRefGoogle Scholar
Chen, G, Xiong, S, Jing, Q, van Gestel, CAM, van Straalen, NM, Roelofs, D, Sun, L and Qiu, H (2023) Maternal exposure to polystyrene nanoparticles retarded fetal growth and triggered metabolic disorders of placenta and fetus in mice. Science of the Total Environment 854, 158666. https://doi.org/10.1016/j.scitotenv.2022.158666.CrossRefGoogle ScholarPubMed
Chen, Q, Yin, D, Jia, Y, Schiwy, S, Legradi, J, Yang, S and Hollert, H (2017) Enhanced uptake of BPA in the presence of nanoplastics can lead to neurotoxic effects in adult zebrafish. Science of the Total Environment 609, 13121321. https://doi.org/10.1016/j.scitotenv.2017.07.144.CrossRefGoogle ScholarPubMed
Chin, AM, Hill, DR, Aurora, M and Spence, JR (2017) Morphogenesis and maturation of the embryonic and postnatal intestine. Seminars in Cell & Developmental Biology 66, 8193. https://doi.org/10.1016/j.semcdb.2017.01.011.CrossRefGoogle ScholarPubMed
Cindrova-Davies, T and Sferruzzi-Perri, AN (2022) Human placental development and function. Seminars in Cell & Developmental Biology 131, 6677. https://doi.org/10.1016/j.semcdb.2022.03.039.CrossRefGoogle ScholarPubMed
Clift, D and Schuh, M (2013) Restarting life: Fertilization and the transition from meiosis to mitosis. Nature Reviews Molecular Cell Biology 14(9), 549562. https://doi.org/10.1038/nrm3643.CrossRefGoogle ScholarPubMed
Cunha, GR, Robboy, SJ, Kurita, T, Isaacson, D, Shen, J, Cao, M and Baskin, LS (2018) Development of the human female reproductive tract. Differentiation 103, 4665. https://doi.org/10.1016/j.diff.2018.09.001.CrossRefGoogle ScholarPubMed
Da Costa Filho, PA, Andrey, D, Eriksen, B, Peixoto, RP, Carreres, BM, Ambühl, ME, Descarrega, JB, Dubascoux, S, Zbinden, P, Panchaud, A and Poitevin, E (2021) Detection and characterization of small-sized microplastics (≥ 5 μm) in milk products. Scientific Reports 11(1), 24046. https://doi.org/10.1038/s41598-021-03458-7.CrossRefGoogle Scholar
da Silva Costa, R, Sainara Maia Fernandes, T, de Sousa Almeida, E, Tomé Oliveira, J, Carvalho Guedes, JA, Julião Zocolo, G, Wagner de Sousa, F and do Nascimento, RF (2021) Potential risk of BPA and phthalates in commercial water bottles: A minireview. Journal of Water and Health 19(3), 411435. https://doi.org/10.2166/wh.2021.202.CrossRefGoogle ScholarPubMed
Daniel, S, Balalian, AA, Insel, BJ, Liu, X, Whyatt, RM, Calafat, AM, Rauh, VA, Perera, FP, Hoepner, LA, Herbstman, J and Factor-Litvak, P (2020) Prenatal and early childhood exposure to phthalates and childhood behavior at age 7 years. Environment International 143, 105894. https://doi.org/10.1016/j.envint.2020.105894.CrossRefGoogle ScholarPubMed
Davis, JD and Wypych, TP (2021) Cellular and functional heterogeneity of the airway epithelium. Mucosal Immunology 14(5), 978990. https://doi.org/10.1038/s41385-020-00370-7.CrossRefGoogle ScholarPubMed
DeBenedictis, B, Guan, H and Yang, K (2016) Prenatal exposure to bisphenol A disrupts mouse fetal liver maturation in a sex-specific manner. Journal of Cellular Biochemistry 117(2), 344350. https://doi.org/10.1002/jcb.25276.CrossRefGoogle Scholar
Demitrack, ES and Samuelson, LC (2016) Notch regulation of gastrointestinal stem cells. The Journal of Physiology 594(17), 47914803. https://doi.org/10.1113/jp271667.CrossRefGoogle ScholarPubMed
Depner, M, Taft, DH, Kirjavainen, PV, Kalanetra, KM, Karvonen, AM, Peschel, S, Schmausser-Hechfellner, E, Roduit, C, Frei, R, Lauener, R, Divaret-Chauveau, A, Dalphin, J-C, Riedler, J, Roponen, M, Kabesch, M, Renz, H, Pekkanen, J, Farquharson, FM, Louis, P, Mills, DA, von Mutius, E, Genuneit, J, Hyvärinen, A, Illi, S, Laurent, L, Pfefferle, PI, Schaub, B, von Mutius, E, Ege, MJ and PASTURE Study Group (2020) Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nature Medicine 26(11), 17661775. https://doi.org/10.1038/s41591-020-1095-x.CrossRefGoogle Scholar
Desai, TJ, Brownfield, DG and Krasnow, MA (2014) Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 507(7491), 190194. https://doi.org/10.1038/nature12930.CrossRefGoogle ScholarPubMed
Dijk, Fv, Song, S, Eck, GWAv, Wu, X, Bos, IST, Boom, DHA, Kooter, IM, Spierings, DCJ, Wardenaar, R, Cole, M, Salvati, A, Gosens, R and Melgert, BN (2021) Inhalable textile microplastic fibers impair lung repair. bioRxiv. https://doi.org/10.1101/2021.01.25.428144.CrossRefGoogle Scholar
Dronjak, L, Exposito, N, Rovira, J, Florencio, K, Emiliano, P, Corzo, B, Schuhmacher, M, Valero, F and Sierra, J (2022) Screening of microplastics in water and sludge lines of a drinking water treatment plant in Catalonia, Spain. Water Research 225, 119185. https://doi.org/10.1016/j.watres.2022.119185.CrossRefGoogle Scholar
Dusza, HM, Katrukha, EA, Nijmeijer, SM, Akhmanova, A, Vethaak, AD, Walker, DI and Legler, J (2022) Uptake, transport, and toxicity of pristine and weathered micro- and nanoplastics in human placenta cells. Environmental Health Perspectives 130(9), 97006. https://doi.org/10.1289/ehp10873.CrossRefGoogle ScholarPubMed
Dusza, HM, van Boxel, J, van Duursen, MBM, Forsberg, MM, Legler, J and Vähäkangas, KH (2023) Experimental human placental models for studying uptake, transport and toxicity of micro- and nanoplastics. Science of the Total Environment 860, 160403. https://doi.org/10.1016/j.scitotenv.2022.160403.CrossRefGoogle ScholarPubMed
Ejaredar, M, Lee, Y, Roberts, DJ, Sauve, R and Dewey, D (2017) Bisphenol A exposure and children’s behavior: A systematic review. Journal of Exposure Science & Environmental Epidemiology 27(2), 175183. https://doi.org/10.1038/jes.2016.8.CrossRefGoogle ScholarPubMed
Engel, A, Buhrke, T, Imber, F, Jessel, S, Seidel, A, Völkel, W and Lampen, A (2017) Agonistic and antagonistic effects of phthalates and their urinary metabolites on the steroid hormone receptors ERα, ERβ, and AR. Toxicology Letters 277, 5463. https://doi.org/10.1016/j.toxlet.2017.05.028.CrossRefGoogle ScholarPubMed
Evangeliou, N, Grythe, H, Klimont, Z, Heyes, C, Eckhardt, S, Lopez-Aparicio, S and Stohl, A (2020) Atmospheric transport is a major pathway of microplastics to remote regions. Nature Communications 11(1), 3381. https://doi.org/10.1038/s41467-020-17201-9.CrossRefGoogle Scholar
Fauser, P, Vorkamp, K and Strand, J (2022) Residual additives in marine microplastics and their risk assessment: A critical review. Marine Pollution Bulletin 177, 113467. https://doi.org/10.1016/j.marpolbul.2022.113467.CrossRefGoogle ScholarPubMed
Feher, J (2012) 9.9 – Female reproductive physiology. In Feher, J (ed.), Quantitative Human Physiology. Boston, MA: Academic Press, pp. 846855.10.1016/B978-0-12-382163-8.00092-XCrossRefGoogle Scholar
