Next Article in Journal
Universal Prime Editing Therapeutic Strategy for RyR1-Related Myopathies: A Protective Mutation Rescues Leaky RyR1 Channel
Previous Article in Journal
Impaired SERPIN–Protease Balance in the Peripheral Lungs of Stable COPD Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Dysfunction-Associated Steatotic Liver Disease Induced by Microplastics: An Endpoint in the Liver–Eye Axis

1
Department of Anatomy, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
2
Department of Pathology and Cytology, General Hospital Pula, 52100 Pula, Croatia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 2837; https://doi.org/10.3390/ijms26072837
Submission received: 30 January 2025 / Revised: 11 March 2025 / Accepted: 20 March 2025 / Published: 21 March 2025
(This article belongs to the Special Issue Nonalcoholic Liver Disease: Mechanisms, Prevention, and Treatment)

Abstract

There is a significant, rather than just anecdotal, connection between the liver and the eyes. This connection is evident in noticeable cases such as jaundice, where the sclera has a yellow tint. But this can be seen through even more subtle indicators, such as molecules known as hepatokines. This relationship is not merely anecdotal; in some studies, it is referred to as the “liver–eye axis”. Ubiquitous environmental contaminants, such as microplastics (MPs), can enter the bloodstream and human body through the conjunctival sac, nasolacrimal duct, and upper respiratory tract mucosa. Once absorbed, these substances can accumulate in various organs and cause harm. Toxic substances from the surface of the eye can lead to local oxidative damage by inducing apoptosis in corneal and conjunctival cells, and irregularly shaped microparticles can exacerbate this effect. Even other toxicants from the ocular surface may be absorbed into the bloodstream and distributed throughout the body. Environmental toxicology presents a challenge because many pollutants can enter the body through the same ocular route as that used by certain medications. Previous research has indicated that the accumulation of MPs may play a major role in the development of chronic liver disease in humans. It is crucial to investigate whether the buildup of MPs in the liver is a potential cause of fibrosis, or simply a consequence of conditions such as cirrhosis and portal hypertension.

1. Microplastics and the Liver

Writing a review based on several articles related to the liver–eye axis and its endpoints was quite daunting and was made even more challenging when the topic of microplastics was included. To tackle this, we conducted a systematic review of the literature using three pairs of keywords in “ANY FIELD”: (1) “liver–eye axis” AND “microplastics”, (2) “steatohepatitis” AND “microplastics”, and (3) “(MAFLD” OR “MASLD”) AND “microplastics”.
To ensure comprehensiveness, we searched three databases via the Internet (PubMed, Web of Science, and Scopus), without any time constraints, from their inception to 6 March 2025. Most of the content accessed was peer-reviewed, i.e., evaluated by another expert in that subject area, so the information should have been of high quality. To prevent the accessing of misleading information that healthcare providers, policymakers, and other decision-makers may face due to an overwhelming volume of research, a systematic review of the scholarly literature was conducted.
Our initial screening identified 421 articles, with 63 removed as duplicates or non-English texts, which narrowed our search down to 358 entries. The study identification process was conducted on three levels: (1) inputs or the identification of studies, (2) processing or screening, and (3) outputs or included studies. This process is showcased in the flowchart in Figure 1.

1.1. Miceoplastics and Nanoplastics

Human development has led to numerous achievements, yet these advancements have often had a significant negative impact: environmental pollution. Critical trajectories of this process, such as food, air, water, and soil, have consistently affected human health. Additionally, the growth of human development has been closely linked to the creation of plastic waste, at least over the past few decades. The production of plastic waste more than doubled between 2000 and 2019, and this trend is continuing, particularly due to the substantial accumulation of plastics in rivers [1,2]. A large portion of this waste originates from plastics with a lifespan of less than five years, with 40% coming specifically from packaging [1].
Regardless of the duration, when plastic waste breaks down, it forms tiny particles known as microplastics (MPs) and nanoplastics (NPs), which are being increasingly found in human and animal tissues. Microplastics and NPs increase cytokine expression and induce enzymatic activity related to inflammation. Exposure to MPs can recruit neutrophils, macrophages, and natural killer cells to the liver [2].
In addition to being formed by the grinding of larger pieces of plastic waste, secondary MPs can also be produced in the form of primary MPs such as resin pellets used in plastic manufacturing. Ordinary consumer products, such as bisphenol A, which was once popular for use in toddlers’ bottles until a consumer boycott occurred [3], or chemicals like phthalates, are the primary sources of microplastic pollution in the environment [4,5]. In some literature, about 80% of the MPs detected were fibrous in shape and were made of polyethylene (25%), polyester (20%), and polyamide (10%) [6]. Microplastics (and NPs) originate from synthetic textiles, urban dust, tires, road markings, marine coatings, personal care products, and engineered plastic pellets [7,8]. Fine particles found in polluted air, such as micro- and nanoplastics, can cause systemic toxicity by penetrating cell membranes [9,10]. There is copious evidence that particle pollutants have caused a number of diseases or influenced existing ones (Table 1). Currently, plastic particles and fibers smaller than 1 μm are classified as nanoplastics, while those ranging from 1 μm to 1 mm are classified as MPs. Fragments that measure between 1 and 5 mm can be referred to as large MPs [11]. Based on all the reviewed literature, MP and NP particles are extremely complex and diverse in terms of their size, shape, density, polymer type, surface properties, and more.
Plastic particle pollutants are reported as risk factors for cardiovascular diseases and stroke. They are implicated in the etiopathogenesis of developmental disruptions, autism, and attention deficit hyperactivity disorder (ADHD) [12,13,14,15]. Plastics, particularly pollutants from the environment, precipitate a series of mental disorders. Additionally, plastic debris from environmental pollution is associated with cognitive decline in Alzheimer’s disease [16].

1.2. Liver Damage

Microplastic and NP particles activate the innate immune system, causing chronic, low-grade inflammation, which also enhances atherogenesis [17]. The NF-κB pathway is then activated [18,19], which enhances the inflammatory response in the liver [20]. Li et al., utilizing qRT-PCR, Western blotting, and immunohistochemical staining, confirmed that NPs/MPs induce liver fibrosis through alpha-smooth muscle actin (α-SMA) [21]. The protein expression level of α-SMA (not of mRNA) significantly increased as the size of the particles increased. It is reasonable to conclude that prolonged exposure to polystyrene microparticles and nanoparticles with larger sizes can significantly induce liver fibrosis.
Many studies have shown that the physicochemical properties of MPs can trigger inflammatory responses and provoke immune reactions when they enter the body and accumulate [22,23]. Once MP and NP particles enter the body, the host’s cells attempt to digest them using their lysosomes, and this ability is not limited to specialized phagocytes [24]. The host’s cells are usually unsuccessful, so plastic particles accumulate in their cytoplasm. Accordingly, after entering the liver tissue, these foreign bodies causes chronic, low-grade inflammation. The nuclear factor–kappa B (NF-κB) pathway is activated, further amplifying the inflammatory response in the liver [20]. When combined with signs of hepatocellular injury, such as the ballooning of hepatocytes, the condition is referred to as metabolic dysfunction-associated steatohepatitis (MASH) [25,26,27].
Regardless of whether this specification is characterized by fibrosis, it is referred to as metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD is often asymptomatic and is usually diagnosed incidentally through abnormal liver enzyme findings or imaging studies that reveal hepatic steatosis. This is the initial stage of steatotic liver disease, which is characterized by more than 5 percent hepatic steatosis and at least one risk factor for cardiometabolic dysfunction, such as dyslipidemia or obesity [28,29,30]. Patients with MASLD should have minimal or no alcohol consumption and no other causes of steatotic liver disease [31]. As MASLD progresses, fat accumulation also continues and results in inflammation that damages the tissue (inflammation progresses from acute to chronic hepatitis), and scarring occurs (fibrosis) [32]. Fibrosis is a gradual process that advances through stages. In the end, it leads to cirrhosis: extensive scarring that disrupts liver architecture and function, significantly raising the risk of hepatocellular carcinoma (HCC).
Notably, the most widely supported theory on the pathogenesis of MASLD implicates insulin resistance coupled with systemic inflammation [33]. The excess accumulation of non-esterified fatty acids and diacylglycerol causes cellular distress and dysfunction, which leads to the overactivation of lipid signaling. This promotes hepatic insulin resistance and endoplasmic reticulum stress, resulting in chronic inflammation, liver fibrosis, cirrhosis, and, ultimately, hepatocarcinogenesis [34].

Chronic Inflammation, Oxidative Stress, and Cellular Senescence

Chronic inflammation, characterized by pro-inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6), combined with oxidative stress, leads to liver damage [35,36]. The presence of polystyrene MPs significantly increases levels of TNF (formerly known as TNF-α), IL-6, and monocyte chemoattractant protein-1 (MCP-1), triggering oxidative stress and inflammation [37,38]. This suggests that a similar mechanism may occur in the liver [39,40]. Increased levels of IL-6 and TNF lead to inflammation and vascular permeability. Additionally, C-reactive protein (CRP), an acute-phase protein produced by the liver, has been linked to the development of both cardiovascular and ocular diseases, thereby connecting systemic inflammation to hepatic damage [36,41].
The pro-inflammatory nature of MPs is likely linked to cellular senescence [42], which is characterized by a permanent state of cell cycle arrest accompanied by the secretion of pro-inflammatory factors and other substances [43]. Various cell types exhibit increased senescence rates after exposure to different types of MPs and nanoplastics [44]. Similarly to oxidative stress and inflammasome activation, cellular senescence is increasingly recognized as a therapeutic target for drug development. Strategies to address this issue can promote the removal of senescent cells by either preventing their formation or inhibiting their harmful pro-inflammatory activity. For instance, Dhakal et al. [45] reported that a class of glucose-lowering medications can reduce senescence induced by MP and NP particles [45]. Notably, this same hypoglycemic medication has also demonstrated cardioprotective effects in various contexts. Another critical factor in cellular senescence is the overexpression of miR-217. This molecule has been shown to regulate critical processes, such as the cell cycle, differentiation, proliferation, and senescence [46]. miR-217 also plays a role in maintaining the structure and function of the extracellular matrix and might be relevant to the integrity of the liver [47]. Additionally, numerous studies have identified other effects associated with MP and NP particles, such as the induction of platelet aggregation, hemolysis, and the activation of immune cells [48].

