1,276
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Cytogenetic damage by vanadium(IV) and vanadium(III) on the bone marrow of mice

ORCID Icon, ORCID Icon, ORCID Icon, &
Received 15 Jun 2023, Accepted 14 Sep 2023, Published online: 05 Oct 2023

Abstract

Vanadium is a strategic metal that has many important industrial applications and is generated by the use of burning fossil fuels, which inevitably leads to their release into the environment, mainly in the form of oxides. The wastes generated by their use represent a major health hazard. Furthermore, it has attracted attention because several genotoxicity studies have shown that some vanadium compounds can affect DNA; among the most studied compounds is vanadium pentoxide, but studies in vivo with oxidation states IV and III are scarce and controversial. In this study, the genotoxic and cytotoxic potential of vanadium oxides was investigated in mouse bone marrow cells using structural chromosomal aberration (SCA) and mitotic index (MI) test systems. Three groups were administered vanadium(IV) tetraoxide (V2O4) intraperitoneally at 4.7, 9.4 or 18.8 mg/kg, and three groups were administered vanadium(III) trioxide (V2O3) at 4.22, 8.46 or 16.93 mg/kg body weight. The control group was treated with sterile water, and the positive control group was treated with cadmium(II) chloride (CdCl2). After 24 h, all doses of vanadium compounds increased the percentage of cells with SCA and decreased the MI. Our results demonstrated that under the present experimental conditions and doses, treatment with V2O4 and V2O3 induces chromosomal aberrations and alters cell division in the bone marrow of mice.

I. Introduction

Vanadium (V) is a strategic metal that has many important industrial applications, used for producing high-quality specialized metal alloys and strengthening steel in the aerospace industry and in the chemical industry, for the production of catalysts, electronics and batteries, e.g., vanadium redox batteries (Huang et al. Citation2015, Hu et al. Citation2018). Among the compounds that are employed in diverse industrial processes are vanadium(III) trioxide (V2O3) and vanadium(IV) tetraoxide (V2O4), which are used in electro-optical switches, aerospace technology and semiconductor manufacturing (Rodríguez-Mercado et al. Citation2010, Zhang et al. Citation2021, Vanitec Citation2023) New uses and applications are continually being discovered for this metal (SCIREA Citation2019).

Vanadium is released into the atmosphere as a consequence of burning fossil fuels and metallurgic industry activities. Measured concentrations in coals worldwide vary from 7 to 100 mg/kg, with an estimated mean of 25 mg/kg. Schlesinger et al. (Citation2017) estimated that extraction associated with coal mining had increased from approximately 100 × 109 g V/year in 2000 to 180–190 x 109 V/year in 2014–16, and their release caused residual oil ash. Levels in air vary by season and location (urban, industrial and rural), and volcanic eruptions, weathering of V-rich rocks and wildfires can lead to their introduction into the ambient environment. The main compounds released consist of V oxides, which accumulate in the soil and water, resulting in constant exposure to organisms (ATSDR Citation2012, Huang et al. Citation2015, Cohen Citation2022).

The most common nonoccupational sources of V exposure are contaminated food (WHO Citation1996, Anke Citation2004, Mehmood et al. Citation2021), because soils contain high concentrations, and from there, they pass to food (Chen et al. Citation2021). For example, following a diesel spill in Oaxaca, Mexico, V levels in sediments were found to range from 95 to 149 μg V/g compared to levels of < 5 μg V/g at distanced reference sites (Salazar-Coria et al. Citation2007). Similarly, Hernandez and Rodríguez (Citation2012) found that levels of V in urban soils of Salamanca, Mexico, reached values of > 600 mg V/kg; these were far higher than in rural soils, but even those were relatively high (i.e., 126 mg V/kg) due partly to the region’s volcanic nature that contains many basalts and rhyolites.

The total ingested in the diet is varied, with an average V intake of 13 μg/day/person in the United Kingdom and an estimated dietary intake between 6 and 18 µg/day in the USA (EFSA Citation2004). In Germany and México, V intake is 10–20 µg/day for women and > 20 µg/day for men (Anke Citation2004, Rodríguez-Mercado et al. Citation2011). Another report indicated that typical daily doses consumed by humans have been estimated at 0.01–0.03 mg V/day (Willsky et al. Citation2011, Pessoa et al. Citation2015). Whole grains, seafood and meats generally contain 5–30 ng/kg of V (EFSA Citation2004), and parsley, mushrooms, black pepper, cereals and shellfish are some of the main sources of V, with concentrations ranging from 0.05 to 2 mg/kg (Tripathi et al. Citation2018). Another important source of V exposure is the use of dietary mineral supplements, which can lead to the consumption of more than 10 mg V/day (Barceloux and Barceloux Citation1999, EFSA Citation2004, Leopardi et al. Citation2005).