Foresta, C, Valente, U, Di Nisio, A, Cacco, N, Magagna, S, Cosci, I, Presciutti, A and Garolla, A (2018) Anogenital distance is associated with genital measures and seminal parameters but not anthropometrics in a large cohort of young adult men. Human Reproduction 33(9), 16281635. https://doi.org/10.1093/humrep/dey249.CrossRefGoogle ScholarPubMed
Fossi, MC, Panti, C, Guerranti, C, Coppola, D, Giannetti, M, Marsili, L and Minutoli, R (2012) Are baleen whales exposed to the threat of microplastics? A case study of the Mediterranean fin whale (Balaenoptera physalus). Marine Pollution Bulletin 64(11), 23742379. https://doi.org/10.1016/j.marpolbul.2012.08.013.CrossRefGoogle Scholar
Fournier, SB, D’Errico, JN, Adler, DS, Kollontzi, S, Goedken, MJ, Fabris, L, Yurkow, EJ and Stapleton, PA (2020) Nanopolystyrene translocation and fetal deposition after acute lung exposure during late-stage pregnancy. Particle and Fibre Toxicology 17(1), 55. https://doi.org/10.1186/s12989-020-00385-9.CrossRefGoogle ScholarPubMed
Fournier, E, Ratel, J, Denis, S, Leveque, M, Ruiz, P, Mazal, C, Amiard, F, Edely, M, Bezirard, V, Gaultier, E, Lamas, B, Houdeau, E, Engel, E, Lagarde, F, Etienne-Mesmin, L, Mercier-Bonin, M and Blanquet-Diot, S (2023) Exposure to polyethylene microplastics alters immature gut microbiome in an infant in vitro gut model. Journal of Hazardous Materials 443, 130383. https://doi.org/10.1016/j.jhazmat.2022.130383.CrossRefGoogle Scholar
Garcia-Alonso, L, Lorenzi, V, Mazzeo, CI, Alves-Lopes, JP, Roberts, K, Sancho-Serra, C, Engelbert, J, Marečková, M, Gruhn, WH, Botting, RA, Li, T, Crespo, B, Van Dongen, S, Kiselev, VY, Prigmore, E, Herbert, M, Moffett, A, Chédotal, A, Bayraktar, OA, Surani, A, Haniffa, M and Vento-Tormo, R (2022) Single-cell roadmap of human gonadal development. Nature 607(7919), 540547. https://doi.org/10.1038/s41586-022-04918-4.CrossRefGoogle ScholarPubMed
Gasperi, J, Wright, SL, Dris, R, Collard, F, Mandin, C, Guerrouache, M, Langlois, V, Kelly, FJ and Tassin, B (2018) Microplastics in air: Are we breathing it in? Current Opinion in Environmental Science & Health 1, 15. https://doi.org/10.1016/j.coesh.2017.10.002.CrossRefGoogle Scholar
Gaston, E, Woo, M, Steele, C, Sukumaran, S and Anderson, S (2020) Microplastics differ between indoor and outdoor air masses: Insights from multiple microscopy methodologies. Applied Spectroscopy 74(9), 10791098. https://doi.org/10.1177/0003702820920652.CrossRefGoogle ScholarPubMed
Giancotti, A, Monti, M, Nevi, L, Safarikia, S, D’Ambrosio, V, Brunelli, R, Pajno, C, Corno, S, Di Donato, V, Musella, A, Chiappetta, MF, Bosco, D, Benedetti Panici, P, Alvaro, D and Cardinale, V (2019) Functions and the emerging role of the foetal liver into regenerative medicine. Cell 8, 914. https://doi.org/10.3390/cells8080914.CrossRefGoogle ScholarPubMed
Gijsman, P and Dozeman, A (1996) Comparison of the UV-degradation chemistry of unstabilized and HALS-stabilized polyethylene and polypropylene. Polymer Degradation and Stability 53(1), 4550. https://doi.org/10.1016/0141-3910(96)00027-4.CrossRefGoogle Scholar
Gouin, T, Cunliffe, D, De France, J, Fawell, J, Jarvis, P, Koelmans, AA, Marsden, P, Testai, EE, Asami, M, Bevan, R, Carrier, R, Cotruvo, J, Eckhardt, A and Ong, CN (2021) Clarifying the absence of evidence regarding human health risks to microplastic particles in drinking-water: High quality robust data wanted. Environment International 150, 106141. https://doi.org/10.1016/j.envint.2020.106141.CrossRefGoogle ScholarPubMed
Grohs, MN, Reynolds, JE, Liu, J, Martin, JW, Pollock, T, Lebel, C and Dewey, D (2019) Prenatal maternal and childhood bisphenol A exposure and brain structure and behavior of young children. Environmental Health 18(1), 85. https://doi.org/10.1186/s12940-019-0528-9.CrossRefGoogle ScholarPubMed
Guerranti, C, Martellini, T, Perra, G, Scopetani, C and Cincinelli, A (2019) Microplastics in cosmetics: Environmental issues and needs for global bans. Environmental Toxicology and Pharmacology 68, 7579. https://doi.org/10.1016/j.etap.2019.03.007.CrossRefGoogle ScholarPubMed
Guilbert, A, Rolland, M, Pin, I, Thomsen, C, Sakhi, AK, Sabaredzovic, A, Slama, R, Guichardet, K and Philippat, C (2021) Associations between a mixture of phenols and phthalates and child behaviour in a French mother–child cohort with repeated assessment of exposure. Environment International 156, 106697. https://doi.org/10.1016/j.envint.2021.106697.CrossRefGoogle Scholar
Hanioka, N, Naito, T and Narimatsu, S (2008) Human UDP-glucuronosyltransferase isoforms involved in bisphenol A glucuronidation. Chemosphere 74(1), 3336. https://doi.org/10.1016/j.chemosphere.2008.09.053.CrossRefGoogle ScholarPubMed
Hartmann, NB, Hüffer, T, Thompson, RC, Hassellöv, M, Verschoor, A, Daugaard, AE, Rist, S, Karlsson, T, Brennholt, N, Cole, M, Herrling, MP, Hess, MC, Ivleva, NP, Lusher, AL and Wagner, M (2019) Are we speaking the same language? Recommendations for a definition and categorization framework for plastic debris. Environmental Science & Technology 53(3), 10391047. https://doi.org/10.1021/acs.est.8b05297.CrossRefGoogle ScholarPubMed
Hass, U, Christiansen, S, Boberg, J, Rasmussen, MG, Mandrup, K and Axelstad, M (2016) Low-dose effect of developmental bisphenol A exposure on sperm count and behaviour in rats. Andrology 4(4), 594607. https://doi.org/10.1111/andr.12176.CrossRefGoogle ScholarPubMed
Healey, A (2012) Embryology of the female reproductive tract. In Mann, GS, Blair, JC and Garden, AS (eds), Imaging of Gynecological Disorders in Infants and Children. Berlin–Heidelberg: Springer, pp. 2130.Google Scholar
Hines, RN (2008) The ontogeny of drug metabolism enzymes and implications for adverse drug events. Pharmacology & Therapeutics 118(2), 250267. https://doi.org/10.1016/j.pharmthera.2008.02.005.CrossRefGoogle ScholarPubMed
Hoelting, L, Scheinhardt, B, Bondarenko, O, Schildknecht, S, Kapitza, M, Tanavde, V, Tan, B, Lee, QY, Mecking, S, Leist, M and Kadereit, S (2013) A 3-dimensional human embryonic stem cell (hESC)-derived model to detect developmental neurotoxicity of nanoparticles. Archives of Toxicology 87(4), 721733. https://doi.org/10.1007/s00204-012-0984-2.CrossRefGoogle ScholarPubMed
Horvatits, T, Tamminga, M, Liu, B, Sebode, M, Carambia, A, Fischer, L, Püschel, K, Huber, S and Fischer, EK (2022) Microplastics detected in cirrhotic liver tissue. eBioMedicine 82, 104147. https://doi.org/10.1016/j.ebiom.2022.104147.CrossRefGoogle ScholarPubMed
Hsu, PC, Jhong, JY, Huang, LP, Lee, KH, Chen, HP and Guo, YL (2021) Transgenerational effects of di(2-ethylhexyl) phthalate on anogenital distance, sperm functions and DNA methylation in rat offspring. International Journal of Molecular Sciences 22(8), 4131. https://doi.org/10.3390/ijms22084131.CrossRefGoogle ScholarPubMed
