Open access peer-reviewed chapter

Cytotoxicity as a Fundamental Response to Xenobiotics

Written By

Grethel León-Mejía, Alvaro Miranda Guevara, Ornella Fiorillo Moreno and Carolina Uribe Cruz

Submitted: 01 October 2020 Reviewed: 27 January 2021 Published: 17 February 2021

DOI: 10.5772/intechopen.96239

From the Edited Volume

Cytotoxicity - New Insights into Toxic Assessment

Edited by Sonia Soloneski and Marcelo L. Larramendy

Chapter metrics overview

464 Chapter Downloads

View Full Metrics

Abstract

Cytotoxicity refers to the ability of a molecule or a compound to cause some type of cellular damage, of which some of the adverse effects that can occur include injuries to some structures or the fundamental processes involved in cell maintenance, such as survival, cell division, cell biochemistry, and the normal cell physiology. The potential for cytotoxicity is one of the first tests that must be performed to determine the effects of drugs, biomolecules, nanomaterials, medical devices, pesticides, heavy metals, and solvents, among others. This potential may be oriented in the mechanism under which it generates cell death, the dose, and the target cells that generate the response. The evaluation of the toxicologic and cytotoxic properties of the chemical substances through in vitro tests has become a competitive alternative to in vivo experimentation as a consequence of ethical considerations. Presently, there are numerous tests conducted to evaluate the cytotoxicity of a certain agent, the selection of which depends on the purpose of the study. In this sense, the present review provides a general overview of the different responses of a cell to xenobiotic agents and the different test that can be useful for evaluation of these responses.

Keywords

  • cytotoxicity
  • xenobiotic
  • apoptosis
  • biomarkers
  • cellular damage

1. Introduction

In the modern world, increasing industrialization continues to pose serious pollution problems [1]. Every year, several countries generate millions of tons of pollutants, which keeps adding to this interrelation among population, technology, resource consumption, and the environment—a situation which is becoming increasingly complex [2]. Although it is true that the reality is alarming, risk quantification and estimation strategies, based on studies of the effects of xenobiotics, are necessary not only for the conservation of the environment but also to acquire the knowledge about the factors that intervene in each specific case to enable foreseeing potential injuries [1]. Humans are exposed to a wide variety of foreign chemical substances, which we collectively call “xenobiotics” that includes natural compounds present in plant foods, such as synthetic compounds in medicines, food additives, and environmental pollutants. At present, the study on the effects of xenobiotics and the elucidation of their mechanisms of action on macromolecules has become extremely important [3, 4].

Cytotoxicity is defined as an alteration of the basic cellular functions that leads to detectable damage [5]. In this sense, the cytotoxicity that a xenobiotic induces may be a relevant point in the elucidation of the mechanism of action of the xenobiotic [6]. The analysis of cytotoxic effects is a fundamental strategy involved in the analysis of the xenobiotic–cell interaction in basic research, industrial development, and in the evaluation of therapeutic and toxic effects of chemical and biological products [5].

Advertisement

2. Cytotoxicity and genotoxicity

Genotoxicity is defined as the capacity of an agent, be it physical, chemical, or biological, to cause damage to the genetic materials or to alter the cellular components that influences the functionality and behavior of the chromosomes within the cell, leading to adverse biological outcomes [7]. Hence, the final cause of cell death may be related to DNA damage [8]. Specific cellular responses include cell cycle arrest and attempts to repair DNA [9]. The use of a specific repair enzyme complex depends on the type of DNA strand break or the chemistry of the adduct formed as well as the repair capacity of the affected cells [9]. If there is excessive DNA damage, an alternative is the action of p53 that activate apoptosis [8].

Apoptosis

One of the most obvious end points of action for several drugs and toxic xenobiotics is cell death [10]. Cell death is divided into two types: i) necrosis, which is characterized by the occurrence under the mechanisms of irreversible cell injury and is considered accidental and ii) apoptosis, which corresponds to programmed cell death that runs under control and is related to homeostasis of tissue growth [11]. The term programmed cell death was introduced for the first time in 1920, and, in 1972, the term apoptosis was coined by Wyllie and Currie, since then the mechanisms and molecular aspects by which this process is conducted have been described [12].

The extrinsic signaling pathway that initiates apoptosis has been so named since it involves interactions mediated by transmembrane receptors, and it has been described that this pathway is initiated by the binding of i) tumor necrosis factor (TNF) ligand to the receptor TNF, ii) TNF-related apoptosis inducing ligand (TRAIL) to death receptor-4 (DR4), and DR5 receptors, or 3) the fatty acid synthetase ligand to the FasR receptor. These associations are known to recruit adapter molecules such as Fas-associated death domain (FADD) or TNF receptor-associated death domain (TRADD), which activates initiator caspases-8 and -10 and, finally, the activation of executor caspases-3, −6, and − 7 that culminates in an apoptotic cell phenotype with characteristic physiological and morphological characteristics [11, 13].

In the intrinsic signaling cascade, a series of intracellular stimuli has been reported specifically in the mitochondria, which induces structural changes in the mitochondrial membrane, mainly due to the opening of the transition pores and alterations in the transmembrane potential entailing a release toward the cytosol of pro-apoptotic substances, which remains within the intermembranal space in a normal state [14]. These released components have been classified into two main groups consisting i) cytochrome c, Smac/diablo (second mitochondrial activator of caspases/direct inhibitors of apoptotic proteins (IAP) binding protein with low Propidium iodide (PI)) and the serine protease HtrA2/Omi (high-temperature requirement): these releases lead the apoptotic cascade via caspase activation. It has been discovered that the release of cytochrome c activates the apoptotic protease activating factor-1 (Apaf-1 protein) and procaspase-9 in addition to ATP, thereby establishing a protein complex called as the apoptosome, which in turn activates caspase-3, initiating the effector pathway of apoptosis. On the other hand, the Smac/Diablo and HtrA2/Om proteins promote apoptosis by inhibiting IAP such as cIAP1, cIAP2, and XIAP. ii) Apoptosis inducing factors, endonuclease G, and caspase-activated DNase: the latter are released into the cytosol, enter the nucleus, and fragment the DNA. The importance of DNA degradation by Ca2+ and Mg2+ dependent endonucleases is that they generate fragments of 180–200 base pairs; this pattern of fragments is highly specific and an extremely clear factor that differentiates this type of programmed cell death with necrosis, which does not present a degradation pattern or specificity in the fragment sizes [11, 13, 14].

Autophagy

Autophagy is an intracellular degradation process that is characterized by the formation of double membrane vesicles called as autophagosomes; these vesicles sequester the cytoplasmic material and later fuse with the lysosome (that contains hydrolytic enzymes), forming the autophagolysosome or autolysosome, where the degradation of the invaginated material occurs [15]. The amino acids and small molecules that are generated through autophagy are returned to the cytoplasm for the generation of energy and for the synthesis of new proteins and biomolecules [16]. The main inducer of autophagy is nutrient deficiency; however, it has been reported that the activation of autophagy is a cell survival mechanism against various stress conditions, including oxidative stress, inflammation, protein aggregation, endoplasmic reticulum stress, metabolic stress, the presence of pathogens, and changes in the mitochondrial function [17]. Autophagy plays an important role in cell and tissue homeostasis by contributing to the generation of energy from degradation events, which plays a role in the quality control of proteins and organelles, in the elimination of long-lived proteins and pathogens, as well as in the regulation of cell death [15, 16].

