Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4924 2023-08-09 21:39:05 |
2 Some sections of the review were significantly rewritten. -939 word(s) 3985 2023-08-10 15:12:12 | |
3 format change + 5 word(s) 3990 2023-08-11 02:53:27 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Moreira, R.; Martins, A.D.; Alves, M.G.; De Lourdes Pereira, M.; Oliveira, P.F. Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance. Encyclopedia. Available online: https://encyclopedia.pub/entry/47856 (accessed on 27 July 2024).
Moreira R, Martins AD, Alves MG, De Lourdes Pereira M, Oliveira PF. Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance. Encyclopedia. Available at: https://encyclopedia.pub/entry/47856. Accessed July 27, 2024.
Moreira, Rúben, Ana D. Martins, Marco G. Alves, Maria De Lourdes Pereira, Pedro F. Oliveira. "Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance" Encyclopedia, https://encyclopedia.pub/entry/47856 (accessed July 27, 2024).
Moreira, R., Martins, A.D., Alves, M.G., De Lourdes Pereira, M., & Oliveira, P.F. (2023, August 09). Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance. In Encyclopedia. https://encyclopedia.pub/entry/47856
Moreira, Rúben, et al. "Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance." Encyclopedia. Web. 09 August, 2023.
Chromium Picolinate on Testicular Steroidogenesis and Antioxidant Balance
Edit

Reduced testosterone (T) levels significantly contribute to male infertility, as this hormone plays a vital role in various functions throughout the male reproductive system. T is produced in the Leydig cells (LC) via testicular steroidogenesis. Dysfunctional LC can impair steroid synthesis and therefore fertility. Endocrine-disrupting chemicals (EDCs) are noteworthy factors influencing steroidogenesis by interfering with hormonal signaling. The heavy metal chromium is classified as an EDC, existing as hexavalent (Cr(VI)) and trivalent chromium (Cr(III)). Despite the debate surrounding Cr(III), chromium picolinate (CrPic3), is used as a nutritional supplement due to its antioxidant and antidiabetic properties. Detrimental effects of CrPic3 on LC encompass enzyme inhibition in steroidogenesis and, while in other cell types, it was found to induce of mutagenesis and apoptosis. Notably, CrPic3 influences male fertility through modifications in reactive oxygen species, T levels, and sperm parameters (e.g., sperm motility and abnormal sperm count). Nonetheless, major gaps and inconsistencies exist in literature concerning its effects on male fertility. Thus, more research is essential to comprehend the mechanisms in which CrPic3 is involved that may be relevant to male fertility, ensuring the safety of this supplement for man.

chromium picolinate trivalent chromium steroidogenesis reactive oxygen species antioxidants testosterone Leydig cells

1. Introduction

Infertility is the inability to achieve pregnancy after 12 months or more of regular unprotected sexual intercourse [1]. This condition can have male and/or female origins, with the male factor alone being responsible for one-third of all infertility cases, as well as one-half of all combined male-female caused cases [2]. In 2015, it was estimated that 30 million men worldwide were infertile [3]. Some of the main contributors to the rise of infertility are Endocrine-disrupting chemicals (EDCs), a group of compounds considered toxic to humans and the environment [4][5]. One of the major characteristics of EDCs is that they interfere with the endocrine signaling of the body [5]. Heavy metals are considered EDCs with known toxicological risk to human health as they affect several factors, including semen quality parameters and the secretory function of accessory sexual glands [6]. This is not surprising, since 2% of men who suffer from infertility present endocrine disruption as the principal cause [7], and EDCs can disrupt the hypothalamus–pituitary–testis axis [8]. Among the hormonal dysfunctions that can occur, the production of testosterone (T), essential for the normal functioning of the male reproductive system and the triggering of spermatogenesis, is sensitive to exposure to various compounds [9]. A group of several heavy metals is included under the heading of EDCs, with chromium (Cr) being a widely used example across several industries. Among the various Cr compounds, chromium picolinate [tris(picolinate)chromium(III)] (CrPic3) has become a very popular supplement [10] to reduce weight or manage blood glucose levels. Although the use of CrPic3 has shown some promising positive effects for human health, its safety is up for debate.

2. Leydig Cells and Testicular Steroidogenesis

Leydig cells (LC) are one of the main somatic cells un the testes [11]. The latter are found in the connective tissue, between the seminiferous tubules, and produce steroids in a process known as testicular steroidogenesis, summarized in Figure 1. T, the main steroid produced, is essential for regulating spermatogenesis, maintaining primary sexual traits (e.g., testicular descent, penis and testes growth), and developing secondary characteristics like male hair patterns and deepening of the voice. [12][13], as well as general androgenic and anabolic effects [11][13]. Testosterone exerts its functions by binding to the androgen receptor (AR) in the cytoplasm, allowing it to bind to specific DNA motifs in the nucleus, regulating the transcription of specific genes [14].
Figure 1. Schematic representation of testicular steroidogenesis that occurs in the Leydig cell.
Testicular steroidogenesis in Leydig cells (LC) is responsible for producing approximately 95% of the circulating levels of T observed in adult males [15]. This process initiates when luteinizing hormone (LH) binds to its receptor (LHR) on the cell membrane, initiating a signaling cascade that leads to the conversion of ATP into cyclic adenosine monophosphate (cAMP), activation of protein kinase A (PKA), and cholesterol release [16]. Cholesterol is then transported to the mitochondria with the help of the steroidogenic acute regulatory protein (StAR) and the peripheral-type benzodiazepine receptor (PBR) [17][18], followed by cleavage by CYP11A1 to form pregnenolone (Preg) [15].  Preg is subsequently transferred to the smooth endoplasmic reticulum (SER) [17], where it undergoes conversion into T through either the classic or backdoor pathway [19]. Herein, the researchers focus the classic pathway, which is known as the main pathway for T synthesis in humans [20]. The key player in this pathway is CYP17A1, which exhibits two enzymatic activities: 17a-hydroxylase and 17,20-lyase [19].
The 17a-hydroxylase activity converts Preg into 17α-hydroxypregnenolone (17OHPreg) and progesterone (Prog) into 17OH-Progesterone (17OHProg). Conversely, the 17,20-lyase activity converts 17OHPreg into dehydroepiandrosterone (DHEA) and 17OHProg into androstenedione. Hydroxysteroid dehydrogenases (HSDs) are crucial to this biosynthesis [21]. 3β-hydroxysteroid dehydrogenase (3β-HSD) catalyzes the conversion of Δ5-steroids (Preg, 17OHPreg, DHEA, and androstenediol) into Δ4-steroids (Prog, 17-OHProg, androstenedione, and T) [19], while 17β-hydroxysteroid dehydrogenase (17β-HSD) facilitates reversible redox reactions, specifically the conversion of DHEA to androstenediol and androstenedione to T [22].

