Skip to main content
  • Research article
  • Open access
  • Published:

Zinc antagonizes iron-regulation of tyrosine hydroxylase activity and dopamine production in Drosophila melanogaster

Abstract

Background

Dopamine (DA) is a neurotransmitter that plays roles in movement, cognition, attention, and reward responses, and deficient DA signaling is associated with the progression of a number of neurological diseases, such as Parkinson’s disease. Due to its critical functions, DA expression levels in the brain are tightly controlled, with one important and rate-limiting step in its biosynthetic pathway being catalyzed by tyrosine hydroxylase (TH), an enzyme that uses iron ion (Fe2+) as a cofactor. A role for metal ions has additionally been associated with the etiology of Parkinson’s disease. However, the way dopamine synthesis is regulated in vivo or whether regulation of metal ion levels is a component of DA synthesis is not fully understood. Here, we analyze the role of Catsup, the Drosophila ortholog of the mammalian zinc transporter SLC39A7 (ZIP7), in regulating dopamine levels.

Results

We found that Catsup is a functional zinc transporter that regulates intracellular zinc distribution between the ER/Golgi and the cytosol. Loss-of-function of Catsup leads to increased DA levels, and we showed that the increased dopamine production is due to a reduction in zinc levels in the cytosol. Zinc ion (Zn2+) negatively regulates dopamine synthesis through direct inhibition of TH activity, by antagonizing Fe2+ binding to TH, thus rendering the enzyme ineffective or non-functional.

Conclusions

Our findings uncovered a previously unknown mechanism underlying the control of cellular dopamine expression, with normal levels of dopamine synthesis being maintained through a balance between Fe2+ and Zn2+ ions. The findings also provide support for metal modulation as a possible therapeutic strategy in the treatment of Parkinson’s disease and other dopamine-related diseases.

Background

Parkinson’s disease (PD) is one of the most common neurodegenerative diseases with clinical manifestation of motor dysfunctions in the form of tremor, rigidity, and bradykinesia. A hallmark of PD is the progressive dopaminergic neuron death mainly in the ventral midbrain substantia nigra pars compacta (SNc) [1]. The exact molecular cause underlying neural cell death in PD has not been fully elucidated, and no cure is available. It has long been believed that dopamine (DA) deficit, resulted from dopaminergic neuron death, is a key culprit of PD. DA is the key signaling neurotransmitter molecule critical to body movement, cognition, attention, and reward response [2]. Besides PD, abnormal dopaminergic signaling has also been implicated in a number of other diseases, including attention deficit hyperactivity disorder, psychosis, schizophrenia, and depression [35]. The entire pathway of DA synthesis has been well established, and the initial and rate-limiting step is catalyzed by the tyrosine hydroxylase (TH) [6], a metal-dependent enzyme that uses Fe2+ as the cofactor. TH activity is known to be regulated by a complex mechanism including phosphorylation [7]. Since alteration in TH activity and DA synthesis/signaling has been found to be associated with PD and other neurological disorders [8], modulation of TH activity or DA signaling has been proposed as a potential therapeutic strategy for the treatment of PD [9, 10].

There are ample evidences indicative of a role for metal ions in PD etiology [11, 12], consistent with the fact that the quintessential TH in DA synthesis is a Fe2+-dependent enzyme. Clinic manifestation of manganism, a disorder of manganese toxicity, resembles idiopathic PD as well as other neurodegenerative diseases such as Lou Gehrig’s disease and multiple sclerosis [13]. In the dopaminergic neurons of PD patients, the iron level was found to be increased in the substantia nigra but decreased in the globus pallidus [14, 15], and zinc in contrast was abnormally accumulated in the region [15]. Feeding rats with excess zinc was found to trigger degeneration of dopaminergic neurons and decreased DA levels [16, 17], implying abnormally high zinc levels in the dopaminergic neurons of PD patients might be causative. Interestingly, iron content in PD patients’ hair was significantly lowered, and zinc content in contrast was increased [18], suggesting a possible alteration and abnormal interplay between these two metal ions in these patients. Whether or not these metal-related defects are part of the underlying cause or aftermath consequences of PD remains to be more affirmatively determined. Furthermore, how these different metal ions might contribute to PD lacks a unified mechanistic understanding.

We have long been interested in understanding how metal ions might contribute to and influence PD and other neurodegenerative diseases. We were particularly interested in a Drosophila mutant named Catecholamines up (Catsup). As the name indicates, in Catsup mutant flies, the DA level was found to be substantially elevated [19]. The corresponding protein Catsup encoded by Catsup/CG10449 has been characterized as a negative regulator of TH activity during catecholamine biosynthesis [19]. Loss of Catsup function was shown to delay the onset of neurological symptoms, dopaminergic neuron death, and morbidity of flies upon paraquat exposure [20]. Catsup has also been found to participate in the synaptic vesicle loading and release of DA [21], the regulation of tracheal morphogenesis [22], the control of sleep behavior in Drosophila [23], and the maintenance of ER/Golgi function [24]. Interestingly, Catsup is homologous to the mammalian zinc transporter ZIP7 protein (also known as SLC39A7) [25], a member of the ZIP (Zrt/Irt-like protein) family [26]. Mammalian ZIP7 is also known to be localized in the ER and functions in mobilizing zinc from ER into the cytosol [2629]. It is therefore possible that the Drosophila Catsup might function as a zinc transporter somehow exerting regulatory control of DA synthesis, and this likely scenario could explain how zinc may contribute to dopaminergic neural degeneration.