The central machinery of the autophagy process is composed of >30 proteins, including the so-called Atg. The autophagy pathway proceeds through five phases: (i) nucleation, which is the formation of a double membrane structure or an isolating membrane called the “phagophore”; (ii) the expansion of the phagophore membrane by the incorporation of the LC3-II protein; (iii) the maturation of this structure in the autophagosome and the sequestration of cytoplasmic material to be degraded; (iv) the fusion of the autophagosome with the lysosomes, which results in the formation of autophagosome and autolysosomes and, finally; (v) the degradation of biological materials sequestered by the hydrolytic enzymes of the lysosome and the recycling of the molecules (above all amino acids, lipids, sugars, and nucleotides) [17, 18].

The nucleation and formation of the phagophore is initiated by the serine and threonine kinase unc-51-like autophagy activating kinase 1 (ULK1). Once the ULK1 complex is activated, it activates the phosphatidylinositol 3-kinase (PtdIns3K) complex that includes members such as Beclin 1, Atg14, Vps15, Vps34, and Ambra1. Two ubiquitin-like conjugation systems participate in the expansion of the phagophore membrane, until the closure of the double membrane vesicle to form the mature autophagosome, which are Atg12-Atg5-Atg16 and Atg4-Atg7-Atg3/LC3-PE (phosphatidylethanolamine) [17, 18]. Eventually, the autophagosome fuses with the lysosome, and the sequestered material is degraded by lysosomal enzymes (such as cathepsins, glucosidases, lipases, and sulphatases). The components of degraded biomolecules, for example amino acids, are returned to the cytoplasm to derive energy and for the synthesis of new biomolecules [17, 18].

Advertisement

3. Cytotoxicity test

Within the battery of in vitro tests that are useful and necessary, alternative toxicology methods for the registration or application of clinical trials of a given substance are referred to as cytotoxicity tests; these tests are capable of detecting, through different known cellular mechanisms, the adverse effects of interference with structure and/or properties essential for cell survival, proliferation, and/or functions [19, 20]. These tests include the integrity of the membrane and the cytoskeleton, metabolism, synthesis, degradation, release of cellular constituents or products, ionic regulation, and cell division [19, 20].

The sensitivity and speed of the damage analysis at the cellular level increases its practical value when simple cytotoxicity markers are used, such as in the determination of viability by exclusion of fluorescent and non-fluorescent dyes. There is a wide availability of markers for intracellular and extracellular structures and functions, as well as to examine several of these markers simultaneously in the same cell to allow analysis at individual cell level [19, 20].

Among the best-known tests are neutral red uptake assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium Bromide (MTT) reduction, clonogenic assay, sulforhodamine B, lactate dehydrogenase (LDH) release colorimetric assay, annexin V/propidium iodide (PI) staining, TUNEL, and kenacid blue and resazurin binding assay. Table 1 shows in vitro key studies on cytotoxicity induced by xenobiotics using different assay.

ReferenceXenobioticCells usedAssay usedOutcome(s)
[21]Pesticides Deltamethrin Fenitrothion, Fipronil, Lambda-cyalothrine, and TeflubenzuronCaco-2 cellsMTT cell viability assayCytotoxic effect of Deltamethrin, Fenitrothion, Fipronil, Lambda-cyalothrine, and Teflubenzuron alone or in combination in human intestinal Caco-2 cells.
[22]MalathionN2a mouse neuroblastoma cellsMTT cell viability and LDH release assayThe non-cholinergic effect of malathion may be mediated by apoptotic cell death via autophagy and lysosomal membrane permeabilization induction in N2a cells.
[23]X-rayHuman hepatocellular carcinoma cellsMTT and clonogenic assaysCell autophagy was significantly increased after ionizing radiation combined with hyperthermia treatment. Autophagic cell death may be due to the increased intracellular ROS.
[24]Gamma radiationHuman breast cancer cell line (MCF-7)Clonogenic cell survival assay, cell viability using trypan blue staining and apoptotic cell death using the TUNEL assayThe dose and time dependence inducing a significant apoptotic death.
[25]Heavy metalsHuman sperm cellsWST-1 and XTTHarmful effect of CuSO4 and CdCl2 on human spermatozoa.
[26]Heavy metalsHT-22 cell lineMTT assay and Annexin V-FITC/ Propidium iodide (PI)Metal mixtures showed higher cytotoxicity compared to individual metals.
[27]Chemotherapeutic drug
Cyclophosphamide
Monocyte Macrophage Cell Line Raw 264.7MTT assayA reduction in cell viability was found in Raw 264.7 cell line indicating the cell cytotoxicity.
[28]Chemotherapeutic drugs
Paclitaxel
Docetaxel
Oxaliplatin
Bicalutamide
Anastrozole
HT-29 and HeLa cellsMTT AssayDose-response cytotoxicity findings. Favorability of in vitro assay for the selection of chemotherapeutic drugs for greater clinical effectiveness.
[29]Aminated polystyrene, zinc oxide, and silver nanoparticlesHeLa cellsMTT, Alamar blue, and neutral red assayAll nanoparticles tested resulted in the decrease in cell viability, increased intracellular ROS production and induction of cell death by caspase-mediated apoptosis.
[30]Copper oxide, copper-iron oxide, and carbon nanoparticlesHuman hepatoma HepG2 cellsMTT and neutral red assaysThere was increased cytotoxicity, mutagenicity, and mitochondrial impairment in the cells treated with higher concentrations of the nanomaterials, especially the copper oxide nanoparticles.
[31]Dental universal adhesivesMonocyte/macrophage peripheral blood cell lineXTT assaySome of the tested adhesives showed significant cytotoxic and genotoxic effects.
[32]Perfluorocarbons for intraocular useBALB/3T3, ARPE-19 cell linesMTT, neutral red uptake, and TUNEL assayQualitative evaluation showed that cytotoxic control induced apoptosis, severe reactivity zones, and cytotoxicity according to ISO 10993-5 in all tested conditions.
[33]Polysiloxane-based polyurethane/lignin elastomersHeLa cellsMTT assayDemonstrate the usefulness of in vitro cytotoxicity studies to improve the response of materials based on polysiloxane-based polyurethane / lignin elastomers.

Table 1.

Overview of in vitro key studies on cytotoxicity induced by xenobiotics using different assay.

Neutral red uptake assay

This test is a measure of the toxicity of a compound in the short or long term, which is determined by the release of a dye (i.e., neutral red) due to the loss of cell viability. In this sense, the fact that a compound is cytotoxic regardless of its action mechanism must be considered, if this interferes in the process of cell division and multiplication [34, 35]. This interference leads to the reduction in the speed of cell growth, which is reflected in the number of cells present in the culture. The degree of growth inhibition related to the concentration of the compound being evaluated is an index of toxicity [34, 35].

Neutral red is taken up by cells (specifically by lysosomes and endosomes) and, as the cell loses viability due to the action of the compound being evaluated, the dye is released into the medium, since only viable cells can retain the dye inside. The amount of neutral red dye that remains after exposure within the cell is then determined and the concentration that produces 50% inhibition of cell growth is then calculated [34, 35].