Workers occupationally exposed to V are at risk of developing respiratory diseases (IARC Citation2006) because the particulates are small, breathable and may penetrate deep into the pulmonary tract (Fortoul et al. Citation2011, Rojas-Lemus et al. Citation2021) and increase heart rates (Magari et al. Citation2002). Some epidemiological studies have indicated that a correlation exists between exposure to airborne V particles and the incidence of cancer in residents of metropolitan areas (Kraus et al. Citation1989). In leukocytes from 52 exposed workers, an increase in the plasma concentration of V and no difference in DNA migration, with comet assay, were reported under standard conditions, but when the authors used a modified technique that detects DNA damage due to the oxidation of DNA bases, they found increased tail lengths, and those workers also had a 2.5-fold higher micronuclei frequency (Ehrlich et al. Citation2008). Vanadium(V) pentoxide (V2O5) has been designated as possibly carcinogenic to humans (Group 2B) by the International Agency for Research on Cancer (IARC 2006), based on evidence of lung cancer in mice published by the NTP (Citation2002), and this chemical and other vanadium compounds have been designated as category 2 by the German Commissions for the Investigation of Health Hazards of Chemical Compounds in the Work Area (Beyersmann and Hartwing Citation2008).

Due to the above, genotoxicity studies of V have attracted attention, and several studies have shown that some V compounds can affect DNA, among other toxic effects. The most studied compound at the cytogenetic level is V2O5, both in vitro and in vivo, which is known to produce DNA single-strand breaks in whole blood leukocytes in vitro (Rojas et al. Citation1996). The results from in vivo studies reported an increased number of cells with damage, with a single-cell gel electrophoresis assay, in liver, kidney, lung, spleen, and heart cells but not in bone marrow (Altamirano-Lozano et al. Citation1999). Additionally, in male mice injected intraperitoneally with V2O5 every 3rd day for 60 days, a DNA damage-dependent dose was reported in testis cells (Altamirano-Lozano et al. Citation1996), in the bone marrow, and no differences were observed in sister chromatid exchanges or in average generational time (Altamirano-Lozano et al. Citation1993). Rojas-Lemus et al. (Citation2014) reported that inhaled V2O5 (0.02 M) 2 hours twice a week increased the frequency of micronuclei in exposed males compared with the control, and no cytotoxic effect was observed.

Studies with other V oxides showed that V2O4 increases the frequency of sister chromatid exchanges and the structural chromosomal aberration (SCA) and induces cytostatic and cytotoxic effects in human cells cultured in vitro (Rodríguez-Mercado et al. Citation2003, Citation2010, Rodríguez-Mercado and Altamirano-Lozano Citation2006), and some authors consider that some vanadium(III) compounds can induce structural chromosome damage (Lloyd et al. Citation1998, Caicedo et al. Citation2008). Nevertheless, we did not find significant differences in the SCA when human lymphocyte cultures were treated with vanadium(III) trioxide (Rodríguez-Mercado et al. Citation2010). However, in vivo studies with vanadium oxides III and IV are scarce and inconclusive. Therefore, the aim of this study was to evaluate the genotoxic and cytotoxic potential of the in vivo intraperitoneal administration of two vanadium oxides, V2O4 and V2O3, in mouse bone marrow cells using the SCA and mitotic index (MI) cytogenetics assay.

II. Materials and methods

2.1. Testing drug

The treatment was performed with vanadium oxides obtained from Aldrich Chemical Company (Milwaukee, Wis., USA), V2O3 (V = 67.98%, O = 32.02%; CAS no. 1314–34-7, 99.6% pure), and V2O4 (V = 61.43%, O = 38.57%; CAS no. 12036–21-4, 99.6% pure), which were macerated and dissolved in injectable water. Cadmium(II) chloride (CdCl2•2.5 H2O CAS 7790–78-5) and KCl were obtained from Sigma–Aldrich (St. Louis, MO, USA). Giemsa was obtained from Hycel of Mexico. Absolute methanol and acetic acid were obtained from JT Baker Mexico.

2.2. Animals

Male CD-1 mice, six to seven weeks old, weighing 25–35 g, were obtained from Bioterium of the FES-Zaragoza, UNAM. The animals were handled in accordance with official regulations in Mexico (NOM-062-ZOO-1999). The animals were housed in polypropylene cages under a controlled temperature of 22 ± 1 °C, and free access to water and food (Harlan 2018S with 18% protein rodent diet) was permitted. The photoperiod consisted of 12 h of artificial light and 12 h of darkness, with a relative humidity of 31%.

2.3. Experimental procedure

The mice were separated and housed in groups of six animals for a total of 8 groups. Three groups were administered 4.7, 9.4 and 18.8 mg/kg body weight (bw) of V2O4, and these values correspond to 1/32, 1/16, and 1/8 of the lethal dose 50 (LD50= 150 mg/kg Aragón and Altamirano-Lozano Citation2001). Three groups were administered 4.22, 8.46 or 16.93 mg/kg bw of V2O3; these doses of V2O3 were calculated based on the amount of vanadium present in V2O4. The vanadium concentration was calculated in both compounds, which were 2.87, 5.75 and 11.5 mg of V in each dose of compounds, respectively. The doses of both compounds contained the same amount of elemental V. The control group was treated with sterile water, and the positive control group was treated with 7 mg/kg bw CdCl2 (Ali Citation2012, El-Habit and Abdel-Moneim Citation2014)

All treatments were injected intraperitoneally (ip) with a volume of 0.1 mL/10 g of mouse body weight. The intraperitoneal route of administration generally offers the second most rapid absorption among the parenteral routes, conferred by the large surface area of the lining of the peritoneal cavity, and the rich blood supply to that area process (Stump et al. Citation1999, Gad Citation2014). Bone marrow was collected 24 h after the treatment. The experimental protocol was approved by the Committee of Ethics and Biosecurity of FES-Zaragoza UNAM (registration number FESZ-CE/21–118-01).