In the current study, we report both genetic and biochemical studies demonstrating that Catsup is indeed a functional zinc transporter participating in zinc homeostasis in D. melanogaster. Catsup knockdown leads to zinc reduction in the cytosol. More importantly, we demonstrated that cytosolic zinc reduction triggered TH activation and DA increase. We further showed that zinc affects TH activity thus DA synthesis by antagonizing Fe2+, the indispensable cofactor of TH. Our study has uncovered a previously unknown mechanism underlying the regulation of DA synthesis. The finding provides a mechanistic framework to understand how zinc and iron ions could affect DA synthesis therefore contribute to PD. Our finding also points to the utility of zinc limitation as a possible novel therapeutic strategy in the treatment of PD and implies a role of metal homeostasis in the regulation of mood or behavior of animals.

Results

Catsup is a zinc transporter functionally analogous to human ZIP7

There are at least ten putative ZIPs in the D. melanogaster genome [3033]. Among them, Catsup/CG10449 shares the highest overall homology with human ZIP7 (hZIP7, also known as SLC39A7, 52% identity and 64% similarity), which functions as a zinc transporter located on the ER/Golgi [26]. Under normal dietary conditions, ubiquitous RNAi of Catsup by daughterless Da-Gal4 produced developmental arrest at the embryonic stage (0% eclosion rate) (Fig. 1A). This eclosion defect resulting from Catsup knockdown could be effectively rescued by hZIP7 from 0 to 83% (Fig. 1A). As a control, expression of dZIP13/CG7816 (also named Zip99C), a close homolog of hZIP7 but previously shown as an iron transporter located on the ER/Golgi [30], had no rescuing effect on Catsup RNAi (Fig. 1A). When Catsup was knocked down in the central nervous system (CNS), directed by the pan-neuronal Elav-Gal4 [34], the flies exhibited early adult death or much reduced lifespan at 25 °C (Fig. 1B). The effects of hZIP7 and dZIP13 overexpression (OE) on the lifespan of Catsup RNAi were also analyzed. hZIP7 exhibited a strong rescuing effect, from ~ 8 to ~ 32 days, on the medium lifespans of Catsup RNAi flies raised at 25 °C, whereas dZIP13 OE had no effect (Fig. 1B). When Catsup was knocked down with Elav-Gal4, blackspots (cell death) appeared in the eyes (Fig. 1C). Consistent with the results described above, the blackspots were also markedly rescued by hZIP7 but not dZIP13 OE (Fig. 1C). The secretory enzyme alkaline phosphatase (ALP) activity is dependent on zinc loading in the Golgi, so its activity is responsive to zinc [35, 36]. In order to test whether zinc level was affected by Catsup in neurons, we tested the ALP activity of Catsup RNAi flies driven by Elav-Gal4 (Fig. 1D). The results indicated that Catsup RNAi in neurons led to zinc elevation in the Golgi.

Fig. 1
figure 1

Catsup is the Drosophila ortholog of hZIP7 and functions as a zinc transporter. A The eclosion defect from ubiquitous Catsup RNA interference (Da-Gal4>Catsup RNAi) could be effectively rescued by hZIP7 expression, but not by dZIP13 OE. Genotypes of the flies used were Da-Gal4> w1118 (control); Da-Gal4>Catsup RNAi; Da-Gal4>Catsup RNAi, hZIP7; Da-Gal4>Catsup RNAi, dZIP13 OE. All values are presented as mean ± SEM; n = 6. ***p < 0.001. B The shortened lifespan of Catsup RNAi in the central nervous system was rescued by hZIP7, but not dZIP13 OE. Genotypes of the flies used were Elav-Gal4> w1118 (control); Elav-Gal4>Catsup RNAi; Elav-Gal4>Catsup RNAi, hZIP7; Elav-Gal4>Catsup RNAi, dZIP13 OE. n = 6.C hZIP7, but not dZIP13 OE, restored the eye blackspots of Catsup RNAi flies. Genotypes of the flies used were Elav-Gal4> w1118 (control); Elav-Gal4>Catsup RNAi; Elav-Gal4>Catsup RNAi, hZIP7; Elav-Gal4>Catsup RNAi, dZIP13 OE. n ≥ 6. D The ALP activity, indicated by absorbance at 405 nm, was increased in the heads of Catsup RNAi flies when driven by the pan-neuronal driver Elav-Gal4, suggesting that Catsup RNAi in neurons led to zinc elevation in the Golgi. Genotypes of the flies used were Elav-Gal4> w1118 (control) and Elav-Gal4>Catsup RNAi. E, F The localization of Catsup in Chinese hamster ovary (CHOK1) cells. Catsup-FLAG was detected by anti-FLAG antibody. This immunofluorescence staining showed that Catsup partially co-localizes with the ER/Golgi in CHOK1 cells

To examine the intracellular position of Catsup, a C-terminal flag-tagged Catsup was introduced into the Chinese hamster ovary (CHO-K1) cells. Immunofluorescence staining indicated that Catsup partially co-localizes with the ER/Golgi markers in CHO-K1 cells (Fig. 1E, F), consistent with the reported mammalian ZIP7 subcellular location [26]. Previous fluorescence imaging with the zinc indicator Zinpyr-1 has shown decreased zinc accumulation in the Golgi compartment in CHO-K1 cells expressing Catsup [37]. Based on the above results, that is, sequence homology, functional substitution, and subcellular localization, as well as roles in subcellular zinc distribution, it is reasonable to conclude that Catsup, like hZIP7, is a zinc transporter that mediates the transport of zinc from the ER/Golgi apparatus to the cytoplasm of the cell.

