Next Article in Journal
Exposure to Bisphenol S and Bisphenol F Alters Gene Networks Related to Protein Translation and Neuroinflammation in SH-SY5Y Human Neuroblastoma Cells
Previous Article in Journal
Mono(2-ethylhexyl) Phthalate Disrupts Mitochondrial Function, Dynamics and Biogenesis in Human Trophoblast Cells at Human Exposure Range Concentrations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxicological Effects of Tartrazine Exposure: A Review of In Vitro and Animal Studies with Human Health Implications

1
Doctoral School of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Carol I Avenue, 20A, 700505 Iași, Romania
2
“Ion Haulica” Institute, Apollonia University, Păcurari Street 11, 700511 Iași, Romania
3
Doctoral School of Geosciences, Faculty of Geography and Geology, “Alexandru Ioan Cuza” University of Iasi, Carol I Avenue, 20A, 700505 Iași, Romania
4
Preclinical Department, Apollonia University, Păcurari Street 11, 700511 Iași, Romania
5
Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Carol I Avenue, 20A, 700505 Iași, Romania
6
Academy of Romanian Scientists, Splaiul Independentei Avenue No. 54, Sector 5, 050094 Bucharest, Romania
7
CENEMED Platform for Interdisciplinary Research, “Grigore T. Popa” University of Medicine and Pharmacy, University Street, No. 16, 700115 Iași, Romania
8
Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iasi, Alexandru Lapusneanu Street, No. 26, 700506 Iași, Romania
9
Physiology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
*
Authors to whom correspondence should be addressed.
Toxics 2025, 13(9), 771; https://doi.org/10.3390/toxics13090771
Submission received: 1 August 2025 / Revised: 9 September 2025 / Accepted: 10 September 2025 / Published: 12 September 2025
(This article belongs to the Section Agrochemicals and Food Toxicology)

Abstract

Tartrazine (TZ, also known as FD&C Yellow No. 5 or E102) is a synthetic, water-soluble yellow food dye widely used in the food and pharmaceutical industries. Some studies have associated TZ with allergic reactions, especially among people with dye sensitivities or pre-existing allergies. Recent research also suggests a possible link between TZ consumption and the worsening of behavioral disorders, especially in children, including symptoms such as hyperactivity, irritability, restlessness, and sleep disturbances. Experimental studies in laboratory animals have highlighted potential risks associated with prolonged or high-dose exposure, including toxic effects on the nervous system and liver function. In addition, increasing evidence indicates that TZ can induce oxidative stress (OS) by increasing the production of reactive oxygen species (ROS), which can contribute to cellular damage and inflammation. Although the evidence remains inconclusive, there are recommendations to limit the intake of synthetic food dyes, especially in children’s diets. The purpose of this review is to examine the potential toxic effects on health of tartrazine by analyzing findings from experimental studies in cell cultures and laboratory animals, as well as correlations observed in humans. We focus on documented adverse reactions, including possible neurotoxic, hepatotoxic, oxidative, and behavioral effects. Through this, we aim to contribute to a more comprehensive understanding of the risks associated with exposure to this synthetic food dye.

Graphical Abstract

1. Introduction

Tartrazine is an azo dye commonly used to impart color to food products [1,2,3,4,5]. Approximately 65% of azo dyes are used as food additives and are found in various products, such as candies, jams, and soft drinks, as well as in pharmaceuticals, cosmetics, and textiles. Although TZ adds no nutritional benefit and has no nutritional value, it remains the most widely used food dye, with a significant presence in the global food industry [6,7,8,9]. According to the World Health Organization (WHO), since 2016, the acceptable daily intake (ADI) has been set at 0–10 mg/kg body weight [10]. Despite being one of the first food additives associated with potential adverse reactions, TZ continues to be approved for use in the European Union (EU) and other regions [11].
Several studies have reported an association between TZ exposure and allergic reactions such as urticaria, asthma, migraines, blurred vision, and pruritus, as well as behavioral disturbances, including hyperactivity in children [12,13,14,15,16,17]. Children appear particularly susceptible, with evidence suggesting potential links between synthetic food dyes and neurobehavioral disorders, including attention deficit/hyperactivity disorder (ADHD), as well as mutagenic or carcinogenic outcomes [18,19]. In a double-blind, placebo-controlled study, some children exposed to varying TZ doses exhibited irritability, restlessness, and sleep disturbances [20].
In recent decades, numerous experimental studies, both in vitro and in animal models, have investigated the effects of TZ on cellular functions, tissue integrity, and behavior. The results suggest this food additive may be involved in inducing OS through the generation of ROS [21,22], activating the inflammatory response through the overproduction of proinflammatory cytokines interleukin-1 (IL-1) and interleukin-6 (IL-6) [23,24], modifying enzymatic activity, and contributing to the onset of neurobehavioral disorders [25], including impairments in learning and memory processes [26,27]. From a biological mechanistic perspective, OS has emerged as a key pathway mediating TZ toxicity, contributing to an imbalance between pro-oxidants and antioxidants in cells [28,29]. This imbalance is reflected in elevated levels of malondialdehyde (MDA), a marker of lipid peroxidation, indicating increased oxidative damage. Concomitantly, endogenous antioxidant enzymes such as superoxide dismutase (SOD) and catalase (CAT) are often significantly reduced, supporting the pro-oxidant shift induced by TZ exposure [30].
However, the data available in scientific literature are heterogeneous, and translating results from preclinical experiments into the context of human exposure remains challenging. At the same time, the food industry is increasingly exploring natural alternatives to mitigate the health side effects of synthetic dyes. Therefore, replacing them with natural alternatives represents a much better option [19]. Some well-known natural alternatives include turmeric, carotenoids, annatto, saffron, and paprika extracts. For example, TZ could potentially be substituted with carotenoids extracted from pequi (Caryocar brasiliense) using high-performance ionic liquids, a modern, sustainable, and efficient extraction method that also enhances safety [31,32].
The aim of this review is to provide a critical and integrative analysis of experimental data from in vitro and animal model studies regarding TZ exposure, with a focus on toxicological mechanisms, including OS, neurotoxicity, and inflammatory responses. By correlating these findings with reported human side effects, this review aims to offer a comprehensive perspective on the potential risks to human health. Thus, this study contributes to clarifying a current issue regarding the safety of synthetic food additives and supports the need to reassess TZ exposure, particularly among children.

2. Health Implications of Tartrazine Exposure

TZ is a synthetic yellow dye widely used in food, pharmaceutical, and non-food products (Table 1). Although it improves product appearance, multiple studies have reported its potential to induce allergic reactions, asthma, or cutaneous hypersensitivity. These adverse effects have led to growing interest in natural alternatives such as annatto or β-carotene [2,25,33,34,35,36,37,38,39,40,41,42,43,44,45].
Because of its widespread use, TZ is strictly regulated. In the United States, the FDA’s Code of Federal Regulations establishes the conditions for its use in foods, drugs, and cosmetics, including batch certification and mandatory labeling to prevent hypersensitivity reactions [46]. In the EU, TZ is regulated under Commission Regulation (EC) No 1333/2006 on food additives, while its use in cosmetics is addressed by Regulation (EC) No 1223/2009 (Annex IV) [47], where it appears as trisodium 5-hydroxy-1-(4-sulphophenyl)-4-((4-sulphophenyl)azo)pyrazole-3-carboxylate and its insoluble lakes, salts, and pigments.
Chemically, TZ is an anionic acid dye that converts to tartrazic acid in acidic conditions [48]. In vivo, TZ is reduced to an aromatic amine, stabilized by its nitroso derivative [49]. This compound can trigger immunological reactions, including fatigue, irritability, clinical depression, headaches, and sleep disturbances. Figure 1 summarizes the main biological pathways through which TZ may affect human health: OS, chronic inflammation, genotoxicity, and neurobehavioral disorders. These mechanisms are primarily observed under repeated or chronic exposure and may contribute to systemic conditions, especially in children and sensitive individuals. Both ingestion and dermal contact may cause hypersensitivity reactions. Some researchers suggest that symptoms may occur even at low exposure levels, with manifestations appearing up to 72 h after contact [45].
Although direct evidence linking TZ to anxiety in humans is limited, preclinical studies suggest that TZ increases OS, negatively affects liver and kidney function, alters lipid levels, and consequently raises the risk of cardiovascular disease [32,50]. Data on neurobehavioral effects are often inconsistent. The results regarding its impact on behavior and neurotoxicity are often contradictory or inconclusive.
TZ has been associated with various adverse reactions in some individuals, including migraines, agitation, asthma attacks, blurred vision, eczema, and other skin rashes. Some studies have also suggested a potentially elevated risk of thyroid cancer [49,51]. A randomized, double-blind, placebo-controlled study reported that administration of a mixture of food colorants (5 mg E110, 2.5 mg E122, 7.5 mg E102, 5 mg E124) along with 45 mg E211 was linked to negative effects on concentration, hyperactivity, and attention in children aged 3 and 8–9 years [52]. Approximately 1% of acute urticaria and/or angioedema cases were attributed to TZ [53]. Moreover, due to concerns regarding hyperactivity and behavioral effects in children, some European Member States have taken regulatory measures to ban or restrict the use of TZ. An overview of the proposed pathways and health effects associated with tartrazine exposure is presented in Figure 1.
Children with suspected hyperactivity exposed to TZ exhibited irritability, restlessness, and sleep disturbances, with severity correlating with dosage [20]. The administration of TZ in atopic patients triggered allergic reactions, predominantly cutaneous and respiratory, which were significantly more frequent compared to placebo. These included angioedema, nasal congestion, rhinorrhea, wheezing, skin rashes, and pruritus, and approximately 5% of participants showed elevated levels of TZ-specific Immunoglobulin E [54].
Data from the literature on the cytotoxic, mutagenic, and genotoxic effects of TZ remain unclear and often contradictory. Although Soares et al. [55] did not report significant cytotoxic effects, they highlighted the potential health risk of prolonged TZ exposure, suggesting it could contribute to carcinogenic processes.
Most preclinical studies demonstrating the toxic effects of TZ have used doses ranging from 100 to 700 mg/kg body weight in rodents [51,56,57,58,59,60,61,62], which is substantially higher than typical human dietary exposure (0–10 mg/kg body weight) [10]. While these high doses are useful for elucidating mechanisms of toxicity, their direct relevance to humans is limited. However, they remain important for risk assessment because cumulative exposure from multiple sources (food, pharmaceuticals, cosmetics) can approach or exceed ADI, especially in children [63]. The acceptable ADI established by JECFA is based on a NOAEL of 984 mg/kg body weight/day from chronic rat studies [10], where body weight reduction was considered the critical endpoint. While this regulatory value provides a safety margin, the in vitro and in vivo evidence summarized in this review demonstrated OS, neurotoxicity, and metabolic alteration at doses approaching or below the NOAEL.
Sensitive populations, such as individuals with allergies or metabolic disorders, may experience adverse effects even at doses below the ADI. EFSA noted that a fraction of the population may develop intolerance reactions at ADI levels [64]. The EFSA assessment took into account additional studies, including those by McCann et al. [52], which observed an increase in hyperactivity in children following the consumption of TZ-containing drinks. EFSA concluded that TZ was negative in long-term carcinogenicity studies, and the DNA migration effects observed in the in vivo Comet assay are not expected to result in carcinogenicity [64].
Children may be particularly vulnerable due to higher consumption of yellow- and orange-colored foods. Although most synthetic dyes were below the ADI, TZ was detected in several products, particularly ice cream, highlighting the need for monitoring and further research to protect this vulnerable population [65]. Taken together, high-dose animal data primarily establish hazards and mechanisms [66] but also provide biological plausibility for human health risks, reinforcing the need for prudent regulation and monitoring.