The MTT reduction assay

This method is simple and is used for the determination of cell viability, given by the number of cells present in the culture, which can be measured based on the formation of a colored compound as a result of a reaction occurring in the mitochondria of the viable cells [35, 36].

MTT is a compound belonging to the family of tetrazolium salts that is soluble in water and a yellow color. The metabolic activity of cells includes mitochondrial succinic dehydrogenase enzyme, but cytosolic reductases or reductases from other subcellular compartments may also be involved. The resulting reduced coenzymes (NADH and NADPH) will convert MTT to its insoluble formazan form [37]. When reduced, MTT becomes a compound purple and insoluble in water. To quantify MTT, it is usually dissolved in an organic solvent such as dimethyl sulfoxide (DMSO). The amount of reduced MTT is quantified by a colorimetric method, since the color changes from yellow to purple as a result of the reaction [35, 37, 38].

Clonogenic assay

This assay enables assessing whether a cell is capable of dividing and forming a colony after being exposed to a treatment [39]. A cell survival curve defines the relationship between the dose of the agent used and the fraction of cells that retain their ability to reproduce. Cell lines of various origins, neoplastic or normal, of human or rodent origin can be used [19, 39].

This test is considered an extremely useful tool owing to its advantages of low cost, reproducibility, and simplicity. It has been used for several decades to evaluate the effects of radiation, chemotherapy, drug development, drug screening, toxicology, and pharmacology [19, 40].

Sulforhodamine B

Sulforhodamine B (SRB) is a bright pink aminoxanthan dye with two negatively charged sulfonic groups ▬SO3▬ capable of electrostatically binding to cations [41]. Under acidic conditions (when dissolved in 1% acetic acid), SRB increases its affinity for the basic amino acids of proteins and binds selectively to them, providing an index of the cellular protein content if the cells were previously fixed with trichloroacetic acid. After removing the unfixed dye, the dye bound to the viable cells is extracted with alkaline medium (Tris solution, pH 10.5) and the absorbance is read at 564 nm [41, 42].

LDH release colorimetric assay

This assay allows the measurement of LDH enzyme activity using a cocktail of reagents containing lactate, NAD+, diaphrose, and the tetrazolium salt INT [43]. LDH catalyzes the reduction of NAD+ to NADH in the presence of L-lactate, and the formation of NADH can be measured by a coupled reaction, in which the tetrazolium salt INT is reduced to a red formazan product that can be measured spectrophotometrically [44]. The increase in the LDH activity in the culture supernatant is proportional to the number of cells lysed [43, 44].

Annexin V/PI staining

Annexin V is a recombinant protein that specifically binds to phosphatidylserine residues, which are exposed on the outer surface of the plasma membrane, and is an effective biomarker in apoptotic cells [45]. Annexin V can be combined with a DNA marker that is not membrane-permeable unless the membrane is compromised in order to distinguish apoptotic cells from necrotic cells [45].

It has been reported that the combination of annexin V-FITC and the cationic marker PI can guarantee this differentiation, registering non-apoptotic cells (annexin V-FICT-negative/PI negative), the cells in early apoptosis (annexin V-FICT positive/PI negative) and necrotic cells (annexin V-FICT positive/PI positive). The samples are analyzed in a cytometer providing an objective and fast quantification [46].

TUNEL assay

A useful method to study apoptosis is the TdT-mediated dUTP-biotin nick end-labeling (TUNEL assay). During apoptosis, nuclear endonucleases digest genomic DNA into oligonucleosomal fragments of approximately 180–200 base pairs. DNA fragments are labeled by the catalytic incorporation of labeled 16-dUTP at the free ends by means of the enzyme terminal deoxynucleotidyl transferase (TdT) [47]. The accessibility of the enzyme to the DNA break points is decreased due to nuclear proteins along with the processes of fixation of the sample and the subsequent fixation with ethanol [48]. The signal increases with a greater number of breaks in the DNA chain, and it can be conducted both by flow cytometry and fluorescence. The TUNEL assay has been widely applied in different types of cells to detect DNA damage produced by different types of xenobiotics [49, 50].

Kenacid blue binding assay

Through this assay, the change in total protein content is measured, which is a reflection of cell proliferation. If a compound is cytotoxic to the cells, it must affect at least one or more processes involved in cell proliferation, such as DNA synthesis, the proper functioning of organelles such as mitochondria and lysosomes or affect the integrity of the membrane or protein synthesis [51]. When the cell growth is affected, the number of cells present in the treated culture must be reduced with respect to the control, such that the measurement of the concentration of proteins present in the culture constitutes an index of toxicity [51]. Generally, the cells are exposed to the product for evaluation for a time period of 72 h, and the product is then removed and the cells are exposed to the dye, which then binds to the cellular proteins. Finally, the amount of kenacid blue retained by the cells is determined and the percent of inhibition of cell growth is quantified [51, 52].

Resazurin binding assay

Resazurin (7-hydroxy-3H-phenoxazin-3-one 10-oxide) is a non-fluorescent blue dye that is commonly used for the measurements of cell viability [53]. Resazurin is reduced to resofurin (a highly fluorescent pink dye) by oxidoreductases detected primarily in the mitochondria of viable cells. Resofurin is excreted into the medium, allowing continuous monitoring of the proliferation and/or cytotoxicity of the substances in human cells, animals, bacteria, and even fungi [54]. This dye is not extremely toxic to cells and allows the continuity of studies in the same cells, which saves time and money, especially in the primary cultures where the cells are extremely scarce and valuable. Furthermore, it is sensitive and highly reproducible. It is therefore possible to determine in samples at 530–580 nm as excitation wavelengths and 570–620 nm as emission wavelengths, since this dye has both chromophoric and fluorophore properties [53, 54].

Biomarkers

The term “biomarker” is applied to measure an interaction between the biological system and a chemical, physical, or biological agent, which was evaluated as a functional or physiological response that occurs at the cellular or molecular level and is associated with the probability of the development of a disease [55].

The main objectives with the use of biomarkers in human and environmental toxicology are to measure the exposure to xenobiotic agents that causes diseases and to predict the toxic response that could possibly occur [55]. This approach allowed an increase in the requirement of regulation for the development of drugs, pesticides, and other compounds that can produce adverse effects on the human health in addition to greater impacts on occupational health [55, 56]. Short- and long-term toxicity studies in vitro systems and in experimental animals are very valuable to demonstrate the association of different substances with the appearance of cytotoxicity and the development of mutagenesis, carcinogenesis, and teratogenesis in order to promote early actions for the protection of human health [56].

Advertisement

4. Cytotoxic drugs

The absorption of a drug depends on its physicochemical properties, its formulation, and its route of administration. A drug must cross several semi-permeable cell barriers before reaching the systemic circulation. The cell membranes act as biological barriers that selectively inhibit the flow of drug molecules [57, 58].

Within toxic events that can compromise cell functions by xenobiotic agents such as nanoparticle-based drugs, cellular oxidative stress is an important biological process that must be considered [59]. Oxidative stress is manifested by the production of reactive oxygen species (ROS). ROS are highly reactive, very small molecules that are produced as a result of the presence of an unpaired valence electron shell; they are highly reactive and possess the ability to interact with macromolecules such as lipids, proteins, and DNA. There are various cellular signals that are conveyed through binding to antioxidant response elements or in response to electrophiles, which regulate the expression and coordinate different genes related to their chemoprotective and detoxifying capacities [59, 60].