2.4. Cytogenetic assay

Twenty-two hours after vanadium compound treatment, the mice were injected with 0.2% colchicine (0.1 mL/10 g bw), to arrest proliferating cells in metaphase (Florian and Mitchison Citation2016), and 2 h later, the animals were sacrificed by cervical dislocation. Bone marrow tissue was obtained from each femur with hypotonic solution (KCl 0.075 M), and samples remained for 1 h at 37 °C. The cells were then fixed with methanol-acetic acid (3:1). To prepare the slides for cytogenetic analysis, 5 drops of the fixed cell suspension were placed on clean glass slides, which were then stained for 30 min with a 5% Giemsa solution.

The MI was calculated as the proportion of cells undergoing cell division. A total of 4000 cells per mouse were assessed, and the MI was calculated using the following formula: MI= (Number of dividing cells, metaphases/total number of bone marrow cells counted) x 100.

The SCA analysis was based on the recommendations of Adler (Citation1984), Savage (Citation2004) and the OECD TG 475 (2013) with slight modifications. The type of aberration scored included chromatid and chromosome breaks, fragments and pulverizations. At least one hundred cells in metaphase per mouse were reviewed, and only metaphases containing 40 ± 2 centromeres were analyzed. The total number and type of aberrations, as well as the percentage of aberrant cells per treatment, were evaluated. All slides were coded and evaluated in a double-blind manner.

2.5. Statistical analysis

The results are expressed as the means ± standard deviations (SD). The Z test was used to assess significant MI differences. Student’s t test was used to compare the group means for SCA. Tukey’s ANOVA was also applied to the difference between groups of the percentage of cells with SCA.

3. Results

The administration of V compounds to mice resulted in no changes in the weight of exposed or control mice. It is important to highlight that the animals treated did not present other symptoms of toxicity.

The SCA and MI results are shown in . The data show a reduction in the MI in the treated groups compared to the negative control group. The control group had an MI of 4.1, while the groups treated with V2O4 showed reductions of 44, 28.5 and 32.6%, respectively; in the groups treated with V2O3, the reductions in the MI were 35.6, 31.4 and 26.8%, respectively.

Table 1. Mitotic index (MI) and structural chromosomal aberrations (SCA) in male mouse bone marrow cells following exposure ip to vanadium(IV) tetraoxide (V2O4) or vanadium(III) trioxide (V2O3).

The percentage of cells with SCA increased in the treated groups. The control group had 4.03% of cells with SCA, while the groups treated with V2O4 showed increases of approximately 3-, 4- and 4.5-fold, respectively; in the groups treated with V2O3, the increases were approximately 3.7-, 3.9- and 4.8-fold, respectively. The most frequent aberration types were fragments and chromatid breaks, and in some concentration cells with 10 or more aberrations, pulverization was observed (). Differences between total chromosome aberrations and the number of cells with aberrations are due to some cells containing or having more than one aberration.

Figure 1. Some of chromosome aberrations in bone marrow cells from CD-1 male mice treated ip with vanadium(IV) or vanadium(III). A) Normal chromosome (control group), B) fragments (dose of 16.9 mg/Kg of V2O3), C) chromatid breaks (dose of 18.8 mg/Kg of V2O4), D) chromosomal breaks (dose of 8.46 mg/Kg of V2O3), E) chromatid breaks, and F) pulverizations (dose of 9.4 mg/Kg of V2O4).

Figure 1. Some of chromosome aberrations in bone marrow cells from CD-1 male mice treated ip with vanadium(IV) or vanadium(III). A) Normal chromosome (control group), B) fragments (dose of 16.9 mg/Kg of V2O3), C) chromatid breaks (dose of 18.8 mg/Kg of V2O4), D) chromosomal breaks (dose of 8.46 mg/Kg of V2O3), E) chromatid breaks, and F) pulverizations (dose of 9.4 mg/Kg of V2O4).

As expected, when mice were treated with cadmium(II) chloride at a dose of 7 mg/kg, a clear reduction in MI and an increase in SCA were observed.

4. Discussion

The results of this work show that there were no differences in mouse weight after intraperitoneal administration of the compounds V2O4 or V2O3. In another work where V2O4 was administered for 60 days to male mice at similar doses (4.7, 9.4 or 18.8 mg/kg bw), no changes in animal weight were observed (Aragón and Altamirano-Lozano Citation2001). However, a reduction in the weight of male CD-1 mice was found when sodium orthovanadate (Na3VO4) was administered orally for 5 weeks at concentrations of 7.5, 75, 750 and 1500 mg/L (Leopardi et al. Citation2005).

Therefore, we can say that the effect of V on the weight of mice depends on the compound, time and the dose administered. Considering that the weight of the whole organism is one of the parameters that provide evidence of toxicity of the different compounds administered (OECD Citation2013), we can conclude that under the present experimental conditions and doses of V2O4 and V2O3 administered in this study do not produce toxicity in mice at this treatment time (24 h).

The data show a reduction in the MI in the groups treated with both compounds compared to the negative control group. MI is a parameter that shows the percentage of cells that reached the metaphase stage of the total of cells that are in mitosis, indicating that the cells are in division, and the reduction of this parameter shows that V2O4 and V2O3 produce an effect on cell proliferation.