3. Oxidative Stress Induced by Tartrazine

Several studies in rodents have associated TZ exposure with the induction of OS [21,51,57,62,67,68] (Table 2). OS represents an imbalance between the production of ROS and the capacity of endogenous antioxidant systems to neutralize them, leading to damage to cellular components such as lipids, proteins, and DNA [69,70].
Experimental studies in Swiss white mice have shown that daily oral administration of TZ at doses as low A 2.5 and 5 mg/kg, from the first day of gestation until 15 days postnatally, induces OS in brain tissue, reflected by an increased MDA level and decreased activities of antioxidant enzymes such as glutathione (GSH) and SOD [71]. Exposure to TZ, even at concentrations considered environmentally relevant (50 mg/L), has been shown to cause significant biochemical changes, such as increased levels of MDA and nitric oxide (NO), along with a decrease in antioxidant enzyme activities (SOD, CAT, GSH) and acetylcholinesterase (AChE), suggesting neurobiochemical dysfunctions [76]. Daily administration of higher doses, such as 320 mg/kg for four weeks, exacerbates oxidative imbalance in the brain by reducing glutathione peroxidase (GPx) activity and increasing lipid peroxidation, reflected in elevated MDA levels [72]. Even significantly lower doses (7.5 mg/kg) administered over a longer period (90 days) induce severe OS, associated with increased MDA and reduced activities of endogenous antioxidant enzymes (CAT, SOD, GPx) and GSH levels [30].
Moreover, the combination of TZ and erythrosine has demonstrated synergistic effects in inducing OS in brain structures such as the striatum, where increases in MDA, AChE, and nitrites, along with reductions in GSH and CAT, suggest disruption of cerebral redox balance and a potential role in neurotoxicity [73].
Repeated exposure to TZ at doses ranging from 10–75 mg/kg over periods of up to 8 weeks has been associated with marked alterations in oxidative balance in key organs such as the liver, kidneys, and pancreas. This occurs through decreased levels of endogenous antioxidants (GSH, SOD, CAT, glutathione reductase (GR)) and increased MDA, reflecting a significant contribution to metabolic and endocrine dysfunctions [1,74,75].
Additional studies have shown that high doses (200–500 mg/kg) exacerbate systemic OS, affecting the liver, kidneys, spleen, and blood. Marked increases in MDA [29,56,57,59,61,62] were observed, along with decreases in GSH, CAT, and SOD [29,56,57,59], TOS, and TAS [59], as well as alterations in histopathological parameters [29,56,57,59,62]. However, Golli et al. [62] reported increases in CAT and GST levels. These imbalances are correlated with marked histopathological changes in the liver, kidneys, and intestines, supporting the idea of systemic toxicity via oxidative mechanisms.
Also, the studies by Amin et al. [68] and Gao et al. [51] highlight the impact of doses ranging from 15 to 700 mg/kg on the liver and brain through the inhibition of GSH, SOD, CAT and the increase in lipid peroxidation [51,68], reflecting the impairment of cognitive functions and potential neurotoxicity. These effects are attributed to the formation of free radicals during the metabolism of TZ, which promotes the activation of inflammatory pathways and increased expression of IL-1 and IL-6, reinforcing the role of chronic inflammation in the toxicity of this substance [51].
Taken together, these studies indicate that TZ disrupts redox homeostasis by increasing ROS and depleting endogenous antioxidants such as SOD, CAT, GPx, and GSH, leading to lipid peroxidation, protein oxidation, and protein DNA damage [1,29,30,51,57,61,67,68,70,72,77,78,79,80]. This oxidative imbalance contributes to neurotoxicity, hepatotoxicity, and systemic organ dysfunction and may interact synergistically with other food additives such as erythrosine. While most data derive from rodent models at doses above human ADI, this finding provides mechanistic insight into potential human health effects, especially in sensitive populations [1,51,56,57]. The close link between OS, inflammation, and metabolic or neurobehavioral alteration underscores the biological plausibility of adverse outcomes in humans [51,68].

4. In Vitro Studies on Tartrazine Toxicity

In vitro experiments provide a controlled environment to assess the direct cellular effects of TZ and to explore potential molecular mechanisms of toxicity. These studies complement in vitro findings and help identify specific cell type susceptibilities. Several in vitro studies have assessed the cytotoxic and genotoxic effects of TZ in different types of human cells and experimental models (Table 3). In human lymphocytes exposed to concentrations ranging from 0.25 to 64.0 mM, TZ did not show cytotoxicity but induced significant genotoxic effects at all tested concentrations, with only partial DNA repair observed [55]. Similarly, human leukocytes exposed to TZ at 5–500 μg/mL showed no cytotoxic or mutagenic effects; however, DNA damage was observed at concentrations ≥70 μg/mL, consistent with in silico toxicity predictions [77].
Taken together, in vitro studies demonstrated that TZ exhibits low cytotoxicity in normal human cells such as lymphocytes [55,80] and fibroblasts [78], even at high concentrations, but consistently induces genotoxic alterations in lymphocytes [55,77,80] and epigenetic modifications in keratinocytes and cancer cell lines [81]. These findings suggest interference with DNA integrity and chromatin regulation. TZ induces stronger cytotoxic effects in some cancer cell types, indicating differential susceptibility depending on cell type. Mechanistically, these effects may involve induction of OS [78], disruption of the DNA repair pathway [55,77], and modulation of epigenetic regulations such as DNA methyltransferases and histone deacetylases [81]. The moderate estrogenic activity observed in breast cancer cells further suggests that TZ could interact with hormonal pathways under certain conditions [79].
An in vitro study on human foreskin fibroblasts evaluated the effects of several dyes, including TZ. Testing revealed that TZ did not induce significant cytotoxic effects, even at high concentrations. In contrast, indigo carmine and chlorophyllin exhibited marked cytotoxicity at elevated doses. Moreover, chlorophyllin also triggered increased ROS production, indicating potential OS. These findings underscore the importance of further research into the safety of dyes commonly used in pharmaceutical and cosmetic formulations [78].
Investigations into the estrogenic, cytotoxic, and genotoxic potential of TZ compared to phytoestrogens using yeast assays and MCF-7 breast cancer cells demonstrated that TZ possesses moderate estrogenic activity, with low cytotoxicity and no genotoxicity observed in short-term exposures [79]. Additionally, TZ tested at concentrations of 2.5, 5, and 10 mM showed no genotoxicity and minimal cytotoxicity in human lymphocytes but induced significant cytotoxic effects in melanoma cells [80]. Exposure to TZ was associated with upregulation of enzymes involved in epigenetic regulation in human cells such as HaCaT, HepG2, and A549, which may influence cell proliferation and survival, potentially favoring activation of oncogenic pathways. Furthermore, significant DNA fragmentation was observed, indicating a potential genotoxic effect. These results suggest that TZ may contribute to epigenetic disruptions and increased cancer risk [81].
Although the concentrations used in vitro are generally higher than typical human dietary exposure, these findings provide biological plausibility for genotoxic and epigenetic effects. Overall, in vitro evidence indicates that, while TZ has limited cytotoxicity in normal human cells, it can induce genotoxic effects and notable cytotoxicity in certain cancer cell lines. Differences in responses between cell types highlight the need to use a variety of cell models when evaluating the safety of this widely used azo dye.