Determining cell viability is essential when analyzing the efficacy of a new drug or treatment [57]. Not all drugs have the same underlying mechanism or the same level of effect, therefore analyzing how they affect cell health can be a key indicator of whether the drugs may work for a specific intended result. The pharmaceutical industry uses a variety of cytotoxicity tests to screen compositions [57]. Chemicals, drugs, and pesticides all affect human cells in different ways, and these tests can uncover the exact mechanisms of how these xenobiotics work in the human body. Cytotoxicity assays can uncover processes such as the destruction of cell membranes, irreversible binding to receptors, impaired protein synthesis, irreversible binding to receptors, and others [19].

Cytotoxic drugs are preferably used to treat neoplastic diseases, these include DNA alkylating agents [61], antimetabolites [62], and microtubule-active agents [63], topoisomerase inhibitors [64], among others. The goal of the pharmaceutical industry is to create increasingly efficient cytotoxic drugs for cancer treatment, with specific targets for certain cellular targets. Nevertheless, they are drugs with high toxicity, mainly utilized for hematopoietic, renal, hepatic, digestive, and dermal ailments [65, 66].

Advertisement

5. Medical devices

According to the WHO, medical devices refer to any instrument, device, implement, machine, implant, reagent for in vitro use, software, material, or other similar or related article. These devices must undergo rigorous tests to determine their biocompatibility when they come into contact with the body, regardless of its mechanical, physical, and chemical properties [67].

With the continuous development of science and technology, if medical devices are new, they must undergo biocompatibility tests, cytotoxicity, sensitization, intradermal irritation, acute systemic toxicity, and a series of tests before entering a clinical setting to ensure that it is safe for use and effective in humans [68]. These cytotoxicity studies are generally quick, simple, and straightforward, and help eliminate materials that may be harmful to the body and further consider whether they need further analysis and evaluation for their safe and effective use [69].

Polyurethanes (PUs) represent a popular and important part of industrial products that are characterized by good flexibility properties, high impact resistance, and durability; these characteristics make them polymers with multiple applications [70]. Their block copolymer character provides them a wide versatility in terms of adapting their physical properties and compatibility; thus, PUs are interesting for internal uses (in vivo), especially in short-term applications, such as in catheters or implants. Similarly, they are interesting for external use applications (in vitro), such as controlled drug release systems. The PUs used as biomedical materials must comply with the mechanical properties for the intended application and must be non-toxic, biodegradable according to the function to be fulfilled, and biocompatible [69, 70].

Biodegradable polymers have gained much attention presently in the medical field in the search for new materials for treating health problems that arise due to their attractive physical properties and good biocompatibility [71].

Advertisement

6. Pesticides

Pesticides, especially herbicides that are used routinely in crop production, have been shown to cause detrimental effects on the human health [72]. These compounds are easily absorbed via different routes, such as the gastrointestinal and respiratory tracts and through the skin. Due to their high stability and affinity for adipose tissues, they can be metabolized and stored in the human organs, mainly in the adipose tissues [72]. Several human diseases have been associated with pesticide exposure, including cancer, hypertension, neurodegenerative diseases (Parkinson’s), and diabetes [73, 74, 75, 76]. Due to the toxicity and extreme persistence of pesticides in the environment, different studies have been conducted on the toxicity of these agents [77].

Glyphosate for example is a broad spectrum post-emergent herbicide used in both agricultural and non-agricultural areas for weed control [78, 79]. Within the investigations in this regard, Nagy et al. [80] compared the cytotoxic and genotoxic potentials of the active ingredient glyphosate using mononuclear white blood cells that were treated with different concentrations of glyphosate and others with three glyphosate-based herbicides and found that glyphosate induces significant cytotoxicity and genotoxicity effects [80].

The cell metabolic activity is an important indicator of cell viability, and succinate dehydrogenase is an enzyme complex found in the inner mitochondrial membrane of eukaryotic cells that can be used to reflect the viability of these cells [81]. In this field, Devi et al. [82] used rotenone and chlorpyrifos to evaluate the interaction of these pesticides with the protein malate dehydrogenase (MDH) and the consequent cytotoxicity induced by these pesticides. The authors found that rotenone and chlorpyrifos bind strongly to MDH, interfering with protein folding and triggering alterations in their secondary structure.

In this sense, it has also been shown that exposure to pesticides can induce oxidative DNA damage, single and double breaks, and adduct formation [83]. Although there are different DNA repair mechanisms for these damages and for the maintenance of cellular integrity, excessive damage or irreparable damage can lead to cell death processes [8, 83, 84].

In fact, it has been described that some pesticides induce cell death through apoptosis signaling pathways in order to maintain cell homeostasis [85, 86]. Pesticide-induced apoptosis can form the basis of several human diseases, such as cancer and neurological diseases [87]. For this reason, studies in this field have been consistently increasing.

Advertisement

7. Radiation

Radiation, according to its energy, can be classified into ionizing and non-ionizing types [88]. In this sense, ionizing radiation corresponds to the radiation of higher energy (shorter wavelength) within the electromagnetic spectrum. These radiations have sufficient energy to remove electrons from the atoms with which they interact to produce ionizations, while the non-ionizing radiations are those that do not have sufficient energy to remove an electron from the atom they interact with, that is, they do not produce ionizations [88]. Figure 1 illustrates the types of radiation.

Figure 1.

Types of radiation.

Ionizing radiation is considered to be more dangerous because it has sufficient energy to alter matter (through ionizing energy). Depending on the environment in which the radiation collides, the mechanism of action of the radiation differs. These types of action mechanism can be classified into a direct-action mechanism, which consists of transferring energy to molecules such as proteins, lipids, or DNA, among others, which causes the bond to break down and, consequently, cause damage to the cells. The indirect mechanism of action occurs when energy is absorbed by water molecules in the body and, consequently, radiolysis occurs. In this process, the free radicals of OH and the release of H+ ions are produced, which recombine and can produce H2, H2O, and H2O2, which when reacting with molecules such as glucose and cholesterol, among others, can cause damage momentary in the metabolism and if they react with DNA, structural damage is generated [89, 90].

These radiations cause DNA damage in different ways, such as double chain breaks, single chain breaks (SSB), hydrogen bond breakage, base dimers, DNA–DNA cross-linking, DNA-protein cross-linking, loss of bases, base modification, and alteration and damage of the repair mechanisms by interaction with cell cycle proteins such as cyclins CDKs and p53 regulator of cell apoptosis, all of which lead to mutations or structural abnormalities that increase the genomic instability [89, 90, 91].

Advertisement

8. Conclusions

The damage at the cellular level, either in some structures or in processes that affect cell maintenance, division, or survival, can lead to processes of cell death. An important point in drug evaluation is to provide an alternative approach to improve the predictive capacity of cytotoxicity assays based on cell analysis through incorporation of more specific parameters and/or more appropriate cellular systems. By means of this approach, cytotoxicity can be defined in an integrated manner, based on genomic, proteomic, and cytomic data, starting from the molecules to cells and from the cells to tissues.