3. Chromium: The Good, the Bad, and the Controversial

Chromium (Cr), the 24th element on the periodic table, occurs in nature in two forms: hexavalent Cr (Cr(VI)) and trivalent Cr (Cr(III)).  In aqueous solutions, Cr(VI) appears as chromate (CrO4-2) or dichromate (Cr2O7-2) ions [23], while Cr(III) salts include compounds like Cr chloride (CrCl3) and Cr picolinate (Cr(C6H4NO2)3) [24]. Understanding their physiological effects, metabolism, and excretion from the body is crucial.

3.1. Cr(VI): A Toxic Form of Cr

Cr(VI) is used in numerous industries, such as leather tanning, metal processing, and chromate production. Improper management of Cr(VI) may cause ingestion, dermal contact, and/or inhalation by the population, which is known to lead to various health issues [25]. Cr(VI) can enter the cell through non-specific membrane anion transporters. In the cytoplasm, antioxidants such as ascorbate [26], cysteine [27], and reduced glutathione (GSH) reduce Cr(VI), generating reactive oxygen species (ROS) [23] and Cr(III) [28]. As the researchers will discuss, part of the mutagenic effect of Cr(VI) is caused by Cr(III), since it is a by-product of its reduction. The health risks of Cr(VI) include nephrotoxicity [29]; hepatotoxicity [30]; and cancer of the lung, nose, and nasal cavity [31]. There are also well documented toxic effects on the male reproductive system and fertility that include testicular effects, such as loss of testes weight [32]; structural effects, namely moderate tubular necrosis, degeneration of LC [32], and disturbance of the germinal epithelium [33]; and molecular effects, in particular the arrest of spermatogenesis, the decrease in androgenesis [34], and the decrease in antioxidant defenses, such as the levels of superoxide dismutase (SOD) [34]. Cr(VI) is credited as the more hazardous form of Cr, and therefore, it has been the focus of the scientific community for a longer period than has Cr(III). Nevertheless, some studies on Cr(III) are raising concerns regarding its safety [25] and even challenging its classification as an essential element.

3.2. Cr(III): A Controversial Essential Element

In 1959, Schwarz and Mertz proposed that Cr(III) should be an essential element due to its role in glucose tolerance [35]. Nevertheless, the debate over this classification continues to divide the scientific and clinical communities. An argument in favor of classifying Cr(III) as an essential element is that it is a part of the glucose tolerance factor (GTF), which is synthesized in vivo after the absorption of dietary Cr [36]. GTF is known to bind to insulin, boosting its activity threefold [36]. However, one study found that there was no correlation between blood levels of Cr and glycemic control [37]. Instead, evidence has shown that there are no symptoms of Cr(III) deficiency when there is glycemic dysfunction, thus challenging the criterion used for Cr to be considered essential [38]. Indeed, this suggests that Cr(III) does not fulfill the requirements to be considered an essential element, although it may have medicinal properties [38]
In 2009, Vincent, who previously considered Cr as essential, published a review to celebrate the 50th anniversary of this classification, stating that it is crucial to identify the biomolecules that form complexes with Cr to understand its biological (side)effects and whether it should actually be considered essential [39]. Even so, since Cr(III) is still considered essential, it is necessary to establish recommended daily doses. In 2014, the European Food Safety Authority (EFSA) determined that the dietary chromium intake should be as follows: 30.1–42.9 μg/day for infants (12 < 36 months), 54.3–71.2 μg/day for children (36 months < 10 years), 63.5–83.4 μg/day for teenagers (10 < 18 years), and 57.3–83.8 μg/day for adults (≥18 years) [40]. However, in 2018, Filippin et al. argued that the daily doses of chromium for adults should be slightly higher, 59.55 μg/day for men and 56.08 μg/day for women, to optimize the nutritional effects and avoid toxicity [41]. Nevertheless, there is still no consensus on the doses of either Cr or CrPic3, or more notably, on the safety of Cr(III).

4. Positive and Adverse Effects of Supplementation with CrPic3

CrPic3 is composed of Cr(III) chelated with picolinic acid (Pic), with the molecular formula Cr(C6H4NO2)3 [42]. Despite the controversy around Cr(III), CrPic3 is already abundantly commercialized as a nutritional supplement, particularly targeted to diabetic and obese patients. It is also sold as a supplement to treat depression [43][44], to protect against heat stress [45], to stimulate ovulation in women with polycystic ovary syndrome, and to improve the lipid profile [46], among other announced benefits.

4.1. Antidiabetic Effects of CrPic3 and Its Implications on Male Fertility

As previously mentioned, CrPic3 is abundantly used as a nutritional supplement because of its antidiabetic properties. Diabetes mellitus is a heterogeneous group of metabolic disorders which includes type 2 diabetes mellitus (T2DM). T2DM is characterized by dysfunction of the insulin-producing pancreatic beta cells, heightened glucagon-producing pancreatic alpha-cell activity, and insulin resistance in the peripheral tissues. When untreated, it leads to hyperglycemia, dyslipidemia, insufficient amino acid uptake, and ATP production [47]
Regarding its antidiabetic effects, CrPic3 is capable of: (1) increasing insulin sensitivity [48][49], (2) increasing glucose tolerance and uptake (both basal and insulin-stimulated) [46][50], and (3) preventing damage caused by hyperglycemia [46]. These effects occur because CrPic3: (a) decreases the phosphorylation of IRS-1, the JNK pathway [51], as well as pro-inflammatory cytokines, mainly TNFα [52][53], which causes the inhibition of IRS-1 through the phosphorylation of Ser307 [54]; (2) increases the presence of the glucose transporter GLUT4 in the cell membrane and stimulates the p38/MAPK pathway [50]; and (3) normalizes the levels of antioxidant enzymes in the liver [46]. CrPic3 also assumes a protective role against dyslipidemia, since it improves the altered lipid profile [46], regulates triglycerides and HDL-c [55][56], and decreases serum cholesterol through the increase in SREBP, a transcription factor responsible for cellular cholesterol homeostasis [57]. However, it does not alter the levels of apolipoproteins ApoA and ApoB [58], markers of cardiovascular risk and metabolic syndrome [59]. Martin et al. (2006) showed evidence that in patients with T2D, between 25–75 years old, CrPic3 attenuated the increase in body weight, favored body fat distribution, and allowed for the decrease in glucose and increase in insulin sensitivity [48]. With that being said, CrPic3 shows promising positive effects regarding diabetes and cardiovascular diseases. However, one must wonder at what cost and if its benefits exceed the possible drawbacks.
Since diabetes mellitus has a strong impact on male fertility, namely a negative effect on sperm parameters (reduced semen volume, count, concentration, and progressive motility) and testosterone levels [60], attempting to ameliorate these effects may be of interest to improve the fertility of men suffering from infertility linked to diabetes. Indeed, Alves et al. found evidence in the literature that led them to believe that, even though the link between diabetes and male infertility is not absolute, there might be mechanisms “of the disease that may affect testicular cells, spermatogenesis, sperm production and sperm maturation” [61]. Interestingly, when Meneses et al. reviewed the effects of metformin, an extensively used antidiabetic drug, on male fertility, they realized that the consensus in the scientific community is that it improves aspects of male fertility, such as levels of FSH, LH, and testosterone; along with sperm concentration, motility, and morphology, among others [62]. This highlights, on one hand, the detrimental effects of diabetes mellitus on male fertility and on the other, the potential benefits of antidiabetic pharmacological substances to ward off those deleterious effects.