Catsup loss results in detrimental organellar zinc accumulation and cytosolic zinc reduction

Catsup functions as a transporter that redistributes zinc from the ER/Golgi to the cytosol. While Catsup reduction is associated with increased DA production, more severe loss of Catsup activity is lethal to the larvae [14]. In fact, ubiquitous Catsup knockdown produced no adult flies (Fig. 1B), and strong loss-of-function Castup mutant alleles have to be maintained in the heterozygous state with the wild-type allele [19]. This lethality could arise, directly or indirectly, from zinc dyshomeostasis either from zinc accumulation in the ER/Golgi and other organelles where Catsup is likely located, or zinc deficiency in the cytosol. In order to distinguish these two possibilities, we checked whether the phenotypes of Catsup RNAi could be rescued or aggravated by other zinc transporters. Catsup was specifically knocked down in the dopaminergic cells with the TH-Gal4 driver [38]. Approximately 63% of these Catsup RNAi flies were able to eclose compared to the control (near 100%), and their lethality could be rescued by hZIP7 (Fig. 2A). If Castup RNAi lethality had been due to cytosolic zinc reduction, we expected that overexpression of dZnT1/CG17723 (also named ZnT63C) [39, 40], a well-established plasma membrane zinc transporter, would exacerbate the toxicity of Catsup knockdown. Contrary to this notion, dZnT1 OE, which should reduce intracellular zinc level, in fact alleviated the lethality of Catsup RNAi (Fig. 2A). As controls, the eclosion of dZnT1 OE or hZIP7 driven by TH-Gal4 showed no defects (Fig. 2A). This suggests that the death from Catsup RNAi came from zinc toxicity, i.e., compartmental zinc accumulation or associated processes such as trafficking in the secretory pathway. Indeed, ER stress has been well noted in previous studies after hZIP7 loss [24, 41].

Fig. 2
figure 2

Catsup knockdown in dopaminergic neurons results in not only zinc but also iron sensitivities due to compartmentalized zinc toxicity. A The decreased eclosion defect of Catsup RNAi flies could be significantly rescued by hZIP7 or dZnT1 OE. Genotypes of the flies used were TH-Gal4> w1118 (control); TH-Gal4>Catsup RNAi; TH-Gal4>Catsup RNAi, dZnT1 OE; TH-Gal4>Catsup RNAi, hZIP7. All values are presented as mean ± SEM; n = 6. ***p < 0.001. B The decreased eclosion defect of Catsup RNAi flies could be significantly influenced by dietary zinc or iron. The eclosion defect became deteriorated after ZnCl2 and FAC supplementation. Genotypes of the flies used were TH-Gal4> w1118 (control) and TH-Gal4>Catsup RNAi. All values are presented as mean ± SEM; n = 6. *p < 0.05, **p < 0.01, ***p < 0.001

The sensitivity to zinc of Catsup knockdown (TH-Gal4>Catsup RNAi) was further confirmed with a dietary zinc supplement. Supplementation of additional ZnCl2 significantly worsened the survival defect from 64 to 39% (Fig. 2B).

TH activity enhancement in Catsup mutants arises from cytosolic zinc reduction instead of organelle zinc stress

Catsup is responsible for intracellular zinc redistribution between the cytosol and the ER/Golgi. As mentioned, the disturbance of this distribution causes lethality as well as elevated DA production. Zinc accumulation in the secretory compartments is the basis for lethality. But what causes the DA increase? Is the observed elevated TH activity due to zinc elevation in the secretory compartments, or zinc reduction in the cytosol, or both? We first confirmed that TH activity is indeed modulated by redistributing intracellular zinc. This is meaningful since it could be argued that Catsup might be associated with some unidentified novel property other than zinc transport. dZnT7 is a well-characterized ER/Golgi-resident zinc transporter which functions in the opposite direction to Catsup [42]. Catsup RNAi and dZnT7 OE both reduce cytosolic zinc and increase ER/Golgi zinc levels. When driven with the dopaminergic neuron driver TH-Gal4, Catsup RNAi and dZnT7 OE exhibited respectively 300% and 200% more TH activity in the head, as compared to the control (Fig. 3A). Consistently, DA levels in the Catsup RNAi and dZnT7 OE fly heads increased nearly 100% as compared to the control (Fig. 3B). This further strengthens the conclusion that increased TH activity in Catsup mutant flies is due to zinc dyshomeostasis, instead of some possible novel property other than zinc transport associated with dZIP7 (Catsup).

Fig. 3
figure 3

TH activity elevation in Catsup RNAi is due to cytosolic zinc reduction. A Either Catsup RNAi or dZnT7 OE in the dopaminergic neurons significantly elevated TH activity. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>dZnT7 OE, TH-Gal4>dZnT7 RNAi, and TH-Gal4>Catsup OE. All values are presented as mean ± SEM and included in Additional file 2: table S1; n = 4. **p < 0.01, ***p < 0.001. B DA level was elevated in Catsup RNAi and dZnT7 OE fly heads. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>dZnT7 OE, TH-Gal4>dZnT7 RNAi, and TH-Gal4>Catsup OE. All values are presented as mean ± SEM and included in Additional file 2: table S2; n ≥ 3. ***p < 0.001. C Expression of zinc exporters enabled resistance to 3-IT, a dopamine synthesis inhibiter. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>dZnT7 OE, and TH-Gal4>dZnT1 OE. All values are presented as mean ± SEM; n = 6. *p < 0.05, **p < 0.01, ***p < 0.001

We then asked whether the zinc accumulation in the secretory compartments or the accompanied cytosolic zinc reduction would enhance DA synthesis. It is known that the lack of DA would lead to death in the flies. Indeed, when the fly was fed with the TH inhibitor 3-iodotyrosine (3-IT), the endogenous DA pool was significantly depleted [20] and the viability strongly suppressed (Fig. 3C). As expected, Catsup RNAi and dZnT7 OE significantly rescued the viability (Fig. 3C). If the effect of Catsup on TH is through cytosolic zinc reduction, we would expect that the expression of the plasma membrane-resident zinc exporter dZnT1 also confers a rescue. Consistent with this notion, dZnT1 OE in the brain enabled more survivals under 3-IT treatment (Fig. 3C). This argues against the model that zinc accumulation in the ER/Golgi enhanced TH activity.