2. Laboratory Assessment

2.1. Histology and Histologic Scoring Systems for Chronic Liver Disease

Liver biopsy remains the gold standard for confirming the diagnosis of MASH and assessing disease severity, and for accurately staging fibrosis [49]. Hepatic steatosis in MASLD—MASH—is characterized by hepatocyte ballooning degeneration and hepatic inflammation [50,51,52]. These histological features may be difficult to distinguish from those seen in alcohol-associated steatohepatitis [53]. Histologically, alcohol-associated steatohepatitis (ASH) is quite similar to MASH, making it challenging to distinguish between the two based solely on a biopsy. In MASLD, (the disease-specific histological criterion), hepatic steatosis features visible hepatocyte ballooning degeneration and hepatic inflammation [52].
Currently, non-invasive methods (non-invasive tests, NITs) for evaluating MASH are being studied. Circulating biomarkers are used in the NITs, and composite scores have been developed to combine the results of various NITs with other clinical parameters to predict the progression from steatohepatitis to fibrosis. However, their sensitivity or accuracy have not yet been proven, at least not enough to replace biopsy [54,55,56]. Only a liver biopsy can confirm a diagnosis of MASH, distinguish it from alcohol-associated steatohepatitis, assess disease severity, and accurately stage fibrosis [49].
In the arm of invasive tests and histologic sampling, non-alcohol-associated fatty liver activity score (NAS) is designed to assess the full range of changes associated with non-alcoholic fatty liver disease (NAFLD) and can be utilized to evaluate the effects of therapy. The total NAS can range from 0 to 8, with fibrosis not included in the score [57,58]. However, it is important to note that the threshold values for NAS do not always align with a diagnosis of MASH and should not be relied upon solely for diagnostic purposes [26]. The semiquantitative histological scoring system, also known as the Alcoholic Hepatitis Histologic Score (AHHS), is associated with the prognosis of patients with alcoholic hepatitis. This system utilizes four histological features: the degree of fibrosis, the degree of neutrophil infiltration, the type of bilirubin stasis, and the presence of megamitochondria. Additional histologic scoring systems for chronic liver disease include fatty liver inhibition and progression, and alcohol-associated liver disease and alcoholic hepatitis.

2.2. Assessing Microplastics

Tissue samples obtained by biopsy should be collected in glass tubes and frozen in liquid nitrogen, fixed in 10% buffered formalin, or fixed in 2.5% glutaraldehyde suitable for electronic microscopy for subsequent analyses.

2.2.1. Macroscopy and Microscopy: Visual Assessment of Microplastics

Microplastics consist of various plastic particles with diverse compositions and sizes. [59]. The visual analysis method involves observing the samples, allowing the MPs to be roughly described and classified according to their color, shape, and size. Interest in quantifying MPs in environmental samples has grown, especially after stool and blood samples have documented exposure to MPs [60,61]. Proper analysis of the quality of MPs is most often performed via analysis using the naked eye or a microscope [62]. Although it cannot provide information on the chemical component of MPs [63,64], this analysis method is simple, inexpensive, and presents a negligible chemical hazard [65].
Data collection related to micro- and nanoplastics can be accomplished using various analytical methods. These include assessing particle number and size, determining polymer and additive identities, measuring mass fraction, and evaluating degradation status. However, no single method provides a complete picture, so a combination of different techniques with their respective limitations and advantages is necessary [64]. This is more challenging for nanoparticles because, without complete chemical information, it is unreliable to determine whether the quantified nanoparticles are nanoplastics at all [66,67]. Studies included in this review that assessed MPs visually identified them as having jagged edges and irregular shapes [68,69,70,71,72,73].

2.2.2. Identifying Microplastic and Nanoplastic Particles

Pyrolysis–gas chromatography–mass spectrometry (Py–GCMS) is a widely favored method for identifying microplastic and nanoplastic particles, primarily due to its ability to detect non-volatile components in solutions, which may be overlooked in conventional GC-MS analyses. Additionally, Py–GC/MS can analyze a broader variety of sample types. This technique is also capable of detecting very small quantities of contaminants, down to microliters or micrograms within liters of sample material [74].
However, this method does not discriminate between microplastics and nanoplastics [75]. Furthermore, Py–GCMS cannot detect certain inorganic substances, and the results may vary slightly with non-homogeneous samples. Additionally, it cannot provide information on the number of particles across different size ranges. For information on particle sizes, Raman micro-spectroscopy or Fourier Transform Infrared (FTIR) spectroscopy are recommended. This variability may sometimes require repeated tests to obtain reliable results [75].
Our review found that studies predominately utilized Py–GCMS, with 83.3% (five out of six) of the included studies employing this method [69,70,71,72,73]. No studies utilized Raman spectroscopy or FTIR. Notably, Raman micro-spectroscopy is a technique that combines a Raman spectrometer, which analyzes the vibrational modes of molecules, with a confocal microscope. This method allows for the chemical analysis of samples with sub-micron spatial resolution, making it particularly effective in identifying MPs. In addition to sample identification, Raman micro-spectroscopy is also employed for quality control, failure analysis, material characterization, and examining physical and chemical properties [76]. FTIR analysis, or FTIR spectroscopy, is an effective method of assessing microplastics by identifying organic, polymeric, and, in some cases, inorganic materials. This method utilizes infrared light to scan test samples and observe their chemical properties. However, it was rarely discussed in the studies included in this review. Therefore, it remains speculative as to whether factors such as costs, staff limitations, the design of the sampling chamber, or the mounting of samples have contributed to the underutilization of FTIR spectroscopy [59,77].
One of the included studies used laser direct infrared imaging (LDIR) [71], which is an innovative infrared microscopy technique that employs a tunable Quantum Cascade Laser (QCL) as its infrared source. This new reflectance-based approach overcomes the coherence artifacts commonly associated with QCLs. It enables the acquisition of large-area, high-definition infrared images and high signal-to-noise point spectra. By extending this technique with attenuated total reflectance (ATR), it is possible to obtain high-fidelity spectra from features smaller than 10 μm.
In summary, Py–GC/MS can be utilized to analyze tissue samples specimens obtained from carotid or coronary arteries and samples from aortas.

2.3. Microplastic-Induced Histopathological Lesions in the Liver

Microplastics can alter the histological structure of tissues, including the liver. Feng et al. [78] conducted histological examinations of the livers of fish subjected to a diet containing varying concentrations of MP particles. They discovered that the fishes’ body weight was significantly reduced, and the antioxidant status of the liver was disrupted. However, in the group receiving the highest concentration of MP particles, only slight disorganization of the liver architecture was observed. The lack of morphological changes in the livers of the fish after MP exposure, which were significantly more pronounced in the MP-fed group than in the control group, indicates that factors other than the direct accumulation of MPs caused liver injury in the fish. More encouraging were the findings of Tian et al. [79], who observed several cellular changes in the H&E staining of liver tissue in zebrafish. They reported a loss of the cells’ polygonal shape, indistinct cell borders, vacuolar degeneration, and the infiltration of inflammatory cells. These lesions were attributed to the cumulative toxicity of MPs in the bloodstream and their subsequent impact on other cellular structures. At last, Zhuang and colleagues [80] identified liver inflammation, cell vacuolation, and karyopyknosis in the adult mice.
Li et al. observed liver injury in mice following exposure to nanoparticles and MPs [21]. They concluded that particle size significantly affected the impact of NPs and MPs on the liver. Notably, long-term exposure significantly decreased both liver weight and the liver-to-body weight ratio as the particle size of NPs and MPs increased. The observations of Li et al. also included hepatocellular vacuolar degeneration and edema, irregularly arranged hepatic cords, cell dikaryon, and inflammation. Similarly, Chen and colleagues found that non-uniform and irregular spherical particles lead to the accumulation of microplastics in duck livers [81].
The effects of microplastic exposure on the human liver were studied using a functionally active 3D liver microtissue model [82]. This model comprised primary human hepatocytes, Kupffer cells, sinusoidal endothelial cells, and hepatic stellate cells. Hematoxylin and eosin (H&E) staining revealed that the typical characteristics of the liver were preserved, with no necrotic core observed over the 21-day exposure period to MPs in the cell culture. This finding indicates adequate access to nutrients and oxygen in the inner zones of the microtissue, highlighting the suitability of this in vitro model for long-term toxicological studies.
Repeated low-dose exposure to smaller MPs led to distorted tissue architecture and the appearance of large extracellular voids, suggesting the presence of dilated canaliculi, a condition occasionally observed in the cholestatic liver. Additionally, intracellular white vacuoles were noted, resembling large lipid droplets, indicative of macrovesicular steatosis. Resident macrophages also accumulated non-biodegradable plastics, represented as uniformly sized intracellular white disks.
Horvatits et al. conducted a proof-of-concept case series that focused on detecting MP particles through the chemical digestion of tissue samples, staining with Nile red, and subsequent examination using fluorescent microscopy and Raman spectroscopy [83]. As a result, classical histopathological assessment was not performed. Their study primarily focused on the MPs, assessing their morphology, size, and the composition of the polymer types, while details regarding liver histoarchitecture were not provided. Nevertheless, the authors identified six different types of MP polymers in the livers of individuals with liver cirrhosis.