4.2. Antioxidant Effects of CrPic3 in Cellular Systems

A possible positive effect that has been attributed to CrPic3 is the increase in antioxidant defenses, which will be explored in this subsection. This is an important feature, since oxidative stress, which results from an imbalance between the production of ROS and antioxidant defense, contributes to the development of numerous pathologies [63]. ROS can, among other effects, disrupt the hormonal crosstalk in the body, causing, for instance, a diminishing of testosterone [64]. Still, it is important to note that the equilibrium of ROS production and elimination is crucial to the functioning of the male reproductive system since low levels of ROS are required for sperm function, but in excess, ROS cause injury that includes lipid peroxidation and DNA damage [65].
Several antioxidant enzymes are more expressed after various treatments with CrPic3, namely GSH, catalase (CAT), SOD, glutathione peroxidase (GPX), and glutathione reductase (GR). Doddigarla et al. found that treating type 2 diabetic rats (high carbohydrate diet induced) orally with 1.4 μg/day of CrPic3 for 8 weeks caused a significant increase in GSH and CAT and a significant decrease in MDA [66]. Likewise, Al-Bishri et al. treated type 2 diabetic rats (streptozotocin induced) with 100 μg/kg body weight of CrPic3. In doing so, they found that the Cr supplement significantly increased GSH, CAT, GPX, and SOD [67]. Using the same model, Kolahian et al. treated rats orally for 4 weeks with 5 mg/kg of CrPic3. This treatment revealed that CrPic3 significantly increased CAT, SOD, and GPX and decreased thiobarbituric acid reactive substances (TBARS) [68]. Similarly, Sundaram et al. also studied the antioxidant effects of a 4-week oral treatment with CrPic3 (1 mg/kg) in streptozotocin-induced diabetic rats, finding that it significantly increased GR, CAT, SOD, and GSH and decreased MDA [46]. Moreover, Jain et al. and Saiyed and Lugo studied the effects of orally administered CrPic3 in human patients from the USA, between 30 to 55 years old, with type 2 diabetes [69][70]. For this purpose, they used 25 (2 men and 23 women) and 43 (sex demographic not disclosed) subjects. In both clinical studies, the subjects were treated orally with 400 μg/day for 3 months [69][70]. While Jain et al. found that CrPic3 decreased protein carbonylation in a non-significant manner [69], Saiyed and Lugo, who performed a similar study, but with a larger group of subjects, showed that this decrease is actually significant [70].
All together, these data show that CrPic3 appears to exhibit antioxidant effects in cellular systems by enhancing the activity of antioxidant enzymes GSH, CAT, SOD, GPX, and GR, and decreasing lipid peroxidation, and protein carbonylation, thus mitigating damage caused by oxidative stress.