Few in vivo studies have evaluated the cytotoxic effect of V oxides. Ciranni et al. (Citation1995) reported that SVO5 (vanadyl sulfate), a compound that contains vanadium(IV), produced an appreciable cytotoxic effect, using polychromatic erythrocytes (PCEs) and normochromatic erythrocytes (NCEs) ratio data after administration of 100 mg/kg intragastrically to male mice.

Previous studies performed with vanadium in oxidation estate +5, which is the main compound studied, show that the administration of 23 mg/kg bw ip of V2O5 to mice decreases the MI, but with the administration of doses of 5.75 and 11.5 mg/kg, MI does not decrease (Altamirano-Lozano et al. Citation1993). Rojas-Lemus et al. (Citation2014) reported a decrease in the reticulocyte count in mice that inhaled V2O5 (0.02 M) 2 h/twice a week, and the PCE/NCE ratio is indicative of cytotoxic effects.

Rodríguez-Mercado et al. (Citation2010) reported that in human leukocytes treated in vitro with three oxides, V2O3, V2O4 or V2O5, decreased MI in all groups treated, so they concluded that the three compounds were capable of inducing cytotoxicity. In another study with the same model, V2O3 caused a decrease in the MI and replication index, which may suggest that V2O3 can alter cell cycle progression (Mateos-Nava et al. Citation2017, Citation2021).

The decrease in MI may be related to the ability of V compounds to alter the activity of different molecules, such as ATPases, protein kinases, ribonucleases and phosphatases, and it has also been shown that V oxides could modify the levels of some proteins responsible for regulating the cell cycle (Attia et al. Citation2005, Mateos-Nava et al. Citation2021, Rojas-Lemus et al. Citation2021). Furthermore, V oxides delay the cell cycle via a mechanism that does not necessarily involve DNA damage, since it has been observed that disrupting cytoskeletal microtubules, hyperosmotic stress and inhibiting histone deacetylases induce the modification of Cdc25C through the p38 MAPK pathway (Zhang et al. Citation2003, Liu et al. Citation2012, Mateos-Nava et al. Citation2021).

Although generally the reduction of MI is a consequence of a reduced rate of cell proliferation, there could also be a contribution from cells that have permanently lost their proliferative capacity since it has been reported that some V compounds have a cytotoxic effect in vitro and that this effect is closely related to its chemical form (Rodriguez-Mercado et al. Citation2010), which increases with the oxidation state and produces this effect only in certain cell types (Rojas-Lemus et al. Citation2014). Guerrero-Palomo et al. (Citation2019) reported that classical apoptotic mechanisms are not related to the cell death induced by some V compounds and showed that higher reactive oxygen species production was induced by SVO5 in the human lung adenocarcinoma cell Line A549.

The MI data do not present adose–response relationship, which can be explained on the one hand by the difference between the concentrations used, which is not large enough to cause more cytotoxicity, but, if SCA is produced, the cell cycle will stop. Alternatively, because metals can have multiple biological effects on cellular signaling pathways, such as adaptation/tolerance mechanisms to low doses (Giani et al. Citation2021, Shi et al. Citation2022), in this work, a single injection of V compounds was administered at doses of 1/32, 1/16 and 1/8 of the LD50. Nevertheless, currently, there is still a gap in knowledge regarding the dose–response relationship, especially for metal compounds (Shahid et al. Citation2020).

Chromosome damage is a very important indicator of genetic damage relevant to environmental studies. In somatic cells, chromosomal aberrations and mutations play a key role in processes leading to malignancies if mutations occur in proto-oncogenes, tumor suppressor genes, and/or DNA damage response genes. The accumulation of DNA damage in somatic cells has also been proposed to play a role in degenerative conditions such as accelerated aging, immune dysfunction and cardiovascular and neurodegenerative diseases (Niida and Nakanishi Citation2006, Doherty Citation2012, Valko et al. Citation2016, Mosesso and Cinelli Citation2019).

The increase in the percentage of cells with SCA in the groups treated with V2O4 found in this study is in accordance with one previous study reported by Ciranni et al. (Citation1995), in which they administered single intragastric doses of three vanadium salts, 100 mg/kg bw SVO5, 75 mg/kg bw sodium orthovanadate (Na3VO4) and 50 mg/kg bw ammonium metavanadate (NH4VO3) (-vanadate in oxidation state V) to male mice. They found an increased frequency of micronucleus by all compounds tested, but only the group treated with SVO5 presented increased chromosome aberrations in bone marrow cells, and the highest effect was observed at 24 h. This chromosome damage was due mainly to breaks and fragments.

Villani et al. (Citation2007) reported that there was no significant difference in comet assays and micronuclei in the PCE ratio in male CD1 mice exposed to doses of 2–1000 mg/L SVO5 in drinking water for 5 weeks, both in the bone marrow and peripheral erythrocytes. The authors concluded that the data indicate scarce bioavailability for orally administered vanadium(IV) and lack of significant genotoxic potential in vivo.