3. Toxic Exposure

Ocular toxicology focuses on the toxic effects of drugs administered through topical, intraocular, or systemic routes. It also includes the evaluation of adverse effects caused by ophthalmic devices, such as contact lenses, intraocular lenses, and glaucoma implants. With the emerging concept of the “liver–eye axis” and an increasing number of hepatokines influencing this relationship, the role of the liver in ocular toxicology is becoming increasingly significant [84,85].
Particulate matter from environmental pollution and other chemical substances (Figure 2), in the form of liquids, aerosols, or vapors (including medications), can breach the eye’s surface and its protective tear film [86]. In a local context, ocular surface damage can be assessed through a variety of ophthalmological symptoms that present in multiple ways. Toxic substances that affect the ocular surface can impair cellular motility and lead to cellular senescence [87,88]. An acute toxicity test used to evaluate the effects of chemicals and their potential to cause eye irritancy or damage is known as the Draize rabbit eye test. In this test, the effects of tested substances are observed using a slit-lamp biomicroscope on enucleated rabbit eyes. The test is not only controversial from the perspective of animal rights but also raises concerns about the subjective scoring of effects and the differences between rabbit and human eyes. For this reason, its scientific validity has been questioned [89]. To be precise, the test observes and records (using a subjective scoring system) effects on the conjunctiva, cornea, and iris. These effects can vary from mild and reversible irritation to severe and irreversible damage that may result in vision loss [90]. To address these shortcomings, a test based on a 3D corneal tissue model was developed and assessed in a series of validation studies. It is currently the subject of OECD test guideline No. 491 [91,92].
These particular pollutants from the air are associated with causes of systemic inflammatory response and cytokine production [93]. Millen et al. [94] performed a scoping review of the scholarly literature to determine whether ambient air pollution was a risk factor for chronic disease of the inner ocular structures and elevated intraocular pressure. They examined 27 identified articles where air pollutants were considered responsible for eye conditions. In their scoping review, Millen et al. [94] examined scholarly literature to assess whether ambient air pollution was a risk factor for chronic diseases affecting the inner ocular structures and elevated intraocular pressure. Their review identified 27 articles that linked air pollutants to various eye conditions. The systemic effects of airborne pollutants that enter the body through the ocular surface are well documented and do not require further discussion here. However, the limited literature on this topic indicates a significant need for additional research.
Environmental toxins lead to oxidative damage and inflammation. Air pollution can reduce the thickness of the tear-film lipid layer, as demonstrated in a study by Wang et al. [95] involving 200 healthy volunteers over a period of three years. This reduction can impair the motility of corneal and conjunctival cells, potentially leading to their senescence or even apoptosis. Consequently, this disruption affects important tissue barriers that regulate ocular drug absorption [96].
Fine particles, such as MPs and NPs, can lead to systemic toxicity by penetrating cell membranes. There is substantial evidence that these particle pollutants are responsible for a variety of diseases or exacerbate existing health conditions. Plastic particle pollutants have been identified as risk factors for cardiovascular diseases and strokes. Additionally, they are associated with developmental disorders, including autism and attention deficit hyperactivity disorder (ADHD) [12,13,14]. Plastic waste, including alternatives to traditional non-degradable plastic polymers, is linked to causes of systemic inflammatory responses and cytokine production [97,98]. Plastic and other environmental pollutants can lead to various mental disorders. Additionally, plastic debris from environmental pollution is linked to cognitive decline in Alzheimer’s disease [16].
Ongoing tests of environmental pollutants and their harmful effects utilize organoids and organoid-on-chip technologies, which leverage their advantages while addressing the limitations of traditional approaches. Although this research is still in its early stages, it demonstrates excellent potential for advancing environmental toxicology [99].
In summary, toxic substances from the eye’s surface can lead to local oxidative damage by inducing apoptosis in corneal and conjunctival cells [100,101]. Irregularly shaped microparticles can exacerbate this effect. Additionally, toxicants from the ocular surface may be absorbed and distributed through the bloodstream. Environmental toxicology presents a challenge because many pollutants can enter the body via the same ocular surface route as certain medications [102].

4. Liver–Eye Axis

There is no irrefutable evidence that MPs or NPs enter the bloodstream over the conjunctiva–environment interface, but this review offers a non-negligible number of reports of environmental pollutants damaging deeper eye structures after being in contact with that interface [97,102,103]. Also, there is a plethora of evidence of the ubiquity of MPs and NP particles [4,104], similar to the growing popularity of fluids from the OS as a biomonitoring tool or a prospect toxicological matrix [85,105]. The recent identification of MPs in the olfactory pathway suggests a potential major entry route for plastic into the brain; thus, the OS should not be dismissed lightly [106].
In that context, the liver–eye axis highlights the connection between these organs as a new area of research that examines the interconnected pathways through which the liver and eyes affect each other’s functions in both health and disease. In recent decades, the number of biomolecules secreted by the liver that regulate systemic metabolism, known as hepatokines, has steadily increased. The effects of these molecules on distant organs often include the eyes [107,108,109]. Hepatokines in the liver–eye axis include fibroblast growth factor 21 (FGF-21), hepatocyte growth factor (HGF), adropin, and angiopoietin-like proteins (ANGPTLs), which have emerged as key factors [110,111,112,113] (Table 2). As inflammation is the underlying cause of MASLD, inflammatory mediators such as CRP, IL-6, and TNF may hold significant importance in the liver–eye axis [33,114]. In the experiment conducted by Zhuang et al. [80], exposure to microplastics in adult mice resulted in increased levels of TNFα and IL-1β, as measured by qRT-PCR.
The connection between the liver and the eyes has recently gained more attention, although it primarily focuses on a one-way relationship. The concept of the “liver–eye axis” is not new nor merely a trend; it is deeply rooted in the theories and practices of Traditional Chinese Medicine (TCM). This perspective could provide a scientific basis for researchers interested in studying the liver–eye connection in a holistic way [135,136]. While this area deserves further investigation, it is also essential to consider the potential liver toxicity associated with ethnomedicine [113,137]. This concept should also be promoted in Western clinical practice. In this way, the latest advances in understanding the liver–eye axis could also be applied to toxicology. This connection has gained recognition even beyond TCM. It can be achieved through various pathways, including inflammation, immunity, metabolism, and oxidative stress [138,139].
The communication that relies on secretory factors and associated cytokines primarily involves hepatokines, molecules secreted exclusively or predominantly by the liver. These molecules are mainly delivered to the liver through paracrine activity or can reach distant organs via the human circulatory system. Additionally, some factors secreted by the eyes can also influence the liver’s condition, acting at a distance. For instance, the receptors of hepatocyte growth factor (HGF) have been detected in the cornea, lens, and retinal tissues [140]. On the other hand, retinal pigment epithelium-derived factor (RPEDF) excreted by the retinal pigment epithelium (RPE) may influence the liver [141]. There have been an increasing number of epidemiological studies indicating that eye disease is associated with protective or risk factors related to liver health. The prompt identification of these factors could improve healthcare outcomes. This review used this information as proof of concept.

5. Conclusions

In summary, toxicants from the eye surface that cause oxidative damage to the ocular surface may be absorbed into the bloodstream and distributed throughout the body. This should be noted when discussing the etiopathogenesis of MASLD and when administering MASLD medications. Consequently, all MASLD medications should be tested in clinical trials that examine the potential liver–eye connection, including any possible negative effects of these drugs on eye health. Improving our understanding of this interconnectedness may ultimately enhance patient outcomes by enabling more precise preventive and therapeutic measures to address the underlying metabolic causes affecting both liver and ocular health.
Future studies are needed to evaluate whether the hepatic accumulation of MP from the ocular surface is a potential cause of fibrosis or a consequence of cirrhosis and portal hypertension. In diagnosis and toxicological assessment, artificial intelligence and deep-learning-based diagnostic tools for detecting conditions related to the liver–eye axis could be a future direction for analytical toxicology.

Author Contributions

Conceptualization, I.Š. and M.P.; methodology, I.Š., M.P. and L.L.; software, L.L. and M.P.; formal analysis, I.Š.; investigation, M.P. and L.L.; resources, I.Š.; data curation, I.Š.; writing—original draft preparation, M.P. and I.Š.; writing—review and editing, M.P., L.L. and I.Š.; visualization, I.Š.; project administration, I.Š.; funding acquisition, I.Š., L.L. and M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The data used in this paper are available upon request.