4.3. Possible Mechanisms for CrPic3 Toxicity

To the researchers' knowledge, no specific mechanism of toxicity has been described for either for Cr(III) alone or CrPic3, but there are some reports focused on the effects of these compounds (Figure 2). CrPic3 circulates in the blood bound to transferrin [71]. When it binds to its receptor on the cell membrane, it induces the endocytosis of CrPic3 [72]. Due to the low pH of the endosome, CrPic3 is separated from transferrin and is reduced by an unknown agent that causes the separation into Pic and four Cr(III) atoms and release of ROS [72]. Cr(III) atoms bind to ApoLMWCr, which becomes HoloLMWCr and exert its effects [72]. It is not clear whether Pic is involved in any mechanism of toxicity [73]. In the LC, Cr(III) is reported to be involved in the suppression of LHR, the decline of StAR activity, and the reduction of the level and/or activity of enzymes such as CYP11A1, CYP17A1, 3βHSD, and 17βHSD [74]. These effects have a possible impact on steroidogenesis by decreasing LH signaling, cholesterol entry into the mitochondria, its conversion to Preg and the reactions that occur in the SER. These effects would ultimately affect T production and all the processes that are dependent on this hormone. This also suggests that the mitochondria may be a target of CrPic3, since it affects mitochondrial enzymes and, as we will see later, it is involved in stress in this organelle.
Figure 2. Possible molecular mechanisms of Leydig cell (LC) damage and steroidogenesis impairment from chromium picolinate (CrPic3). 
Interestingly, CrPic3 affects cytokine production in a way that has been described to cause a decrease in IL-6 [52] and TNF-α [52][53]. Contrarily, others report that it increases TNF-α and IL-2 cytokines [75]. This is an important issue, since if pro-inflammatory cytokines are indeed increased, CrPic3 could affect steroid production, as these cytokines impact negatively steroidogenesis [76]. Indeed, two authors reported that TNF-α acts as a transcription inhibitor for genes involved in steroidogenesis, such as StAR, with Suescun and collaborators hinting at the possibility of the involvement of the TNFR1 pathway [77][78]. Hales et al. and Wang et al. also highlighted this suppressive behavior of IL-1β and IL-6, respectively, but no pathway has been proposed [78][79].
As mentioned earlier, it is suggested that part of the toxicity of Cr(VI) is caused by Cr(III). After Cr(VI) is reduced to Cr(III) in the cell, it enters the nucleus, where it unwinds the DNA and binds to it, forming Cr-DNA adducts [80]. Cr(III) binds to an N7 atom of a guanine, forming two types of complexes in the major grove: binary complexes, comprised of Cr(III) and DNA, or tertiary complexes, with a third molecule [81]. This molecule can be histidine or ascorbate. The histidine complex does not harm the cell [82], while the ascorbate complex crosslinks with DNA, causing strand breaks [26]. Furthermore, the replacement rate of Cr(III)-DNA adducts is low, which means that these can have permanent consequences [81].
CrPic3 causes signs of apoptosis in the ovarian cell line CHO AA8 when administered at 80 μg/cm2, for 48 h [73]. One of these indicators of apoptosis was the mitochondrial swelling and degradation of the cristae in a dose-dependent manner [73]. A recent study has demonstrated that this chromium compound increased the levels of caspase-8 and caspase-3 in the blood of Wistar rats orally treated with 0.3 mg/kg body weight for 8 weeks [75]. Caspase-8 is involved in the extrinsic pathway of apoptosis, while caspase-3 is involved in the intrinsic and extrinsic pathways [83]. In human peripheral blood lymphocytes, a treatment with 50 μM of CrPic3 increased expression of caspase 3 by 1.8- and 2.2-fold compared to the control after 24 h and 48 h, respectively. In these cells, 24 h of exposure to 100 μM of CrPic3 also increases the BAX/Blc-2 ratio, causes the collapse of the mitochondrial membrane potential, and triggers the displacement of cytochrome c into the cytoplasm, which was 3.2-fold that observed in the control condition. Furthermore, the authors concluded that the cytotoxicity of CrPic3 in the lymphocytes is centered around the mitochondria and oxidative stress caused by intracellular ROS [84]. CrPic3 also causes apoptosis by increasing BAX in HBL-100 human mammary epithelial cells after treatment with 10 μg/L for 6 h [85]. Additionally, another study used 25 ppm potassium dichromate as a source of Cr(III), rather than CrPic3, to study apoptosis in the offspring germ cells of rats after gestational exposure to drinking water containing this chromium compound, from day 9.5 to day 14.5 of gestation [86]. The observed effects included the upregulation of pro-apoptotic cascades p53/p27-BAX-Caspase-3 and p53-SOD2 and a decrease in the expression of anti-apoptotic proteins pAKT, pERK, and XIAP [86]. The p53 tumor suppressor is a pro-apoptotic gene, as well as a protein that is involved in pathways that cause cell death [87]. Regarding p27, it has an ambiguous effect on apoptosis, so it is necessary to clarify if this increase induced by CrPic3 is pro- or anti-apoptotic [88]. BAX then causes the release of cytochrome c from the mitochondria [89], which ends up activating caspase-3 [90]. The anti-apoptotic protein pAKT [91] is involved in the phosphoinositide 3-kinase (PI3K)/Akt pathway, which interrupts the progression of the cell cycle [92][93]. pERK and XIAP block apoptosis downstream of the mitochondria, with ERK being activated in phosphorylation by MEK and thus activating XIAP, which inhibits the effector caspases [94]. Hence, Cr(III) appears to have a pro-apoptotic role in the described cells. However, 200 ppb of CrPic3 was described to have no negative effect on the proliferation of the myoblast cell line C2C12 after 5 days of exposure, indicating that it is not an inducer of apoptosis in all cell types [95]. In various cell types, including ovary cells, lymphocytes, epithelial cells, and germ cells, CrPic3 seems to exhibit pro-apoptotic effects, potentially mediated by BAX, caspase-3, and -8, depending on the exposure conditions. However, these effects appear to be specific to each cell type. It's important to note that no research has yet examined the potential apoptotic impact of CrPic3 on LC.

4.4. Impact of CrPic3 on Male Fertility

The influence of CrPic3 has on male fertility is still not fully understood. In Table 1, the researchers summarize the main effects found in the literature.
Two studies identified damage in LC after treatment with CrPic3 (8 mg and 15 mg/kg body weight for 90 days in albino rats [96] and 0.250, 0.375, and 0.500 mg/animal/day for 84 days in Santa Inês lambs [97]). However, one of these studies suggested that this was an artifact, so more research is needed to clarify these results [97]. Contradictory results were found regarding the effects of CrPic3 in LC function. Of four papers that explored this topic, two have reported that the supplement decreases T production, LH levels, and the expression of enzymes involved in steroidogenesis, especially HSDs [96][98]. Contrarily, others state that CrPic3 increases T production [56][99]. These contrasting data may be a result of different study designs, which emphasizes the importance of further research to determine the effects on humans.
Table 1. Main effects of chromium picolinate (CrPic3) on male fertility.
Sperm cells may be harmed by CrPic3, particularly due to the degeneration of spermatids it causes [96]. It has been hypothesized that this may be due to ROS accumulation; however, the researchers hypothesize that if T levels are indeed reduced (also described in the entry), this may cause the detachment of spermatids from the Sertoli cells [102], which would decrease the number of cells that progress to spermatozoa. In spite of this, some sperm parameters are improved after diet supplementation with CrPic3, specifically sperm motility and abnormal sperm count. Indeed, the total and progressive motility of spermatozoa was increased in rabbit bucks treated with CrPic3 (0.009 mg/kg body weight for 63 days) [100]. Furthermore, abnormal sperm count decreased in rabbit bucks (0.009 mg/kg body weight for 63 days) [100] and breeding boars (0.08181 mg/kg of food) [101].

Collectively, these studies suggest that CrPic3 has the potential to influence both LC and sperm cells, yet its precise mechanism of action remains largely unexplored.

5. Conclusions

In summary, Chromium (Cr) falls under the category of EDCs due to its potential to disrupt endocrine signaling. The role of Cr(III) is a subject of ongoing debate, with discussions about whether it should be considered essential or hazardous. Despite this controversy, Cr(III), often in the form of CrPic3, is used as a nutritional supplement, primarily for its antioxidative and antidiabetic properties, among others. Regarding male fertility, CrPic3 seems to impact the expression of a receptor and certain enzymes involved in steroidogenesis within LC, which could potentially lead to a reduction in T levels. Additionally, Cr(III) has been observed to induce changes in the production of inflammatory cytokines, notably TNF-α, exhibit mutagenic and genotoxic effects, and increase apoptosis in various cell types. Should these effects be confirmed in LC, it is plausible that they could interfere with steroid production, ultimately affecting male fertility.