5. Tartrazine Toxicity in Experimental Animal Models

Experimental studies in animal models indicate that TZ exerts multisystem toxicity, particularly under chronic or high-dose exposure (Table 4). Data obtained from Danio rerio show increased sensitivity to TZ during embryonic development. According to the study by Joshi and Katti [82], exposure to concentrations ≥10 mM was associated with the occurrence of deformities, edema, cardiovascular dysfunctions, delayed hatching, and increased mortality. At concentrations ≥75 mM, embryonic development was completely inhibited, highlighting the significant toxic potential of the substance on embryonic development. Similar findings were reported by Jiang et al. [83], who observed reduced embryonic survival, delayed hatching, swelling of the heart, and deformities along the body axis, with estimated effective concentration for 50% of the population and lethal concentration for 50% of the population values of 42.66 mM and 47.10 mM, respectively. Gupta et al. [84] further noted that exposure to TZ in embryonic water, especially at concentrations of 0.5% and above, significantly increased hatching rates and altered SOD1 gene expression during early developmental stages. Additionally, Thanh et al. [85] identified a vascular toxicity profile manifested by hemorrhages, edema, and abnormal vessel branching, correlated with altered migration and proliferation of endothelial cells. Adverse effects were also observed under chronic exposure conditions. Linskens et al. [86] demonstrated that prolonged administration of TZ (22 μM) caused cognitive deficits in adult fish, reflected by decreased learning capacity and cognitive flexibility. These changes were not present with limited exposure during the early post-embryonic period, suggesting a duration-dependent manifestation of neurotoxicity.
In rodents, oral administration of TZ induces significant structural and functional changes. Meena and Meena [87] reported that TZ caused a dose-dependent increase in body weight, deformation of seminiferous tubules, decreased Leydig and Sertoli cell numbers, and reduced spermatozoa. Similarly, Arefin et al. [88] reported evidence of hepatotoxicity and nephrotoxicity at doses of 200–400 mg/kg, including significant increases in bilirubin, creatinine, and body weight. TZ significantly affects glucose homeostasis and pancreatic endocrine function, being associated with increased blood glucose levels and lipase activity, as well as decreased insulin, calcium, and magnesium levels after 30 days of administration [1]. Other histopathological effects include dilation of perineural spaces and severe degeneration of Purkinje cells, edema, and increased astrocyte populations. Subchronic exposure to TZ has been correlated with alterations in hepatic and renal parameters, evidenced by increases in alanine aminotransferase (ALT), aspartate aminotransferase (AST), urea, and total protein [61], as well as transaminases, lactate dehydrogenase (LDH), creatinine, and uric acid, with decreases in total proteins, albumin, and globulins [51,72,73].
At the neurobehavioral level, TZ induces hyperactivity, anxiety, and cognitive deficits in behavioral tests, alongside cerebral histological changes, including neuronal apoptosis and vascular congestion, highlighting its neurotoxic effect [51,72,73]. In a complementary study, Kamel and El-Lathey [89] reported anxious and depressive effects, as well as decreased social interaction in male Wistar rats, suggesting a cumulative negative impact on mental health.
Chronic exposure (7.5–500 mg/kg) has been correlated with increases in hepatic enzymes (ALT, AST, alkaline phosphatase (ALP)) [26,30,56,60,68,74,90,91], renal markers (urea, creatinine, uric acid) [56,60,74,90,91], and lipid parameters (low-density lipoprotein (LDL), triglycerides) [30,59,74,90,91]. These changes were accompanied by histopathological lesions in vital organs such as the liver, kidneys, testes, stomach, colon, heart, and central nervous system [30,56,57,74,90,91,92,93,94]. Hematological and immunological alterations included leukocytosis, increased tumor necrosis factor alpha (TNF-α), and splenic changes, alongside decreases in hemoglobin and platelets [93,95]. Endocrine disorders, electrolyte imbalances, and neuroinflammation were also reported [73].
Altinoz et al. [57] reported severe intestinal lesions characterized by reduced intestinal antioxidant levels and increases in lipid peroxidation. Additionally, cardiovascular impairment was suggested by elevated cardiac troponin, creatine kinase, and non-high-density lipoprotein (HDL) cholesterol [92], parameters commonly used as early indicators of myocardial injury, suggesting a potential cardiovascular risk under prolonged exposure conditions.
A significant number of studies have investigated the protective role of natural compounds. Among these, Curcumin, crocin, Nigella sativa oil, chlorophyll, and spinach fruit extract have proven effective in reducing OS, normalizing biochemical parameters, and alleviating tissue damage [30,56,57,58,59,74,75,91,93,94,97]. Al-Seeni et al. [74] reported that administration of 10 mg/kg for 8 weeks resulted hepatic and renal dysfunctions, histopathological lesions, and alterations in lipid profile and oxidative markers, with partial improvement following administration of Nigella sativa oil. Comparatively, natural yellow coloring agents such as curcumin and β-carotene not only provide coloration but also exhibit antioxidant and anti-inflammatory properties [98,99,100,101,102], highlighting their potential as a safer alternative to TZ. For instance, curcumin and β-carotene have been shown in multiple studies to exhibit antioxidant and anti-inflammatory effects, with curcumin reducing OS and inflammation in cell and animal models [98,99,100], and β-carotene potentially modulating pathways such as TNF and sphingomyelin signaling [101,102]. Integrating these natural compounds into foods could mitigate the risks associated with synthetic azo dyes while maintaining product appeal.
Available data consistently highlights the systemic toxic effects of TZ, manifested through hepatic, renal, hematological, metabolic, and neurological dysfunctions, confirmed by both biochemical changes and histopathological lesions. Prolonged exposure also induces hematological and immunological disorders, disturbances in glucose and electrolyte metabolism, and negative effects on the central nervous system, including anxiety, depression, reduced locomotor activity, and neuroinflammation [30,56,73,90,93,95,96].

6. Conclusions and Future Perspectives

Tartrazine remains a widely used synthetic food additive; yet, accumulating scientific evidence raises serious concerns about its safety. Preclinical studies consistently report toxic effects such as oxidative stress, inflammation, neurotoxicity, metabolic disruption, hepatotoxicity, and potential genotoxicity. Even though these findings, such as antioxidant system impairment and activation of inflammatory pathways are mostly from animal models, these results are relevant to human health. Although direct clinical evidence is limited, epidemiological data suggests associations between tartrazine exposure and adverse effects like hyperactivity, allergies, migraines, and behavioral issues, particularly in children. Considering the potential for cumulative exposure, individual variability, and unclear thresholds for chronic toxicity, a re-evaluation of tartrazine’s safety is both timely and necessary. Replacing tartrazine with safer, natural alternatives should be prioritized to protect public health, especially for sensitive populations. To better understand the risks associated with tartrazine, future studies should focus on long-term clinical investigation, the identification of specific biomarkers, and clarification of the epigenetic mechanisms involved. Simultaneously, developing safe natural alternatives and reassessing current regulations remain essential priorities to ensure public health protection.