Acknowledgments

I.Š. acknowledges the University of Rijeka, Faculty of Medicine, for their constant support. We appreciate all our patients, colleagues, and teachers.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. European Commission. Proposal for a Regulation of the European Parliament and of the Council. Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/HTML/?uri=CELEX:52022PC0677 (accessed on 3 March 2025).
  2. Chiang, C.C.; Yeh, H.; Shiu, R.F.; Chin, W.C.; Yen, T.H. Impact of microplastics and nanoplastics on liver health: Current understanding and future research directions. World J. Gastroenterol. 2024, 30, 1011–1017. [Google Scholar] [CrossRef] [PubMed]
  3. Jeon, G.W. Bisphenol A leaching from polycarbonate baby bottles into baby food causes potential health issues. Clin. Exp. Pediatr. 2022, 65, 450–452. [Google Scholar] [CrossRef] [PubMed]
  4. Rani, A. Types and Sources of Microplastics; The Ubiquitous Environment Contaminant: A Review. J. Polym. Mater. 2022, 39, 17–35. [Google Scholar] [CrossRef]
  5. Mirani, A.; Kianfar, E.; Maleknia, L.; Javanbakht, M. Recent advances in nicotine electrochemical biosensors: A review. Case Stud. Chem. Environ. Eng. 2024, 9, 100753. [Google Scholar] [CrossRef]
  6. Sarkar, S.; Diab, H.; Thompson, J. Microplastic Pollution: Chemical Characterization and Impact on Wildlife. Int. J. Environ. Res. Public Health 2023, 20, 1745. [Google Scholar] [CrossRef]
  7. Sharma, P.; Sharma, P. Micro(nano)plastics: Invisible compounds with a visible impact. F1000Research 2024, 13, 69. [Google Scholar] [CrossRef]
  8. Ashrafy, A.; Liza, A.A.; Islam, M.N.; Billah, M.M.; Arafat, S.T.; Rahman, M.M.; Rahman, S.M. Microplastics Pollution: A Brief Review of Its Source and Abundance in Different Aquatic Ecosystems. J. Hazard. Mater. Adv. 2023, 9, 100215. [Google Scholar] [CrossRef]
  9. Lai, H.; Liu, X.; Qu, M. Nanoplastics and Human Health: Hazard Identification and Biointerface. Nanomaterials 2022, 12, 1298. [Google Scholar] [CrossRef]
  10. Dehghanian, Z.; Asgari Lajayer, B.; Biglari Quchan Atigh, Z.; Nayeri, S.; Ahmadabadi, M.; Taghipour, L.; Senapathi, V.; Astatkie, T.; Price, G.W. Micro (nano) plastics uptake, toxicity and detoxification in plants: Challenges and prospects. Ecotoxicol. Environ. Saf. 2023, 268, 115676. [Google Scholar] [CrossRef]
  11. Ivleva, N.P. Chemical Analysis of Microplastics and Nanoplastics: Challenges, Advanced Methods, and Perspectives. Chem. Rev. 2021, 121, 11886–11936. [Google Scholar] [CrossRef]
  12. Moore, S.; Paalanen, L.; Melymuk, L.; Katsonouri, A.; Kolossa-Gehring, M.; Tolonen, H. The Association between ADHD and Environmental Chemicals-A Scoping Review. Int. J. Environ. Res. Public Health 2022, 19, 2849. [Google Scholar] [CrossRef] [PubMed]
  13. Botelho, R.M.; Silva, A.L.M.; Borbely, A.U. The Autism Spectrum Disorder and Its Possible Origins in Pregnancy. Int. J. Environ. Res. Public Health 2024, 21, 244. [Google Scholar] [CrossRef] [PubMed]
  14. Zaheer, J.; Kim, H.; Ko, I.O.; Jo, E.K.; Choi, E.J.; Lee, H.J.; Shim, I.; Woo, H.J.; Choi, J.; Kim, G.H.; et al. Pre/post-natal exposure to microplastic as a potential risk factor for autism spectrum disorder. Environ. Int. 2022, 161, 107121. [Google Scholar] [CrossRef] [PubMed]
  15. Thi, L.A.P.; Nguyen, V.-H.; Do, X.D.; Dang, T.H.L.; Do, H.T.; Nguyen, T.L. Microplastics occurrence, contamination, and effects on human health—A critical review. In Microplastics; Elsevier: Amsterdam, The Netherlands, 2025; pp. 451–479. [Google Scholar]
  16. Wang, G.; Lin, Y.; Shen, H. Exposure to Polystyrene Microplastics Promotes the Progression of Cognitive Impairment in Alzheimer’s Disease: Association with Induction of Microglial Pyroptosis. Mol. Neurobiol. 2024, 61, 900–907. [Google Scholar] [CrossRef]
  17. Liang, J.; Ji, F.; Abdullah, A.L.B.; Qin, W.; Zhu, T.; Tay, Y.J.; Li, Y.; Han, M. Micro/nano-plastics impacts in cardiovascular systems across species. Sci. Total Environ. 2024, 942, 173770. [Google Scholar] [CrossRef]
  18. Cheng, Y.; Yang, Y.; Bai, L.; Cui, J. Microplastics: An often-overlooked issue in the transition from chronic inflammation to cancer. J. Transl. Med. 2024, 22, 959. [Google Scholar] [CrossRef]
  19. Zhu, M.; Li, P.; Xu, T.; Zhang, G.; Xu, Z.; Wang, X.; Zhao, L.; Yang, H. Combined exposure to lead and microplastics increased risk of glucose metabolism in mice via the Nrf2/NF-kappaB pathway. Environ. Toxicol. 2024, 39, 2502–2511. [Google Scholar] [CrossRef]
  20. Xu, R.; Cao, J.W.; Lv, H.L.; Geng, Y.; Guo, M.Y. Polyethylene microplastics induced gut microbiota dysbiosis leading to liver injury via the TLR2/NF-kappaB/NLRP3 pathway in mice. Sci. Total Environ. 2024, 917, 170518. [Google Scholar] [CrossRef]
  21. Li, Q.; Zhu, K.; Huang, L.; Niu, X.; Li, L.; Gao, L.; Xia, Z. Polystyrene microplastics induce liver fibrosis and lipid deposition in mice through three hub genes revealed by the RNA-seq. Sci. Rep. 2025, 15, 2583. [Google Scholar] [CrossRef]
  22. Li, Y.; Chen, L.; Zhou, N.; Chen, Y.; Ling, Z.; Xiang, P. Microplastics in the human body: A comprehensive review of exposure, distribution, migration mechanisms, and toxicity. Sci. Total Environ. 2024, 946, 174215. [Google Scholar] [CrossRef]
  23. Lee, Y.; Sung, M.; Sung, S.E.; Choi, J.H.; Kang, K.K.; Park, J.W.; Kim, Y.J.; Lee, S. The histopathological and functional consequences of microplastic exposure. Discov. Appl. Sci. 2025, 7, 72. [Google Scholar] [CrossRef]
  24. Yee, M.S.; Hii, L.W.; Looi, C.K.; Lim, W.M.; Wong, S.F.; Kok, Y.Y.; Tan, B.K.; Wong, C.Y.; Leong, C.O. Impact of Microplastics and Nanoplastics on Human Health. Nanomater 2021, 11, 496. [Google Scholar] [CrossRef]
  25. Alpízar Salazar, M.; Olguín Reyes, S.E.; Medina Estévez, A.; Saturno Lobos, J.A.; De Aldecoa Castillo, J.M.; Carrera Aguas, J.C.; Alaniz Monreal, S.; Navarro Rodríguez, J.A.; Alpízar Sánchez, D.M. Natural History of Metabolic Dysfunction-Associated Steatotic Liver Disease: From Metabolic Syndrome to Hepatocellular Carcinoma. Medicina 2025, 61, 88. [Google Scholar] [CrossRef] [PubMed]
  26. Samy, A.M.; Kandeil, M.A.; Sabry, D.; Abdel-Ghany, A.A.; Mahmoud, M.O. From NAFLD to NASH: Understanding the spectrum of non-alcoholic liver diseases and their consequences. Heliyon 2024, 10, e30387. [Google Scholar] [CrossRef]
  27. Stoess, C.; Choi, Y.K.; Onyuru, J.; Friess, H.; Hoffman, H.M.; Hartmann, D.; Feldstein, A.E. Cell Death in Liver Disease and Liver Surgery. Biomedicines 2024, 12, 559. [Google Scholar] [CrossRef]
  28. Fan, J.G.; Xu, X.Y.; Yang, R.X.; Nan, Y.M.; Wei, L.; Jia, J.D.; Zhuang, H.; Shi, J.P.; Li, X.Y.; Sun, C.; et al. Guideline for the Prevention and Treatment of Metabolic Dysfunction-associated Fatty Liver Disease (Version 2024). J. Clin. Transl. Hepatol. 2024, 12, 955–974. [Google Scholar] [CrossRef]
  29. Yang, A.H.; Tincopa, M.A.; Tavaglione, F.; Ajmera, V.H.; Richards, L.M.; Amangurbanova, M.; Butcher, C.; Hernandez, C.; Madamba, E.; Singh, S.; et al. Prevalence of steatotic liver disease, advanced fibrosis and cirrhosis among community-dwelling overweight and obese individuals in the USA. Gut 2024, 73, 2045–2053. [Google Scholar] [CrossRef]
  30. Panganiban, J.; Kehar, M.; Ibrahim, S.H.; Hartmann, P.; Sood, S.; Hassan, S.; Ramirez, C.M.; Kohli, R.; Censani, M.; Mauney, E.; et al. Metabolic dysfunction-associated steatotic liver disease (MASLD) in children with obesity: An Obesity Medicine Association (OMA) and expert joint perspective 2025. Obes. Pillars 2025, 100164. [Google Scholar] [CrossRef]
  31. Leal-Lassalle, H.; Estévez-Vázquez, O.; Cubero, F.J.; Nevzorova, Y.A. Metabolic and alcohol-associated liver disease (MetALD): A representation of duality. NPJ Gut Liver 2025, 2, 1. [Google Scholar] [CrossRef]
  32. Mladenic, K.; Lenartic, M.; Marinovic, S.; Polic, B.; Wensveen, F.M. The “Domino effect” in MASLD: The inflammatory cascade of steatohepatitis. Eur. J. Immunol. 2024, 54, e2149641. [Google Scholar] [CrossRef]
  33. Sandireddy, R.; Sakthivel, S.; Gupta, P.; Behari, J.; Tripathi, M.; Singh, B.K. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front. Cell Dev. Biol. 2024, 12, 1433857. [Google Scholar] [CrossRef]