Catsup loss may affect TH activity independent of TH phosphorylation and BH4 synthesis

Protein phosphorylation is one of the major mechanisms so far reported regulating TH activity [43]. Drosophila TH is encoded by the gene named pale [44, 45]. Bioinformatics analysis showed that Drosophila TH is similar to human TH [44, 46], sharing 54% identity and 74% similarity (Fig. 4A). The lack of commercial antibodies able to recognize phosphorylated Drosophila TH, so we tested in human neuroblastoma cells (SH-SY5Y cells) the effect of metals on mammalian TH phosphorylation at Ser40, the major player on TH activity among the three phosphorylation sites and the only one conserved between the fly and mammals [43, 47]. Western analysis indicated that the phosphorylation of TH at Ser40 was not prominently and consistently affected by zinc, iron, and chelators (Fig. 4B, Additional file 1: Figure S1 and Additional file 1: Figure S6). In other words, the regulatory effect of zinc or iron on TH activity may not be attributed to changes in phosphorylation.

Fig. 4
figure 4

The in vivo effects of Catsup on TH appear primarily not due to TH phosphorylation and BH4 synthesis. A Sequence comparison among the fruit fly, human, and rat THs. Phosphorylation positions in mammalian TH and their corresponding amino acids in Drosophila TH are indicated with yellow arrowheads. Only the critical third phosphorylation site (Ser40) is conserved in the fly. Predicted metal-binding residues are marked with red arrows. B TH phosphorylation in SH-SY5Y cells under different metal or metal chelator treatments. No obvious and consistent changes were observed. An anti-phosphorylated TH antibody was used. Actin was used as a loading control. C The eclosion defect of Da-Gal4>GTPCH RNAi flies was rescued by DMPH4 supplementation in the food. Genotypes of the flies used were Da-Gal4>GTPCH RNAi. All values are presented as mean ± SEM; n = 6. ***p < 0.001. D DMPH4 supplementation in the food had no effect on the activity of TH. Genotype of the flies used was w1118. All values are presented as mean ± SEM; n = 6

TH requires tetrahydrobiopterin (BH4) as a cofactor for its activity [48]. Catsup mutations resulted in strongly elevated BH4 levels [21]. So, we wondered if the effect of zinc on TH activity is via its influence on BH4. The enzyme GTP cyclohydrolase I (GTPCH) is the rate-limiting enzyme for BH4 biosynthesis [21]. In Drosophila, GTP cyclohydrolase I (GTPCH) is encoded by Punch (Pu) [49]. Ubiquitous RNAi of Pu by Da-Gal4 led to eclosion defects in Drosophila, which could be rescued by supplement of 6, 7-dimethyl 5, 6, 7, 8-tetrahydropterin (DMPH4), a synthetic cofactor for TH similar to but more stable than the natural cofactor (6R)-5,6,7,8-tetrahydrobiopterin (BH4) (Fig. 4C) [19, 50]. This indicates that dietary DMPH4 is physiologically accessible to TH in vivo. Consistently, it is also reported that pre-feeding flies with BH4 (0.34 mg/mL) could inhibit the firing of the NPF neurons [51]. Nevertheless, for normal flies reared on normal diet, we found that TH activity remained unaffected with additional BH4 supplementation (Fig. 4D). We consider this negative finding supporting the hypothesis that the amount of endogenous BH4 is probably saturated for the activation of TH under normal conditions, and additional BH4 may not further improve the TH enzyme activity. In the case of Catsup mutation, although TH activity is elevated, TH protein level is not [19]. This suggests that the observed increase in TH activity in Catsup loss flies may at least partially come from another route different from the increased BH4 synthesis.

Zinc and iron antagonize with each other and directly impact TH activity

Based on our in vivo work as described above, we hypothesized an interaction between zinc and iron in regulating TH activity. Literature search revealed that it had previously been shown with in vitro biochemistry experiments that iron has a positive effect, while other divalent metal ions such as zinc, cobalt, and nickel ions have negative effects on recombinant human TH activity [52, 53]. However, it is not known whether these metal ions are relevant in vivo in regulating the activity of TH enzyme, especially when considering that intracellular labile metal ions are normally present at very low concentrations.