On the other hand, in in vitro studies with human isolated lymphocytes, Wozniak and Blasiak (Citation2004) found that DNA damage was induced with 0.5 or 1 mM SVO5 within one hour of exposure. Studies conducted by our working group reported that treatment of human peripheral lymphocyte cultures with 1, 2, 4 or 8 µg/mL V2O4 produced an increase in chromatid breaks and acentric fragments as well as dicentrics (Rodríguez-Mercado et al. Citation2010). Additionally, vanadium(IV) induces DNA double-strand breaks (Rodríguez-Mercado et al. Citation2011), and this damage to the DNA can lead to chromosome lesions since it is generally accepted that mechanisms involved in repairing double-strand breaks and the genetic processes of recombination are mainly responsible for forming chromosomal aberrations (Obe and Durante Citation2010, Yamamura et al. Citation2018). It is likely that the increase in the SCA that we observed in this work is caused by this type of damage. Our results confirm that V2O4 causes DNA damage in both in vivo and in vitro cytogenetic tests and in different cell types.

Few studies have been carried out to assess the genotoxic potential of V2O3. Owusu-Yaw et al. (Citation1990) reported an increase in SCA frequencies induced by V2O3 in CHO cells. In human blood leukocytes treated in vitro with the same compound produced DNA-single-strand breaks at all of the concentrations and treatment times tested (Rodríguez-Mercado et al. Citation2011) but showed no significant increase in the frequency of SCA (Rodríguez-Mercado et al. Citation2010).

In this study, we found that both V oxides produce SCA in non-dose dependent manner. The most frequent aberration types were fragments and chromatid breaks in both compounds, although it should be noted that some chromosome-type breaks and pulverisation were also observed. The increase in the percentage of cells with SCA with treatment with V2O4 and V2O3 may be based on different mechanisms. One of the modes of action in the induction of genotoxic effects of V compounds is the generation of ROS and/or DNA–protein cross-links because vanadium(IV) has the capability to bind DNA through interactions with guanine and adenine N-7 atoms and backbone PO2 groups, resulting in the formation of 8-hydroxy-2′-deoxyguanosine, which is a common oxidative DNA adduct, and multiple and accumulative DNA single- and double-strand breaks cause clastogenicity (Rodríguez-Mercado et al. Citation2011, Espinosa-Zurutuza et al. Citation2018) or its ability to react with hydrogen peroxide and generate hydroxyl radicals (•OH) (Prousek Citation2007, He et al. Citation2022).

Unrepaired or misrepaired breaks may very well result in chromosomal damage, a lesion in the DNA, which is converted to a single-strand break during repair when a section of DNA is removed. If DNA synthesis occurs before the repair is complete, this single-strand break can be converted to a double-strand break, leading to the production of a visible aberration. For this reason, the production of chromosome aberrations requires cells to pass through S-phase for aberration formation. In addition, these aberrations will be chromatid type at the first metaphase after development (Danford Citation2012). Consequently, most of these aberrations may contribute to mutagenesis and carcinogenesis. Chromosome-type and chromatid-type aberrations and micronuclei can predict cancer risk (Doherty Citation2012). Therefore, the possibility that V oxides may produce cancer or chronic degenerative disease should not be ruled out.

The results show that both oxides produce a similar increase in SCA, despite having different oxidation states, which can be explained because we administered the same amount of elemental V, as stated in the material and methods section, and the applied doses of both compounds contain the same amount of elemental V. However, there is also the possibility that part of the vanadium(III) administered could be oxidized to vanadium(IV). Since V compounds that enter the bloodstream are subjected to speciation, it is known that vanadyl bound to transferrin is distributed to the body tissues and bones; vanadate (V) and oxidovanadium (IV) bound to transferrin can enter the intracellular space by endocytosis, analogous to Fe3+, and vanadium (IV) is not easily reduced to vanadium (III) under physiological conditions, but vanadium (III) can be oxidized to vanadium (IV) (Rehder Citation2013, Citation2015, López-Valdez et al. Citation2022).

Many environmental factors can damage DNA, and understanding the mechanisms associated with the outcomes of this process will provide researchers with tools to understand DNA environmental insults and ways to prevent or avoid them. Our results contribute to a better understanding of the mechanisms by which metals induce genotoxic effects. Finally, we want to note that the cytogenetic damage that V compounds produce is similar to that produced by other metals, such as Cd (Fahmy et al. Citation2000, Celik et al. Citation2005, Citation2009, El-Habit and Abdel-Moneim Citation2014).

5. Conclusions

In conclusion, our results showed that V2O4 and V2O3 treatment in mice reduced the MI, so it altered cell proliferation and increased the proportion of cells with SCA in bone marrow.

Acknowledgements

We thank MVZ. Adriana Altamirano Bautista and MVZ. Roman Hernández Meza for their technical support in the care and handling of the animals.

Disclosure statement

The authors declare no conflicts of interest.

Data availability statement

The authors confirm that the data supporting the findings of this study are available within the article.

Additional information

Funding

During the development of this work, partial support was provided by Dirección General de Asuntos del Personal Académico, Universidad Nacional Autónoma de México PAPIIT IA201123 and IN210324.