  34. Vesković, M.; Šutulović, N.; Hrnčić, D.; Stanojlović, O.; Macut, D.; Mladenović, D. The Interconnection between Hepatic Insulin Resistance and Metabolic Dysfunction-Associated Steatotic Liver Disease—The Transition from an Adipocentric to Liver-Centric Approach. Curr. Issues Mol. Biol. 2023, 45, 9084–9102. [Google Scholar] [CrossRef] [PubMed]
  35. Bhol, N.K.; Bhanjadeo, M.M.; Singh, A.K.; Dash, U.C.; Ojha, R.R.; Majhi, S.; Duttaroy, A.K.; Jena, A.B. The interplay between cytokines, inflammation, and antioxidants: Mechanistic insights and therapeutic potentials of various antioxidants and anti-cytokine compounds. Biomed. Pharmacother. 2024, 178, 117177. [Google Scholar] [CrossRef] [PubMed]
  36. Scarlata, G.G.M.; Colaci, C.; Scarcella, M.; Dallio, M.; Federico, A.; Boccuto, L.; Abenavoli, L. The Role of Cytokines in the Pathogenesis and Treatment of Alcoholic Liver Disease. Diseases 2024, 12, 69. [Google Scholar] [CrossRef]
  37. Bridgeman, L.; Cimbalo, A.; López-Rodríguez, D.; Pamies, D.; Frangiamone, M. Exploring toxicological pathways of microplastics and nanoplastics: Insights from animal and cellular models. J. Hazard. Mater. 2025, 490, 137795. [Google Scholar] [CrossRef]
  38. Feng, L.; Chen, C.; Xiong, X.; Wang, X.; Li, X.; Kuang, Q.; Wei, X.; Gao, L.; Niu, X.; Li, Q.; et al. PS-MPs promotes the progression of inflammation and fibrosis in diabetic nephropathy through NLRP3/Caspase-1 and TGF-β1/Smad2/3 signaling pathways. Ecotoxicol. Environ. Saf. 2024, 273, 116102. [Google Scholar] [CrossRef]
  39. Liu, Y.; Xu, K.; Xiang, Y.; Ma, B.; Li, H.; Li, Y.; Shi, Y.; Li, S.; Bai, Y. Role of MCP-1 as an inflammatory biomarker in nephropathy. Front. Immunol. 2023, 14, 1303076. [Google Scholar] [CrossRef]
  40. Takenoshita, Y.; Tokito, A.; Jougasaki, M. Inhibitory Effects of Eicosapentaenoic Acid on Vascular Endothelial Growth Factor-Induced Monocyte Chemoattractant Protein-1, Interleukin-6, and Interleukin-8 in Human Vascular Endothelial Cells. Int. J. Mol. Sci. 2024, 25, 2749. [Google Scholar] [CrossRef]
  41. Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef]
  42. de Magalhaes, J.P. Cellular senescence in normal physiology. Science 2024, 384, 1300–1301. [Google Scholar] [CrossRef]
  43. Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular mechanisms and diseases. Signal Transduct. Target. Ther. 2023, 8, 200. [Google Scholar] [CrossRef]
  44. Mahmud, F.; Sarker, D.B.; Jocelyn, J.A.; Sang, Q.A. Molecular and Cellular Effects of Microplastics and Nanoplastics: Focus on Inflammation and Senescence. Cells 2024, 13, 1788. [Google Scholar] [CrossRef] [PubMed]
  45. Dhakal, B.; Shiwakoti, S.; Park, E.Y.; Kang, K.W.; Schini-Kerth, V.B.; Park, S.H.; Ji, H.Y.; Park, J.S.; Ko, J.Y.; Oak, M.H. SGLT2 inhibition ameliorates nano plastics-induced premature endothelial senescence and dysfunction. Sci. Rep. 2023, 13, 6256. [Google Scholar] [CrossRef] [PubMed]
  46. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 645593. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, Z.; Ming, J.; Liu, Y.; Hu, G.; Liao, Q. Epigenetic modification of miR-217 promotes intervertebral disc degeneration by targeting the FBXO21-ERK signalling pathway. Arthritis. Res. Ther. 2022, 24, 261. [Google Scholar] [CrossRef]
  48. Lett, Z.; Hall, A.; Skidmore, S.; Alves, N.J. Environmental microplastic and nanoplastic: Exposure routes and effects on coagulation and the cardiovascular system. Environ. Pollut. 2021, 291, 118190. [Google Scholar] [CrossRef]
  49. Palmer, M.; Kleiner, D.E.; Goodman, Z.; Brunt, E.; Avigan, M.I.; Regev, A.; Hayashi, P.H.; Lewis, J.H.; Mehta, R.; Harrison, S.A.; et al. Liver biopsy for assessment of suspected drug-induced liver injury in metabolic dysfunction-associated steatohepatitis clinical trials: Expert consensus from the Liver Forum. Aliment. Pharmacol. Ther. 2024, 59, 201–216. [Google Scholar] [CrossRef]
  50. Wang, S.; Friedman, S.L. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci. Transl. Med. 2023, 15, eadi0759. [Google Scholar] [CrossRef]
  51. Wang, D.K.; Miao, J.X.; Zhang, L.H.; Zhang, L. Research advances in the diagnosis and treatment of MASLD/MASH. Ann. Med. 2025, 57, 2445780. [Google Scholar] [CrossRef]
  52. Ampuero, J.; Aller, R.; Gallego-Duran, R.; Crespo, J.; Calleja, J.L.; Garcia-Monzon, C.; Gomez-Camarero, J.; Caballeria, J.; Lo Iacono, O.; Ibanez, L.; et al. The biochemical pattern defines MASLD phenotypes linked to distinct histology and prognosis. J. Gastroenterol. 2024, 59, 586–597. [Google Scholar] [CrossRef]
  53. Sergi, C.M. NAFLD (MASLD)/NASH (MASH): Does It Bother to Label at All? A Comprehensive Narrative Review. Int. J. Mol. Sci. 2024, 25, 8462. [Google Scholar] [CrossRef] [PubMed]
  54. Tseng, M.; Syed, T.; Siddiqui, M. Non-invasive diagnosis of metabolic dysfunction associated steatotic liver disease (MASLD). In Hepatology; Elsevier: Amsterdam, The Netherlands, 2025; pp. 111–140. [Google Scholar]
  55. Sanyal, A.J.; Shankar, S.S.; Yates, K.P.; Bolognese, J.; Daly, E.; Dehn, C.A.; Neuschwander-Tetri, B.; Kowdley, K.; Vuppalanchi, R.; Behling, C.; et al. Diagnostic performance of circulating biomarkers for non-alcoholic steatohepatitis. Nat. Med. 2023, 29, 2656–2664. [Google Scholar] [CrossRef] [PubMed]
  56. Stefanakis, K.; Mingrone, G.; George, J.; Mantzoros, C.S. Accurate non-invasive detection of MASH with fibrosis F2-F3 using a lightweight machine learning model with minimal clinical and metabolomic variables. Metabolism 2025, 163, 156082. [Google Scholar] [CrossRef] [PubMed]
  57. Brunt, E.M.; Kleiner, D.E.; Wilson, L.A.; Belt, P.; Neuschwander-Tetri, B.A.; Network, N.C.R. Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: Distinct clinicopathologic meanings. Hepatology 2011, 53, 810–820. [Google Scholar] [CrossRef]
  58. Hjelkrem, M.; Stauch, C.; Shaw, J.; Harrison, S.A. Validation of the non-alcoholic fatty liver disease activity score. Aliment. Pharmacol. Ther. 2011, 34, 214–218. [Google Scholar] [CrossRef]
  59. Huang, Z.; Hu, B.; Wang, H. Analytical methods for microplastics in the environment: A review. Environ. Chem. Lett. 2023, 21, 383–401. [Google Scholar] [CrossRef]
  60. Leslie, H.A.; van Velzen, M.J.M.; Brandsma, S.H.; Vethaak, A.D.; Garcia-Vallejo, J.J.; Lamoree, M.H. Discovery and quantification of plastic particle pollution in human blood. Environ. Int. 2022, 163, 107199. [Google Scholar] [CrossRef]
  61. Schwabl, P.; Koppel, S.; Konigshofer, P.; Bucsics, T.; Trauner, M.; Reiberger, T.; Liebmann, B. Detection of Various Microplastics in Human Stool: A Prospective Case Series. Ann. Intern. Med. 2019, 171, 453–457. [Google Scholar] [CrossRef]
  62. Karlsson, T.M.; Karrman, A.; Rotander, A.; Hassellov, M. Comparison between manta trawl and in situ pump filtration methods, and guidance for visual identification of microplastics in surface waters. Environ. Sci. Pollut. Res. Int. 2020, 27, 5559–5571. [Google Scholar] [CrossRef]
  63. Lavers, J.L.; Oppel, S.; Bond, A.L. Factors influencing the detection of beach plastic debris. Mar. Environ. Res. 2016, 119, 245–251. [Google Scholar] [CrossRef]
  64. Caputo, F.; Vogel, R.; Savage, J.; Vella, G.; Law, A.; Della Camera, G.; Hannon, G.; Peacock, B.; Mehn, D.; Ponti, J.; et al. Measuring particle size distribution and mass concentration of nanoplastics and microplastics: Addressing some analytical challenges in the sub-micron size range. J. Colloid Interface Sci. 2021, 588, 401–417. [Google Scholar] [CrossRef] [PubMed]
  65. von Moos, N.; Burkhardt-Holm, P.; Kohler, A. Uptake and effects of microplastics on cells and tissue of the blue mussel Mytilus edulis L. after an experimental exposure. Environ. Sci. Technol. 2012, 46, 11327–11335. [Google Scholar] [CrossRef] [PubMed]
  66. Cai, H.W.; Xu, E.G.; Du, F.N.; Li, R.L.; Liu, J.F.; Shi, H.H. Analysis of environmental nanoplastics: Progress and challenges. Chem. Eng. J. 2021, 410, 128208. [Google Scholar] [CrossRef]
  67. Schwaferts, C.; Niessner, R.; Elsner, M.; Ivleva, N.P. Methods for the analysis of submicrometer- and nanoplastic particles in the environment. Trac-Trends Anal. Chem. 2019, 112, 52–65. [Google Scholar] [CrossRef]
  68. Huang, Y.; Liang, B.; Li, Z.; Zhong, Y.; Wang, B.; Zhang, B.; Du, J.; Ye, R.; Xian, H.; Min, W.; et al. Polystyrene nanoplastic exposure induces excessive mitophagy by activating AMPK/ULK1 pathway in differentiated SH-SY5Y cells and dopaminergic neurons in vivo. Part. Fibre Toxicol. 2023, 20, 44. [Google Scholar] [CrossRef]