We hypothesized that zinc and iron may confer direct and opposite effects on TH within a physiological context. TH is a cytosolic enzyme. From this perspective, the notion that the effect of Catsup mutation on TH arises from its cytosolic zinc reduction is reasonable. To formally investigate this possibility and exclude other likely interpretations, Catsup was heterologously expressed in yeast cells, a foreign, arguably relatively clean, and physiologically relevant system. Native yeast lacks the TH counterpart as well as BH4 and DA synthesis. We reasoned that if zinc and iron could impact TH independent of phosphorylation or BH4 variations, we might be able to detect metal-modulated TH activities in the yeast cells. We first tested whether Catsup indeed functions as a zinc transporter in the yeast. To do that, the ZHY3 strain lacking the Zrt1 and Zrt2 proteins for zinc uptake was used [54]. We considered that by using ZHY3 strain, a zinc-deficient cellular state would be created, and Catsup expression might be able to alleviate this cytosolic zinc deficiency. Interestingly, on the normal media ZHY3 with induced Catsup expression could not grow well (Fig. 5A). This defect could be attributed either to higher zinc in the cytosol or lower zinc in the secretory compartment. However, growth was restored with the cell-permeable zinc chelator TPEN, suggesting the defect arose from zinc accumulation in the cytosol. Consistently, Catsup expression rendered ZHY3 overtly sensitive to zinc stresses (Fig. 5A). These results are consistent with the expected role of Castup in redistributing zinc between the cytosol and the secretory compartments, only the stress in this case primarily arising from zinc accumulation in the cytosol in the presence of toxic amounts of zinc. As an alternative measurement, we used zinc-responsive Zrt1-luciferase reporter to indicate the cytoplasmic zinc pool. Double knockout of Zrt1 and Zrt2 zinc transporter genes results in a low cytoplasmic zinc pool in the yeast cell. When the cytoplasmic zinc pool is low, the transcriptional activity of Zrt1 is upregulated, resulting in a higher luciferase activity. The expression of Catsup in ZHY3 led to a 60% decrease of luciferase activity compared with that in the control, suggesting an increased zinc level in the cytoplasm (Fig. 5B). The activity of ALP is very sensitive to zinc deficiency [55]. As expected, Catsup expression in ZHY3 significantly reduced the activity of ALP, indicating a decreased zinc level in the secretory pathway (Fig. 5C).

Fig. 5
figure 5

Zinc and iron potently but oppositely affect TH activity when expressed and assayed in yeast. A The growth defect of ZHY3 yeast expressing Catsup is exacerbated by zinc while rescued by TPEN. Transgenes could be induced on the synthetic galactose and raffinose (SGR) media but not on the synthetic dextrose (SD) media. n = 6. B The promoter activity of ZRT1 is decreased when Catsup was expressed in ZHY3 yeast. All values are presented as mean ± SEM and included in Additional file 2: table S3; n = 6.***p < 0.001. C The ALP activity was reduced when Catsup was expressed in ZHY3 yeast. All values are presented as mean ± SEM and included in Additional file 2: table S4; n = 6. ***p < 0.001. D L-dopa production in the TH-expressing ZHY3 yeast could be inhibited by Catsup expression. Six hours and 18 h refer to the yeast incubation time for L-dopa production. All values are presented as mean ± SEM and included in Additional file 2: table S5; n = 4. **p < 0.01,***p < 0.001. E The generation of L-dopa in ZHY3 was elevated by iron and inhibited by zinc added in the media. All values are presented as mean ± SEM and included in Additional file 2: table S6; n = 3. **p < 0.01,***p < 0.001. F The activity of TH in ZHY3 was inhibited by Catsup expression or zinc while elevated by iron. All values are presented as mean ± SEM and included in Additional file 2: table S7; n = 3. *p < 0.05, **p < 0.01,***p < 0.001

To analyze zinc’s effect on the activity of TH in yeast, Drosophila TH was then introduced into the ZHY3 strain. The yeast was incubated in the presence of 1 mM DMPH4 as yeast lacks endogenous BH4 synthesis. As shown in Fig. 5D, ZHY3 expressing TH synthesized L-dopa, and the expression of Catsup in ZHY3 resulted in a 40% decrease of the L-dopa level. We then tested the influence of exogeneous zinc and iron on the TH activity by measuring L-dopa production (Fig. 5E). The amount of L-dopa synthesized in ZHY3 expressing TH increased with iron and decreased with zinc supplementation (Fig. 5E). We also directly measured the TH activity with the radioisotope (Fig. 5F). The results indicated that the TH activity was inhibited by Catsup and zinc, whereas induced by iron (Fig. 5F). Given the unexpected growth inhibitory effect of Catsup on ZHY3, the effect of zinc and iron on L-dopa synthesis was further examined with wild-type INVSc1 strain expressing TH, and similar results were obtained (Additional file 1: Figure S2). These results indicated that the activity of TH heterologously expressed in the yeast can be influenced by the intracellular iron or zinc available to cells. Noteworthy is that since the yeast is unable to synthesize BH4, the fact that under a fixed BH4 level zinc and iron could still markedly affect TH activity indicates that the zinc and iron effect on TH could be direct.

We also tested TH responsiveness to iron and zinc in E. coli. Native E. coli also lacks TH and DA synthesis, as well as the essential cofactor BH4. Drosophila TH was expressed in E. coli and provided with exogenous DMPH4. As shown in Fig. 6A, E. coli-expressing Drosophila TH synthesized L-dopa, and the amount of L-dopa synthesized increased with incremental elevations of iron and decreased with zinc. The expression of TH, on the other hand, appeared not much affected by the addition of zinc and iron (Additional file 1: Figure S3). Therefore, both the experiments in the yeast and E. coli indicate that TH activities within a cell could be modulated robustly and possibly directly by iron and zinc homeostasis, and that iron and zinc antagonize each other in regulating TH activity.