The advent of new and improved cytotoxicity assays that are safe, robust, and affordable has been instrumental in advancing the pharmaceutical development process. With these analyzes, presently, research has begun to rely less on animal testing and more on studies that are more economical.

References

  1. 1. Rai, P.K., 2016. Impacts of particulate matter pollution on plants: Implications for environmental biomonitoring. Ecotoxicology and Environmental Safety 129, 120-136. https://doi.org/10.1016/j.ecoenv.2016.03.012
  2. 2. Al-Thani, H., Koç, M., Isaifan, R.J., 2018. A review on the direct effect of particulate atmospheric pollution on materials and its mitigation for sustainable cities and societies. Environ Sci Pollut Res 25, 27839-27857. https://doi.org/10.1007/s11356-018-2952-8
  3. 3. Oesch, F., Fabian, E., Guth, K., Landsiedel, R., 2014. Xenobiotic-metabolizing enzymes in the skin of rat, mouse, pig, guinea pig, man, and in human skin models. Arch Toxicol 88, 2135-2190. https://doi.org/10.1007/s00204-014-1382-8
  4. 4. Tabrez, S., Shakil, S., Urooj, M., Damanhouri, G.A., Abuzenadah, A.M., Ahmad, M., 2011. Genotoxicity Testing and Biomarker Studies on Surface Waters: An Overview of the Techniques and Their Efficacies. Journal of Environmental Science and Health, Part C 29, 250-275. https://doi.org/10.1080/10590501.2011.601849
  5. 5. Mukherjee, P.K., 2019. Bioassay-Guided Isolation and Evaluation of Herbal Drugs, in: Quality Control and Evaluation of Herbal Drugs. Elsevier, pp. 515-537. https://doi.org/10.1016/B978-0-12-813374-3.00013-2
  6. 6. Pagano, M., Faggio, C., 2015. The use of erythrocyte fragility to assess xenobiotic cytotoxicity: Cell Death and Erythrocytes. Cell Biochem Funct 33, 351-355. https://doi.org/10.1002/cbf.3135
  7. 7. Yan, J.S., Chen, M., Wang, Q ., 2013. New Drug Regulation and Approval in China, in: A Comprehensive Guide to Toxicology in Preclinical Drug Development. Elsevier, pp. 713-723. https://doi.org/10.1016/B978-0-12-387815-1.00029-0
  8. 8. Jarvis, I.W.H., Dreij, K., Mattsson, Å., Jernström, B., Stenius, U., 2014. Interactions between polycyclic aromatic hydrocarbons in complex mixtures and implications for cancer risk assessment. Toxicology 321, 27-39. https://doi.org/10.1016/j.tox.2014.03.012
  9. 9. De Zio, D., Cianfanelli, V., Cecconi, F., 2013. New Insights into the Link Between DNA Damage and Apoptosis. Antioxidants & Redox Signaling 19, 559-571. https://doi.org/10.1089/ars.2012.4938
  10. 10. Wnuk, A., Kajta, M., 2017. Steroid and Xenobiotic Receptor Signalling in Apoptosis and Autophagy of the Nervous System. IJMS 18, 2394. https://doi.org/10.3390/ijms18112394
  11. 11. D’Arcy, M.S., 2019. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int 43, 582-592. https://doi.org/10.1002/cbin.11137
  12. 12. Wyllie, A.H., Kerr, J.F.R., Currie, A.R., 1980. Cell Death: The Significance of Apoptosis, in: International Review of Cytology. Elsevier, pp. 251-306. https://doi.org/10.1016/S0074-7696(08)62312-8
  13. 13. Elmore, S., 2007. Apoptosis: A Review of Programmed Cell Death. Toxicol Pathol 35, 495-516. https://doi.org/10.1080/01926230701320337
  14. 14. Jeong, S.-Y., Seol, D.-W., 2008. The role of mitochondria in apoptosis. BMB Reports 41, 11-22. https://doi.org/10.5483/BMBRep.2008.41.1.011
  15. 15. Mukhopadhyay, S., Panda, P.K., Sinha, N., Das, D.N., Bhutia, S.K., 2014. Autophagy and apoptosis: where do they meet? Apoptosis 19, 555-566. https://doi.org/10.1007/s10495-014-0967-2
  16. 16. Saha, S., Panigrahi, D.P., Patil, S., Bhutia, S.K., 2018. Autophagy in health and disease: A comprehensive review. Biomedicine & Pharmacotherapy 104, 485-495. https://doi.org/10.1016/j.biopha.2018.05.007
  17. 17. Glick, D., Barth, S., Macleod, K.F., 2010. Autophagy: cellular and molecular mechanisms. J. Pathol. 221, 3-12. https://doi.org/10.1002/path.2697
  18. 18. Levy, J.M.M., Towers, C.G., Thorburn, A., 2017. Targeting autophagy in cancer. Nat Rev Cancer 17, 528-542. https://doi.org/10.1038/nrc.2017.53
  19. 19. Adan, A., Kiraz, Y., Baran, Y., 2016. Cell Proliferation and Cytotoxicity Assays. CPB 17, 1213-1221. https://doi.org/10.2174/1389201017666160808160513
  20. 20. Caldas, I.P., Alves, G.G., Barbosa, I.B., Scelza, P., de Noronha, F., Scelza, M.Z., 2019. In vitro cytotoxicity of dental adhesives: A systematic review. Dental Materials 35, 195-205. https://doi.org/10.1016/j.dental.2018.11.028
  21. 21. Ilboudo, S., Fouche, E., Rizzati, V., Toé, A.M., Gamet-Payrastre, L., Guissou, P.I., 2014. In vitro impact of five pesticides alone or in combination on human intestinal cell line Caco-2. Toxicology Reports 1, 474-489. https://doi.org/10.1016/j.toxrep.2014.07.008
  22. 22. Venkatesan, R., Park, Y.U., Ji, E., Yeo, E.-J., Kim, S.Y., 2017. Malathion increases apoptotic cell death by inducing lysosomal membrane permeabilization in N2a neuroblastoma cells: a model for neurodegeneration in Alzheimer’s disease. Cell Death Discov. 3, 17007. https://doi.org/10.1038/cddiscovery.2017.7
  23. 23. Yuan, G.-J., Deng, J.-J., Cao, D.-D., Shi, L., Chen, X., Lei, J.-J., Xu, X.-M., 2017. Autophagic cell death induced by reactive oxygen species is involved in hyperthermic sensitization to ionizing radiation in human hepatocellular carcinoma cells. WJG 23, 5530. https://doi.org/10.3748/wjg.v23.i30.5530
  24. 24. Fazel, M., Mehnati, P., Baradaran, B., Pirayesh, J., 2016. Evaluation of gamma radiation-induced cytotoxicity of breast cancer cells: Is there a time-dependent dose with high efficiency? Indian J Cancer 53, 25. https://doi.org/10.4103/0019-509X.180862
  25. 25. Hardneck, F., Israel, G., Pool, E., Maree, L., 2018. Quantitative assessment of heavy metal effects on sperm function using computer-aided sperm analysis and cytotoxicity assays. Andrologia 50, e13141. https://doi.org/10.1111/and.13141