  69. Marfella, R.; Prattichizzo, F.; Sardu, C.; Fulgenzi, G.; Graciotti, L.; Spadoni, T.; D’Onofrio, N.; Scisciola, L.; La Grotta, R.; Frige, C.; et al. Microplastics and Nanoplastics in Atheromas and Cardiovascular Events. N. Engl. J. Med. 2024, 390, 900–910. [Google Scholar] [CrossRef]
  70. Huynh, K. Presence of microplastics in carotid plaques linked to cardiovascular events. Nat. Rev. Cardiol. 2024, 21, 279. [Google Scholar] [CrossRef]
  71. Wang, T.; Yi, Z.; Liu, X.; Cai, Y.; Huang, X.; Fang, J.; Shen, R.; Lu, W.; Xiao, Y.; Zhuang, W.; et al. Multimodal detection and analysis of microplastics in human thrombi from multiple anatomically distinct sites. EBioMedicine 2024, 103, 105118. [Google Scholar] [CrossRef]
  72. Liu, S.; Wang, C.; Yang, Y.; Du, Z.; Li, L.; Zhang, M.; Ni, S.; Yue, Z.; Yang, K.; Wang, Y.; et al. Microplastics in three types of human arteries detected by pyrolysis-gas chromatography/mass spectrometry (Py-GC/MS). J. Hazard. Mater. 2024, 469, 133855. [Google Scholar] [CrossRef]
  73. Yang, Y.; Zhang, F.; Jiang, Z.; Du, Z.; Liu, S.; Zhang, M.; Jin, Y.; Qin, Y.; Yang, X.; Wang, C.; et al. Microplastics are associated with elevated atherosclerotic risk and increased vascular complexity in acute coronary syndrome patients. Part. Fibre Toxicol. 2024, 21, 34. [Google Scholar] [CrossRef]
  74. Anuar, S.T.; Altarawnah, R.S.; Mohd Ali, A.A.; Lee, B.Q.; Khalik, W.; Yusof, K.; Ibrahim, Y.S. Utilizing Pyrolysis-Gas Chromatography/Mass Spectrometry for Monitoring and Analytical Characterization of Microplastics in Polychaete Worms. Polymers 2022, 14, 3054. [Google Scholar] [CrossRef] [PubMed]
  75. Brits, M.; van Velzen, M.J.M.; Sefiloglu, F.Ö.; Scibetta, L.; Groenewoud, Q.; Garcia-Vallejo, J.J.; Vethaak, A.D.; Brandsma, S.H.; Lamoree, M.H. Quantitation of micro and nanoplastics in human blood by pyrolysis-gas chromatography–mass spectrometry. Microplastics Nanoplastics 2024, 4, 12. [Google Scholar] [CrossRef]
  76. Vasudeva, M.; Warrier, A.K.; Kartha, V.B.; Unnikrishnan, V.K. Advances in microplastic characterization: Spectroscopic techniques and heavy metal adsorption insights. TrAC Trends Anal. Chem. 2025, 183, 118111. [Google Scholar] [CrossRef]
  77. Campanale, C.; Savino, I.; Massarelli, C.; Uricchio, V.F. Fourier Transform Infrared Spectroscopy to Assess the Degree of Alteration of Artificially Aged and Environmentally Weathered Microplastics. Polymers 2023, 15, 911. [Google Scholar] [CrossRef] [PubMed]
  78. Feng, S.; Zeng, Y.; Cai, Z.; Wu, J.; Chan, L.L.; Zhu, J.; Zhou, J. Polystyrene microplastics alter the intestinal microbiota function and the hepatic metabolism status in marine medaka (Oryzias melastigma). Sci. Total Environ. 2021, 759, 143558. [Google Scholar] [CrossRef]
  79. Tian, R.; Guan, M.; Chen, L.; Wan, Y.; He, L.; Zhao, Z.; Gao, T.; Zong, L.; Chang, J.; Zhang, J. Mechanism insights into the histopathological changes of polypropylene microplastics induced gut and liver in zebrafish. Ecotoxicol. Environ. Saf. 2024, 280, 116537. [Google Scholar] [CrossRef]
  80. Zhuang, J.; Chen, Q.; Xu, L.; Chen, X. Combined exposure to polyvinyl chloride and polystyrene microplastics induces liver injury and perturbs gut microbial and serum metabolic homeostasis in mice. Ecotoxicol. Environ. Saf. 2023, 267, 115637. [Google Scholar] [CrossRef]
  81. Chen, Y.; Jin, H.; Ali, W.; Zhuang, T.; Sun, J.; Wang, T.; Song, J.; Ma, Y.; Yuan, Y.; Bian, J.; et al. Co-exposure of polyvinyl chloride microplastics with cadmium promotes nonalcoholic fatty liver disease in female ducks through oxidative stress and glycolipid accumulation. Poult. Sci. 2024, 103, 104152. [Google Scholar] [CrossRef]
  82. Guraka, A.; Souch, G.; Duff, R.; Brown, D.; Moritz, W.; Kermanizadeh, A. Microplastic-induced hepatic adverse effects evaluated in advanced quadruple cell human primary models following three weeks of repeated exposure. Chemosphere 2024, 364, 143032. [Google Scholar] [CrossRef]
  83. Horvatits, T.; Tamminga, M.; Liu, B.; Sebode, M.; Carambia, A.; Fischer, L.; Puschel, K.; Huber, S.; Fischer, E.K. Microplastics detected in cirrhotic liver tissue. EBioMedicine 2022, 82, 104147. [Google Scholar] [CrossRef]
  84. Patton, G.N.; Lee, H.J. Chemical Insights into Topical Agents in Intraocular Pressure Management: From Glaucoma Etiopathology to Therapeutic Approaches. Pharmaceutics 2024, 16, 274. [Google Scholar] [CrossRef] [PubMed]
  85. Kaluzhny, Y.; Klausner, M. In vitro reconstructed 3D corneal tissue models for ocular toxicology and ophthalmic drug development. In Vitro Cell Dev. Biol. Anim. 2021, 57, 207–237. [Google Scholar] [CrossRef] [PubMed]
  86. Rauchman, S.H.; Locke, B.; Albert, J.; De Leon, J.; Peltier, M.R.; Reiss, A.B. Toxic External Exposure Leading to Ocular Surface Injury. Vision 2023, 7, 32. [Google Scholar] [CrossRef] [PubMed]
  87. Wu, D.; Lim, B.X.H.; Seah, I.; Xie, S.; Jaeger, J.E.; Symons, R.K.; Heffernan, A.L.; Curren, E.E.M.; Leong, S.C.Y.; Riau, A.K.; et al. Impact of Microplastics on the Ocular Surface. Int. J. Mol. Sci. 2023, 24, 3928. [Google Scholar] [CrossRef]
  88. Kitazawa, K.; Numa, K.; Patel, S.K.; King, C.D.; Matsumoto, A.; Sotozono, C.; Desprez, P.Y.; Schilling, B.; Campisi, J. Cellular senescence exacerbates features of aging in the eyes. Aging Biol. 2023, 1, 20230014. [Google Scholar] [CrossRef]
  89. Hub, E.S. Eye Irritation: In Vivo Rabbit Eye Test Template for Pre-Existing Data. Available online: https://joint-research-centre.ec.europa.eu/reference-measurement/european-union-reference-laboratories/eu-reference-laboratory-alternatives-animal-testing-eurl-ecvam/alternative-methods-toxicity-testing/validated-test-methods-health-effects/eye-irritationserious-eye-damage/eye-irritation-vivo_en (accessed on 17 January 2025).
  90. OECD. In Environment Directorate Joint Meeting of the Chemicals Committee and the Working Party on Chemicals, Pesticides and Biotechnology; OECD: Paris, France, 2013.
  91. Yamamoto, N.; Hiramatsu, N.; Kato, Y.; Sato, A.; Kojima, H. Development of an Eye Irritation Test Method Using an In-House Fabrication of a Reconstructed Human Cornea-like Epithelium Model for Eye Hazard Identification. Bioengineering 2024, 11, 302. [Google Scholar] [CrossRef]
  92. OECD. Test No. 491: Short Time Exposure In Vitro Test Method for Identifying (i) Chemicals Inducing Serious Eye Damage and (ii) Chemicals Not Requiring Classification for Eye Irritation or Serious Eye Damage. Available online: https://www.oecd.org/en/publications/test-no-491-short-time-exposure-in-vitro-test-method-for-identifying-i-chemicals-inducing-serious-eye-damage-and-ii-chemicals-not-requiring-classification-for-eye-irritation-or-serious-eye-damage_9789264242432-en.html (accessed on 17 January 2025).
  93. Ma, J.; Chiu, Y.F.; Kao, C.C.; Chuang, C.N.; Chen, C.Y.; Lai, C.H.; Kuo, M.L. Fine particulate matter manipulates immune response to exacerbate microbial pathogenesis in the respiratory tract. Eur. Respir. Rev. 2024, 33, 230259. [Google Scholar] [CrossRef]
  94. Millen, A.E.; Dighe, S.; Kordas, K.; Aminigo, B.Z.; Zafron, M.L.; Mu, L. Air Pollution and Chronic Eye Disease in Adults: A Scoping Review. Ophthalmic Epidemiol. 2024, 31, 1–10. [Google Scholar] [CrossRef]
  95. Wang, H.; Jia, H.; Han, J.; Zhang, Z.; Yin, X.; Mu, N.; Zhu, Y.; Li, M. Correlation Between Air Quality Index and Tear Film Lipid Layer Thickness: Comparison Between Patients with Sjogren’s Syndrome and with Meibomian Gland Dysfunction. Curr. Eye Res. 2023, 48, 447–455. [Google Scholar] [CrossRef]
  96. Vitar, R.M.L.; Arana, A.G.H.; Marchini, T.O.; Evelson, P.A.; Ferreira, S.M. The Ocular Surface as a Target of Air Pollution. In Environmental Stressors and OxInflammatory Tissues Responses; CRC Press: Boca Raton, FL, USA, 2024; pp. 80–88. [Google Scholar]
  97. Lin, C.C.; Chiu, C.C.; Lee, P.Y.; Chen, K.J.; He, C.X.; Hsu, S.K.; Cheng, K.C. The Adverse Effects of Air Pollution on the Eye: A Review. Int. J. Environ. Res. Public Health 2022, 19, 1186. [Google Scholar] [CrossRef]
  98. Woo, S.H.; Kim, D.Y.; Choi, J.H. Roles of Vascular Smooth Muscle Cells in Atherosclerotic Calcification. J. Lipid Atheroscler. 2023, 12, 106–118. [Google Scholar] [CrossRef] [PubMed]