Fig. 6
figure 6

Zinc ion interferes with ferrous iron binding to TH. A The production of L-dopa in E. coli-expressing TH was elevated by iron and inhibited by zinc added in the media. All values are presented as mean ± SEM and included in Additional file 2: table S8; n = 3. *p < 0.05, **p < 0.01. B Binding of iron to Drosophila TH. Apo-TH was incubated with 15 μM ferrous iron and different concentrations of zinc. Free and bound metal ions were separated by rapid ultrafiltration, and the iron level was determined by BPS-based colorimetric assays. The amount of bound iron decreased with increased zinc concentration in the buffer. All values presented are included in Additional file 2: table S12

It is known that iron is a cofactor of TH and the redox state of the iron is essential for its function [52]. Site-directed mutagenesis studies and crystal structure analysis revealed that the side chains of two histidine residues and one glutamate residue in the active site of mammalian TH are directly involved in iron coordination [56]. Some other divalent metal ions also could bind with TH and inhibit its activity in vitro [52, 53]. Not surprisingly, Drosophila TH and human TH are well conserved in these iron-binding sites (Fig. 4A). We hypothesized zinc could compete with iron binding with Drosophila TH and subsequently inhibit its activity. To this end, we performed a metal-binding assay. Drosophila TH was expressed in E. coli and purified. The purified recombinant Drosophila TH was incubated with Fe2+ and Zn2+. The bound and free iron fractions were then separated and measured by BPS-based colorimetric assays (Fig. 6B). We observed that Drosophila TH contained about 0.8 iron atom/subunit when incubated with ferrous iron, and the amount of bound iron decreased with increasing concentrations of zinc in the incubation buffer (Fig. 6B). The competing binding of iron and zinc on TH was further confirmed by ICP-MS (Additional file 1: Figure S4). Notably, zinc appears to be able to bind TH at multiple sites, likely as a result of non-specific binding in particular at high concentrations. These results indicate that zinc regulates Drosophila TH activity by affecting TH binding with iron.

Modulation of zinc and iron homeostasis alters phenotypes associated with PD

PD is a neurodegenerative disorder characterized in its late phase by sustained loss of dopaminergic neurons from the substantia nigra pars compacta (SNpc) and other brainstem regions [57]. Since DA loss is a cardinal feature of PD and L-dopa is utilized as an initial therapy to treat PD, we wondered whether the effect of zinc and iron on TH could influence PD phenotypes. Rotenone is a specific high-affinity inhibitor of mitochondrial complex I [58] and has been shown to be an environmental toxin (along with other chemicals such as MPTP and paraquat) contributing significantly towards the occurrence of sporadic PD in humans and animal models [59]. Rotenone exposure caused serious mobility impairment and reduced lifespan for Drosophila at 25 °C (Fig. 7A, B). When raised at 25 °C for 7 days in the presence of rotenone, about 30% of flies reached the subjectively designated height (8 cm) within a set time (7 s) (Fig. 7A). Catsup RNAi and dZnT7 OE were significantly rescued, whereas Catsup OE exacerbated, the mobility impairment (Fig. 7A). Consistent with the results described above, Catsup RNAi and dZnT7 OE extended the reduced longevity of rotenone-treated flies, whereas Catsup OE shortened it (Fig. 7 B). The overexpression of Malvolio (Mvl), an iron importer which presumably increases the iron level in the cytoplasm, also exhibited dramatic rescuing effects on the mobility impairment and the reduced lifespan (Fig. 7A, B). Catsup RNAi, dZnT7 OE, Catsup OE, and Mvl OE all exhibited mobility and survival defects when raised on normal food (Additional file 1: Figure S5). However, their effects on roteone-induced PD are diverse. Catsup OE, which on normal food survived better than Catsup RNAi and dZnT7 OE (Additional file 1: Figure S5), faired much worse on rotenone (Fig. 7A, B). In fact, although Catsup RNAi and dZnT7 OE were harmful on their own in the normal context (Additional file 1: Figure S5), they displayed a strong rescuing effect on rotenone-treated flies (Fig. 7A, B).

Fig. 7
figure 7

Modulation of zinc and iron homeostasis is an effective means to alter a spectrum of phenotypes associated with PD. A The mobility defect of PD flies (exposing to rotenone) could be rescued by Catsup RNAi, dZnT7 OE, or Mvl OE. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>Catsup OE, TH-Gal4>dZnT7 OE, and TH-Gal4>Mvl OE. All values are presented as mean ± SEM and included in Additional file 2: table S9; n ≥ 6. ***p < 0.001. B The shortened lifespan of PD flies (exposing to rotenone) could be modified by adjusting zinc transporter expressions. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>Catsup OE, TH-Gal4>dZnT7 OE, and TH-Gal4>Mvl OE. C The mobility rescue effect of Catsup RNAi on PD flies (exposing to rotenone) could be neutralized by hZIP7 expression. The exacerbated defect of Catsup OE on PD flies could be rescued by dZnT7 OE and Mvl OE. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>Catsup RNAi, hZIP7 OE, TH-Gal4>Catsup OE, TH-Gal4> Catsup OE;dZnT7 OE, TH-Gal4> Catsup OE, and Mvl OE. All values are presented as mean ± SEM and included in Additional file 2: table S10; n ≥ 6.. ***p < 0.001. One-way analysis of variance (ANOVA) was used for multiple groups. D The lifespan rescue of Catsup RNAi on PD flies (exposing to rotenone) could be neutralized by hZIP7 expression. The exacerbated defect of Catsup OE on PD flies could be rescued by dZnT7 OE and Mvl OE. Genotypes of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>Catsup RNAi, hZIP7 OE, TH-Gal4>Catsup OE, TH-Gal4> Catsup OE, dZnT7 OE, TH-Gal4> Catsup OE, and Mvl OE. All values are presented as mean ± SEM and included in Additional file 2: table S11; n ≥ 4. ***p < 0.001

Further study indicated that the rescue effect of Catsup RNAi on flies with rotenone exposure could be inhibited by hZIP7 OE, and the exacerbation effect of Catsup OE on these flies could be rescued by dZnT7 OE and Mvl OE (Fig. 7C, D). Taken together, these results indicated that modulating the zinc and iron levels in the Drosophila brain could modify a whole range of symptoms of rotenone-treated flies.