References

  1. Zegers-Hochschild, F.; Adamson, G.D.; de Mouzon, J.; Ishihara, O.; Mansour, R.; Nygren, K.; Sullivan, E.; Vanderpoel, S.; International Committee for Monitoring Assisted Reproductive Technologies; World Health Organization. International Committee for Monitoring Assisted Reproductive Technology (ICMART) and the World Health Organization (WHO) revised glossary of ART terminology, 2009. Fertil. Steril. 2009, 92, 1520–1524.
  2. Sharlip, I.D.; Jarow, J.P.; Belker, A.M.; Lipshultz, L.I.; Sigman, M.; Thomas, A.J.; Schlegel, P.N.; Howards, S.S.; Nehra, A.; Damewood, M.D.; et al. Best practice policies for male infertility. Fertil. Steril. 2002, 77, 873–882.
  3. Agarwal, A.; Mulgund, A.; Hamada, A.; Chyatte, M.R. A unique view on male infertility around the globe. Reprod. Biol. Endocrinol. 2015, 13, 37.
  4. Ma, Y.; He, X.; Qi, K.; Wang, T.; Qi, Y.; Cui, L.; Wang, F.; Song, M. Effects of environmental contaminants on fertility and reproductive health. Int. J. Environ. Sci. Technol. 2019, 77, 210–217.
  5. Yilmaz, B.; Terekeci, H.; Sandal, S.; Kelestimur, F. Endocrine disrupting chemicals: Exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev. Endocr. Metab. Disord. 2020, 21, 127–147.
  6. Badr, F.M.; El-Habit, O. Heavy Metal Toxicity Affecting Fertility and Reproduction of Males. In Bioenvironmental Issues Affecting Men’s Reproductive and Sexual Health; Elsevier: Amsterdam, The Netherlands, 2018; Volume 35, pp. 293–304.
  7. Concepcion-Zavaleta, M.; Paz Ibarra, J.L.; Ramos-Yataco, A.; Coronado-Arroyo, J.; Concepcion-Urteaga, L.; Roseboom, P.J.; Williams, C.A. Assessment of hormonal status in male infertility. An update. Diabetes Metab. Syndr. Clin. Res. Rev. 2022, 16, 102447.
  8. Carnegie, C. Diagnosis of Hypogonadism: Clinical Assessments and Laboratory Tests. Rev. Urol. 2004, 6, S3–S8.
  9. Ohlander, S.J.; Lindgren, M.C.; Lipshultz, L.I. Testosterone and Male Infertility. Urol. Clin. N. Am. 2016, 43, 195–202.
  10. Maret, W. Chromium Supplementation in Human Health, Metabolic Syndrome, and Diabetes; De Gruyter: Berlin, Germany; Boston, MA, USA, 2019; Volume 19, pp. 231–251.
  11. Huhtaniemi, I.; Teerds, K. Leydig Cells. In Encyclopedia of Reproduction, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 1, pp. 30–38.
  12. Chapin, R.E. Reproductive System, Male. In Encyclopedia of Toxicology; Elsevier: Amsterdam, The Netherlands, 2014; Volume 4, pp. 82–92.
  13. Nassar, G.N.; Leslie, S.W. Physiology, Testosterone; StatPearls Publishing: Treasure Island, FL, USA, 2022.
  14. Walker, W.H. Molecular mechanisms of testosterone action in spermatogenesis. Steroids 2009, 74, 602–607.
  15. Flück, C.E.; Pandey, A.V. Testicular Steroidogenesis. In Endocrinology of the Testis and Male Reproduction; Springer: Berlin/Heidelberg, Germany, 2017; pp. 343–371.
  16. Rone, M.B.; Fan, J.; Papadopoulos, V. Cholesterol transport in steroid biosynthesis: Role of protein–protein interactions and implications in disease states. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2009, 1791, 646–658.
  17. Haider, S.G. Cell Biology of Leydig Cells in the Testis; Elsevier: Amsterdam, The Netherlands, 2004; Volume 233.
  18. Hauet, T.; Yao, Z.X.; Bose, H.S.; Wall, C.T.; Han, Z.; Li, W.; Hales, D.B.; Miller, W.L.; Culty, M.; Papadopoulos, V. Peripheral-type benzodiazepine receptor-mediated action of steroidogenic acute regulatory protein on cholesterol entry into leydig cell mitochondria. Mol. Endocrinol. 2005, 19, 540–554.
  19. Grinspon, R.P.; Bergada, I.; Rey, R.A. Male Hypogonadism and Disorders of Sex Development. Front. Endocrinol. 2020, 11, 211–225.
  20. Lee, H.G.; Kim, C.J. Classic and backdoor pathways of androgen biosynthesis in human sexual development. Ann. Pediatr. Endocrinol. Metab. 2022, 27, 83–89.
  21. Grinspon, R.P.; Bergada, I.; Rey, R.A. Male Hypogonadism and Disorders of Sex Development. Front. Endocrinol. 2020, 11, 211–225.
  22. Simpson, J.L. Disorders of the Gonads, Genital Tract, and Genitalia. In Emery and Rimoin’s Principles and Practice of Medical Genetics; Academic Press: Cambridge, MA, USA, 2013; pp. 1–45.
  23. Genchi, G.; Lauria, G.; Catalano, A.; Carocci, A.; Sinicropi, M.S. The Double Face of Metals: The Intriguing Case of Chromium. Appl. Sci. 2021, 11, 638.
  24. Bagchi, D.; Stohs, S.; Downs, B.; Bagchi, M.; Preuss, H. Cytotoxicity and oxidative mechanisms of different forms of chromium. Toxicology 2002, 180, 5–22.
  25. Pure Earth. World’s Worst Pollution Problems: The New Top Six Toxic Threats: A Priority List for Remediation. 2015. Available online: https://www.worstpolluted.org/docs/WWP15.pdf (accessed on 15 July 2023).
  26. Quievryn, G.; Messer, J.; Zhitkovich, A. Carcinogenic Chromium(VI) Induces Cross-Linking of Vitamin C to DNA in Vitro and in Human Lung A549 Cells. Biochemistry 2002, 41, 3156–3167.
  27. Quievryn, G.; Goulart, M.; Messer, J.; Zhitkovich, A. Reduction of Cr (VI) by cysteine Significance in human lymphocytes and formation of DNA damage in reactions with variable reduction rates. Mol. Cell. Biochem. 2001, 222, 107–118.
  28. Zablon, H.; VonHandorf, A.; Puga, A. Chromium exposure disrupts chromatin architecture upsetting the mechanisms that regulate transcription. Exp. Biol. Med. 2019, 244, 752–757.