Author Contributions

Conceptualization, M.V., A.S. and A.C.; methodology, M.V., V.R. and C.A.; validation, V.B., M.H. and A.C.; formal analysis, M.V. and A.S.; investigation, resources, data curation, M.V., A.S., V.R., V.B., G.P., C.I., A.C., C.A. and M.H.; writing—original draft preparation, M.V., A.S., V.R. and I.-M.B.; writing—review and editing, G.P. and C.A.; visualization, V.R., C.I. and I.-M.B.; supervision, V.B. and M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rehman, K.; Ashraf, A.; Azam, F.; Hamid Akash, M.S. Effect of Food Azo-Dye Tartrazine on Physiological Functions of Pancreas and Glucose Homeostasis. Turk. J. Biochem. 2019, 44, 197–206. [Google Scholar] [CrossRef]
  2. Leulescu, M.; Rotaru, A.; Pălărie, I.; Moanţă, A.; Cioateră, N.; Popescu, M.; Morîntale, E.; Bubulică, M.V.; Florian, G.; Hărăbor, A.; et al. Tartrazine: Physical, Thermal and Biophysical Properties of the Most Widely Employed Synthetic Yellow Food-Colouring Azo Dye. J. Therm. Anal. Calorim. 2018, 134, 209–231. [Google Scholar] [CrossRef]
  3. Chung, K.T. Azo Dyes and Human Health: A Review. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2016, 34, 233–261. [Google Scholar] [CrossRef] [PubMed]
  4. Ambroziewicz, Z.M.; Siemiątkowski, R.; Łata, M.; Dowgiert, S.; Sikorska, M.; Kamiński, J.; Więcław, K.; Grabowska, H.; Chruściel, J.; Mąsior, G. Long-Term Health Effects of Artificially Colored Foods in Adults and Children: A Review of Scientific Literature on Attention Deficits, Carcinogenicity, and Allergy Risks. J. Educ. Health Sport 2024, 76, 56522. [Google Scholar] [CrossRef]
  5. Kamat, P.; Shetti, P.P.; Paranjape, R.; Shetti, P.P. Techniques for Detection and Measurement of Tartrazine in Food Products 2008 to 2022: A Review. J. Chem. Health Risks 2024, 14, 721–729. Available online: https://jchr.org/index.php/JCHR/article/view/6813 (accessed on 17 July 2025).
  6. Mehedi, N.; Ainad-Tabet, S.; Mokrane, N.; Addou, S.; Zaoui, C.; Kheroua, O.; Saidi, D. Reproductive Toxicology of Tartrazine (FD and C Yellow No. 5) in Swiss Albino Mice. Am. J. Pharmacol. Toxicol. 2009, 4, 130–135. [Google Scholar] [CrossRef]
  7. Barciela, P.; Perez-Vazquez, A.; Prieto, M.A. Azo Dyes in the Food Industry: Features, Classification, Toxicity, Alternatives, and Regulation. Food Chem. Toxicol. 2023, 178, 113935. [Google Scholar] [CrossRef]
  8. Alshehrei, F. Role of Microorganisms in Biodegradation of Food Additive Azo Dyes: A Review. Afr. J. Biotechnol. 2020, 19, 799–805. [Google Scholar] [CrossRef]
  9. Aali, R.; Yari, A.R.; Ghafuri, Y.; Behnamipour, S. Health Risk Assessment of Synthetic Tartrazine Dye in Some Food Products in Qom Province (Iran). Curr. Nutr. Food Sci. 2024, 20, 726–733. [Google Scholar] [CrossRef]
  10. World Health Organization. JECFA Chemical Evaluation: Chemical ID 3885. 2016. Available online: https://apps.who.int/food-additives-contaminants-jecfa-database/Home/Chemical/3885 (accessed on 11 July 2025).
  11. Vander Leek, T.K. Food Additives and Reactions: Antioxidants, Benzoates, Parabens, Colorings, Flavorings, Natural Protein-Based Additives. Encycl. Food Allergy 2024, 1, 862–881. [Google Scholar] [CrossRef]
  12. Wróblewska, B. Influence of Food Additives and Contaminants (Nickel and Chromium) on Hypersensitivity and Other Adverse Health Reactions—A Review. Pol. J. Food Nutr. Sci. 2009, 59, 287–294. Available online: https://journal.pan.olsztyn.pl/pdf-98219-30948?filename=30948.pdf (accessed on 11 July 2025).
  13. Pestana, S.; Moreira, M.; Olej, B. Safety of Ingestion of Yellow Tartrazine by Double-Blind Placebo Controlled Challenge in 26 Atopic Adults. Allergol. Immunopathol. 2010, 38, 142–146. [Google Scholar] [CrossRef]
  14. Banc, R.; Filip, L.; Cozma-Petruț, A.; Ciobârcă, D.; Miere, D. Yellow and Red Synthetic Food Dyes and Potential Health Hazards: A Mini Review. Bull. Univ. Agric. Sci. Vet. Med. Cluj-Napoca Food Sci. Technol. 2024, 81, 1–17. [Google Scholar] [CrossRef]
  15. Bateman, B.J. The Behaviour of Three Year Olds in Relation to Allergy and Exposure to Artificial Additives. Doctoral Thesis, University of Southampton, Southampton, UK, 2004. Available online: https://eprints.soton.ac.uk/465625/ (accessed on 11 July 2025).
  16. Tuormaa, T.E. The Adverse Effects of Food Additives on Health: A Review of the Literature with Special Emphasis on Childhood Hyperactivity. J. Orthomol. Med. 1994, 9, 225–243. Available online: https://orthomolecular.org/library/jom/1994/articles/1994-v09n04-p225.shtml (accessed on 21 July 2025).
  17. Feketea, G.; Tsabouri, S. Common Food Colorants and Allergic Reactions in Children: Myth or Reality? Food Chem. 2017, 230, 578–588. [Google Scholar] [CrossRef]
  18. Arnold, L.E.; Lofthouse, N.; Hurt, E. Artificial Food Colors and Attention-Deficit/Hyperactivity Symptoms: Conclusions to Dye For. Neurotherapeutics 2012, 9, 599–609. [Google Scholar] [CrossRef]
  19. Silva, M.M.; Reboredo, F.H.; Lidon, F.C. Food Colour Additives: A Synoptical Overview on Their Chemical Properties, Applications in Food Products and Health Side Effects. Foods 2022, 11, 379. [Google Scholar] [CrossRef]
  20. Rowe, K.S.; Rowe, K.J. Synthetic Food Coloring and Behavior: A Dose Response Effect in a Double-Blind, Placebo-Controlled, Repeated-Measures Study. J. Pediatr. 1994, 125, 691–698. [Google Scholar] [CrossRef]
  21. Visweswaran, B. Oxidative Stress by Tartrazine in the Testis of Wistar Rats. IOSR J. Pharm. Biol. Sci. 2012, 2, 44–49. [Google Scholar] [CrossRef]
  22. El-Desoky, G.E.; Wabaidur, S.M.; Alothman, Z.A.; Habila, M.A. Regulatory Role of Nano-Curcumin against Tartrazine-Induced Oxidative Stress, Apoptosis-Related Genes Expression, and Genotoxicity in Rats. Molecules 2020, 25, 5801. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, L.; Lv, X.; Zhang, Y.; Xin, Q.; Zou, Y.; Li, X. Tartrazine Exposure Results in Histological Damage, Oxidative Stress, Immune Disorders and Gut Microbiota Dysbiosis in Juvenile Crucian Carp (Carassius carassius). Aquat. Toxicol. 2021, 241, 105998. [Google Scholar] [CrossRef]
  24. Wu, L.; Xu, Y.; Lv, X.; Chang, X.; Ma, X.; Tian, X.; Shi, X.; Li, X.; Kong, X. Impacts of an Azo Food Dye Tartrazine Uptake on Intestinal Barrier, Oxidative Stress, Inflammatory Response and Intestinal Microbiome in Crucian Carp (Carassius auratus). Ecotoxicol. Environ. Saf. 2021, 223, 112551. [Google Scholar] [CrossRef]
  25. Elhkim, M.O.; Héraud, F.; Bemrah, N.; Gauchard, F.; Lorino, T.; Lambré, C.; Frémy, J.M.; Poul, J.M. New Con-siderations Regarding the Risk Assessment on Tartrazine. An Update Toxicological Assessment, Intolerance Reactions and Maximum Theoretical Daily Intake in France. Regul. Toxicol. Pharmacol. 2007, 47, 308–316. [Google Scholar] [CrossRef] [PubMed]
  26. Khayyat, L.; Essawy, A.; Sorour, J.; Soffar, A. Tartrazine Induces Structural and Functional Aberrations and Genotoxic Effects in Vivo. PeerJ 2017, 5, e3041. [Google Scholar] [CrossRef] [PubMed]
  27. Tanaka, T. Reproductive and Neurobehavioural Toxicity Study of Tartrazine Administered to Mice in the Diet. Food Chem. Toxicol. 2006, 44, 179–187. [Google Scholar] [CrossRef] [PubMed]
  28. Mandelker, L. Oxidative Stress, Free Radicals, and Cellular Damage. In Studies in Veterinary Medicine; Humana Press: Totowa, NJ, USA, 2011; pp. 1–17. [Google Scholar] [CrossRef]
  29. Aldaamy, A.M.Z.; Al-Zubiady, N.M.H. Study on Toxic Effect of Tartrazine Pigment on Oxidative Stress in Male Albino Rats. Biochem. Cell Arch. 2021, 21, 1021–1026. Available online: https://connectjournals.com/03896.2021.21.1021 (accessed on 18 July 2025).
  30. El-Desoky, G.E.; Abdel-Ghaffar, A.; Al-Othman, Z.A.; Habila, M.A.; Al-Sheikh, Y.A.; Ghneim, H.K.; Giesy, J.P.; Aboul-Soud, M.A.M. Curcumin Protects against Tartrazine-Mediated Oxidative Stress and Hepatotoxicity in Male Rats. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 635–645. Available online: https://pubmed.ncbi.nlm.nih.gov/28239801/ (accessed on 18 July 2025).
  31. Ramos-Souza, C.; Nass, P.; Jacob-Lopes, E.; Zepka, L.Q.; Braga, A.R.C.; De Rosso, V.V. Changing Despicable Me: Potential Replacement of Azo Dye Yellow Tartrazine for Pequi Carotenoids Employing Ionic Liquids as High-Performance Extractors. Int. Food Res. J. 2023, 174, 113593. [Google Scholar] [CrossRef]
  32. Amchova, P.; Siska, F.; Ruda-Kucerova, J. Safety of Tartrazine in the Food Industry and Potential Protective Factors. Heliyon 2024, 10, e38111. [Google Scholar] [CrossRef]
  33. Lee, M.; Fondriest Gentry, A.; Schwartz, R.; Bauman, J. Tartrazine-Containing Drugs. Drug Intell. Clin. Pharm. 1981, 15, 782–788. [Google Scholar] [CrossRef]
  34. Miller, K. Sensitivity to Tartrazine. Br. Med. J. 1982, 285, 1597–1598. [Google Scholar] [CrossRef][Green Version]
  35. Pennington, C.R. Diet and Disease. In Therapeutic Nutrition; Springer: Boston, MA, USA, 1988; pp. 226–262. [Google Scholar] [CrossRef]
  36. Moutinho, I.L.D.; Bertges, L.C.; Assis, R.V.C. Prolonged Use of the Food Dye Tartrazine (FD&C Yellow N° 5) and Its Effects on the Gastric Mucosa of Wistar Rats. Braz. J. Biol. 2007, 67, 141–145. [Google Scholar] [CrossRef]
  37. Skypala, I. Other Causes of Food Hypersensitivity. In Food Hypersensitivity: Diagnosing and Managing Food Allergies and Intolerance; Blackwell Publisher: Hoboken, NJ, USA, 2009; Available online: https://onlinelibrary.wiley.com/doi/book/10.1002/9781444312119#page=219 (accessed on 18 July 2025).