Discussion

Zinc and iron are critical to a variety of proteins and are essential for normal CNS development and function [60]. Previous studies showed that Catsup mutants have significantly elevated TH activity, resulting in abnormally high levels of catecholamines [19]. In this study, we confirmed that Catsup is indeed functionally analogous to hZIP7, transporting zinc from the secretory compartments into the cytoplasm. We found that manipulating Catsup in the brains can greatly modulate DA synthesis and PD progress by modulating TH activity through the interference of iron. We provided in vivo evidence that TH activity is affected by a balance of zinc and iron levels. Further study indicated that manipulation of zinc levels and iron levels in PD brains may be a novel therapeutic strategy. This may hold great significance for the prevention and treatment of PD and even other diseases caused by catecholamine dysfunction.

Ubiquitous manipulation of zinc and iron homeostasis often resulted in reduced viability. For this reason, dramatic modulation of metal homeostasis may not be feasible in PD rescue. We have primarily relied on genetic measures to modulate regional metal homeostasis such as in the brain or TH-positive cells to achieve a significant effect. It is envisioned that in clinical cases, targeted modification of metal homeostasis may be necessary to obtain satisfactory results. This is particularly so when the blood-brain barrier is taken into account, which may partially insulate the brain from the circulation system. Indeed, in our case, the nutritional supplement of metals in the food was much less efficient to reach the kind of rescue we saw with tissue-specific genetic interventions.

Catsup is a member of the ZIP (Slc39A) transporter family [30]. Most ZIP family members transport zinc, but a few have been found to transport other metals, including ZIP8, ZIP13, and ZIP14 [61]. Catsup shares the highest overall homology with human ZIP7 [25, 30], which is an identified zinc transporter [26]. Simultaneously, Catsup also shares high homology with human dZIP13, which fulfills the iron effluxing role in Drosophila [30]. In this study, we studied the function of Catsup and verified that it is a zinc transporter located in the secretory compartments. In a previous study, we provided in vivo functional analysis to differentiate different transporting functions of these two ZIPs; the HNXXD motif is required for zinc transport activities for Catsup [37].

The activity of many enzymes is regulated by metal ions. For example, the activity of superoxide dismutase (SOD) can be induced by copper but inhibited by iron or cadmium [62]; cytochrome c oxidase needs iron and copper to execute its activity, but the presence of zinc inhibits its activity [63]. So, it seems important to balance these metal ions in vivo to regulate the activity of these enzymes. In this study, we showed that in vivo iron stimulates while zinc inhibits TH activity, consistent with the previous report that the activity of recombinant human TH was rapidly activated by incubation with iron and inhibited by some other metal ions in vitro [52].

Protein phosphorylation is reported as one major mechanism for controlling the activity of TH [43]. TH phosphorylation is regulated by several protein kinases and protein phosphatases [43]. It is well known that zinc plays an important role in affecting phosphorylation signaling [64, 65]. Here, we found that the TH phosphorylation is not appreciably affected by zinc or iron modulation, so the effect of Catsup on TH activity seems through a route other than phosphorylation.

TH requires BH4 as a cofactor [48], and the enzyme activity is catalyzed by ferrous iron [66]. As a result, the synthesis of catecholamines is not only dependent on TH activity but also on the level of BH4 and iron. Because the TH activity is not increased in flies feeding with DMPH4, we proposed that under normal conditions, the BH4 level in the body may be sufficient or saturated, so that excess BH4 in vivo has no further contribution to the TH activity. Expression of TH within the yeast and E. coli, both of which lack BH4 synthesis and likely some other regulatory pathways tuning TH activity, indicated that TH activity is robustly responsive to iron and zinc modulation. Altogether, we propose that iron in the cytoplasm binds TH to catalyze its activity, and zinc can compete with iron binding in TH. Since Catsup is responsible for zinc release from the secretary pathway to cytoplasm, Catsup mutations lead to zinc deficiency in the cytoplasm, and in consequence, more iron could bind to TH, resulting in the increased TH activity (Fig. 8).

Fig. 8
figure 8

A model to explain Catsup’s effect on TH activity. Zinc appears to be an important regulator of TH activity or dopamine production in vivo. When Catsup is mutated, zinc fails to be effectively redistributed between ER/Golgi and cytosol, leading to zinc elevation in the secretory pathway but reduction in the cytosol. Zinc normally antagonizes iron in regulating TH activity. When zinc release is blocked, TH has better access to iron and its activity is elevated

Increasing amounts of evidence show that imbalance of trace metal homeostasis is strongly related to the pathological process of PD [67, 68]. Here, we revealed that there exists a new way to regulate DA synthesis in vivo through modulating metal homeostasis. The knowledge gained in this study appeared to be adaptable in ameliorating PD features in a fly model. It is reasonable to speculate that cells could physiologically modulate dopamine synthesis through zinc control, by maintaining a balance between zinc storage and release to adjust the cytosolic zinc levels. Indeed, mammalian ZIP7 was reported to be regulated by phosphorylation [69]. Therefore, it is expected that cells could adjust DA levels through modifying expression or posttranslational modifications of ZIP7 in response to physiological changes.

These results may suggest new strategies for the prevention and treatment of PD and some other diseases closely related to DA. Considering zinc depletion and iron increase may achieve similar end results, in our opinion, it may be preferable to use iron limitation strategy when depressing dopamine levels is intended or zinc limitation in the case of boosting dopamine production. This is because iron may in short term raise dopamine levels; it may also simultaneously stimulate elevated production of reactive oxygen species, heightening dopaminergic neuron damages in the long run. Besides PD, DA is closely related to pain, drug addiction, attention deficit hyperactivity disorder, and many kinds of mental diseases such as psychosis, schizophrenia, and depression [2, 5]. Unfortunately, the etiology of many of these diseases remains less well clarified. It would be interesting to see how the balance of zinc and iron could modulate the disease progression for some of these physiological disorders that might be DA-related.