References

  • Adler, I. D., 1984. Cytogenetic test in mammals: In: V.S. and P.J.M., eds. Mutagenicity testing: a practical approach. Washington DC, IRL Press, 275–306. https://docs.google.com/document/d/1mXrOpwO-L-c4bloE3ryRED_JqI_PV18K/edit?usp=sharing&ouid=115074128154034679310&rtpof=true&sd=true
  • Ali, T.H., 2012. Determination of the lethal dose 50% (LD50) of cadmium chloride and the histopathological changes in male mice liver and kidneys. Journal of Education and Science, 25 (3), 27–38.
  • Altamirano-Lozano, M. A., et al., 1993. Cytogenetic and teratogenic effects of vanadium pentoxide on mice. Medical Science Research, 21 (19),711–713.
  • Altamirano-Lozano, M.A., et al., 1996. Reprotoxic and genotoxic studies of vanadium pentoxide in male mice. Teratogenesis, Carcinogenesis, and Mutagenesis, 16 (1), 7–17.
  • Altamirano-Lozano, M.A., et al., 1999. Genotoxic studies of vanadium pentoxide (V2O5) in male mice. II. Effects in several mouse tissues. Teratogenesis, Carcinogenesis, and Mutagenesis, 19 (4), 243–255.
  • Anke, M., 2004. Vanadium – an element both essential and toxic to plants, animals and humans? Anales de la Real Academia Nacional de Farmacia, 70, 961–999.
  • Aragón, M.A., and Altamirano-Lozano, M., 2001. Sperm and testicular modifications induced by sub-chronic treatments with vanadium (IV) in CD-1 mice. Reproductive Toxicology (Elmsford, N.Y.), 15 (2), 145–151.
  • ATSDR, Agency for Toxic Substances and Disease Registry 2012., Toxicological Profile for Vanadium. Department of Health and Human Services. Public Health Service, Atlanta Georgia.
  • Attia, S.M., et al., 2005. Orthovanadate increased the frequency of aneuploid mouse sperm without micronucleus induction in mouse bone marrow erythrocytes at the same dose level. Mutation Research, 583 (2), 158–167.
  • Barceloux, G.D., and Barceloux, D., 1999. Vanadium. Journal of Toxicology, 37 (2), 265–278.
  • Beyersmann, D., and Hartwig, A., 2008. Carcinogenic metal compounds: recent insight into molecular and cellular mechanisms. Archives of Toxicology, 82 (8), 493–512.
  • Caicedo, M., et al., 2008. Analysis of metal ion-induced DNA damage, apoptosis, and necrosis in human (Jurkat) T-cells demonstrates Ni2+ and V3+ are more toxic than other metals: Al3+, Be2+, Co2+, Cr3+, Cu2+, Fe3+, Mo5+, Nb5+, Zr2+. Journal of Biomedical Materials Research. Part A, 86 (4), 905–913.
  • Celik, A., et al., 2005. A study on the investigation of cadmium chloride genotoxicity in rat bone marrow using micronucleus test and chromosome aberration analysis. Toxicology and Industrial Health, 21 (10), 243–248.
  • Celik, A., et al., 2009. Assessment of cadmium genotoxicity in peripheral blood and bone marrow tissues of male Wistar rats. Toxicology Mechanisms and Methods, 19 (2), 135–140.
  • Chen, L., et al., 2021. Vanadium in soil-plant system: Source, fate, toxicity, and bioremediation. Journal of Hazardous Materials, 405, 124200.
  • Ciranni, R.L., et al., 1995. Vanadium salts induce cytogenetic effects in in vivo treated mice. Mutation Research, 343 (1), 53–60.
  • Cohen, M. D., 2022. Vanadium. In: G. F. Nordberg and M. Costa, eds. Handbook on the Toxicology of metals. NY, USA: Elsevier B.V., 937–961.
  • Danford, N., 2012. The interpretation and analysis of cytogenetic data. In: J. Parry and E. Parry, eds. Methods in Molecular Biology. NY, USA: Springer, 817, 93–120.
  • Doherty, A. T., 2012. Cytogenetic in vivo assays in somatic cells. In: J. Parry and E. Parry, eds. Methods in Molecular Biology. NY, USA: Springer, 817, 271–304.
  • EFSA, European Food Safety Authority, 2004. Opinion of the scientific panel on dietetic products, nutrition and allergies related to the tolerable upper intake level of vanadium. EFSA Journal, 33, 1–22.
  • Ehrlich, A., et al., 2008. Inhalative exposure to vanadium pentoxide causes DNA damage in workers: results of a multiple end point study. Environmental Health Perspectives, 116 (12), 1689–1693.
  • El-Habit, O.H., and Abdel-Moneim, A.E., 2014. Testing the genotoxicity, cytotoxicity, and oxidative stress of cadmium and nickel and their additive effect in male mice. Biological Trace Element Research, 159 (1-3), 364–372.
  • Espinosa-Zurutuza, M., et al., 2018. Oxidative stress as a mechanism involved in kidney damage after subchronic exposure to vanadium inhalation and oral sweetened beverages in a mouse model. International Journal of Toxicology, 37 (1), 45–52.
  • Fahmy, M.A., and Aly, F.A., 2000. In vivo and in vitro studies on the genotoxicity of cadmium chloride in mice. Journal of Applied Toxicology, 20 (3), 231–238.
  • Florian, S., and Mitchison, T. J., 2016. Anti-Microtubule Drugs. In: P. Chang and O. Ryoma, eds. Methods and Protocols. Methods in Molecular Biology, New York: Springer Science, 403–421.