  26. 26. Karri, V., Kumar, V., Ramos, D., Oliveira, E., Schuhmacher, M., 2018. An in vitro cytotoxic approach to assess the toxicity of heavy metals and their binary mixtures on hippocampal HT-22 cell line. Toxicology Letters 282, 25-36. https://doi.org/10.1016/j.toxlet.2017.10.002
  27. 27. Yadav, A., Mandal, M.K., Dubey, K.K., 2020. In Vitro Cytotoxicity Study of Cyclophosphamide, Etoposide and Paclitaxel on Monocyte Macrophage Cell Line Raw 264.7. Indian J Microbiol 60, 511-517. https://doi.org/10.1007/s12088-020-00896-1
  28. 28. Florento, L., Matias, R., Tuaño, E., Santiago, K., Dela Cruz, F., Tuazon, A., 2012. Comparison of Cytotoxic Activity of Anticancer Drugs against Various Human Tumor Cell Lines Using In Vitro Cell-Based Approach. Int J Biomed Sci 8, 76-80
  29. 29. Sharma, A., Gorey, B., Casey, A., 2019. In vitro comparative cytotoxicity study of aminated polystyrene, zinc oxide and silver nanoparticles on a cervical cancer cell line. Drug and Chemical Toxicology 42, 9-23. https://doi.org/10.1080/01480545.2018.1424181
  30. 30. Adeyemi, J.A., Machado, A.R.T., Ogunjimi, A.T., Alberici, L.C., Antunes, L.M.G., Barbosa, F., 2020. Cytotoxicity, mutagenicity, oxidative stress and mitochondrial impairment in human hepatoma (HepG2) cells exposed to copper oxide, copper-iron oxide and carbon nanoparticles. Ecotoxicology and Environmental Safety 189, 109982. https://doi.org/10.1016/j.ecoenv.2019.109982
  31. 31. Wawrzynkiewicz, A., Rozpedek-Kaminska, W., Galita, G., Lukomska-Szymanska, M., Lapinska, B., Sokolowski, J., Majsterek, I., 2020. The Cytotoxicity and Genotoxicity of Three Dental Universal Adhesives—An In Vitro Study. IJMS 21, 3950. https://doi.org/10.3390/ijms21113950
  32. 32. Romano, M.R., Ferrara, M., Gatto, C., Ferrari, B., Giurgola, L., D’Amato Tóthová, J., 2019. Evaluation of Cytotoxicity of Perfluorocarbons for Intraocular Use by Cytotoxicity Test In Vitro in Cell Lines and Human Donor Retina Ex Vivo. Trans. Vis. Sci. Tech. 8, 24. https://doi.org/10.1167/tvst.8.5.24
  33. 33. Xu, C., Chen, G., Tan, Z., Hu, Z., Qu, Z., Zhang, Q ., Lu, Maoping, Wu, K., Lu, Mangeng, Liang, L., 2020. Evaluation of cytotoxicity in vitro and properties of polysiloxane-based polyurethane/lignin elastomers. Reactive and Functional Polymers 149, 104514. https://doi.org/10.1016/j.reactfunctpolym.2020.104514
  34. 34. Ates, G., Vanhaecke, T., Rogiers, V., Rodrigues, R.M., 2017. Assaying Cellular Viability Using the Neutral Red Uptake Assay, in: Gilbert, D.F., Friedrich, O. (Eds.), Cell Viability Assays, Methods in Molecular Biology. Springer New York, New York, NY, pp. 19-26. https://doi.org/10.1007/978-1-4939-6960-9_2
  35. 35. Mello, D.F., Trevisan, R., Rivera, N., Geitner, N.K., Di Giulio, R.T., Wiesner, M.R., Hsu-Kim, H., Meyer, J.N., 2020. Caveats to the use of MTT, neutral red, Hoechst and Resazurin to measure silver nanoparticle cytotoxicity. Chemico-Biological Interactions 315, 108868. https://doi.org/10.1016/j.cbi.2019.108868
  36. 36. Pascua-Maestro, R., Corraliza-Gomez, M., Diez-Hermano, S., Perez-Segurado, C., Ganfornina, M.D., Sanchez, D., 2018. The MTT-formazan assay: Complementary technical approaches and in vivo validation in Drosophila larvae. Acta Histochemica 120, 179-186. https://doi.org/10.1016/j.acthis.2018.01.006
  37. 37. van Meerloo, J., Kaspers, G.J.L., Cloos, J., 2011. Cell Sensitivity Assays: The MTT Assay, in: Cree, I.A. (Ed.), Cancer Cell Culture, Methods in Molecular Biology. Humana Press, Totowa, NJ, pp. 237-245. https://doi.org/10.1007/978-1-61779-080-5_20
  38. 38. Sylvester, P.W., 2011. Optimization of the Tetrazolium Dye (MTT) Colorimetric Assay for Cellular Growth and Viability, in: Satyanarayanajois, S.D. (Ed.), Drug Design and Discovery, Methods in Molecular Biology. Humana Press, Totowa, NJ, pp. 157-168. https://doi.org/10.1007/978-1-61779-012-6_9
  39. 39. Rafehi, H., Orlowski, C., Georgiadis, G.T., Ververis, K., El-Osta, A., Karagiannis, T.C., 2011. Clonogenic Assay: Adherent Cells. JoVE 2573. https://doi.org/10.3791/2573
  40. 40. Nuryadi, E., Mayang Permata, T.B., Komatsu, S., Oike, T., Nakano, T., 2018. Inter-assay precision of clonogenic assays for radiosensitivity in cancer cell line A549. Oncotarget 9, 13706-13712. https://doi.org/10.18632/oncotarget.24448
  41. 41. Orellana, E., Kasinski, A., 2016. Sulforhodamine B (SRB) Assay in Cell Culture to Investigate Cell Proliferation. BIO-PROTOCOL 6. https://doi.org/10.21769/BioProtoc.1984
  42. 42. Vichai, V., Kirtikara, K., 2006. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc 1, 1112-1116. https://doi.org/10.1038/nprot.2006.179
  43. 43. Parhamifar, L., Andersen, H., Moghimi, S.M., 2019. Lactate Dehydrogenase Assay for Assessment of Polycation Cytotoxicity, in: Ogris, M., Sami, H. (Eds.), Nanotechnology for Nucleic Acid Delivery, Methods in Molecular Biology. Springer New York, New York, NY, pp. 291-299. https://doi.org/10.1007/978-1-4939-9092-4_18
  44. 44. Kumar, P., Nagarajan, A., Uchil, P.D., 2018. Analysis of Cell Viability by the Lactate Dehydrogenase Assay. Cold Spring Harb Protoc 2018, pdb.prot095497. https://doi.org/10.1101/pdb.prot095497
  45. 45. Crowley, L.C., Marfell, B.J., Scott, A.P., Waterhouse, N.J., 2016a. Quantitation of Apoptosis and Necrosis by Annexin V Binding, Propidium Iodide Uptake, and Flow Cytometry. Cold Spring Harb Protoc 2016, pdb.prot087288. https://doi.org/10.1101/pdb.prot087288
  46. 46. Duensing, T.D., Watson, S.R., 2018. Assessment of Apoptosis (Programmed Cell Death) by Flow Cytometry. Cold Spring Harb Protoc 2018, pdb.prot093807. https://doi.org/10.1101/pdb.prot093807