  38. Bush, R.K.; Taylor, S.L. Reactions to Food and Drug Additives. In Middleton’s Allergy: Principles and Practice, 8th ed.; Mosby: Maryland Heights, MO, USA, 2014; Volume 2, pp. 1340–1356. [Google Scholar] [CrossRef]
  39. Breitkreutz, J.; Tuleu, C. Pediatric and Geriatric Pharmaceutics and Formulation. In Modern Pharmaceutics, 5th ed.; CRC Press: Boca Raton, FL, USA, 2016; Volume 2, Available online: https://www.taylorfrancis.com/chapters/edit/10.1201/b14445-32/pediatric-geriatric-pharmaceutics-formulation (accessed on 18 July 2025).
  40. Rovina, K.; Siddiquee, S.; Shaarani, S.M. A Review of Extraction and Analytical Methods for the Determination of Tartrazine (E 102) in Foodstuffs. Crit. Rev. Anal. Chem. 2017, 47, 309–324. [Google Scholar] [CrossRef]
  41. Sezgin, A.; Ayyildiz, S. Food Additives: Colorants. In Science Within Food: Up-to-Date Advance s on Research and Educational Ideas. Researchgate 2018, 122, 87–94. Available online: https://www.researchgate.net/publication/322404775_Food_Additives_Colorants (accessed on 28 July 2025).
  42. Sharma, R.; Bamola, S.; Kumar Verma, S. Effects of Acid Yellow 23 Food Dye on Environment and Its Removal on Various Surfaces-A Mini Review. Int. Res. J. Eng. Technol. 2020, 7, 4550–4573. [Google Scholar]
  43. Koop, B.L.; Maciel, A.G.; Soares, L.S.; Monteiro, A.R.; Valencia, G.A. Natural Colorants. In Natural Additives in Foods; Springer: Cham, Switzerland, 2022; pp. 87–122. Available online: https://link.springer.com/chapter/10.1007/978-3-031-17346-2_4 (accessed on 18 July 2025).
  44. Athira, N.; Jaya, D.S. Effects of Tartrazine on Growth and Brain Biochemistry of Indian Major Carps on Long-Term Exposure. Int. J. Adv. Biochem. Res. 2022, 6, 17–25. [Google Scholar] [CrossRef]
  45. Hegazy, A.A.; Awad Hegazy, A.; Haliem, W.A.; Haliem, R.A.; Mamdouh El-Bestawy, E.; Mohammad, G.; Ali, E. Brief Overview about Tartrazine Effects on Health. Eur. Chem. Bull. 2023, 12, 4698–4707. Available online: https://www.academia.edu/106433462/Brief_Overview_about_Tartrazine_Effects_on_Health (accessed on 14 June 2025).
  46. Food and Drug Administration. Title 21 Code of Federal Regulations. A Point Time in eCFR System. § 74.1705 FD&C Yellow No. 5. Available online: https://www.ecfr.gov/current/title-21/chapter-I/subchapter-A/part-74/subpart-B/section-74.1705?utm_source (accessed on 1 September 2025).
  47. Regulation (EC) No 1223/2009 of the European Parliament and of the Council of 30 November 2009 on Cosmetic Products (Recast) (Text with EEA Relevance). 2009. Available online: https://eur-lex.europa.eu/eli/reg/2009/1223/oj/eng?utm_source (accessed on 1 September 2025).
  48. Pay, R.; Sharrock, A.V.; Elder, R.; Maré, A.; Bracegirdle, J.; Torres, D.; Malone, N.; Vorster, J.; Kelly, L.; Ryan, A.; et al. Preparation, Analysis and Toxicity Characterisation of the Redox Metabolites of the Azo Food Dye Tartrazine. Food Chem. Toxicol. 2023, 182, 114193. [Google Scholar] [CrossRef]
  49. Kamal, A.A.; Fawzia, S.E.-S. Toxicological and Safety Assessment of Tartrazine as a Synthetic Food Additive on Health Biomarkers: A Review. Afr. J. Biotechnol. 2018, 17, 139–149. [Google Scholar] [CrossRef]
  50. dos Santos, J.R.; Soares, L.d.S.; Soares, B.M.; de Gomes Farias, M.; de Oliveira, V.A.; de Sousa, N.A.B.; Negreiros, H.A.; da Silva, F.C.C.; Peron, A.P.; Pacheco, A.C.L.; et al. Cytotoxic and mutagenic effects of the food additive tartrazine on eukaryotic cells. BMC Pharmacol. Toxicol. 2022, 23, 95. [Google Scholar] [CrossRef]
  51. Gao, Y.; Li, C.; Shen, J.; Yin, H.; An, X.; Jin, H. Effect of Food Azo Dye Tartrazine on Learning and Memory Functions in Mice and Rats, and the Possible Mechanisms Involved. J. Food Sci. 2011, 76, T125–T129. [Google Scholar] [CrossRef]
  52. McCann, D.; Barrett, A.; Cooper, A.; Crumpler, D.; Dalen, L.; Grimshaw, K.; Kitchin, E.; Lok, K.; Porteous, L.; Prince, E.; et al. Food Additives and Hyperactive Behaviour in 3-Year-Old and 8/9-Year-Old Children in the Community: A Randomised, Double-Blinded, Placebo-Controlled Trial. Lancet 2007, 370, 1560–1567. [Google Scholar] [CrossRef]
  53. Nettis, E.; Colanardi, M.C.; Ferrannini, A.; Tursi, A. Suspected Tartrazine-Induced Acute Urticar-ia/Angioedema Is Only Rarely Reproducible by Oral Rechallenge. Clin. Exp. Allergy 2003, 33, 1725–1729. [Google Scholar] [CrossRef] [PubMed]
  54. Araújo Caldas, L.; Caldas Marmo, F.; da Costa, P.V.; Viana Jacobson, L.S. Hormesis in Tartrazine Allergic Responses of Atopic Patients: An Overview of Clinical Trials and a Raw Data Revision. Environ. Dis. 2020, 5, 59. [Google Scholar] [CrossRef]
  55. Soares, B.M.; Araújo, T.M.T.; Ramos, J.A.B.; Pinto, L.C.; Khayat, B.M.; De Oliveira Bahia, M.; Montenegro, R.C.; Burbano, R.M.R.G.; Khayat, A.S. Effects on DNA Repair in Human Lymphocytes Exposed to the Food Dye Tartrazine Yellow. Anticancer. Res. 2015, 35, 1465–1474. Available online: https://pubmed.ncbi.nlm.nih.gov/25750299/ (accessed on 18 July 2025). [PubMed]
  56. Velioglu, C.; Erdemli, M.E.; Gul, M.; Erdemli, Z.; Zayman, E.; Bag, H.G.; Altinoz, E. Protective Effect of Crocin on Food Azo Dye Tartrazine-Induced Hepatic Damage by Improving Biochemical Parameters and Oxidative Stress Biomarkers in Rats. Gen. Physiol. Biophys. 2019, 38, 73–82. [Google Scholar] [CrossRef]
  57. Altinoz, E.; Erdemli, M.E.; Gül, M.; Erdemli, Z.; Gül, S.; Turkoz, Y. Prevention of Toxic Effects of Orally Administered Tartrazine by Crocin in Wistar Rats. Toxicol. Environ. Chem. 2021, 103, 184–198. [Google Scholar] [CrossRef]
  58. Erdemli, M.E.; Gul, M.; Altinoz, E.; Zayman, E.; Aksungur, Z.; Bag, H.G. The Protective Role of Crocin in Tartrazine Induced Nephrotoxicity in Wistar Rats. Biomed. Pharmacother. 2017, 96, 930–935. [Google Scholar] [CrossRef]
  59. Erdemli, Z.; Altinoz, E.; Erdemli, M.E.; Gul, M.; Bag, H.G.; Gul, S. Ameliorative Effects of Crocin on Tartrazine Dye–Induced Pancreatic Adverse Effects: A Biochemical and Histological Study. Environ. Sci. Pollut. Res. Int. 2021, 28, 2209–2218. [Google Scholar] [CrossRef]
  60. Aldaamy, A.; Merza Hamza, N.; Masikh Zebalah Al-Daamy, A.; Marza Hamza Al-Zubiady, N. Study of The Toxic Effect of Tartrazine Dye on Some Biochemical Parameters in Male Albino Rats. Sci. J. Med. Res. 2020, 4, 111–117. Available online: https://sjomr.org.in/index.php/SJOMR/article/view/134 (accessed on 18 July 2025).
  61. Abdelgayed, S.S. Toxicological and Histopathological Studies on the Effect of Tartrazine in Male Albino Rats. Int. J. Agric. Biol. Eng. 2016, 10, 469–474. Available online: https://www.researchgate.net/publication/309791778 (accessed on 7 June 2025).
  62. El Golli, N.; Bini-Dhouib, I.; Jrad, A.; Boudali, I.; Nasri, B.; Belhadjhmida, N.; El Fazaa, S. Toxicity Induced after Subchronic Administration of the Synthetic Food Dye Tartrazine in Adult Rats, Role of Oxidative Stress. Recent. Adv. Biol. Med. 2016, 2, 20. [Google Scholar] [CrossRef]
  63. Refai, H.M.; Mahmoud, M.A.A.; Ghuniem, M.M. Elevated Health Risks for Children and Adolescents: Quantification of Artificial Sweeteners and Synthetic Dyes in Powdered Beverages Reveals Regulatory Gaps. J. Food Compos. Anal. 2025, 148, 108239. [Google Scholar] [CrossRef]
  64. Scientific Opinion on the re-evaluation Tartrazine (E 102). EFSA J. 2009, 7, 1331. Available online: https://www.efsa.europa.eu/en/efsajournal/pub/1331 (accessed on 18 July 2025). [CrossRef]
  65. Rahnama, H.; Mazloomi, S.M.; Berizi, E.; Abbasi, A.; Gholami, Z. Identification of Tartrazine adulteration and evaluating exposure to synthetic dyes of sunset yellow and Quinoline yellow through consumption of food products among children. Food Sci. Nutr. 2022, 10, 3781–3788. [Google Scholar] [CrossRef]
  66. Miller, M.D.; Steinmaus, C.; Golub, M.S.; Castorina, R.; Thilakartne, R.; Bradman, A.; Marty, M.A. Potential impacts of synthetic food dyes on activity and attention in children: A review of the human and animal evidence. Environ. Health. 2022, 21, 45. [Google Scholar] [CrossRef]
  67. Boussada, M.; Lamine, J.A.; Bini, I.; Abidi, N.; Lasrem, M.; El-Fazaa, S.; El-Golli, N. Assessment of a Sub-Chronic Consumption of Tartrazine (E102) on Sperm and Oxidative Stress Features in Wistar Rat. Int. Food Res. J. 2017, 24, 1473–1481. [Google Scholar]
  68. Amin, K.A.; Abdel Hameid, H.; Abd Elsttar, A.H. Effect of Food Azo Dyes Tartrazine and Carmoisine on Bi-ochemical Parameters Related to Renal, Hepatic Function and Oxidative Stress Biomarkers in Young Male Rats. Food Chem. Toxicol. 2010, 48, 2994–2999. [Google Scholar] [CrossRef]
  69. Ozougwu, J.C. Physiology of the Liver. Indian J. Res. Pharm. Biotechnol. 2017, 4, 13–24. Available online: https://www.academia.edu/38598619/Physiology_of_the_liver?auto=download (accessed on 18 July 2025).
  70. Kiran, T.R.; Otlu, O.; Karabulut, A.B. Oxidative Stress and Antioxidants in Health and Disease. J. Lab. Med. 2023, 47, 1–11. [Google Scholar] [CrossRef]
  71. Albasher, G.; Maashi, N.; Alfarraj, S.; Almeer, R.; Albrahim, T.; Alotibi, F.; Bin-Jumah, M.; Mahmoud, A.M. Perinatal exposure to tartrazine triggers oxidative stress and neurobehavioral alterations in mice offspring. Antioxidants 2020, 9, 53. [Google Scholar] [CrossRef]
  72. Hosieny, N.A.; Mona, E.; Ahmed, S.M.; Zayed, M. Toxic Effects of Food Azo Dye Tartrazine on the Brain of Young Male Albino Rats: Role of Oxidative Stress. Zagazig J. Forensic Med. 2020, 19, 60–73. [Google Scholar] [CrossRef]