  • Fortoul, T.I., et al., 2011. Vanadium inhalation in a mouse model for the understanding of air-suspended particle systemic repercussion. Journal of Biomedicine & Biotechnology, 2011, 951043–11.
  • Gad, C. S., 2014. Rodents model for toxicity testing and biomarkers. In: R. C. Gupta ed. Biomarkers in Toxicology. Amsterdam, The Netherlands: Elsevier, 7–69.
  • Gianì, F., et al., 2021. Heavy metals in the environment and thyroid cancer. Cancers, 13 (16), 4052.
  • Guerrero-Palomo, G., et al., 2019. Vanadium compounds and cellular death mechanisms in the A549 cell line: The relevance of the compound valence. Journal of Applied Toxicology, 39 (3), 540–552.
  • He, X., et al., 2022. Vanadium pentoxide induced oxidative stress and cellular senescense in human lung fibroblasts. Redox Biology, 55, 102409.
  • Hernández, H., and Rodríguez, R., 2012. Geochemical evidence for the origin of vanadium in an urban environment. Environmental Monitoring and Assessment, 184, 5327–5342.
  • Hu, X., et al., 2018. Release kinetics of vanadium from vanadium (III, IV and V) oxides: Effect of pH, temperature and oxide dose. Journal of Environmental Sciences, 67, 96–103.
  • Huang, J.H., et al., 2015. Vanadium (bio) geochemistry. Chemical Geology, 417, 68–89.
  • IARC, Working Groupon the evaluation of carcinogenic Risks to humans, 2006. Cobalt in hard-metals and cobalt sulfate, gallium arsenide, indium phosphide and vanadium pentoxide. IARC Monographs on the Evaluation of Carcinogenic Risks Humans, 86, 1–294.
  • Kraus, T., et al., 1989. Quantitation of manganese and vanadium levels in the lungs of normal and occupationally exposed persons. Zentralbl Hyg Umwelimed, 188, 108–126.
  • Leopardi, P., et al., 2005. Assessment of the in vivo genotoxicity of vanadate: analysis of micronuclei and DNA damage induced in mice by oral exposure. Toxicology Letters, 158 (1), 39–49.,
  • Liu, T., et al., 2012. Reactive-oxygen-species mediated Cdc25C degradation results in differential antiproliferative activities of vanadate, tungstate, and molybdate in the PC-3 human prostate cancer cell line. Journal of Biological Inorganic Chemistry, 17 (2), 311–320.
  • Lloyd, D.R., et al., 1998. Comparison of formation of 8-hydroxy-2’-deoxiguanosine and single- and double-strand breaks in DNA mediated by Fenton reactions. Chemical Research in Toxicology, 11 (5), 420–427.
  • López-Valdez, N., et al., 2022. Sex differences in vanadium inhalation effects in non-ciliated bronchiolar cells. Histology and Histopathology, 30, 18566.
  • Magari, S.R., et al., 2002. The association of particulate air metal concentrations with heart rate variability. Environmental Health Perspectives, 110 (9), 875–880.
  • Mateos-Nava, R.A., et al., 2017. Premature chromatid separation and altered proliferation of human leukocytes treated with vanadium (III) oxide. Drug and Chemical Toxicology, 40 (4), 457–462.
  • Mateos-Nava, R.A., et al., 2021. Vanadium oxides modify the expression levels of the p21, p53, and Cdc25C proteins in human lymphocytes treated in vitro. Environmental Toxicology, 36 (8), 1536–1543.
  • Mehmood, S., et al., 2021. Comparative efficacy of raw and HNO3- modified biochar derived from rice straw on vanadium transformation and its uptake by rice (Oryza sativa L.): Insights from photosynthesis, antioxidative response, and gene-expression profile. Environmental Pollution, 289, 117916.
  • Mosesso, P., and Cinelli, S., 2019. In Vitro Cytogenetic Assays: Chromosomal Aberrations and Micronucleus Tests. Methods in Molecular Biology, 2031, 79–104.
  • Niida, H., and Nakanishi, M., 2006. DNA damage checkpoints in mammals. Mutagenesis, 21 (1), 3–9.
  • NOM 062-ZOO-1999., Norma Oficial Mexicana 1999., Especificación Técnica para la producción, cuidado y uso de animales de laboratorio México. SAGARPA, Diario Oficial de México.
  • NTP. National Toxicology Program, 2002. NTP toxicology and carcinogenesis studies of vanadium pentoxide (CAS No. 1314-62-1) in F344/N rats and B6C3F1 mice (inhalation). National Toxicology Program Technical Report Sereries, 507, 1–343.
  • Obe, G., and Durante, M., 2010. DNA double strand breaks and chromosomal aberrations. Cytogenetic and Genome Research, 128 (1-3), 8–16.
  • OECD, 2013. Mammalian bone marrow chromosomal aberration test. Organization for Economic Cooperation and Development, Guidelines for Testing of Chemical, Paris, DRAFT TG 475.
  • Owusu-Yaw, J., et al., 1990. An assessment of the genotoxicity of vanadium. Toxicology Letters, 50 (2-3), 327–336.
  • Pessoa, J.C., et al., 2015. Vanadium compounds in medicine. Coordination Chemistry Reviews, 301, 24–48.