  99. Hu, C.; Yang, S.; Zhang, T.; Ge, Y.; Chen, Z.; Zhang, J.; Pu, Y.; Liang, G. Organoids and organoids-on-a-chip as the new testing strategies for environmental toxicology-applications & advantages. Environ. Int. 2024, 184, 108415. [Google Scholar] [CrossRef] [PubMed]
  100. Anwar, K.N.; Soleimani, M.; Ashraf, M.J.; Moghtader, A.; Koganti, R.; Ghalibafan, S.; Baharnoori, M.; Arabpour, Z.; Cheraqpour, K.; Sebhat, A.M.; et al. Senescence and Stress Signaling Pathways in Corneal Cells After Nitrogen Mustard Injury. Cells 2024, 13, 2021. [Google Scholar] [CrossRef] [PubMed]
  101. Dammak, A.; Pastrana, C.; Martin-Gil, A.; Carpena-Torres, C.; Peral Cerda, A.; Simovart, M.; Alarma, P.; Huete-Toral, F.; Carracedo, G. Oxidative Stress in the Anterior Ocular Diseases: Diagnostic and Treatment. Biomedicines 2023, 11, 292. [Google Scholar] [CrossRef]
  102. Iqbal, S.; Ramini, A.; Kaja, S. Impact of particulate matter and air pollution on ocular surface disease: A systematic review of preclinical and clinical evidence. Ocul. Surf. 2025, 35, 100–116. [Google Scholar] [CrossRef]
  103. Alijagic, A.; Suljević, D.; Fočak, M.; Sulejmanović, J.; Šehović, E.; Särndahl, E.; Engwall, M. The triple exposure nexus of microplastic particles, plastic-associated chemicals, and environmental pollutants from a human health perspective. Environ. Int. 2024, 188, 108736. [Google Scholar] [CrossRef]
  104. Gruber, E.S.; Stadlbauer, V.; Pichler, V.; Resch-Fauster, K.; Todorovic, A.; Meisel, T.C.; Trawoeger, S.; Hollóczki, O.; Turner, S.D.; Wadsak, W.; et al. To Waste or Not to Waste: Questioning Potential Health Risks of Micro- and Nanoplastics with a Focus on Their Ingestion and Potential Carcinogenicity. Expo. Health 2023, 15, 33–51. [Google Scholar] [CrossRef]
  105. Yu, C.H.; Riker, C.D.; Lu, S.E.; Fan, Z.T. Biomonitoring of emerging contaminants, perfluoroalkyl and polyfluoroalkyl substances (PFAS), in New Jersey adults in 2016–2018. Int. J. Hyg. Environ. Health 2020, 223, 34–44. [Google Scholar] [CrossRef]
  106. Amato-Lourenco, L.F.; Dantas, K.C.; Junior, G.R.; Paes, V.R.; Ando, R.A.; de Oliveira Freitas, R.; da Costa, O.; Rabelo, R.S.; Soares Bispo, K.C.; Carvalho-Oliveira, R.; et al. Microplastics in the Olfactory Bulb of the Human Brain. JAMA Netw. Open 2024, 7, e2440018. [Google Scholar] [CrossRef]
  107. Qin, L.; Wu, J.; Sun, X.; Huang, X.; Huang, W.; Weng, C.; Cai, J. The regulatory role of metabolic organ-secreted factors in the nonalcoholic fatty liver disease and cardiovascular disease. Front. Cardiovasc. Med. 2023, 10, 1119005. [Google Scholar] [CrossRef]
  108. Wang, F.; So, K.F.; Xiao, J.; Wang, H. Organ-organ communication: The liver’s perspective. Theranostics 2021, 11, 3317–3330. [Google Scholar] [CrossRef] [PubMed]
  109. Fritsche, K.; Zikova-Kloas, A.; Marx-Stoelting, P.; Braeuning, A. Metabolism-Disrupting Chemicals Affecting the Liver: Screening, Testing, and Molecular Pathway Identification. Int. J. Mol. Sci. 2023, 24, 2686. [Google Scholar] [CrossRef] [PubMed]
  110. Milani, I.; Codini, M.; Guarisco, G.; Chinucci, M.; Gaita, C.; Leonetti, F.; Capoccia, D. Hepatokines and MASLD: The GLP1-Ras-FGF21-Fetuin-A Crosstalk as a Therapeutic Target. Int. J. Mol. Sci. 2024, 25, 10795. [Google Scholar] [CrossRef] [PubMed]
  111. Kim, T.H.; Hong, D.G.; Yang, Y.M. Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism. Biomedicines 2021, 9, 1903. [Google Scholar] [CrossRef]
  112. Orfanidou, M.; Polyzos, S.A. Retinopathy in Metabolic Dysfunction-Associated Steatotic Liver Disease. Medicina 2025, 61, 38. [Google Scholar] [CrossRef]
  113. Wu, J.; Duan, C.; Yang, Y.; Wang, Z.; Tan, C.; Han, C.; Hou, X. Insights into the liver-eyes connections, from epidemiological, mechanical studies to clinical translation. J. Transl. Med. 2023, 21, 712. [Google Scholar] [CrossRef]
  114. Mouliou, D.S. C-Reactive Protein: Pathophysiology, Diagnosis, False Test Results and a Novel Diagnostic Algorithm for Clinicians. Diseases 2023, 11, 132. [Google Scholar] [CrossRef]
  115. Chen, L.; Lu, J.; Hu, J.; Gong, X. Unveiling the multifaceted role of adropin in various diseases (Review). Int. J. Mol. Med. 2024, 54, 90. [Google Scholar] [CrossRef]
  116. Pennisi, G.; Maurotti, S.; Ciociola, E.; Jamialahmadi, O.; Bertolazzi, G.; Mirarchi, A.; Bergh, P.O.; Scionti, F.; Mancina, R.M.; Spagnuolo, R.; et al. ANGPTLownregulation Increases Intracellular Lipids by Reducing Energy Utilization. Arter. Thromb. Vasc. Biol. 2024, 44, 1086–1097. [Google Scholar] [CrossRef]
  117. Lin, Y.H.; Chen, C.Y.; Chi, H.C.; Wu, M.H.; Lai, M.W.; Yeh, C.T. ANGPTL3 overcomes sorafenib resistance via suppression of SNAI1 and CPT1A in liver cancer. Transl. Oncol. 2025, 52, 102250. [Google Scholar] [CrossRef]
  118. Xu, J.; Wu, F.; Zhu, Y.; Wu, T.; Cao, T.; Gao, W.; Liu, M.; Qian, W.; Feng, G.; Xi, X.; et al. ANGPTL4 regulates ovarian cancer progression by activating the ERK1/2 pathway. Cancer Cell Int. 2024, 24, 54. [Google Scholar] [CrossRef] [PubMed]
  119. Baron-Menguy, C.; Freneau, M.; Mrad, M.-A.; Lebot, C.; Rio, M.; L’allinec, V.; Quillard, T.; Desal, H.; Redon, R.; Bourcier, R. ANGPTL6 variant induces cerebral vascular dysfunction and predisposes to intracranial aneurysm in mice. bioRxiv 2025. [Google Scholar] [CrossRef]
  120. Xu, F.; Wang, N.; Li, G.; Tian, D.; Shi, X. ANGPTL8/Betatrophin Improves Glucose Tolerance in Older Mice and Metabolomic Analysis Reveals Its Role in Insulin Resistance in HepG2 Cells. Diabetes Metab. Syndr. Obes. 2021, 14, 4209–4221. [Google Scholar] [CrossRef] [PubMed]
  121. Xiao, Z.; Yeung, C.L.S.; Yam, J.W.P.; Mao, X. An update on the role of complement in hepatocellular carcinoma. Front. Immunol. 2022, 13, 1007382. [Google Scholar] [CrossRef]
  122. Jensen-Cody, S.O.; Potthoff, M.J. Hepatokines and metabolism: Deciphering communication from the liver. Mol. Metab. 2021, 44, 101138. [Google Scholar] [CrossRef]
  123. Lee-Odegard, S.; Ueland, T.; Thorsby, P.M.; Aukrust, P.; Michelsen, A.E.; Halvorsen, B.; Drevon, C.A.; Birkeland, K.I. Fetuin-A mediates the difference in adipose tissue insulin resistance between young adult pakistani and norwegian patients with type 2 diabetes. BMC Endocr. Disord. 2022, 22, 208. [Google Scholar] [CrossRef]
  124. Dogru, T.; Kirik, A.; Gurel, H.; Rizvi, A.A.; Rizzo, M.; Sonmez, A. The Evolving Role of Fetuin-A in Nonalcoholic Fatty Liver Disease: An Overview from Liver to the Heart. Int. J. Mol. Sci. 2021, 22, 6627. [Google Scholar] [CrossRef]
  125. Yakout, S.M.; Hussein, S.; Al-Attas, O.S.; Hussain, S.D.; Saadawy, G.M.; Al-Daghri, N.M. Hepatokines fetuin A and fetuin B status in women with/without gestational diabetes mellitus. Am. J. Transl. Res. 2023, 15, 1291–1299. [Google Scholar]
  126. Pasmans, K.; Goossens, G.H.; Groenhuijzen, E.; Kemper, E.J.; Reijnders, D.; Most, J.; Blaak, E.E.; Watt, M.J.; Meex, R.C. Fetuin B in white adipose tissue induces inflammation and is associated with peripheral insulin resistance in mice and humans. Obesity 2024, 32, 517–527. [Google Scholar]
  127. De Sousa-Coelho, A.; Rodriguez-Rodriguez, R.; Softic, S.; Jonker, J.; Relat, J. FGF21 as a therapeutic target for obesity and insulin resistance: From rodent models to humans. Front. Endocrinol. 2023, 14, 1253675. [Google Scholar]
  128. Bielka, W.; Przezak, A.; Pawlik, A. Follistatin and follistatin-like 3 in metabolic disorders. Prostaglandins Other Lipid Mediat. 2023, 169, 106785. [Google Scholar] [CrossRef] [PubMed]
  129. Crisan, D.; Avram, L.; Morariu-Barb, A.; Grapa, C.; Hiriscau, I.; Craciun, R.; Donca, V.; Nemes, A. Sarcopenia in MASLD-Eat to Beat Steatosis, Move to Prove Strength. Nutrients 2025, 17, 178. [Google Scholar] [CrossRef] [PubMed]
  130. Elbasiony, E.; Cho, W.; Mittal, S.K.; Chauhan, S.K. Suppression of lipopolysaccharide-induced corneal opacity by hepatocyte growth factor. Sci. Rep. 2022, 12, 494. [Google Scholar] [CrossRef] [PubMed]
  131. Yuan, T.H.; Yue, Z.S.; Zhang, G.H.; Wang, L.; Dou, G.R. Beyond the Liver: Liver-Eye Communication in Clinical and Experimental Aspects. Front. Mol. Biosci. 2021, 8, 823277. [Google Scholar] [CrossRef]
  132. Takata, N.; Ishii, K.A.; Takayama, H.; Nagashimada, M.; Kamoshita, K.; Tanaka, T.; Kikuchi, A.; Takeshita, Y.; Matsumoto, Y.; Ota, T.; et al. LECT2 as a hepatokine links liver steatosis to inflammation via activating tissue macrophages in NASH. Sci. Rep. 2021, 11, 555. [Google Scholar] [CrossRef]