  73. Wopara, I.; Adebayo, O.G.; Umoren, E.B.; Aduema, W.; Iwueke, A.V.; Etim, O.E.; Pius, E.A.; James, W.B.; Wodo, J. Involvement of Striatal Oxido-Inflammatory, Nitrosative and Decreased Cholinergic Activity in Neurobehavioral Alteration in Adult Rat Model with Oral Co-Exposure to Erythrosine and Tartrazine. Heliyon 2021, 7, e08454. [Google Scholar] [CrossRef] [PubMed]
  74. Al-Seeni, M.N.; El Rabey, H.A.; Al-Hamed, A.M.; Zamazami, M.A. Nigella sativa Oil Protects against Tartrazine Toxicity in Male Rats. Toxicol. Rep. 2018, 5, 146–155. [Google Scholar] [CrossRef] [PubMed]
  75. Abd-Elhakim, Y.M.; Moustafa, G.G.; Hashem, M.M.; Ali, H.A.; Abo-EL-Sooud, K.; El-Metwally, A.E. Influence of the Long-Term Exposure to Tartrazine and Chlorophyll on the Fibrogenic Signalling Pathway in Liver and Kidney of Rats: The Expression Patterns of Collagen 1-α, TGFβ-1, Fibronectin, and Caspase-3 Genes. Environ. Sci. Pollut. Res. Int. 2019, 26, 12368–12378. [Google Scholar] [CrossRef] [PubMed]
  76. Haridevamuthu, B.; Murugan, R.; Seenivasan, B.; Meenatchi, R.; Pachaiappan, R.; Almutairi, B.O.; Arokiyaraj, S.; Kathiravan, M.K.; Arockiaraj, J. Synthetic Azo-Dye, Tartrazine Induces Neurodevelopmental Toxicity via Mitochondria-Mediated Apoptosis in Zebrafish Embryos. J. Hazard. Mater. 2024, 461, 132524. [Google Scholar] [CrossRef]
  77. Floriano, J.M.; da Rosa, E.; Amaral, Q.D.F.d.; Zuravski, L.; Chaves, P.E.E.; Machado, M.M.; de Oliveir, L.F.S. Is Tartrazine Really Safe? In Silico and Ex Vivo Toxicological Studies in Human Leukocytes: A Question of Dose. Toxicol. Res. 2018, 7, 1128–1134. [Google Scholar] [CrossRef]
  78. Pasdaran, A.; Azarpira, N.; Heidari, R.; Nourinejad, S.; Zare, M.; Hamedi, A. Effects of Some Cosmetic Dyes and Pigments on the Proliferation of Human Foreskin Fibroblasts and Cellular Oxidative Stress; Potential Cytotoxicity of Chlorophyllin and Indigo Carmine on Fibroblasts. J. Cosmet. Dermatol. 2022, 21, 3979–3985. [Google Scholar] [CrossRef]
  79. Nasri, A.; Pohjanvirta, R. In Vitro Estrogenic, Cytotoxic, and Genotoxic Profiles of the Xenoestrogens 8-Prenylnaringenine, Genistein and Tartrazine. Environ. Sci. Pollut. Res. 2021, 28, 27988–27997. [Google Scholar] [CrossRef]
  80. Haverić, A.; Inajetović, D.; Vareškić, A.; Hadžić, M.; Haverić, S. In vitro Analysis of Tartrazine Genotoxicity and Cytotoxicity. Genet. Appl. 2017, 1, 37–43. [Google Scholar] [CrossRef]
  81. Zand, A.; Macharia, J.M.; Szabó, I.; Gerencsér, G.; Molnár, Á.; Raposa, B.L.; Varjas, T. The Impact of Tartrazine on DNA Methylation, Histone Deacetylation, and Genomic Stability in Human Cell Lines. Nutrients 2025, 17, 913. [Google Scholar] [CrossRef]
  82. Joshi, V.; Katti, P. Developmental Toxicity Assay for Food Additive Tartrazine Using Zebrafish (Danio rerio) Embryo Cultures. Int. J. Toxicol. 2018, 37, 38–44. [Google Scholar] [CrossRef] [PubMed]
  83. Jiang, L.L.; Li, K.; Yan, D.L.; Yang, M.F.; Ma, L.; Xie, L.Z. Toxicity Assessment of 4 Azo Dyes in Zebrafish Embryos. Int. J. Toxicol. 2020, 39, 115–123. [Google Scholar] [CrossRef] [PubMed]
  84. Gupta, R.; Ranjan, S.; Yadav, A.; Verma, B.; Malhotra, K.; Madan, M.; Chopra, O.; Jain, S.; Gupta, S.; Joshi, A.; et al. Toxic Effects of Food Colorants Erythrosine and Tartrazine on Zebrafish Embryo Development. Curr. Res. Nutr. Food Sci. 2019, 7, 876–885. [Google Scholar] [CrossRef]
  85. Thanh, D.D.; Bich-Ngoc, N.; Paques, C.; Christian, A.; Herkenne, S.; Struman, I.; Muller, M. The Food Dye Tartrazine Disrupts Vascular Formation Both in Zebrafish Larvae and in Human Primary Endothelial Cells. Sci. Rep. 2024, 14, 30367. [Google Scholar] [CrossRef]
  86. Linskens, A. The Long Term Effects of Tartrazine (FD&C Yellow No. 5) on Learning, Cognitive Flexibility, and Memory of Zebrafish (Danio rerio) Embryos into Adulthood. Available online: https://cpb-us-w2.wpmucdn.com/sites.uwm.edu/dist/8/202/files/2018/06/Linskens_paper_LM_Seymour_2018-1lxiur8.pdf (accessed on 24 July 2025).
  87. Meena, G.; Meena, B. Evaluation of Possible Toxic Effect of Tartrazine Food dye on Swiss Albino Mice, and Histology of Testis. Int. J. Innov. Res. Multidiscip. Field 2020, 6, 100–105. Available online: https://www.researchgate.net/publication/353750136_Evaluation_of_Possible_Toxic_Effect_of_Tartrazine_Food_dye_on_Swiss_Albino_Mice_and_Histology_of_Testis (accessed on 9 September 2025).
  88. Arefin, S.; Hossain, M.S.; Neshe, S.A.; Rashid, M.O.; Amin, M.T.; Hussain, S. Tartrazine Induced Changes in Physiological and Biochemical Parameters in Swiss Albino Mice, Mus Musculus. Marmara Pharm. J. 2017, 21, 564–569. [Google Scholar] [CrossRef]
  89. Kamel, M.M.; El-Lethey, H.S. The Potential Health Hazard of Tartrazine and Levels of Hyperactivity, Anxiety-Like Symptoms, Depression and Anti-Social Behaviour in Rats. Am. J. Sci. 2011, 7, 1211–1218. Available online: https://www.scirp.org/reference/referencespapers?referenceid=3298281 (accessed on 24 July 2025).
  90. Usman, J.N.; Muhammad, G.A. Sub-acute toxicity study on tartrazine in male albino rats. Dutse J. Pure Appl. Sci. 2022, 8, 97–105. [Google Scholar] [CrossRef]
  91. El-Desoky, G.E.; Wabaidur, S.M.; AlOthman, Z.A.; Habila, M.A. Evaluation of Nano-Curcumin Effects against Tartrazine-Induced Abnormalities in Liver and Kidney Histology and Other Biochemical Parameters. Food Sci. Nutr. 2022, 10, 1344–1356. [Google Scholar] [CrossRef]
  92. Iroh, G.; Weli, B.O.; Adele, U.A.; Briggs, O.N.; Waribo, H.A.; Elekima, I. Assessment of Atherogenic Indices and Markers of Cardiac Injury in Albino Rats Orally Administered with Tartrazine Azo Dye. J. Adv. Med. Pharm. Sci. 2020, 22, 51–61. [Google Scholar] [CrossRef]
  93. Abd-Elhakim, Y.M.; Hashem, M.M.; El-Metwally, A.E.; Anwar, A.; Abo-EL-Sooud, K.; Moustafa, G.G.; Ali, H.A. Comparative Haemato-Immunotoxic Impacts of Long-Term Exposure to Tartrazine and Chlorophyll in Rats. Int. Immunopharmacol. 2018, 63, 145–154. [Google Scholar] [CrossRef] [PubMed]
  94. Balta, I.; Sevastre, B.; Mireşan, V.; Taulescu, M.; Raducu, C.; Longodor, A.L.; Marchiş, Z.; Mariş, C.S.; Coroian, A. Protective Effect of Blackthorn Fruits (Prunus spinosa) against Tartrazine Toxicity Development in Albino Wistar Rats. BMC Chem. 2019, 13, 104. [Google Scholar] [CrossRef]
  95. Himri, I.; Bellahcen, S.; Souna, F.; Belmakki, F.; Aziz, M.; Bnouham, M.; Zoheir, J.; Berkia, Z.; Mekhfi, H.; Saalaoui, E. A 90-Day Oral Toxicity Study of Tartrazine, a Synthetic Food Dye, in Wistar Rats. Int. J. Pharm. Pharm. Sci. 2011, 3, 159–169. Available online: https://www.scirp.org/journal/paperinformation?paperid=20481 (accessed on 15 July 2025).
  96. El-Sakhawy, M.A.; Mohamed, D.W.; Ahmed, Y.H. Histological and Immunohistochemical Evaluation of the Effect of Tartrazine on the Cerebellum, Submandibular Glands, and Kidneys of Adult Male Albino Rats. Environ. Sci. Pollut. Res. Int. 2019, 26, 9574–9584. [Google Scholar] [CrossRef]
  97. Abd, B.E.; Naby, E.; Shalaby, R.A.; Fouda, F.M.; Ebiya, R.A. Evaluation of biochemical effects of tartrazine and curcumin in male albino rats. World J. Pharm. Res. 2022, 11, 1472–1486. [Google Scholar] [CrossRef]
  98. Boroumand, N.; Samarghandian, S.; Isaac Hashemy, S. Immunomodulatory, anti-inflammatory, and antioxidant effects of curcumin. J. Herbmed. Pharmacol. 2018, 7, 211–219. [Google Scholar] [CrossRef]
  99. Jurenka, J.S. Anti-inflammatory Properties of Curcumin, a Major Constituent of Curcuma longa: A Review of Preclinical and Clinical Research. Altern. Med. Rev. 2009, 14, 141–153. Available online: https://pubmed.ncbi.nlm.nih.gov/19594223/ (accessed on 15 July 2025).
  100. Peng, Y.; Ao, M.; Dong, B.; Jiang, Y.; Yu, L.; Chen, Z.; Hu, C.; Xu, R. Anti-inflammatory effects of curcumin in the inflammatory diseases: Status, limitations and countermeasures. Drug Des. Dev. Ther. 2021, 15, 4503–4525. [Google Scholar] [CrossRef]
  101. Ciccone, M.M.; Cortese, F.; Gesualdo, M.; Carbonara, S.; Zito, A.; Ricci, G.; De Pascalis, F.; Scicchitano, P.; Riccioni, G. Dietary Intake of Carotenoids and Their Antioxidant and Anti-Inflammatory Effects in Cardiovascular Care. Mediat. Inflamm. 2013, 2013, 782137. [Google Scholar] [CrossRef] [PubMed]
  102. Wu, S.; Chen, R.; Chen, J.; Yang, N.; Li, K.; Zhang, Z.; Zhang, R. Study of the Anti-Inflammatory Mechanism of β-Carotene Based on Network Pharmacology. Molecules 2023, 28, 7540. [Google Scholar] [CrossRef]
Figure 1. Overview of biological responses linked to Tartrazine Exposure (Figure partially created using BioRender resources (https://www.biorender.com/)).
Figure 1. Overview of biological responses linked to Tartrazine Exposure (Figure partially created using BioRender resources (https://www.biorender.com/)).
Toxics 13 00771 g001
Table 1. Common Uses of Tartrazine and Relevant Toxicological Concerns.
Table 1. Common Uses of Tartrazine and Relevant Toxicological Concerns.
CategoryProduct ExamplesReferences
Food productsTZ is widely used to impart an intense yellow color in various food products such as bread, beverages, cereals, peanuts, candies, jellies, chewing gum, flavored chips, creams, ice cream, yogurts, cakes, instant desserts, soups, sauces, jams, flavored rice, and pasta. Due to potential adverse effects, there is a growing tendency to replace it with natural pigments like annatto or β-carotene.[2,35,37,40,41,42,43,45]
PharmaceuticalsTZ is used as a coloring excipient in multivitamins, gelatin capsules, tablets, syrups, and pediatric medicines. In sensitive individuals, it may cause allergic reactions or asthma. [33,36,38,39,40]
Non-food