  47. 47. Crowley, L.C., Marfell, B.J., Waterhouse, N.J., 2016b. Detection of DNA Fragmentation in Apoptotic Cells by TUNEL. Cold Spring Harb Protoc 2016, pdb.prot087221. https://doi.org/10.1101/pdb.prot087221
  48. 48. Kyrylkova, K., Kyryachenko, S., Leid, M., Kioussi, C., 2012. Detection of Apoptosis by TUNEL Assay, in: Kioussi, C. (Ed.), Odontogenesis, Methods in Molecular Biology. Humana Press, Totowa, NJ, pp. 41-47. https://doi.org/10.1007/978-1-61779-860-3_5
  49. 49. Li, C., Zhao, K., Zhang, H., Liu, L., Xiong, F., Wang, K., Chen, B., 2018. Lead exposure reduces sperm quality and DNA integrity in mice. Environmental Toxicology 33, 594-602. https://doi.org/10.1002/tox.22545
  50. 50. Yang, L., Tu, D., Zhao, Z., Cui, J., 2017. Cytotoxicity and apoptosis induced by mixed mycotoxins (T-2 and HT-2 toxin) on primary hepatocytes of broilers in vitro. Toxicon 129, 1-10. https://doi.org/10.1016/j.toxicon.2017.01.001
  51. 51. Clothier, R., Gottschalg, E., Casati, S., Balls, M., 2006. The FRAME Alternatives Laboratory Database. 1. In Vitro Basal Cytotoxicity determined by the Kenacid Blue Total Protein Assay. Altern Lab Anim 34, 151-175. https://doi.org/10.1177/026119290603400203
  52. 52. Hurst, H.S., Clothier, R.H., Pratten, M., 2007. An Evaluation of a Novel Chick Cardiomyocyte Micromass Culture Assay with Two Teratogens/Embryotoxins Associated with Heart Defects. Altern Lab Anim 35, 505-514. https://doi.org/10.1177/026119290703500510
  53. 53. Csepregi, R., Lemli, B., Kunsági-Máté, S., Szente, L., Kőszegi, T., Németi, B., Poór, M., 2018. Complex Formation of Resorufin and Resazurin with Β-Cyclodextrins: Can Cyclodextrins Interfere with a Resazurin Cell Viability Assay? Molecules 23, 382. https://doi.org/10.3390/molecules23020382
  54. 54. Silva, F.S.G., Starostina, I.G., Ivanova, V.V., Rizvanov, A.A., Oliveira, P.J., Pereira, S.P., 2016. Determination of Metabolic Viability and Cell Mass Using a Tandem Resazurin/Sulforhodamine B Assay: SRB/Resazurin Assay for Cell Viability and Metabolism. Current Protocols in Toxicology 68, 2.24.1-2.24.15. https://doi.org/10.1002/cptx.1
  55. 55. Santonen, T., Aitio, A., Fowler, B.A., Nordberg, M., 2015. Biological Monitoring and Biomarkers, in: Handbook on the Toxicology of Metals. Elsevier, pp. 155-171. https://doi.org/10.1016/B978-0-444-59453-2.00008-1
  56. 56. Aronson, J.K., Ferner, R.E., 2017. Biomarkers—A General Review. Current Protocols in Pharmacology 76. https://doi.org/10.1002/cpph.19
  57. 57. Hilde Bohets, Pieter Annaert, Geert Mannens, Ludy van Beijsterveldt, Katelijne Anciaux, Peter Verboven, Willem Meuldermans, Karel Lavrijsen, 2001. Strategies for Absorption Screening in Drug Discovery and Development. CTMC 1, 367-383. https://doi.org/10.2174/1568026013394886
  58. 58. Lee, Y., Choi, S.Q ., 2019. Quantitative analysis for lipophilic drug transport through a model lipid membrane with membrane retention. European Journal of Pharmaceutical Sciences 134, 176-184. https://doi.org/10.1016/j.ejps.2019.04.020
  59. 59. Yang, B., Chen, Y., Shi, J., 2019. Reactive Oxygen Species (ROS)-Based Nanomedicine. Chem. Rev. 119, 4881-4985. https://doi.org/10.1021/acs.chemrev.8b00626
  60. 60. Abdal Dayem, A., Hossain, M., Lee, S., Kim, K., Saha, S., Yang, G.-M., Choi, H., Cho, S.-G., 2017. The Role of Reactive Oxygen Species (ROS) in the Biological Activities of Metallic Nanoparticles. IJMS 18, 120. https://doi.org/10.3390/ijms18010120
  61. 61. Cheng, S.-Y., Seo, J., Huang, B.T., Napolitano, T., Champeil, E., 2016. Mitomycin C and decarbamoyl mitomycin C induce p53-independent p21WAF1/CIP1 activation. International Journal of Oncology 49, 1815-1824. https://doi.org/10.3892/ijo.2016.3703
  62. 62. Petaccia, M., Gentili, P., Bešker, N., D’Abramo, M., Giansanti, L., Leonelli, F., La Bella, A., Gradella Villalva, D., Mancini, G., 2016. Kinetics and mechanistic study of competitive inhibition of thymidine phosphorylase by 5-fluoruracil derivatives. Colloids and Surfaces B: Biointerfaces 140, 121-127. https://doi.org/10.1016/j.colsurfb.2015.12.020
  63. 63. Wei, C., Pan, Y., Huang, H., Li, Y.-P., 2018. Estramustine phosphate induces prostate cancer cell line PC3 apoptosis by down-regulating miR-31 levels. European Review for Medical and Pharmacological Sciences 22, 40-45. https://doi.org/10.26355/eurrev_201801_14098
  64. 64. Kovács E.R., Tóth S., Erdélyi D.J., 2018. Az etopozid szervezeten belüli eloszlásában és metabolizmusában szerepet játszó epigenetikai hatások. Orvosi Hetilap 159, 1295-1302. https://doi.org/10.1556/650.2018.31162
  65. 65. Kitao, H., Iimori, M., Kataoka, Y., Wakasa, T., Tokunaga, E., Saeki, H., Oki, E., Maehara, Y., 2018. DNA replication stress and cancer chemotherapy. Cancer Sci 109, 264-271. https://doi.org/10.1111/cas.13455
  66. 66. Lin, A., Giuliano, C.J., Palladino, A., John, K.M., Abramowicz, C., Yuan, M.L., Sausville, E.L., Lukow, D.A., Liu, L., Chait, A.R., Galluzzo, Z.C., Tucker, C., Sheltzer, J.M., 2019. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl. Med. 11, eaaw8412. https://doi.org/10.1126/scitranslmed.aaw8412
  67. 67. Who - World Health Organization, 2017, ISBN 9789241512350 https://www.who.int/health-topics/medical-devices#tab=tab_1
  68. 68. Reeve, L., Baldrick, P., 2017. Biocompatibility assessments for medical devices – evolving regulatory considerations. Expert Review of Medical Devices 14, 161-167. https://doi.org/10.1080/17434440.2017.1280392
  69. 69. Bernard, M., Jubeli, E., Pungente, M.D., Yagoubi, N., 2018. Biocompatibility of polymer-based biomaterials and medical devices – regulations, in vitro screening and risk-management. Biomater. Sci. 6, 2025-2053. https://doi.org/10.1039/C8BM00518D