  • Pfeiffer, P., et al., 2004. Pathways of DNA double-strand break repair and their impact on the prevention and formation of chromosomal aberrations. Cytogenetic and Genome Research, 104 (1-4), 7–13.
  • Prousek, J., 2007. Fenton chemistry in biology and medicine. Pure and Applied Chemistry, 79 (12), 2325–2338.
  • Rehder, D., 2013. Vanadium. Its role for humans. In: A. Sigel, H. Sigel and N.K.O. Sigel, eds. Interrelations between Essential Metal Ions and Human Diseases, Metal Ions in Life Sciences. Springer, 13,139–169.
  • Rehder, D., 2015. The role of vanadium in biology. Metallomics, 7 (5), 730–742.
  • Rodríguez-Mercado, J.J., et al., 2003. Genotoxic effects of vanadium(IV) in human peripheral blood cells. Toxicology Letters, 144 (3), 359–369.
  • Rodriguez-Mercado, J.J., and Altamirano-Lozano, M.A., 2006. Vanadium: pollution, metabolism and genotoxicity (Vanadio: contaminación, metabolismo y genotoxicidad). Revista Internacional de Contaminación Ambiental, 22, 173–189.
  • Rodríguez-Mercado, J.J., et al., 2010. Chromosomal damage induced by vanadium oxides in human peripheral lymphocytes. Drug and Chemical Toxicology, 33 (1), 97–102.
  • Rodríguez-Mercado, J.J., et al., 2011. DNA damage induction in human cells exposed to vanadium oxides in vitro. Toxicology in Vitro, 25 (8), 1996–2002.
  • Rojas, E., et al., 1996. Genotoxicity of vanadium pentoxide evaluate by the single cell gel electrophoresis assay in human lymphocytes. Mutation Research, 359 (2), 77–84.
  • Rojas-Lemus, M., et al., 2014. Sex differences in blood genotoxic and cytotoxic effects as a consequence of vanadium inhalation: micronucleus assay evaluation. Journal of Applied Toxicology, 34 (3), 258–264.
  • Rojas-Lemus, M., et al., 2021. Toxic effects of inhaled vanadium attached to particulate matter: a literature review. International Journal of Environmental Research and Public Health, 18 (16), 8457.
  • Salazar-Coria, L., et al., 2007. Polyaromatic hydrocarbons (PAHs) and metal evaluation after a diesel spill in Oaxaca, Mexico. Bulletin of Environmental Contamination and Toxicology, 79 (4), 462–467.
  • Savage, J.R., 2004. On the nature of visible chromosomal gaps and breaks. Cytogenetic and Genome Research, 104 (1-4), 46–55.
  • Schlesinger, W.H., et al., 2017. Global biogeochemical cycle of vanadium. Proceedings of the National Academy of Sciences, 114 (52), E11092–E11100.
  • SCIREA. Org. Journal of Environment, 2019. Vanadium and its main industry: A review and the resource potential in Canada. Available from: https://article.scirea.org/pdf/61046.pdf
  • Shahid, M., et al., 2020. Trace elements-induced phytohormesis: A critical review and mechanistic interpretation. Critical Reviews in Environmental Science and Technology, 50 (19), 1984–2015.
  • Shi, P., et al ., 2022. Identification of the hormetic dose-response and regulatory network of multiple metals co-exposure-related hypertension via integration of metallomics and adverse outcome pathways. Science of the Total Environment, 15(817), 153039.
  • Stump, D.G., et al., 1999. Comparative effects of single intraperitoneal or oral doses of sodium arsenate or arsenic trioxide during in utero development. Teratology, 60 (5), 283–291.
  • Tripathi, D., et al., 2018. Vanadium in biosphere and its role in biological processes. Biological Trace Element Research, 186 (1), 52–67.
  • Valko, M., et al., 2016. Redox- and non-redox-metal-induced formation of free radicals and their role in human disease. Archives of Toxicology, 90 (1), 1–37.
  • Vanitec, 2023. Using V. Available from: https://vanitec.org//vanadium/using-vanadium [Accesed 11 2023].
  • Villani, P., et al., 2007. Evaluation of genotoxicity of oral exposure to tetravalent vanadium in vivo. Toxicology Letters, 170 (1), 11–18.
  • WHO, World Health Organization 1996., Trace elements in human nutrition and health. World Health Organization. Geneva, 180–183.
  • Willsky, R.G., et al., 2011. Anti-diabetic effects of a series of vanadium dipicolinate complexes in rats with streptozotocin-induced diabetes. Coordination Chemistry Reviews, 255 (19-20), 2258–2269.
  • Wozniak, K., and Blasiak, J., 2004. Vanadyl sulfate can differentially damage DNA in human lymphocytes and HeLa cells. Archives of Toxicology, 78 (1), 7–15.
  • Yamamura, E., et al., 2018. Correlation between the results of in vitro and in vivo chromosomal damage tests in consideration of exposure levels of test chemicals. Genes and Environment, 40 (1), 640–646.
  • Zhang, W., et al., 2021. Prenatal and postnatal exposure to vanadium and the immune function of children. Journal of Trace Elements in Medicine and Biology, 67, 126787.
  • Zhang, Z., et al., 2003. Role of reactive oxygen species and MAPKs in vanadate-induced G2/M phase arrest. Free Radical Biology & Medicine, 34 (10), 1333–1342.