Hua, T, Kiran, S, Li, Y and Sang, Q-XA (2022) Microplastics exposure affects neural development of human pluripotent stem cell-derived cortical spheroids. Journal of Hazardous Materials 435, 128884. https://doi.org/10.1016/j.jhazmat.2022.128884.CrossRefGoogle ScholarPubMed
Huang, T, Zhang, W, Lin, T, Liu, S, Sun, Z, Liu, F, Yuan, Y, Xiang, X, Kuang, H, Yang, B and Zhang, D (2022) Maternal exposure to polystyrene nanoplastics during gestation and lactation induces hepatic and testicular toxicity in male mouse offspring. Food and Chemical Toxicology 160, 112803. https://doi.org/10.1016/j.fct.2021.112803.CrossRefGoogle ScholarPubMed
Hunt, PA, Lawson, C, Gieske, M, Murdoch, B, Smith, H, Marre, A, Hassold, T and Vandevoort, CA (2012) Bisphenol A alters early oogenesis and follicle formation in the fetal ovary of the rhesus monkey. Proceedings of the National Academy of Sciences 109(43), 1752517530. https://doi.org/10.1073/pnas.1207854109.CrossRefGoogle ScholarPubMed
Ibrahim, YS, Tuan Anuar, S, Azmi, AA, Wan Mohd Khalik, WMA, Lehata, S, Hamzah, SR, Ismail, D, Ma, ZF, Dzulkarnaen, A, Zakaria, Z, Mustaffa, N, Tuan Sharif, SE and Lee, YY (2021) Detection of microplastics in human colectomy specimens. JGH Open 5(1), 116121. https://doi.org/10.1002/jgh3.12457.CrossRefGoogle ScholarPubMed
Im, G-B, Kim, YG, Jo, I-S, Yoo, TY, Kim, S-W, Park, HS, Hyeon, T, G-R, Yi and Bhang, SH (2022) Effect of polystyrene nanoplastics and their degraded forms on stem cell fate. Journal of Hazardous Materials 430, 128411. https://doi.org/10.1016/j.jhazmat.2022.128411.CrossRefGoogle ScholarPubMed
Jenner, LC, Rotchell, JM, Bennett, RT, Cowen, M, Tentzeris, V and Sadofsky, LR (2022) Detection of microplastics in human lung tissue using μFTIR spectroscopy. Science of the Total Environment 831, 154907. https://doi.org/10.1016/j.scitotenv.2022.154907.CrossRefGoogle ScholarPubMed
Jenner, LC, Sadofsky, LR, Danopoulos, E and Rotchell, JM (2021) Household indoor microplastics within the Humber region (United Kingdom): Quantification and chemical characterisation of particles present. Atmospheric Environment 259, 118512. https://doi.org/10.1016/j.atmosenv.2021.118512.CrossRefGoogle Scholar
Jeong, B, Baek, JY, Koo, J, Park, S, Ryu, Y-K, Kim, K-S, Zhang, S, Chung, C, Dogan, R, Choi, H-S, Um, D, Kim, T-K, Lee, WS, Jeong, J, Shin, W-H, Lee, J-R, N-S, Kim and Lee, DY (2022) Maternal exposure to polystyrene nanoplastics causes brain abnormalities in progeny. Journal of Hazardous Materials 426, 127815. https://doi.org/10.1016/j.jhazmat.2021.127815.CrossRefGoogle ScholarPubMed
Jin, M, Wang, X, Ren, T, Wang, J and Shan, J (2021) Microplastics contamination in food and beverages: Direct exposure to humans. Journal of Food Science 86(7), 28162837. https://doi.org/10.1111/1750-3841.15802.CrossRefGoogle ScholarPubMed
Josso, N, Lamarre, I, Picard, J-Y, Berta, P, Davies, N, Morichon, N, Peschanski, M and Jeny, R (1993) Anti-Müllerian hormone in early human development. Early Human Development 33(2), 9199. https://doi.org/10.1016/0378-3782(93)90204-8.CrossRefGoogle ScholarPubMed
Karavan, JR and Pepling, ME (2012) Effects of estrogenic compounds on neonatal oocyte development. Reproductive Toxicology 34(1), 5156. https://doi.org/10.1016/j.reprotox.2012.02.005.CrossRefGoogle ScholarPubMed
Kiyokawa, H and Morimoto, M (2020) Notch signaling in the mammalian respiratory system, specifically the trachea and lungs, in development, homeostasis, regeneration, and disease. Development, Growth & Differentiation 62(1), 6779. https://doi.org/10.1111/dgd.12628.CrossRefGoogle ScholarPubMed
Klaeger, F, Tagg, AS, Otto, S, Bienmüller, M, Sartorius, I and Labrenz, M (2019) Residual monomer content affects the interpretation of plastic degradation. Scientific Reports 9(1), 2120. https://doi.org/10.1038/s41598-019-38685-6.CrossRefGoogle ScholarPubMed
Koelmans, AA, Mohamed Nor, NH, Hermsen, E, Kooi, M, Mintenig, SM and De France, J (2019) Microplastics in freshwaters and drinking water: Critical review and assessment of data quality. Water Research 155, 410422. https://doi.org/10.1016/j.watres.2019.02.054.CrossRefGoogle ScholarPubMed
Koelmans, AA, Redondo-Hasselerharm, PE, Nor, NHM, De Ruijter, VN, Mintenig, SM and Kooi, M (2022) Risk assessment of microplastic particles. Nature Reviews Materials 7(2), 138152. https://doi.org/10.1038/s41578-021-00411-y.CrossRefGoogle Scholar
Koopman, P, Gubbay, J, Vivian, N, Goodfellow, P and Lovell-Badge, R (1991) Male development of chromosomally female mice transgenic for Sry. Nature 351(6322), 117121. https://doi.org/10.1038/351117a0.CrossRefGoogle ScholarPubMed
Lee, H-S, Amarakoon, D, C-I, Wei, Choi, KY, Smolensky, D and Lee, S-H (2021) Adverse effect of polystyrene microplastics (PS-MPs) on tube formation and viability of human umbilical vein endothelial cells. Food and Chemical Toxicology 154, 112356. https://doi.org/10.1016/j.fct.2021.112356.CrossRefGoogle ScholarPubMed
Leslie, HA and Depledge, MH (2020) Where is the evidence that human exposure to microplastics is safe? Environment International 142, 105807. https://doi.org/10.1016/j.envint.2020.105807.CrossRefGoogle ScholarPubMed
Leslie, HA, van Velzen, MJM, Brandsma, SH, Vethaak, AD, Garcia-Vallejo, JJ and Lamoree, MH (2022) Discovery and quantification of plastic particle pollution in human blood. Environment International 163, 107199. https://doi.org/10.1016/j.envint.2022.107199.CrossRefGoogle ScholarPubMed
Levardon, H, Yonker, LM, Hurley, BP and Mou, H (2018) Expansion of airway basal cells and generation of polarized epithelium. Bio-Protocol 8(11), e2877. https://doi.org/10.21769/BioProtoc.2877.CrossRefGoogle ScholarPubMed
Lewandowski, TA, Hayes, AW and Beck, BD (2005) Risk evaluation of occupational exposure to methylene dianiline and toluene diamine in polyurethane foam. Human & Experimental Toxicology 24(12), 655662. https://doi.org/10.1191/0960327105ht587oa.CrossRefGoogle ScholarPubMed
Li, F, He, J, Wei, J, Cho, WC and Liu, X (2015) Diversity of epithelial stem cell types in adult lung. Stem Cells International 2015, 728307. https://doi.org/10.1155/2015/728307.CrossRefGoogle ScholarPubMed
Li, J, Liu, H and Paul Chen, J (2018) Microplastics in freshwater systems: A review on occurrence, environmental effects, and methods for microplastics detection. Water Research 137, 362374. https://doi.org/10.1016/j.watres.2017.12.056.CrossRefGoogle ScholarPubMed
Li, D, Shi, Y, Yang, L, Xiao, L, Kehoe, DK, Gun’Ko, YK, Boland, JJ and Wang, JJ (2020) Microplastic release from the degradation of polypropylene feeding bottles during infant formula preparation. Nature Food 1(11), 746754. https://doi.org/10.1038/s43016-020-00171-y.CrossRefGoogle ScholarPubMed
Liu, S, Guo, J, Liu, X, Yang, R, Wang, H, Sun, Y, Chen, B and Dong, R (2023) Detection of various microplastics in placentas, meconium, infant feces, breastmilk and infant formula: A pilot prospective study. Science of the Total Environment 854, 158699. https://doi.org/10.1016/j.scitotenv.2022.158699.CrossRefGoogle ScholarPubMed