  29. Wu, Y.H.; Lin, J.C.; Wang, T.Y.; Lin, T.J.; Yen, M.C.; Liu, Y.H.; Wu, P.L.; Chen, F.W.; Shih, Y.L.; Yeh, I.J. Hexavalent chromium intoxication induces intrinsic and extrinsic apoptosis in human renal cells. Mol. Med. Rep. 2020, 21, 851–857.
  30. Handa, K.; Jindal, R. Estimating the hepatotoxic impact of hexavalent chromium on Ctenopharyngodon idellus through a multi-biomarker study. Environ. Adv. 2021, 5, 100108.
  31. Yatera, K.; Morimoto, Y.; Ueno, S.; Noguchi, S.; Kawaguchi, T.; Tanaka, F.; Suzuki, H.; Higashi, T. Cancer Risks of Hexavalent Chromium in the Respiratory Tract. J. UOEH 2018, 40, 157–172.
  32. Chandra, A.K.; Chatterjee, A.; Ghosh, R.; Sarkar, M.; Chaube, S.K. Chromium induced testicular impairment in relation to adrenocortical activities in adult albino rats. Reprod. Toxicol. 2007, 24, 388–396.
  33. Abbas, T.; Khawaja, A.; Qaisar, F.; Sajjad, A. Effects of chromium on testes and protective role of mulberry. J. Sheikh Zayed Med. Coll. 2017, 8, 1200–1204.
  34. Chandra, A.K.; Chatterjee, A.; Ghosh, R.; Sarkar, M. Vitamin E-supplementation protect chromium (VI)-induced spermatogenic and steroidogenic disorders in testicular tissues of rats. Food Chem. Toxicol. 2010, 48, 972–979.
  35. Schwarz, K.; Mertz, W. Chromium(III) and the glucose tolerance factor. Arch. Biochem. Biophys. 1959, 85, 292–295.
  36. McCarty, M. The therapeutic potential of glucose tolerance factor. Med. Hypotheses 1980, 6, 1177–1189.
  37. Lin, C.C.; Tsweng, G.J.; Lee, C.F.; Chen, B.H.; Huang, Y.L. Magnesium, zinc, and chromium levels in children, adolescents, and young adults with type 1 diabetes. Clin. Nutr. 2016, 35, 880–884.
  38. Stearns, D.M. Is chromium a trace essential metal? Biofactors 2000, 11, 149–162.
  39. Vincent, J.B. Chromium: Celebrating 50 years as an essential element? Dalton Trans. 2010, 39, 3787–3794.
  40. Casalegno, C.; Schifanella, O.; Zennaro, E.; Marroncelli, S.; Briant, R. Collate literature data on toxicity of Chromium (Cr) and Nickel (Ni) in experimental animals and humans. EFSA Support. Publ. 2015, 12, 478E.
  41. Filippini, T.; Cilloni, S.; Malavolti, M.; Violi, F.; Malagoli, C.; Tesauro, M.; Bottecchi, I.; Ferrari, A.; Vescovi, L.; Vinceti, M. Dietary intake of cadmium, chromium, copper, manganese, selenium and zinc in a Northern Italy community. J. Trace Elem. Med. Biol. 2018, 50, 508–517.
  42. Witkamp, R. Biologically Active Compounds in Food Products and Their Effects on Obesity and Diabetes. In Chemistry, Molecular Sciences and Chemical Engineering; Elsevier: Amsterdam, The Netherlands, 2010; Volume 3, pp. 509–545.
  43. Komorowski, J.R.; Tuzcu, M.; Sahin, N.; Juturu, V.; Orhan, C.; Ulas, M.; Sahin, K. Chromium picolinate modulates serotonergic properties and carbohydrate metabolism in a rat model of diabetes. Biol. Trace Elem. Res. 2012, 149, 50–56.
  44. McLeod, M.; Golden, R. Chromium treatment of depression. Int. J. Neuropsychopharmacol. 2000, 3, 311–314.
  45. Sahin, K.; Tuzcu, M.; Orhan, C.; Sahin, N.; Kucuk, O.; Ozercan, I.H.; Juturu, V.; Komorowski, J.R. Anti-diabetic activity of chromium picolinate and biotin in rats with type 2 diabetes induced by high-fat diet and streptozotocin. Br. J. Nutr. 2013, 110, 197–205.
  46. Sundaram, B.; Aggarwal, A.; Sandhir, R. Chromium picolinate attenuates hyperglycemia-induced oxidative stress in streptozotocin-induced diabetic rats. J. Trace Elem. Med. Biol. 2013, 27, 117–121.
  47. Reed, J.; Bain, S.; Kanamarlapudi, V. A Review of Current Trends with Type 2 Diabetes Epidemiology, Aetiology, Pathogenesis, Treatments and Future Perspectives. Diabetes Metab. Syndr. Obes. Targets Ther. 2021, 14, 3567–3602.
  48. Martin, J.; Wang, Z.Q.; Zhang, X.H.; Wachtel, D.; Volaufova, J.; Matthews, D.E.; Cefalu, W.T. Chromium Picolinate Supplementation Attenuates Body Weight Gain and Increases Insulin Sensitivity in Subjects with Type 2 Diabetes. Diabetes Care 2006, 29, 1826–1832.
  49. Dong, F.; Kandadi, M.; Ren, J.; Sreejayan, N. Chromium (D-phenylalanine)3 Supplementation Alters Glucose Disposal, Insulin Signaling, and Glucose Transporter-4 Membrane Translocation in Insulin-Resistant Mice. J. Nutr. 2008, 138, 1846–1851.
  50. Wang, Y.Q.; Yao, M.H. Effects of chromium picolinate on glucose uptake in insulin-resistant 3T3-L1 adipocytes involve activation of p38 MAPK. J. Nutr. Biochem. 2009, 20, 982–991.
  51. Chen, W.Y.; Chen, C.J.; Liu, C.H.; Mao, F.C. Chromium supplementation enhances insulin signalling in skeletal muscle of obese KK/HlJ diabetic mice. Diabetes Obes. Metab. 2009, 11, 293–303.
  52. Jain, S.K.; Rains, J.L.; Croad, J.L. Effect of chromium niacinate and chromium picolinate supplementation on lipid peroxidation, TNF-alpha, IL-6, CRP, glycated hemoglobin, triglycerides, and cholesterol levels in blood of streptozotocin-treated diabetic rats. Free Radic. Biol. Med. 2007, 43, 1124–1131.
  53. Imanparasta, F.; Javaheric, J.; Kamankesha, F.; Rafieid, F.; Salehie, A.; Mollaaliakbaria, Z.; Rezaeia, F.; Rahimif, A.; Abbasig, E. The effects of chromium and vitamin D3 co-supplementation on insulin resistance and tumor necrosis factor-alpha in type 2 diabetes a randomized placebo-controlled trial. Appl. Physiol. Nutr. Metab. 2020, 45, 471–477.