products
TZ is also present in non-food products such as soaps, cosmetics, shampoos, hair conditioners, pastels, crayons, and stamp dyes. Skin contact may cause hypersensitivity reactions.[25,34,44,45]
TZ—Tartrazine.
Table 2. Evidence of Oxidative Imbalance Following Tartrazine Exposure in Rodent Models.
Table 2. Evidence of Oxidative Imbalance Following Tartrazine Exposure in Rodent Models.
Experimental Organismn=Species/StrainAge/Weight DoseTimeMethod of AdministrationSampleEffectRef.
Mice60Swiss white mice25–30 g2.5 and 5 mg/kgday one of pregnancy to day 15 after birth.oral gavageBrain

tissue
↑ MDA
↓ GSH
↓ SOD
[71]
Rats18Young male albino rats28 days, 60–80 g320 mg/kg tartrazine in 1 mL distilled water, daily4 weeksoral gavageBrain↓ GPx
↑ MDA
[72]
50Wistar male albino rats180 and 200 g7.5 mg/kg90 daysdiets containing dry massLiver↑ MDA
↓ GSH
[30]
Serum↓ SOD
↓ CAT
↓ GPx
24Male Wistar rats10–12 weeks, 180–200 g2, 6, 10 mg/kg (erythrosine + TZ 50:50 mix)6 weeksoral gavageBrain tissue↑ MDA
↓ GSH
↓ CAT
↑ ACHe
[73]
18Male albino rats175–185 g10 mg/kg (+3.75 mg/kg sulfanilic acid)8 weeksoral administrationSerum, liver, and kidney tissue homogenate↑ MDA
↓ GSH
↓ SOD
↓ CAT
↓ GR
[74]
18White albino rats -Low (10 mg/kg) and high (50 mg/kg) doses15 and 30 daysoral administrationSerum↓ SOD[1]
30Sprague–Dawley male albino rats150–200 g75 mg/kg90 daysoral administration by orogastric gavageHepatic and renal tissue homogenate↑ MDA
↓ GSH
↓ SOD
↓ CAT
[75]
18Male albino rats10–15 weeks,
190–250 g
400 mg/kg30 daysoral administrationSerum↑ MDA
↓ GSH
↓ SOD
↓ CAT
↓ GPx
[29]
40Female Wistar albino rats225–250 g500 mg/kg21 daysoral gavageTissue homogen
ates
↑ MDA
↑ SOD
↑ TOS
↓ GSH
↓ CAT
↓ TAS
[56]
40Adult female Wistar rats225–250 g, 8–10 weeks500 mg/kg3 weeksoral gavageTissue homogenates↑ MDA
↑ TOS
↓ GSH
↓ SOD
↓ CAT
↓ TAS
[57]
30Adult male Sprague–Dawley rats120–150 g200 mg/kg60 daysoral administrationTissue homogenate↑ MDA[61]
40Female Wistar rats225–250 g500 mg/kg3 weeksoral gavageTissue homogenate↑ MDA
↑ TOS
↓ GSH
↓ TAS
↓ SOD
↓ CAT
[59]
20Male Wistar rats 130 ± 40 g300 mg30 daysoral administrationTissue homogenate↑ MDA
↑ CAT
↑ GST
[62]
36Young male albino rats60–80 glow doses of TZ 15 mg/kg bw30 daysoral administrationLiver tissue homogenate↑ MDA
↓ CAT
↓ SOD
[68]
high doses were 500 mg/kg bw↑ MDA
↓ CAT
↓ SOD
↓ GSH
40Sprague–Dawley rats70 ± 10 g0, 175, 350, and 700 mg/kg bw30 daysoral gavageBrain tissue↓ GSH
↓ SOD
↑ MDA
[51]
ACHe—Acetylcholinesterase; bw—Body Weight; CAT—Catalase; GPx—Glutathione peroxidase; GR—Glutathione reductase; GSH—Glutathione; GST—Glutathione S-transferase; MDA—Malondialdehyde; SOD—Superoxide dismutase; TAS—Total Antioxidant Status; TOS—Total Oxidant Status; TZ—Tartrazine, ↑—significantly increased; ↓—significantly decreased.
Table 3. Summary of Experimental Studies Assessing the Cytotoxic and Genotoxic Effects of Tartrazine in Different Human Cell Types and Assays.
Table 3. Summary of Experimental Studies Assessing the Cytotoxic and Genotoxic Effects of Tartrazine in Different Human Cell Types and Assays.
Cell TypeConcentration TestedTests PerformedKey FindingsRef.
Human lymphocytes0.25–64.0 mMMTT assay, alkaline comet assayNo cytotoxicity; genotoxic at all doses; partial DNA repair.[55]
Human leukocytes5–500 μg/mLTrypan Blue viability, Micronucleus test, Comet assay, Cytogenetics, In silicoNo cytotoxicity/mutagenicity; DNA damage at ≥70 μg/mL; supported by in silico models.[77]
Human foreskin fibroblasts10, 100, 250, 500, 1000, and 2000 μg/mL for various dyesMTT assay, ROS, lipid peroxidation, LDHTZ: no effect; indigo carmine/chlorophyllin cytotoxic at high doses[78]
Yeast assay, MCF-7 breast cancer cellsNot specified (short-term)Estrogenic activity, LDH release, micronucleus test8-PN showed the strongest estrogenic effect, followed by TZ and genistein; all exhibited low cytotoxicity and no genotoxicity.[79]
Human lymphocytes, GR-M melanoma cells2.5, 5, and 10 mMChromosome aberration, CBMN assay, trypan blue testNo genotoxicity in lymphocytes; low cytotoxicity in lymphocytes; high cytotoxicity in melanoma cells[80]
HaCaT20 µM, 40 µM, and 80 µMqRT-PCR
Alkaline Comet Assay
Upregulated DNMT and HDAC genes with increased DNA fragmentation, indicating epigenetic and genotoxic effects.[81]
HepG2
A549
8-PN—8-Prenylnaringenin; CBMN assay—Cytokinesis-Block Micronucleus Assay; DNA—Deoxyribonucleic Acid; DNMT—DNA Methyltransferase; HaCaT—immortalized human keratinocyte; HDAC—Histone Deacetylase; HepG2—Human liver cancer cell line; LDH—Lactate Dehydrogenase; MTT—Methyl Thiazolyl Tetrazolium; qRT-PCR—Quantitative Real-Time Polymerase Chain Reaction; ROS—reactive oxygen species; TZ—Tartrazine.
Table 4. Effects of Tartrazine on Behavior and Biochemistry in Animal Models.
Table 4. Effects of Tartrazine on Behavior and Biochemistry in Animal Models.
Experimental ModelNumber (n=)Method of Administration TimeDoseAnalysisEffectRef.
Zebrafish
Zebrafish embryo280 (20/concentration)Exposed to E3 medium with varying TZ concentrations in Petri dishes24, 48, 72, 96, 120, 144, 168 hpf0, 0.1, 1, 2, 3, 4, 5, 10, 20, 30, 40, 50, 75, 100 mMDevelopmental anomalies (heart rate, edema, tail distortion, hatching, mortality) observed via bright field microscopy.Control embryos hatched normally; ≥10 mM caused early hatching with deformities and ≥40 mM increased mortality.[82]
25 embryos/well Embryos exposed in 6-well plates with E3 medium supplemented with TZ3–4 h post-fertilization to 4 dpf0, 5, 10, 20, 50 g/LZebrafish embryo toxicity and vascular defects.Dose-dependent vascular defects: hemorrhage, edema, small eye, vessel abnormalities.[85]
20 embryos/wellExposed in E3 medium72 h (hpf)5–100 mM (various concentrations)Developmental and cardiac toxicity parametersTZ caused dose-dependent drops in survival, hatching, cardiac/yolk sac edema, spinal defects, and heart rate.[83]
9Exposure via aquatic media6 months to a year22 μM Behavioral tests: T-maze test, cognitive flexibility, memory, learning, perseverance, consistency in choices.Learning, memory, and flexibility impaired
task completion and perseverance reduced.
[86]
100Exposed to varying erythrosine and TZ levels in embryo water.Up to 10 dpfErythrosine: 0.001–0.1%; TZ: 0.01–0.5%Biochemical and genetic analysesHigh TZ (≥0.5%) boosted hatching (55% at 48 hpf, 100% at 72 hpf) and triggered SOD1 expression via OS.[84]
Mice
KunMing mouse (20 ± 2 g)40Oral gavage30 days0, 175, 350, and mg/kg body massBehavioral (Step-through, Morris maze) and biochemical testsTZ negatively affects learning and memory in mice, increasing escape time and reducing reaction time in tests.[51]
Male Swiss albino mice (4 weeks)15Oral administration72 days100 and 200 mg/kgHistological analysesIncreased bw, mild deformation of seminiferous tubules, moderate reduction of Leydig and Sertoli cells, fewer spermatozoa[87]
Swiss albino mice
(25–30 g)
15Oral administration25 days200 mg/kg Physiological and biochemical analysesInsignificant decrease in the cholesterol level, no significant in triglyceride, significantly increased bilirubin and creatinine[88]
400 mg/kgInsignificant decrease in the cholesterol level, significant increase in triglycerides, bilirubin, and creatinine
Rats
Sprague–Dawley rats (70 ±
10 g)
40Oral gavage30 days175, 350, 700 mg/kg body massBehavioral tests: Open-field test.
Biochemical analyses
TZ increases activity and anxiety in rats, also causing histopathological changes in the brain.[51]
Male Wistar rats (40–50 g)45Dissolved in tap drinking water16 weeks0%, 1% (low dose) and 2.5% (high dose)Behavioral tests: Open field behaviour test
Elevated plus maze test
Light-Dark transition task
Forced swim test
Social interaction test
The study highlights the harmful effects of TZ on anxiety and depression, highlighting the risks of long-term exposure to food dyes on mental health.[89]
Young male albino rats (28 days old, 60–80 g)18Oral gavage4 weeks320 mg/kg TZ in 1 mL distilled water, once daily.Neurobiological and histological analysis:
Brains were harvested and analyzed for histological changes.
TZ has a neurotoxic effect, evidenced by histological changes such as neuronal apoptosis and vascular congestion.[72]
White albino rats of either sex18Oral administration15 and 30 daysLow dose: 10 mg/kg
High dose: 50 mg/kg
Biochemical, hormonal, and histological analysesTZ disrupts glucose balance, damages pancreas, alters endocrine function.
Increases glucose, lipase decreases insulin, Ca, Mg
[1]
Male albino rats18Oral administration8 weeks10 mg/kg (+3.75 mg/kg sulfanilic acid)Biochemical and histological It caused liver and kidney dysfunction with lesions.
Increased cholesterol, triglycerides, LDL, VLDL, ALT, AST, ALP, bilirubin, creatinine, urea, uric acid.
Decreased HDL, total protein.
[74]
Wistar male albino rats50Diets containing dry mass90 days7.5 mg/kgBiochemical and histological analysesTZ raised lipids, liver enzymes, kidney function.
Increased total cholesterol, triglycerides, LDL, ALT, AST, ALP, LDH.
[30]
Female Wistar albino rats (225–250 g)40Oral gavage21 days500 mg/kgBiochemical analyses and histopathological examinationsIncreased AST, ALT, ALP indicating liver damage.[56]
Male albino rats (65–80 g)12Oral administration7 weeks7.5 and 75 mg/kgBiochemical and histopathological analysesStudy showed harmful lipids, biochemical changes, and liver/kidney damage.
Increased cholesterol, triglycerides, LDL, VLDL, ALT, AST, ALP, creatinine, urea, uric acid.
[90]
Male Wistar albino rats (200–250 g)40Oral administration50 days7.5 mg/kgBiochemical and histopathological analysesTZ impaired liver/kidney, altered histology, lipids, glucose.
Increased ALT, AST, ALP, GGT, urea, uric acid, creatinine, protein, cholesterol, triglycerides, LDL
decreased HDL.
[91]
Adult female Wistar rats (225–250 g, 8–10 weeks old)40Oral gavage3 weeks500 mg/kgBiochemical and histopathological analysesTZ caused degenerative and metaplastic changes in ileum and colon epithelium.[57]
Wistar albino rats (146–153 g)20Dissolved in 1 mL of distilled water30 days7.5 mg/kg bwBiochemical, histological, and ultrastructural analysesTZ raised AST, ALT, ALP, uric acid, urea, creatinine, reduced antioxidants, and caused liver and kidney damage.[26]
Male Wistar rats (10–12 weeks old, 180–200 g)24Oral gavage6 weeks2, 6, 10 mg/kg (50:50 erythrosine-TZ)Behavioral (open field test, forced swimming test, tail suspension test), biochemical and enzymatic analyses Increased nitrite, TNF-α worsened anxiety and depression.[73]
Sprague–Dawley male albino rats (150–200 g)30Oral administration by orogastric gavage90 days75 mg/kgBiochemical, genetic, immunohistochemical, histology analysesIncreased AST, ALT, urea, creatinine
liver and kidney damage.
[75]
Female Wistar albino rats (225–250 g)40Oral gavage21 days500 mg/kgBiochemical and histopathological analysesTZ caused kidney glomerular collapse, inflammation, congestion.[58]
Albino rats (~0.2 kg)63Oral administration30 and 60 days7.5 mg/kgBiochemical and histopathological analysesTZ damages heart, raises nHDL and creatine kinase, increasing cardiovascular risk.[92]
Male rats (10–15 weeks old, 190–250 g)18Oral administration30 days400 mg/kgBiochemical analysesIncreased ALT, AST, ALP, urea, uric acid, creatinine decreased Na, K, Ca.[60]
Adult male Sprague–Dawley albino rats30Oral administration90 days1.35 mg/kgHematological, immunological, and histopathological analysesDecreased hemoglobin, RBC, PCV%, platelets
increased WBC, neutrophils, lymphocytes, monocytes.
[93]
Female Wistar rats (225–250 g)40Oral gavage3 weeks500 mg/kgBiochemical and histopathological analysesIncreased total cholesterol, glucose, triglycerides, LDL, VLDL
decreased HDL.
[59]
Albino Wistar rats20Oral administration7 weeks75 mg/250 mL water
100 mg/250 mL water
Biochemical, hematological, and histopathological analysesTZ damaged liver, kidneys, spleen
no change in cholesterol, triglycerides, ALT.
Increased AST, creatinine, WBC, neutrophils, lymphocytes.
[94]
Adult male Sprague–Dawley rats (120–150 g)30Oral administration60 days200 mg/kgBiochemical, histological, and physiological analysesSubchronic TZ affects liver and kidney parameters and induces OS.
Increased ALT, AST, urea, total protein.
[61]
Male and female Wistar rats (170–200 g)30Oral administration13 weeks5 mg/kgHematological and histopathological analysesNo effect[95]
7.5 mg/kgDecreased platelets
increased neutrophils, basophils, and mean platelet volume.
10 mg/kgNo effect
Adult male albino rats (120–150 g)40Oral gavage30 days7.5 mg/kg bwHistopathological and Immunohistochemical analysesTZ causes structural damage in cerebellum, glands, kidneys, with edema, congestion, neuron vacuolization, and cell deformation.[96]
15 mg/kg bwEdema, dilated perineural spaces, and degenerating Purkinje cells.
100 mg/kg bwSevere Purkinje cell degeneration, gray matter vacuolization, edema, nuclear pyknosis, vessel engorgement, increased astrocytes
Male Wistar rats (130 ± 40 g)20Oral administration30 days300 mgBiochemical and histopathological analyses Increased transaminases, LDH, creatinine, uric acid, kidney proteins
decreased total protein, albumin, globulin
HDL unchanged.
[62]
Young male albino rats (Rattus norvegicus), 60–80 g36Oral administration30 daysLow dose: 15 mg/kg bwBiochemical analysesIncreased ALT, AST, ALP, total protein, albumin, globulin, creatinine, urea
decreased serum cholesterol.
[68]
High dose: 500 mg/kg bwIncreased ALP, total protein, albumin, creatinine, urea
ALP—Alkaline Phosphatase; ALT—Alanine Aminotransferase; AST—Aspartate Aminotransferase; bw—Body weight; Ca—Calcium; dpf—Days Post Fertilization, GGT—Gamma-Glutamyl Transferase; HDL—High-Density Lipoprotein; hpf—Hours Post Fertilization; LDH—Lactate Dehydrogenase; LDL—Low-Density Lipoprotein; MPV—Mean Platelet Volume; Na—Sodium; OS—Oxidative Stress; PCV%—Packed Cell Volume Percentage (hematocrit); RBC—Red Blood Cells; SOD1—Superoxide Dismutase 1; TNF-α—Tumor Necrosis Factor-alpha; TZ—Tartrazine; VLDL—Very Low-Density Lipoprotein; WBC—White Blood Cells.
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