Liu, X, Shi, H, Xie, B, Dionysiou, DD and Zhao, Y (2019) Microplastics as both a sink and a source of bisphenol A in the marine environment. Environmental Science & Technology 53(17), 1018810196. https://doi.org/10.1021/acs.est.9b02834.CrossRefGoogle Scholar
Long, Z, Fan, J, Wu, G, Liu, X, Wu, H, Liu, J, Chen, Y, Su, S, Cheng, X, Xu, Z, Su, H, Cao, M, Zhang, C, Hai, C and Wang, X (2021) Gestational bisphenol A exposure induces fatty liver development in male offspring mice through the inhibition of HNF1b and upregulation of PPARγ. Cell Biology and Toxicology 37(1), 6584. https://doi.org/10.1007/s10565-020-09535-3.CrossRefGoogle ScholarPubMed
López-Vázquez, J, Rodil, R, Trujillo-Rodríguez, MJ, Quintana, JB, Cela, R and Miró, M (2022) Mimicking human ingestion of microplastics: Oral bioaccessibility tests of bisphenol A and phthalate esters under fed and fasted states. Science of the Total Environment 826, 154027. https://doi.org/10.1016/j.scitotenv.2022.154027.CrossRefGoogle ScholarPubMed
Lu, IC, Chao, H-R, Mansor, W-N-W, Peng, C-W, Hsu, Y-C, Yu, T-Y, Chang, W-H and Fu, L-M (2021) Levels of phthalates, bisphenol-A, nonylphenol, and microplastics in fish in the estuaries of northern Taiwan and the impact on human health. Toxics 9(10), 246. https://doi.org/10.3390/toxics9100246.CrossRefGoogle ScholarPubMed
Lu, X, Zeng, F, Wei, S, Gao, R, Abdurahman, A, Wang, H and Liang, W (2022) Effects of humic acid on Pb2+ adsorption onto polystyrene microplastics from spectroscopic analysis and site energy distribution analysis. Scientific Reports 12(1), 8932. https://doi.org/10.1038/s41598-022-12776-3.CrossRefGoogle ScholarPubMed
Lucaccioni, L, Trevisani, V, Passini, E, Righi, B, Plessi, C, Predieri, B and Iughetti, L (2021) Perinatal exposure to phthalates: From endocrine to neurodevelopment effects. International Journal of Molecular Sciences 22(8), 4063. https://doi.org/10.3390/ijms22084063.CrossRefGoogle ScholarPubMed
Luo, T, Wang, C, Pan, Z, Jin, C, Fu, Z and Jin, Y (2019a) Maternal polystyrene microplastic exposure during gestation and lactation altered metabolic homeostasis in the dams and their F1 and F2 offspring. Environmental Science & Technology 53(18), 1097810992. https://doi.org/10.1021/acs.est.9b03191.CrossRefGoogle ScholarPubMed
Luo, T, Zhang, Y, Wang, C, Wang, X, Zhou, J, Shen, M, Zhao, Y, Fu, Z and Jin, Y (2019b) Maternal exposure to different sizes of polystyrene microplastics during gestation causes metabolic disorders in their offspring. Environmental Pollution 255, 113122. https://doi.org/10.1016/j.envpol.2019.113122.CrossRefGoogle ScholarPubMed
Ma, T, Yin, X, Han, R, Ding, J, Zhang, H, Han, X and Li, D (2017) Effects of in utero exposure to di-n-butyl phthalate on testicular development in rat. International Journal of Environmental Research and Public Health 14(10), 1284. https://doi.org/10.3390/ijerph14101284.CrossRefGoogle ScholarPubMed
Maitre, L, Bustamante, M, Hernández-Ferrer, C, Thiel, D, Lau, C-HE, Siskos, AP, Vives-Usano, M, Ruiz-Arenas, C, Pelegrí-Sisó, D, Robinson, O, Mason, D, Wright, J, Cadiou, S, Slama, R, Heude, B, Casas, M, Sunyer, J, Papadopoulou, EZ, Gutzkow, KB, Andrusaityte, S, Grazuleviciene, R, Vafeiadi, M, Chatzi, L, Sakhi, AK, Thomsen, C, Tamayo, I, Nieuwenhuijsen, M, Urquiza, J, Borràs, E, Sabidó, E, Quintela, I, Carracedo, Á, Estivill, X, Coen, M, González, JR, Keun, HC and Vrijheid, M (2022) Multi-omics signatures of the human early life exposome. Nature Communications 13(1), 7024. https://doi.org/10.1038/s41467-022-34422-2.CrossRefGoogle ScholarPubMed
Makiyan, Z (2016) Studies of gonadal sex differentiation. Organogenesis 12(1), 4251. https://doi.org/10.1080/15476278.2016.1145318.CrossRefGoogle ScholarPubMed
Mammadov, E, Uncu, M and Dalkan, C (2018) High prenatal exposure to bisphenol A reduces anogenital distance in healthy male newborns. Journal of Clinical Research in Pediatric Endocrinology 10(1), 2529. https://doi.org/10.4274/jcrpe.4817.CrossRefGoogle ScholarPubMed
Maranghi, F, Lorenzetti, S, Tassinari, R, Moracci, G, Tassinari, V, Marcoccia, D, Di Virgilio, A, Eusepi, A, Romeo, A, Magrelli, A, Salvatore, M, Tosto, F, Viganotti, M, Antoccia, A, Di Masi, A, Azzalin, G, Tanzarella, C, Macino, G, Taruscio, D and Mantovani, A (2010) In utero exposure to di-(2-ethylhexyl) phthalate affects liver morphology and metabolism in post-natal CD-1 mice. Reproductive Toxicology 29(4), 427432. https://doi.org/10.1016/j.reprotox.2010.03.002.CrossRefGoogle ScholarPubMed
McCracken, KW and Wells, JM (2017) Mechanisms of embryonic stomach development. Seminars in Cell & Developmental Biology 66, 3642. https://doi.org/10.1016/j.semcdb.2017.02.004.CrossRefGoogle ScholarPubMed
Mei, W, Chen, G, Bao, J, Song, M, Li, Y and Luo, C (2020) Interactions between microplastics and organic compounds in aquatic environments: A mini review. Science of the Total Environment 736, 139472. https://doi.org/10.1016/j.scitotenv.2020.139472.CrossRefGoogle ScholarPubMed
Min, K, Cuiffi, JD and Mathers, RT (2020) Ranking environmental degradation trends of plastic marine debris based on physical properties and molecular structure. Nature Communications 11(1), 727. https://doi.org/10.1038/s41467-020-14538-z.CrossRefGoogle ScholarPubMed
Mok, S, Jeong, Y, Park, M, Kim, S, Lee, I, Park, J, Kim, S, Choi, K and Moon, H-B (2021) Exposure to phthalates and bisphenol analogues among childbearing-aged women in Korea: Influencing factors and potential health risks. Chemosphere 264, 128425. https://doi.org/10.1016/j.chemosphere.2020.128425.CrossRefGoogle ScholarPubMed
Najahi, H, Alessio, N, Squillaro, T, Conti, GO, Ferrante, M, Di Bernardo, G, Galderisi, U, Messaoudi, I, Minucci, S and Banni, M (2022) Environmental microplastics (EMPs) exposure alter the differentiation potential of mesenchymal stromal cells. Environmental Research 214, 114088. https://doi.org/10.1016/j.envres.2022.114088.CrossRefGoogle ScholarPubMed
Nassar, GN and Leslie, SW (2023) Physiology, testosterone. In StatPearls. Treasure Island, FL: StatPearls Publishing. Available at: https://www.ncbi.nlm.nih.gov/books/NBK526128/ (Accessed 3 Dec 2022).Google Scholar
Nesan, D, Sewell, LC and Kurrasch, DM (2018) Opening the black box of endocrine disruption of brain development: Lessons from the characterization of bisphenol A. Hormones and Behavior 101, 5058. https://doi.org/10.1016/j.yhbeh.2017.12.001.CrossRefGoogle ScholarPubMed
Olajuyin, AM, Zhang, X and Ji, H-L (2019) Alveolar type 2 progenitor cells for lung injury repair. Cell Death Discovery 5(1), 63. https://doi.org/10.1038/s41420-019-0147-9.CrossRefGoogle ScholarPubMed