  54. Rui, L.; Aguirre, V.; Kim, J.K.; Shulman, G.I.; Lee, A.; Corbould, A.; Dunaif, A.; White, M.F. Insulin/IGF-1 and TNF-alpha stimulate phosphorylation of IRS-1 at inhibitory Ser307 via distinct pathways. J. Clin. Investig. 2001, 107, 181–189.
  55. Suksomboon, N.; Poolsup, N.; Yuwanakorn, A. Systematic review and meta-analysis of the efficacy and safety of chromium supplementation in diabetes. J. Clin. Pharm. Ther. 2014, 39, 292–306.
  56. Mehrim, A.I. Physiological, biochemical and histometric responses of Nile tilapia (Oreochromis niloticus L.) by dietary organic chromium (chromium picolinate) supplementation. J. Adv. Res. 2014, 5, 303–310.
  57. Pattar, G.R.; Tackett, L.; Liu, P.; Elmendorf, J.S. Chromium picolinate positively influences the glucose transporter system via affecting cholesterol homeostasis in adipocytes cultured under hyperglycemic diabetic conditions. Mutat. Res. 2006, 610, 93–100.
  58. Shahinfar, H.; Amini, M.R.; Sheikhhossein, F.; Djafari, F.; Jafari, A.; Shab-Bidar, S. The effect of chromium supplementation on apolipoproteins: A systematic review and meta-analysis of randomized clinical trials. Clin. Nutr. ESPEN 2020, 40, 34–41.
  59. Dominiczak, M.H.; Caslake, M.J. Apolipoproteins: Metabolic role and clinical biochemistry applications. Ann. Clin. Biochem. 2011, 48, 498–515.
  60. Zhong, O.; Ji, L.; Wang, J.; Lei, X.; Huang, H. Association of diabetes and obesity with sperm parameters and testosterone levels: A meta-analysis. Diabetol. Metab. Syndr. 2021, 13, 109.
  61. Alves, M.G.; Oliveira, P. Diabetes Mellitus and male reproductive function: Where we stand? Int. J. Diabetol. Vasc. Dis. Res. 2023, 1, 1–2.
  62. Meneses, M.J.; Sousa, M.; Alves, M.; Oliveira, P. The Antidiabetic Drug Metformin and Male Reproductive Function: An Overview. Int. J. Diabetol. Vasc. Dis. Res. 2015, 1, 1–2.
  63. Vona, R.; Pallotta, L.; Cappelletti, M.; Severi, C.; Matarrese, P. The Impact of Oxidative Stress in Human Pathology: Focus on Gastrointestinal Disorders. Antioxidants 2021, 10, 201.
  64. Darbandi, M.; Darbandi, S.; Agarwal, A.; Sengupta, P.; Durairajanayagam, D.; Henkel, R.; Sadeghi, M.R. Reactive oxygen species and male reproductive hormones. Reprod. Biol. Endocrinol. 2018, 16, 87.
  65. Rato, L.; Oliveira, P.F.; Sousa, M.; Silva, B.M.; Alves, M.G. Chapter 2.6—Role of Reactive Oxygen Species in Diabetes-Induced Male Reproductive Dysfunction. In Oxidants, Antioxidants and Impact of the Oxidative Status in Male Reproduction; Henkel, R., Samanta, L., Agarwal, A., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 135–147.
  66. Doddigarla, Z.; Parwez, I.; Abidi, S.; Ahmad, J. Effect of Chromium Picolinate and Melatonin either in Single or in a Combination in Alloxan Induced Male Wistar Rats. J. Biomed. Sci. 2017, 6.
  67. Al-Bishri, W.M. Attenuating impacts of chromium and nano resveratrol against hyperglycemia induced oxidative stress in diabetic rats. Int. J. Pharm. Res. Allied Sci. 2017, 6, 61–69.
  68. Kolahian, S.; Sadri, H.; Shahbazfar, A.A.; Amani, M.; Mazadeh, A.; Mirani, M. The Effects of Leucine, Zinc, and Chromium Supplements on Inflammatory Events of the Respiratory System in Type 2 Diabetic Rats. PLoS ONE 2015, 10, e0133374.
  69. Jain, S.K.; Kahlon, G.; Morehead, L.; Dhawan, R.; Lieblong, B.; Stapleton, T.; Caldito, G.; Hoeldtke, R.; Levine, S.N.; Bass III, P.F. Effect of chromium dinicocysteinate supplementation on circulating levels of insulin, TNF-α, oxidative stress, and insulin resistance in type 2 diabetic subjects: Randomized, double-blind, placebo-controlled study. Mol. Nutr. Food Res. 2012, 56, 1333–1341.
  70. Saiyed, Z.M.; Lugo, J.P. Impact of chromium dinicocysteinate supplementation on inflammation, oxidative stress, and insulin resistance in type 2 diabetic subjects: An exploratory analysis of a randomized, double-blind, placebo-controlled study. Food Nutr. Res. 2016, 60, 31762.
  71. Clodfelder, B.J.; Vincent, J.B. The time-dependent transport of chromium in adult rats from the bloodstream to the urine. J. Biol. Inorg. Chem. 2005, 10, 383–393.
  72. Sun, Y.; Ramirez, J.; Woski, S.A.; Vincent, J.B. The binding of trivalent chromium to low-molecular-weight chromium-binding substance (LMWCr) and the transfer of chromium from transferrin and chromium picolinate to LMWCr. JBIC J. Biol. Inorg. Chem. 2000, 5, 129–136.
  73. Manygoats, K.R.; Yazzie, M.; Stearns, D.M. Ultrastructural damage in chromium picolinate-treated cells: A TEM study. Transmission electron microscopy. J. Biol. Inorg. Chem. 2002, 7, 791–798.
  74. Navin, A.K.; Aruldhas, M.M.; Navaneethabalakrishnan, S.; Mani, K.; Michael, F.M.; Srinivasan, N.; Banu, S.K. Prenatal exposure to hexavalent chromium disrupts testicular steroidogenic pathway in peripubertal F(1) rats. Reprod. Toxicol. 2021, 101, 63–73.
  75. Dworzanski, W.; Sembratowicz, I.; Cholewinska, E.; Tutaj, K.; Fotschki, B.; Juskiewicz, J.; Ognik, K. Effects of Different Chromium Compounds on Hematology and Inflammatory Cytokines in Rats Fed High-Fat Diet. Front. Immunol. 2021, 12, 614000.
  76. Leisegang, K.; Henkel, R. The in vitro modulation of steroidogenesis by inflammatory cytokines and insulin in TM3 Leydig cells. Reprod. Biol. Endocrinol. 2018, 16, 26.