  133. Nono Nankam, P.A.; Bluher, M. Retinol-binding protein 4 in obesity and metabolic dysfunctions. Mol. Cell Endocrinol. 2021, 531, 111312. [Google Scholar] [CrossRef]
  134. Montgomery, M.K.; Bayliss, J.; Devereux, C.; Bezawork-Geleta, A.; Roberts, D.; Huang, C.; Schittenhelm, R.B.; Ryan, A.; Townley, S.L.; Selth, L.A.; et al. SMOC1 is a glucose-responsive hepatokine and therapeutic target for glycemic control. Sci. Transl. Med. 2020, 12, eaaz8048. [Google Scholar] [CrossRef]
  135. Xi, X.; Han, L.; Ding, M.; Li, J.; Qiao, C.; Liu, Z.; Qie, S. Exploring the relationship between intestinal flora and the pathological mechanism of myopia in adolescents from the perspective of Chinese and Western medicine: A review. Medicine 2023, 102, e33393. [Google Scholar] [CrossRef]
  136. Wang, M.; Yu, X. A bibliographic study of “liver-eye” related research: A correlation function analytic research between age-related macular degeneration (AMD) and traditional chinese medicine (TCM) liver wind internal movement syndrome. Adv. Clin. Med. 2023, 13, 6342. [Google Scholar] [CrossRef]
  137. Liu, Z.-W.; Shu, J.; Tu, J.-Y.; Zhang, C.-H.; Hong, J. Liver in the Chinese and Western Medicine. Integr. Med. Int. 2017, 4, 39–45. [Google Scholar] [CrossRef]
  138. Toragall, V.; Baskaran, V. Chitosan-sodium alginate-fatty acid nanocarrier system: Lutein bioavailability, absorption pharmacokinetics in diabetic rat and protection of retinal cells against H(2)O(2) induced oxidative stress in vitro. Carbohydr. Polym. 2021, 254, 117409. [Google Scholar] [CrossRef] [PubMed]
  139. Kang, K.H.; Shin, D.; Ryu, I.H.; Kim, J.K.; Lee, I.S.; Koh, K.; Yoo, T.K. Association between cataract and fatty liver diseases from a nationwide cross-sectional study in South Korea. Sci. Rep. 2024, 14, 77. [Google Scholar] [CrossRef]
  140. Wilson, S.E.; Walker, J.W.; Chwang, E.L.; He, Y.G. Hepatocyte growth factor, keratinocyte growth factor, their receptors, fibroblast growth factor receptor-2, and the cells of the cornea. Investig. Ophthalmol. Vis. Sci. 1993, 34, 2544–2561. [Google Scholar]
  141. Pączek, S.; Zajkowska, M.; Mroczko, B. Pigment Epithelial-Derived Factor in Pancreatic and Liver Cancers—From Inflammation to Cancer. Biomedicines 2024, 12, 2260. [Google Scholar] [CrossRef]
Figure 1. This diagram illustrates the study selection process and is based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. It is important to note that the International Prospective Register of Systematic Reviews (PROSPERO) does not accept scoping reviews, literature reviews, or mapping reviews. Therefore, this literature review was not registered with PROSPERO.
Figure 1. This diagram illustrates the study selection process and is based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. It is important to note that the International Prospective Register of Systematic Reviews (PROSPERO) does not accept scoping reviews, literature reviews, or mapping reviews. Therefore, this literature review was not registered with PROSPERO.
Ijms 26 02837 g001
Figure 2. Schematic of substances entering the system through the eye and then being distributed after the first-pass effect. (BioRender licensed to the University of Rijeka, Croatia, https://www.biorender.com (accessed on 29 January 2025), was used to make the figure).
Figure 2. Schematic of substances entering the system through the eye and then being distributed after the first-pass effect. (BioRender licensed to the University of Rijeka, Croatia, https://www.biorender.com (accessed on 29 January 2025), was used to make the figure).
Ijms 26 02837 g002
Table 1. The classification of plastic waste depending on size.
Table 1. The classification of plastic waste depending on size.
Plastic Debris NameParticle Size
Macroplastics>5 mm
Larger microplastics1–5 mm
Microplastics67–500 µm, 1–5000 µm, 20–5000 µm, or more broadly, as <5000 µm [definition supported by the National Oceanic and Atmospheric Administration (NOAA)]
Nanoplastics<20 µm to as small as 1 nm
Table 2. Hepatokines with the annotation of those involved in the liver–eye axis.
Table 2. Hepatokines with the annotation of those involved in the liver–eye axis.
HepatokinesCorrelated with (Biological Functions)Liver Is the Target Organ/Paracrine Mechanism?Impacts the Liver–Eye Axis? Possible Key Factors for Liver–Eye ContactReferences
AdropinMacronutrient intake and estrogen.YesYes[112,113,115]
ANGPTL3Increases lipid production in the liver and plasma lipid levels, promotes lipogenesis and the liver’s inflammatory response, and decreases glucose uptake.YesYes[112,113,116,117]
ANGPTL4Inhibits lipoprotein lipase and activates cAMP-stimulated lipolysis in adipocytes. Yes[112,113,118]
ANGPTL6Enhances insulin signaling in skeletal muscle and mitochondrial oxygen consumption in white adipose tissue. Inhibits gluconeogenesis in the liver.YesYes[112,113,119]
ANGPTL8/betatrophinDubious action on beta-cell proliferation.YesYes[112,113,120]
CFHA complement inhibitor inhibits its excessive activation and is a key player in maintaining complement homeostasis. It is present as a soluble protein and is also attached to cell surfaces throughout the human body.YesYes[110,111,121,122]
Fetuin-AIncreases inflammation and insulin resistance.Yes [123,124,125]
Fetuin-BIncreases hepatic steatosis and mediates impaired insulin action and glucose intolerance.Yes [125,126]
FGF-21An insulin-sensitizing hormone/metabolic actions.YesYes[112,113,127]
FollistatinIncreases insulin resistance, promotes thermogenesis, and induces the differentiation of brown adipocytes. Enhances glucose levels and the uptake of free fatty acids after exercise training. Inhibits FSH production and suppresses skeletal muscle growth.Yes [128,129]
GDF15Increases energy metabolism and lowers body weight; it is possibly implicated in the pathogenesis of anorexia. Stimulates thermogenic and lipolytic genes.
Improves glucose tolerance and insulin sensitivity. Prevents liver steatosis.
Yes [110,111,113,122]
HepassocinPromotes insulin resistance and adipogenesis.Yes [110,111,122]
HGFParacrine cellular growth, motility, and morphogenic factor.YesYes[110,111,112,113,122,130,131]
LECT2Promotes the accumulation of lipids and inflammation in the liver and the development of insulin resistance in skeletal muscle.Yes [110,111,122,132]
RBP4Depending on the source, the effect may be controversial. It increases lipolysis in adipocytes, is associated with insulin resistance and components of metabolic syndrome, promotes hepatic mitochondrial dysfunction and hepatic steatosis, and impairs insulin signaling.??[110,111,112,122,133]
Selenoprotein PInhibits hepatic gluconeogenesis and decreases glucose uptake in the skeletal muscle.Yes [122]
SMOC1Improves glycemic control via inhibiting gluconeogenesis and glucose output from the liver.Yes [110,111,122,134]
ANGPTLs: antiopoietin-like proteins; CFH: complement factor H; FSH: follicle-stimulating hormone; GDF15: growth differentiation factor 15; HGF: hepatocyte growth factor; LECT2: leukocyte cell-derived chemotaxin 2; RBP4: retinol-binding protein 4; SMOC1: SPARC-related modular calcium-binding protein 1.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Šoša, I.; Labinac, L.; Perković, M. Metabolic Dysfunction-Associated Steatotic Liver Disease Induced by Microplastics: An Endpoint in the Liver–Eye Axis. Int. J. Mol. Sci. 2025, 26, 2837. https://doi.org/10.3390/ijms26072837

AMA Style

Šoša I, Labinac L, Perković M. Metabolic Dysfunction-Associated Steatotic Liver Disease Induced by Microplastics: An Endpoint in the Liver–Eye Axis. International Journal of Molecular Sciences. 2025; 26(7):2837. https://doi.org/10.3390/ijms26072837

Chicago/Turabian Style

Šoša, Ivan, Loredana Labinac, and Manuela Perković. 2025. "Metabolic Dysfunction-Associated Steatotic Liver Disease Induced by Microplastics: An Endpoint in the Liver–Eye Axis" International Journal of Molecular Sciences 26, no. 7: 2837. https://doi.org/10.3390/ijms26072837

APA Style

Šoša, I., Labinac, L., & Perković, M. (2025). Metabolic Dysfunction-Associated Steatotic Liver Disease Induced by Microplastics: An Endpoint in the Liver–Eye Axis. International Journal of Molecular Sciences, 26(7), 2837. https://doi.org/10.3390/ijms26072837

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

Article Metrics

Back to TopTop