Park, E-J, Han, J-S, Park, E-J, Seong, E, Lee, G-H, Kim, D-W, Son, H-Y, H-Y, Han and Lee, B-S (2020) Repeated-oral dose toxicity of polyethylene microplastics and the possible implications on reproduction and development of the next generation. Toxicology Letters 324, 7585. https://doi.org/10.1016/j.toxlet.2020.01.008.CrossRefGoogle ScholarPubMed
Pask, A (2016) The reproductive system. In Wilhelm, D and Bernard, P (eds), Non-coding RNA and the Reproductive System. Dordrecht: Springer, pp. 112.Google Scholar
Peñalver, R, Arroyo-Manzanares, N, López-García, I and Hernández-Córdoba, M (2020) An overview of microplastics characterization by thermal analysis. Chemosphere 242, 125170. https://doi.org/10.1016/j.chemosphere.2019.125170.CrossRefGoogle ScholarPubMed
Pittenger, MF, Discher, DE, Péault, BM, Phinney, DG, Hare, JM and Caplan, AI (2019) Mesenchymal stem cell perspective: Cell biology to clinical progress. npj Regenerative Medicine 4(1), 22. https://doi.org/10.1038/s41536-019-0083-6.CrossRefGoogle ScholarPubMed
Plastics Europe (2021) Plastics: The Facts 2021. Plastics Europe. Available at https://plasticseurope.org/knowledge-hub/plastics-the-facts-2021/ (accessed 11 July 2023).Google Scholar
Pleasure, JR, Pleasure, D and Pleasure, SJ (2017) 133: Trophic Factor, nutritional, and hormonal regulation of brain development. In Polin, RA, Abman, SH, Rowitch, DH, Benitz, WE and Fox, WW (eds), Fetal and Neonatal Physiology, 5th Edn. Elsevier, pp. 13261333.e1323.10.1016/B978-0-323-35214-7.00133-5CrossRefGoogle Scholar
Priskorn, L, Bang, AK, Nordkap, L, Krause, M, Mendiola, J, Jensen, TK, Juul, A, Skakkebaek, NE, Swan, SH and Jørgensen, N (2019) Anogenital distance is associated with semen quality but not reproductive hormones in 1106 young men from the general population. Human Reproduction 34(1), 1224. https://doi.org/10.1093/humrep/dey326.CrossRefGoogle Scholar
Priskorn, L, Petersen, JH, Jørgensen, N, Kyhl, HB, Andersen, MS, Main, KM, Andersson, A-M, Skakkebaek, NE and Jensen, TK (2018) Anogenital distance as a phenotypic signature through infancy. Pediatric Research 83(3), 573579. https://doi.org/10.1038/pr.2017.287.CrossRefGoogle ScholarPubMed
Ragusa, A, Matta, M, Cristiano, L, Matassa, R, Battaglione, E, Svelato, A, De Luca, C, D’Avino, S, Gulotta, A, Rongioletti, MCA, Catalano, P, Santacroce, C, Notarstefano, V, Carnevali, O, Giorgini, E, Vizza, E, Familiari, G and Nottola, SA (2022a) Deeply in plasticenta: Presence of microplastics in the intracellular compartment of human placentas. International Journal of Environmental Research and Public Health 19(18), 11593. https://doi.org/10.3390/ijerph191811593.CrossRefGoogle ScholarPubMed
Ragusa, A, Notarstefano, V, Svelato, A, Belloni, A, Gioacchini, G, Blondeel, C, Zucchelli, E, De Luca, C, D’Avino, S, Gulotta, A, Carnevali, O and Giorgini, E (2022b) Raman microspectroscopy detection and characterisation of microplastics in human breastmilk. Polymers 14(13), 2700. https://doi.org/10.3390/polym14132700.CrossRefGoogle ScholarPubMed
Ragusa, A, Svelato, A, Santacroce, C, Catalano, P, Notarstefano, V, Carnevali, O, Papa, F, Rongioletti, MCA, Baiocco, F, Draghi, S, D’Amore, E, Rinaldo, D, Matta, M and Giorgini, E (2021) Plasticenta: First evidence of microplastics in human placenta. Environment International 146, 106274. https://doi.org/10.1016/j.envint.2020.106274.CrossRefGoogle ScholarPubMed
Rahman, MS, Kwon, W-S, Karmakar, PC, Yoon, S-J, Ryu, B-Y and Pang, M-G (2017) Gestational exposure to bisphenol A affects the function and proteome profile of F1 spermatozoa in adult mice. Environmental Health Perspectives 125(2), 238245. https://doi.org/10.1289/ehp378.CrossRefGoogle ScholarPubMed
Relaix, F and Zammit, PS (2012) Satellite cells are essential for skeletal muscle regeneration: The cell on the edge returns centre stage. Development 139(16), 28452856. https://doi.org/10.1242/dev.069088.CrossRefGoogle ScholarPubMed
Repouskou, A, Panagiotidou, E, Panagopoulou, L, Bisting, PL, Tuck, AR, Sjödin, MOD, Lindberg, J, Bozas, E, Rüegg, J, Gennings, C, Bornehag, C-G, Damdimopoulou, P, Stamatakis, A and Kitraki, E (2019) Gestational exposure to an epidemiologically defined mixture of phthalates leads to gonadal dysfunction in mouse offspring of both sexes. Scientific Reports 9(1), 6424. https://doi.org/10.1038/s41598-019-42377-6.CrossRefGoogle Scholar
Rodriguez, AK, Mansoor, B, Ayoub, G, Colin, X and Benzerga, AA (2020) Effect of UV-aging on the mechanical and fracture behavior of low density polyethylene. Polymer Degradation and Stability 180, 109185. https://doi.org/10.1016/j.polymdegradstab.2020.109185.CrossRefGoogle Scholar
Rodríguez, HA, Santambrosio, N, Santamaría, CG, Muñoz-de-Toro, M and Luque, EH (2010) Neonatal exposure to bisphenol A reduces the pool of primordial follicles in the rat ovary. Reproductive Toxicology 30(4), 550557. https://doi.org/10.1016/j.reprotox.2010.07.008.CrossRefGoogle ScholarPubMed
Roduit, C, Frei, R, Ferstl, R, Loeliger, S, Westermann, P, Rhyner, C, Schiavi, E, Barcik, W, Rodriguez-Perez, N, Wawrzyniak, M, Chassard, C, Lacroix, C, Schmausser-Hechfellner, E, Depner, M, von Mutius, E, Braun-Fahrländer, C, Karvonen, AM, Kirjavainen, PV, Pekkanen, J, Dalphin, JC, Riedler, J, Akdis, C, Lauener, R and O’Mahony, L (2019) High levels of butyrate and propionate in early life are associated with protection against atopy. Allergy 74(4), 799809. https://doi.org/10.1111/all.13660.CrossRefGoogle ScholarPubMed
Rolfo, A, Nuzzo, AM, De Amicis, R, Moretti, L, Bertoli, S and Leone, A (2020) Fetal–maternal exposure to endocrine disruptors: Correlation with diet intake and pregnancy outcomes. Nutrients 12(6), 1744. https://doi.org/10.3390/nu12061744.CrossRefGoogle ScholarPubMed
Rolland, M, Lyon-Caen, S, Thomsen, C, Sakhi, AK, Sabaredzovic, A, Bayat, S, Slama, R, Méary, D and Philippat, C (2023) Effects of early exposure to phthalates on cognitive development and visual behavior at 24 months. Environmental Research 219, 115068. https://doi.org/10.1016/j.envres.2022.115068.CrossRefGoogle ScholarPubMed
Rossant, J and Tam, PPL (2022) Early human embryonic development: Blastocyst formation to gastrulation. Developmental Cell 57(2), 152165. https://doi.org/10.1016/j.devcel.2021.12.022.CrossRefGoogle ScholarPubMed
Roswall, J, Olsson, LM, Kovatcheva-Datchary, P, Nilsson, S, Tremaroli, V, Simon, M-C, Kiilerich, P, Akrami, R, Krämer, M, Uhlén, M, Gummesson, A, Kristiansen, K, Dahlgren, J and Bäckhed, F (2021) Developmental trajectory of the healthy human gut microbiota during the first 5 years of life. Cell Host & Microbe 29(5), 765776.e763. https://doi.org/10.1016/j.chom.2021.02.021.CrossRefGoogle ScholarPubMed
Rubin, BS (2011) Bisphenol A: An endocrine disruptor with widespread exposure and multiple effects. The Journal of Steroid Biochemistry and Molecular Biology 127(1), 2734. https://doi.org/10.1016/j.jsbmb.2011.05.002.CrossRefGoogle ScholarPubMed