Visternicu, M.; Săvucă, A.; Rarinca, V.; Burlui, V.; Plavan, G.; Ionescu, C.; Ciobica, A.; Balmus, I.-M.; Albert, C.; Hogas, M. Toxicological Effects of Tartrazine Exposure: A Review of In Vitro and Animal Studies with Human Health Implications. Toxics 2025, 13, 771. https://doi.org/10.3390/toxics13090771

AMA Style

Visternicu M, Săvucă A, Rarinca V, Burlui V, Plavan G, Ionescu C, Ciobica A, Balmus I-M, Albert C, Hogas M. Toxicological Effects of Tartrazine Exposure: A Review of In Vitro and Animal Studies with Human Health Implications. Toxics. 2025; 13(9):771. https://doi.org/10.3390/toxics13090771

Chicago/Turabian Style

Visternicu, Malina, Alexandra Săvucă, Viorica Rarinca, Vasile Burlui, Gabriel Plavan, Cătălina Ionescu, Alin Ciobica, Ioana-Miruna Balmus, Cristina Albert, and Mihai Hogas. 2025. "Toxicological Effects of Tartrazine Exposure: A Review of In Vitro and Animal Studies with Human Health Implications" Toxics 13, no. 9: 771. https://doi.org/10.3390/toxics13090771

APA Style

Visternicu, M., Săvucă, A., Rarinca, V., Burlui, V., Plavan, G., Ionescu, C., Ciobica, A., Balmus, I.-M., Albert, C., & Hogas, M. (2025). Toxicological Effects of Tartrazine Exposure: A Review of In Vitro and Animal Studies with Human Health Implications. Toxics, 13(9), 771. https://doi.org/10.3390/toxics13090771

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