  77. Suescun, M.O.; Rival, C.; Theas, M.S.; Calandra, R.S.; Lustig, L. Involvement of tumor necrosis factor-α in the pathogenesis of autoimmune orchitis in rats. Biol. Reprod. 2003, 68, 2114–2121.
  78. Hales, D.B. Testicular macrophage modulation of Leydig cell steroidogenesis. J. Reprod. Immunol. 2002, 57, 3–18.
  79. Wang, Y.; Chen, L.; Xie, L.; Li, L.; Li, X.; Li, H.; Liu, J.; Chen, X.; Mao, B.; Song, T. Interleukin 6 inhibits the differentiation of rat stem Leydig cells. Mol. Cell. Endocrinol. 2018, 472, 26–39.
  80. Blankert, S.A.; Coryell, V.H.; Picard, B.T.; Wolf, K.K.; Lomas, R.E.; Stearns, D.M. Characterization of Nonmutagenic Cr(III)-DNA Interactions. Chem. Res. Toxicol. 2003, 16, 847–854.
  81. Brown, S.; Lockart, M.M.; Thomas, C.S.; Bowman, M.K.; Woski, S.A.; Vincent, J.B. Molecular Structure of Binary Chromium(III)-DNA Adducts. ChemBioChem 2020, 21, 628–631.
  82. Lankford, E.; Thomas, C.S.; Marchi, S.; Brown, S.; Woski, S.A.; Vincent, J.B. Examining the Potential Formation of Ternary Chromium-Histidine-DNA Complexes and Implications for Their Carcinogenicity. Biol. Trace Elem. Res. 2022, 200, 1473–1481.
  83. Xu, G.; Shi, Y. Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 2007, 17, 759–771.
  84. Jana, M.; Rajaram, A.; Rajaram, R. Chromium picolinate induced apoptosis of lymphocytes and the signaling mechanisms thereof. Toxicol. Appl. Pharmacol. 2009, 237, 331–344.
  85. Dębski, B.; Lamparska-Przybysz, M.; Gajewska, M. Influence of Cr(III)-picolinate, and Cr(III)-nicotinate on apoptosis induction in HBL-100 human mammary epithelial cells. Probl. Hig. Epidemiol. 2016, 97, 95–99.
  86. Sivakumar, K.K.; Stanley, J.A.; Arosh, J.A.; Pepling, M.E.; Burghardt, R.C.; Banu, S.K. Prenatal exposure to chromium induces early reproductive senescence by increasing germ cell apoptosis and advancing germ cell cyst breakdown in the F1 offspring. Dev. Biol. 2014, 388, 22–34.
  87. Shen, Y.; White, E. p53-Dependent Apoptosis Pathways. Adv. Cancer Res. 2001, 82, 55–84.
  88. Abbastabar, M.; Kheyrollah, M.; Azizian, K.; Bagherlou, N.; Tehrani, S.S.; Maniati, M.; Karimian, A. Multiple functions of p27 in cell cycle, apoptosis, epigenetic modification and transcriptional regulation for the control of cell growth: A double-edged sword protein. DNA Repair 2018, 69, 63–72.
  89. Pena-Blanco, A.; Garcia-Saez, A.J. Bax, Bak and beyond–mitochondrial performance in apoptosis. FEBS J. 2018, 285, 416–431.
  90. Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S.M.; Ahmad, M.; Alnemri, E.S.; Wang, X. Cytochrome c and dATP-Dependent Formation of Apaf-1/Caspase-9 Complex Initiates an Apoptotic Protease Cascade. Cell 1997, 91, 479–489.
  91. Kim, A.H.; Khursigara, G.; Sun, X.; Franke, T.F.; Chao, M.V. Akt phosphorylates and negatively regulates apoptosis signal-regulating kinase 1. Mol. Cell. Biol. 2001, 21, 893–901.
  92. Chang, F.; Lee, J.T.; Navolanic, P.M.; Steelman, L.S.; Shelton, J.G.; Blalock, W.L.; Franklin, R.A.; McCubrey, J.A. Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: A target for cancer chemotherapy. Leukemia 2003, 17, 590–603.
  93. Franke, T.F.; Hornik, C.P.; Segev, L.; Shostak, G.A.; Sugimoto, C. PI3K/Akt and apoptosis: Size matters. Oncogene 2003, 22, 8983–8998.
  94. Gardai, S.J.; Whitlock, B.B.; Xiao, Y.Q.; Bratton, D.B.; Henson, P.M. Oxidants inhibit ERK/MAPK and prevent its ability to delay neutrophil apoptosis downstream of mitochondrial changes and at the level of XIAP. J. Biol. Chem. 2004, 279, 44695–44703.
  95. Tsa, M.C.; Lien, T.F. Chromium Picolinate did not Effect on the Proliferation and Differentiation of Myoblasts. Am. J. Anim. Vet. Sci. 2007, 2, 79–83.
  96. Rehab, M.E.; Ashraf, M.E. The protective effect of Panax ginseng against chromium picolonate induced testicular changes. Afr. J. Pharm. Pharmacol. 2014, 8, 346–355.
  97. Dallago, B.S.L.; Braz, S.; Marçola, T.G.; McManus, C.; Caldeira, D.F.; Campeche, A.; Gomes, E.F.; Paim, T.P.; Borges, B.O.; Louvandini, H. Blood Parameters and Toxicity of Chromium Picolinate Oral Supplementation in Lambs. Biol. Trace Elem. Res. 2015, 168, 91–102.
  98. Zakaria, A.D.; Fayed, A.H.; Hedaya, S.A.; Gad, S.B.; Hafez, M.H. Effect of chromium Picolinate on some reproductive aspects in male rats. Alex. J. Vet. Sci. 2011, 34, 113–124.
  99. Ezzat, W. Effect of supplementing diet with sodium bentonite and/or organic chromium on productive, physiological performance and immune response of matrouh chickens strain. 1- during growth period. Egypt. Poult. Sci. J. 2016, 36, 841–857.
  100. Dorra, T.; El-Serwy, A.; Ismail, F.; Nasif, A. Physical semen characteristics of california rabbit bucks administrated with different chromium levels. J. Anim. Poult. Prod. 2007, 32, 8931–8941.
  101. Horký, P.; Jančíková, P.; Zeman, L. The effect of a supplement of chromium (picolinate) on the level of blood glucose, insulin activity and changes in laboratory evaluation of the ejaculate of breeding boars. Acta Univ. Agric. Silvic. Mendel. Brun. 2013, 60, 49–56.
  102. Walker, W.H. Non-classical actions of testosterone and spermatogenesis. Philos. Trans. R. Soc. B Biol. Sci. 2010, 365, 1557–1569.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 171
Revisions: 3 times (View History)
Update Date: 11 Aug 2023
1000/1000
Video Production Service