  70. 70. Zuber, M., Zia, F., Zia, K.M., Tabasum, S., Salman, M., Sultan, N., 2015. Collagen based polyurethanes—A review of recent advances and perspective. International Journal of Biological Macromolecules 80, 366-374. https://doi.org/10.1016/j.ijbiomac.2015.07.001
  71. 71. Kenry, Liu, B., 2018. Recent Advances in Biodegradable Conducting Polymers and Their Biomedical Applications. Biomacromolecules 19, 1783-1803. https://doi.org/10.1021/acs.biomac.8b00275
  72. 72. Jabłońska-Trypuć, A., Wydro, U., Wołejko, E., Butarewicz, A., 2019. Toxicological Effects of Traumatic Acid and Selected Herbicides on Human Breast Cancer Cells: In Vitro Cytotoxicity Assessment of Analyzed Compounds. Molecules 24, 1710. https://doi.org/10.3390/molecules24091710
  73. 73. Jemal, A., Bray, F., Center, M.M., Ferlay, J., Ward, E., Forman, D., 2011. Global cancer statistics. CA: A Cancer Journal for Clinicians 61, 69-90. https://doi.org/10.3322/caac.20107
  74. 74. Thapa, K., Pant, B.R., 2014. Pesticides in vegetable and food commodities: environment and public health concern. J Nepal Health Res Counc 12, 208-210
  75. 75. Ball, N., Teo, W.-P., Chandra, S., Chapman, J., 2019. Parkinson’s Disease and the Environment. Front. Neurol. 10, 218. https://doi.org/10.3389/fneur.2019.00218
  76. 76. Brust, R.S., Oliveira, L.P.M. de, Silva, A.C.S.S. da, Regazzi, I.C.R., Aguiar, G.S. de, Knupp, V.M. de A.O., 2019. Epidemiological profile of farmworkers from the state of Rio de Janeiro. Rev. Bras. Enferm. 72, 122-128. https://doi.org/10.1590/0034-7167-2017-0555
  77. 77. Bilal, M., Iqbal, H.M.N., Barceló, D., 2019. Persistence of pesticides-based contaminants in the environment and their effective degradation using laccase-assisted biocatalytic systems. Science of The Total Environment 695, 133896. https://doi.org/10.1016/j.scitotenv.2019.133896
  78. 78. Chikoye, D., Lum, A.F., Udensi, U.E., 2010. Efficacy of a new glyphosate formulation for weed control in maize in southwest Nigeria. Crop Protection 29, 947-952. https://doi.org/10.1016/j.cropro.2010.06.011
  79. 79. Uren Webster, T.M., Laing, L.V., Florance, H., Santos, E.M., 2014. Effects of Glyphosate and its Formulation, Roundup, on Reproduction in Zebrafish ( Danio rerio ). Environ. Sci. Technol. 48, 1271-1279. https://doi.org/10.1021/es404258h
  80. 80. Nagy, K., Tessema, R.A., Budnik, L.T., Ádám, B., 2019. Comparative cyto- and genotoxicity assessment of glyphosate and glyphosate-based herbicides in human peripheral white blood cells. Environmental Research 179, 108851. https://doi.org/10.1016/j.envres.2019.108851
  81. 81. Stockert, J.C., Horobin, R.W., Colombo, L.L., Blázquez-Castro, A., 2018. Tetrazolium salts and formazan products in Cell Biology: Viability assessment, fluorescence imaging, and labeling perspectives. Acta Histochemica 120, 159-167. https://doi.org/10.1016/j.acthis.2018.02.005
  82. 82. Devi, S., Karsauliya, K., Srivastava, T., Raj, R., Kumar, D., Priya, S., 2021. Pesticide interactions induce alterations in secondary structure of malate dehydrogenase to cause destability and cytotoxicity. Chemosphere 263, 128074. https://doi.org/10.1016/j.chemosphere.2020.128074
  83. 83. Benedetti, D., Lopes Alderete, B., de Souza, C.T., Ferraz Dias, J., Niekraszewicz, L., Cappetta, M., Martínez-López, W., Da Silva, J., 2018. DNA damage and epigenetic alteration in soybean farmers exposed to complex mixture of pesticides. Mutagenesis 33, 87-95. https://doi.org/10.1093/mutage/gex035
  84. 84. Kapka-Skrzypczak, L., Czajka, M., Sawicki, K., Matysiak-Kucharek, M., Gabelova, A., Sramkova, M., Bartyzel-Lechforowicz, H., Kruszewski, M., 2019. Assessment of DNA damage in Polish children environmentally exposed to pesticides. Mutation Research/Genetic Toxicology and Environmental Mutagenesis 843, 52-56. https://doi.org/10.1016/j.mrgentox.2018.12.012
  85. 85. Jiang, J., Wu, S., Liu, X., Wang, Y., An, X., Cai, L., Zhao, X., 2015. Effect of acetochlor on transcription of genes associated with oxidative stress, apoptosis, immunotoxicity and endocrine disruption in the early life stage of zebrafish. Environmental Toxicology and Pharmacology 40, 516-523. https://doi.org/10.1016/j.etap.2015.08.005
  86. 86. Zerin, T., Song, H.-Y., Kim, Y.-S., 2015. Extracellular signal-regulated kinase pathway play distinct role in acetochlor-mediated toxicity and intrinsic apoptosis in A549 cells. Toxicology in Vitro 29, 85-92. https://doi.org/10.1016/j.tiv.2014.09.011
  87. 87. Mattson, M.P., 2000. Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol 1, 120-130. https://doi.org/10.1038/35040009
  88. 88. Hamada, N., Fujimichi, Y., 2014. Classification of radiation effects for dose limitation purposes: history, current situation and future prospects. Journal of Radiation Research 55, 629-640. https://doi.org/10.1093/jrr/rru019
  89. 89. Rak, J., Chomicz, L., Wiczk, J., Westphal, K., Zdrowowicz, M., Wityk, P., Żyndul, M., Makurat, S., Golon, Ł., 2015. Mechanisms of Damage to DNA Labeled with Electrophilic Nucleobases Induced by Ionizing or UV Radiation. J. Phys. Chem. B 119, 8227-8238. https://doi.org/10.1021/acs.jpcb.5b03948
  90. 90. Runge, R., Arlt, J., Oehme, L., Freudenberg, R., Kotzerke, J., 2017. Comparison of clonogenic cell survival and DNA damage induced by 188Re and X-rays in rat thyroid cells. Nuklearmedizin 56, 47-54. https://doi.org/10.3413/Nukmed-0842-16-08
  91. 91. Zhang, X., Ye, C., Sun, F., Wei, W., Hu, B., Wang, J., 2016. Both Complexity and Location of DNA Damage Contribute to Cellular Senescence Induced by Ionizing Radiation. PLoS ONE 11, e0155725. https://doi.org/10.1371/journal.pone.0155725

Written By

Grethel León-Mejía, Alvaro Miranda Guevara, Ornella Fiorillo Moreno and Carolina Uribe Cruz

Submitted: 01 October 2020 Reviewed: 27 January 2021 Published: 17 February 2021