Salian, S, Doshi, T and Vanage, G (2009) Perinatal exposure of rats to bisphenol A affects the fertility of male offspring. Life Sciences 85(21), 742752. https://doi.org/10.1016/j.lfs.2009.10.004.CrossRefGoogle ScholarPubMed
Sansom, SN, Griffiths, DS, Faedo, A, Kleinjan, D-J, Ruan, Y, Smith, J, Van Heyningen, V, Rubenstein, JL and Livesey, FJ (2009) The level of the transcription factor Pax6 is essential for controlling the balance between neural stem cell self-renewal and neurogenesis. PLoS Genetics 5(6), e1000511. https://doi.org/10.1371/journal.pgen.1000511.CrossRefGoogle ScholarPubMed
Schittny, JC (2017) Development of the lung. Cell and Tissue Research 367(3), 427444. https://doi.org/10.1007/s00441-016-2545-0.CrossRefGoogle ScholarPubMed
Schwartz, CL, Christiansen, S, Vinggaard, AM, Axelstad, M, Hass, U and Svingen, T (2019) Anogenital distance as a toxicological or clinical marker for fetal androgen action and risk for reproductive disorders. Archives of Toxicology 93(2), 253272. https://doi.org/10.1007/s00204-018-2350-5.CrossRefGoogle ScholarPubMed
Sessa, F, Polito, R, Monda, V, Scarinci, A, Salerno, M, Carotenuto, M, Cibelli, G, Valenzano, A, Campanozzi, A, Mollica, MP, Monda, M and Messina, G (2021) Effects of a plastic-free lifestyle on urinary bisphenol A levels in school-aged children of southern Italy: A pilot study. Frontiers in Public Health 9, 626070. https://doi.org/10.3389/fpubh.2021.626070.CrossRefGoogle ScholarPubMed
Sheaffer, KL and Kaestner, KH (2012) Transcriptional networks in liver and intestinal development. Cold Spring Harbor Perspectives in Biology 4(9), a008284. https://doi.org/10.1101/cshperspect.a008284.CrossRefGoogle ScholarPubMed
Shen, F, Li, D, Guo, J and Chen, J (2022) Mechanistic toxicity assessment of differently sized and charged polystyrene nanoparticles based on human placental cells. Water Research 223, 118960. https://doi.org/10.1016/j.watres.2022.118960.CrossRefGoogle ScholarPubMed
Shengchen, W, Jing, L, Yujie, Y, Yue, W and Shiwen, X (2021) Polystyrene microplastics-induced ROS overproduction disrupts the skeletal muscle regeneration by converting myoblasts into adipocytes. Journal of Hazardous Materials 417, 125962. https://doi.org/10.1016/j.jhazmat.2021.125962.CrossRefGoogle ScholarPubMed
Shi, J, Dong, Y, Shi, Y, Yin, T, He, W, An, T, Tang, Y, Hou, X, Chong, S, Chen, D, Qin, K and Lin, H (2022) Groundwater antibiotics and microplastics in a drinking-water source area, northern China: Occurrence, spatial distribution, risk assessment, and correlation. Environmental Research 210, 112855. https://doi.org/10.1016/j.envres.2022.112855.CrossRefGoogle Scholar
Shin, D and Monga, SPS (2013) Cellular and molecular basis of liver development. Comprehensive Physiology 3, 799815. https://doi.org/10.1002/cphy.c120022.CrossRefGoogle ScholarPubMed
Sol, CM, Santos, S, Duijts, L, Asimakopoulos, AG, Martinez-Moral, M-P, Kannan, K, Jaddoe, VWV and Trasande, L (2020) Fetal phthalates and bisphenols and childhood lipid and glucose metabolism. A population-based prospective cohort study. Environment International 144, 106063. https://doi.org/10.1016/j.envint.2020.106063.CrossRefGoogle ScholarPubMed
Spence, JR, Lauf, R and Shroyer, NF (2011) Vertebrate intestinal endoderm development. Developmental Dynamics 240(3), 501520. https://doi.org/10.1002/dvdy.22540.CrossRefGoogle ScholarPubMed
Stiemsma, LT and Turvey, SE (2017) Asthma and the microbiome: Defining the critical window in early life. Allergy, Asthma & Clinical Immunology 13(1), 3. https://doi.org/10.1186/s13223-016-0173-6.CrossRefGoogle ScholarPubMed
Strakovsky, RS, Wang, H, Engeseth, NJ, Flaws, JA, Helferich, WG, Pan, Y-X and Lezmi, S (2015) Developmental bisphenol A (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicology and Applied Pharmacology 284(2), 101112. https://doi.org/10.1016/j.taap.2015.02.021.CrossRefGoogle ScholarPubMed
Sun, X, Li, D, Liang, H, Miao, M, Song, X, Wang, Z, Zhou, Z and Yuan, W (2018) Maternal exposure to bisphenol A and anogenital distance throughout infancy: A longitudinal study from Shanghai, China. Environment International 121, 269275. https://doi.org/10.1016/j.envint.2018.08.055.CrossRefGoogle ScholarPubMed
Susiarjo, M, Hassold, TJ, Freeman, E and Hunt, PA (2007) Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genetics 3(1), e5. https://doi.org/10.1371/journal.pgen.0030005.CrossRefGoogle ScholarPubMed
Swan, SH, Main, KM, Liu, F, Stewart, SL, Kruse, RL, Calafat, AM, Mao, CS, Redmon, JB, Ternand, CL, Sullivan, S and Teague, JL (2005) Decrease in anogenital distance among male infants with prenatal phthalate exposure. Environmental Health Perspectives 113(8), 10561061. https://doi.org/10.1289/ehp.8100.CrossRefGoogle ScholarPubMed
Tang, Z-R, Xu, X-L, Deng, S-L, Z-X, Lian and Yu, K (2020) Oestrogenic endocrine disruptors in the placenta and the fetus. International Journal of Molecular Sciences 21(4), 1519. https://doi.org/10.3390/ijms21041519.CrossRefGoogle ScholarPubMed
Thakurela, S, Tiwari, N, Schick, S, Garding, A, Ivanek, R, Berninger, B and Tiwari, VK (2016) Mapping gene regulatory circuitry of Pax6 during neurogenesis. Cell Discovery 2(1), 15045. https://doi.org/10.1038/celldisc.2015.45.CrossRefGoogle ScholarPubMed
Tiwari, SK, Agarwal, S, Chauhan, LKS, Mishra, VN and Chaturvedi, RK (2019) Correction to: bisphenol-A impairs myelination potential during development in the hippocampus of the rat brain. Molecular Neurobiology 56(7), 52705271. https://doi.org/10.1007/s12035-019-1611-5.CrossRefGoogle ScholarPubMed
Uccelli, A, Moretta, L and Pistoia, V (2008) Mesenchymal stem cells in health and disease. Nature Reviews Immunology 8(9), 726736. https://doi.org/10.1038/nri2395.CrossRefGoogle ScholarPubMed
Van Camp, JK, Beckers, S, Zegers, D and Van Hul, W (2014) Wnt signaling and the control of human stem cell fate. Stem Cell Reviews and Reports 10(2), 207229. https://doi.org/10.1007/s12015-013-9486-8.CrossRefGoogle ScholarPubMed
Van Den Dries, MA, Guxens, M, Spaan, S, Ferguson, KK, Philips, E, Santos, S, Jaddoe, VWV, Ghassabian, A, Trasande, L, Tiemeier, H and Pronk, A (2020) Phthalate and bisphenol exposure during pregnancy and offspring nonverbal IQ. Environmental Health Perspectives 128(7), 077009. https://doi.org/10.1289/ehp6047.CrossRefGoogle ScholarPubMed
Vethaak, AD and Legler, J (2021) Microplastics and human health. Science 371(6530), 672674. https://doi.org/10.1126/science.abe5041.CrossRefGoogle ScholarPubMed
Vilela, J, Hartmann, A, Silva, EF, Cardoso, T, Corcini, CD, Varela-Junior, AS, Martinez, PE and Colares, EP (2014) Sperm impairments in adult vesper mice (Calomys laucha) caused by in utero exposure to bisphenol A. Andrologia 46(9), 971978. https://doi.org/10.1111/and.12182.CrossRefGoogle ScholarPubMed
Warburton, D, Bellusci, S, De Langhe, S, Del Moral, P-M, Fleury, V, Mailleux, A, Tefft, D, Unbekandt, M, Wang, K and Shi, W (2005) Molecular mechanisms of early lung specification and branching morphogenesis. Pediatric Research 57(7), 2637. https://doi.org/10.1203/01.PDR.0000159570.01327.ED.CrossRefGoogle ScholarPubMed
Wardman, T, Koelmans, AA, Whyte, J and Pahl, S (2021) Communicating the absence of evidence for microplastics risk: Balancing sensation and reflection. Environment International 150, 106116. https://doi.org/10.1016/j.envint.2020.106116.CrossRefGoogle ScholarPubMed
Warner, GR, Dettogni, RS, Bagchi, IC, Flaws, JA and Graceli, JB (2021) Placental outcomes of phthalate exposure. Reproductive Toxicology 103, 117. https://doi.org/10.1016/j.reprotox.2021.05.001.CrossRefGoogle ScholarPubMed
Weis, JS and Palmquist, KH (2021) Reality check: Experimental studies on microplastics lack realism. Applied Sciences 11, 8529. https://doi.org/10.3390/app11188529.CrossRefGoogle Scholar
Wernroth, M-L, Peura, S, Hedman, AM, Hetty, S, Vicenzi, S, Kennedy, B, Fall, K, Svennblad, B, Andolf, E, Pershagen, G, Theorell-Haglöw, J, Nguyen, D, Sayols-Baixeras, S, Dekkers, KF, Bertilsson, S, Almqvist, C, Dicksved, J and Fall, T (2022) Development of gut microbiota during the first 2 years of life. Scientific Reports 12(1), 9080. https://doi.org/10.1038/s41598-022-13009-3.CrossRefGoogle ScholarPubMed
Wijesekara, H, Bolan, NS, Bradney, L, Obadamudalige, N, Seshadri, B, Kunhikrishnan, A, Dharmarajan, R, Ok, YS, Rinklebe, J, Kirkham, MB and Vithanage, M (2018) Trace element dynamics of biosolids-derived microbeads. Chemosphere 199, 331339. https://doi.org/10.1016/j.chemosphere.2018.01.166.CrossRefGoogle ScholarPubMed
Winkler, AS, Cherubini, A, Rusconi, F, Santo, N, Madaschi, L, Pistoni, C, Moschetti, G, Sarnicola, ML, Crosti, M, Rosso, L, Tremolada, P, Lazzari, L and Bacchetta, R (2022) Human airway organoids and microplastic fibers: A new exposure model for emerging contaminants. Environment International 163, 107200. https://doi.org/10.1016/j.envint.2022.107200.CrossRefGoogle ScholarPubMed
Wu, P, Cai, Z, Jin, H and Tang, Y (2019) Adsorption mechanisms of five bisphenol analogues on PVC microplastics. Science of the Total Environment 650, 671678. https://doi.org/10.1016/j.scitotenv.2018.09.049.CrossRefGoogle ScholarPubMed
Xie, S, Zhang, R, Li, Z, Liu, C, Chen, Y and Yu, Q (2023) Microplastics perturb colonic epithelial homeostasis associated with intestinal overproliferation, exacerbating the severity of colitis. Environmental Research 217, 114861. https://doi.org/10.1016/j.envres.2022.114861.CrossRefGoogle ScholarPubMed
Yang, D, Zhu, J, Zhou, X, Pan, D, Nan, S, Yin, R, Lei, Q, Ma, N, Zhu, H, Chen, J, Han, L, Ding, M and Ding, Y (2022) Polystyrene micro- and nano-particle coexposure injures fetal thalamus by inducing ROS-mediated cell apoptosis. Environment International 166, 107362. https://doi.org/10.1016/j.envint.2022.107362.CrossRefGoogle ScholarPubMed
Yin, H, Price, F and Rudnicki, MA (2013) Satellite cells and the muscle stem cell niche. Physiological Reviews 93(1), 2367. https://doi.org/10.1152/physrev.00043.2011.CrossRefGoogle ScholarPubMed
Zhang, X, Huang, CT, Chen, J, Pankratz, MT, Xi, J, Li, J, Yang, Y, Lavaute, TM, Li, X-J, Ayala, M, Bondarenko, GI, Du, Z-W, Jin, Y, Golos, TG and Zhang, S-C (2010) Pax6 is a human neuroectoderm cell fate determinant. Cell Stem Cell 7(1), 90100. https://doi.org/10.1016/j.stem.2010.04.017.CrossRefGoogle ScholarPubMed
Zhang, Y, Jiang, M, Kim, E, Lin, S, Liu, K, Lan, X and Que, J (2017) Development and stem cells of the esophagus. Seminars in Cell & Developmental Biology 66, 2535. https://doi.org/10.1016/j.semcdb.2016.12.008.CrossRefGoogle ScholarPubMed
Zhang, J, Wang, L and Kannan, K (2020) Microplastics in house dust from 12 countries and associated human exposure. Environment International 134, 105314. https://doi.org/10.1016/j.envint.2019.105314.CrossRefGoogle ScholarPubMed
Zhang, J, Wang, L, Trasande, L and Kannan, K (2021) Occurrence of polyethylene terephthalate and polycarbonate microplastics in infant and adult feces. Environmental Science & Technology Letters 8(11), 989994. https://doi.org/10.1021/acs.estlett.1c00559.CrossRefGoogle Scholar
Zhang, Y, Wang, X, Zhao, Y, Zhao, J, Yu, T, Yao, Y, Zhao, R, Yu, R, Liu, J and Su, J (2023) Reproductive toxicity of microplastics in female mice and their offspring from induction of oxidative stress. Environmental Pollution 327, 121482. https://doi.org/10.1016/j.envpol.2023.121482.CrossRefGoogle ScholarPubMed
Zhang, K, Zhang, Y, Gu, L, Lan, M, Liu, C, Wang, M, Su, Y, Ge, M, Wang, T, Yu, Y, Liu, C, Li, L, Li, Q, Zhao, Y, Yu, Z, Wang, F, Li, N and Meng, Q (2018) Islr regulates canonical Wnt signaling-mediated skeletal muscle regeneration by stabilizing Dishevelled-2 and preventing autophagy. Nature Communications 9(1), 5129. https://doi.org/10.1038/s41467-018-07638-4.CrossRefGoogle ScholarPubMed
Zhao, Q, Zhu, L, Weng, J, Jin, Z, Cao, Y, Jiang, H and Zhang, Z (2023) Detection and characterization of microplastics in the human testis and semen. Science of the Total Environment 877, 162713. https://doi.org/10.1016/j.scitotenv.2023.162713.CrossRefGoogle ScholarPubMed
Zhou, R, Chen, F, Feng, X, Zhou, L, Li, Y and Chen, L (2015) Perinatal exposure to low-dose of bisphenol A causes anxiety-like alteration in adrenal axis regulation and behaviors of rat offspring: A potential role for metabotropic glutamate 2/3 receptors. Journal of Psychiatric Research 64, 121129. https://doi.org/10.1016/j.jpsychires.2015.02.018.CrossRefGoogle ScholarPubMed
Zhou, Y, Wang, J, Zou, M, Jia, Z, Zhou, S and Li, Y (2020) Microplastics in soils: A review of methods, occurrence, fate, transport, ecological and environmental risks. Science of the Total Environment 748, 141368. https://doi.org/10.1016/j.scitotenv.2020.141368.CrossRefGoogle ScholarPubMed
Zhu, L, Zhu, J, Zuo, R, Xu, Q, Qian, Y and An, L (2023) Identification of microplastics in human placenta using laser direct infrared spectroscopy. Science of the Total Environment 856, 159060. https://doi.org/10.1016/j.scitotenv.2022.159060.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Microplastics exposure routes. An overview of different microplastic exposure routes. Primary sources include clothes and cosmetics, whereas secondary sources include larger pieces of plastic. Microbeads from cosmetics, microfibers from clothes and smaller plastic particles derived from plastic degradation can enter humans directly via food and/or drinks or via the natural environment. When pregnant women are exposed, a developing fetus can be exposed too. Image created with BioRender.com.

Figure 1

Figure 2. Early human development. An overview of human development at different time points. First, a sperm cell fuses with an egg cell during fertilization to form a zygote and this time point is referred to as gestational day 0. The zygote develops further into a blastocyte, consisting of an inner cell mass (purple cells) and trophoblasts (pink cells) on day 5. The inner cell mass further differentiates into ectoderm (blue cells), mesoderm (red cells) and endoderm (yellow cells) on day 15 and is called a gastrula. The embryo will then further develop and is called a fetus after week 8. Image created with BioRender.com.

Figure 2

Figure 3. Effects of microplastics on various organs and tissues. Overview of effects of microplastics exposure on various organs and tissues of a developing fetus. Microplastics have detrimental effects on development of the placenta, central nervous system, liver, intestines, lungs, reproductive system and stem cells. Image created with BioRender.com.

Author comment: Microplastics: A threat for developing and repairing organs? — R0/PR1

Comments

No accompanying comment.

Review: Microplastics: A threat for developing and repairing organs? — R0/PR2

Conflict of interest statement

Reviewer declares none.

Comments

Dear authors

I enjoyed reading your MS describing the effects of NPs and MPs on human development and repair. I have comments, that could be useful to improve the MS before it is published here or elsewhere.

General comment

My major concern is that this review only listed articles indicating that MPs and NPs are deleterious to humans, although this topic is still controversial and there are papers showing lack of adverse effects or the effects were detected in extreme concentrations that are not realistic. This kind of papers should be included in this review, to provide a critical analysis of the theme. Some of them were listed in other reviews, for example those presented below:

https://doi.org/10.1016/j.tifs.2018.12.009

https://doi.org/10.3390/ijerph17041212

https://doi.org/10.3390/app11188529

https://doi.org/10.1016/j.envint.2020.105807

https://doi.org/10.1016/j.watres.2019.02.054

https://doi.org/10.1016/j.envint.2020.106116

https://doi.org/10.1016/j.envint.2020.106141

Specific comments

Some references citations differ from the journal’s guidelines, please check them.

Line 41: Perhaps “Environmental disruptors” may be more aproppriate here

Line 143: remove the dot after conditions.

Lines 257-258: I suggest you to include that MPs and NPs can be toxic by direct or indirect ways (as some effects are due to chemicals leachated from plastics).

Review: Microplastics: A threat for developing and repairing organs? — R0/PR3

Conflict of interest statement

Reviewer declares none.

Comments

The review on “Microplastics: a threat for developing and repairing organs?” is interesting, but not very informative at this stage. The authors can improve by doing a more critical analysis of the data published in the literature.

Since Bisphenol A and phthalates are considered as polymer additives (or leachates), please add a section after section 2 to define their function and list some of the common additives used in plastics.

Throughout the review, there is a mix up of toxicity due to nanoplastic particles and common additives in polymers such as phthalates and bisphenol-A. Can the authors clearly separate these two major factors in the review?

In addition, results from in vivo studies using animal models must be separated from in vitro studies using cell lines and the authors should compare them.

In almost all cases examined, the authors just stated simple conclusions from the published literature. It would be great if the authors can do more critical evaluations. For example, there are many factors, which influences the interaction of nanoparticles with the organs and none of them are really touched up on in this review. The size, shape, concentration and time of exposure etc. are critical towards including different toxicity response in organs. Yet, the authors did not include any of these parameters. Another important aspect is how the particles induce the observed developmental issues in different organs and regeneration of tissues. Mechanistic in site will make the review much more useful to the audience.

Overall, the review is very shallow and only touched on the surface of the topics covered here. The authors are encouraged to expand the document based on my above comments.

Recommendation: Microplastics: A threat for developing and repairing organs? — R0/PR4

Comments

Dear Authors,

We have now received the required number of reviews for your manuscript for developing and repairing organs?'. I am giving a recommendation of Major Revision based on that being the recommendation from one fo the reviewers. However, after reading the review I also see that they are asking for quite extensive revisions and additions to the content of the manuscript, when these reviews are expected to be shorter (4000-5000 words) than conventional review articles. Therefore, I acknowedlge that it can be a challenge to go into the depth that they reviewer is asking for here.

That said, I think they do have some good points that could at least be highlighted in places throughout the manuscript, if not covered in detail.

Kind regards

Andy Booth

Decision: Microplastics: A threat for developing and repairing organs? — R0/PR5

Comments

No accompanying comment.

Author comment: Microplastics: A threat for developing and repairing organs? — R1/PR6

Comments

No accompanying comment.

Review: Microplastics: A threat for developing and repairing organs? — R1/PR7

Conflict of interest statement

Reviewer declares none.

Comments

Dear authors

I am satisfied with the revisions made and think the MS can be published now. Well done!

Review: Microplastics: A threat for developing and repairing organs? — R1/PR8

Conflict of interest statement

I have no conflict or competing interest with the current review.

Comments

The review on “Microplastics: a threat for developing and repairing organs?” is interesting and well written. Such compilation of data is helpful for both experts working in the area and those young researchers entering this highly relevant and fast moving area of research. The review talks about the impact of MNPs and additives on different organ development, but little on the features of MNPs or exposure quantities or exposure time or mechanism of action. All of these are more important towards learning the overall impact on organ developments or human health. Often multiple factors acts in a synergistic way to cause health issues. Thus, current review is qualitative in nature and can be improved significantly. Pease see the following comments for improving the quality.

1. Separate the discussion on MNPs from additives, because the mechanism of action for these items is different? Is the toxicity observed from MNPs occurring due to the presence of additives or is it due to the nature of polymers? Not very clear.

2. Why only PS and PE MNPs? How about other polymers? There are many papers on other polymers too.

3. Include a section on key variables of MNPs (e.g. size, shape, concentration, chemical nature, exposure time, animal or cellular models tested etc.), which show impact on human health

4. Add more details on exposure conditions or how the impact was determined under each conditions/organs need to be included.

5. Impacts of bisphenol A and phthalates on organ development or overall human health is widely studied. Here also, many factors influence the outcome. The authors should provide some quantitative information and not just qualitative.

Overall, the review is too preliminary and qualitative in character. Improve it by adding critical data mentioned above.

Recommendation: Microplastics: A threat for developing and repairing organs? — R1/PR9

Comments

Apologies for the slow response to your revised manuscript submission. One of the original reviewers came back with a long list of revisions to the revised version, most of which were issues that they had not raised in their first evaluation. It seemed like they had not realised this was a revised version fo a manuscript that they had reviewed alread. After repeated attempts to contact the review to to try and clarify the situation, I am yet to receive a response. As the other reviewer of the manuscript was very happy with your revisions and recommend an accept decision, I have now decided to proceed with an accept decision.

Once again, thank you for your pateience while I tried to sort out this situation and congratulations on your manuscript.

Kind regards

Andy Booth

Decision: Microplastics: A threat for developing and repairing organs? — R1/PR10

Comments

No accompanying comment.