Conclusions

Analyses of dopamine elevation in Catsup mutation/knock-down revealed an important role of zinc homeostasis in regulating TH activity. Zinc antagonizes ferrous iron, a cofactor of the TH enzyme, to exert a negative effect on TH. This work enriches our understanding of dopamine regulation in vivo and may help our fight against PD and other forms of dopamine-related diseases.

Methods

Drosophila strains, yeast strains, and maintenance

Drosophila strains used in this study are listed as follows: Da-Gal4 (Bloomington #8641), Elav-Gal4 (Bloomington #458), and TH-Gal4 (Bloomington #8848), all from the Bloomington Drosophila Stock Center. p Catsup RNAi (VDRC #100095) and dZnT7 RNAi (VDRC #107388) were obtained from the Vienna Drosophila RNAi Center. Transgenic fly strains used in this report have been previously described, including dZIP13 OE [37], dZIP1 OE [42], dZnT1 OE [39]. pUAST-Malvolio construct (Mvl OE) was injected into w1118 background flies following standard protocols. One line with relatively more severe neuronal phenotype when directed by ELav-Gal4 was chosen for the studies reported here.

All flies were maintained on standard cornmeal media at 25 °C with 60% humidity under a 12-h light–dark cycle unless otherwise stated. Concentrations of supplemented metals or metal chelators used were as follows: 5 mM ferric ammonium citrate (FAC; Sigma-Aldrich), 150 μM bathophenanthroline disulfonate (BPS; Sigma-Aldrich), 50 μM N,N,N′,N′-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN; Sigma-Aldrich), and 2 mM ZnCl2 (Beijing Yili Fine Chemicals Ltd. Co., Beijing, China). For inhibitor studies, concentration of 3-Iodo-L-tyrosine (3-IT; Sigma-Aldrich) used was 75 μM. For the rotenone feeding experiment, 2-day-old adult flies were transferred to vials with instant Drosophila medium and exposed to 150 μM freshly prepared rotenone (Sigma-Aldrich) for 7 days. The medium was replaced every day to ensure that the rotenone was effective.

The ZHY3 strain lacking the Zrt1 and Zrt2 proteins for zinc uptake was used in this study [54].

Cell culture and plasmid transfection

For subcellular localization experiments, CHO-K1 cells were maintained in DMEM (Invitrogen) containing 10% fetal bovine serum (FBS, Gibco BRL, Gaithersburg, MD, USA) at 37 °C under 5% CO2 atmosphere. When the cells reached 80% confluence, they were transfected with pIRESneo-Catsup-FLAG using Lipofectamine™ 3000 (Invitrogen) in accordance with the manufacturer’s instructions. After 24 h, cells were fixed, stained with the FLAG antibody (1:500) and the Golgi marker (anti-GM130, 1:500) or ER marker (anti-PDI, 1:500), and imaged with the ZEISS LSM780 microscope. Anti-FLAG mouse monoclonal antibody (ab18230), anti-FLAG rabbit polyclonal antibody (ab1162), anti-GM130 rabbit polyclonal antibody (ab30637), and anti-PDI mouse monoclonal antibody (ab2792) were obtained from Abcam (Cambridge, MA, USA). Secondary antibodies include cy3-conjugated goat anti-mouse and cy3-conjugated goat anti-rabbit IgG (Zhongshan Goldenbridge Biotechnology, Beijing, China). The experiments were repeated three times.

SH-SY5Y cells were used to test the effect of metals on mammalian TH phosphorylation at Ser40. SH-SY5Y cells were maintained in DMEM (Invitrogen) with 10% FBS and 1% PS under an atmosphere of 95% air and 5% CO2 at 37 °C. To test metals’ effects, 1 μM BPS, 50 μM ZnCl2, 1 μM TPEN, and 10 μM FeCl2 (2 mg/mL vitamin C were added in the medium to inhibit iron oxidation) were respectively added in the medium. The experiments were repeated three times.

Eclosion, mobility, and longevity assays

For eclosion assays, Gal4 lines were crossed with various transgenic lines for ~ 3 days, as indicated in each experiment, and allowed to lay eggs on juice-agar plates for ~ 24 h. Newly hatched first-instar larvae were transferred to normal food or food supplemented with metals or metal chelators, as indicated. The density of each vial was controlled to 60–100 larvae, and the total number of emerging adults of each genotype was counted [30]. Assays were done with six replicates for each group, and the experiments were repeated at least three times. The results from all experiment runs were pooled together for analyses.

To assay the mobility of PD flies, 20 females were placed in a vertical glass vial. After a 1-h recovery from CO2 exposure, flies were gently tapped to the bottom of the column. The number of flies reaching the indicated height (~ 8 cm) was counted after 7 s of climbing under red light. Six parallel group tests were conducted for each genotype, and the experiments were repeated at least three times [34]. The presented data are from the results of all experiments.

For longevity assays, 3-day-old adult males or females were kept in vials (20 individuals of each vial) containing cornmeal medium and maintained at 25 °C. Flies were transferred to fresh media every 2–3 days, and the number of live flies was recorded every alternate day. Six parallel group tests were conducted for each genotype, and the experiments were repeated at least three times [34]. The data were presented as Kaplan–Meier survival distributions, and significance was determined by log-rank tests.

Eye morphology analysis

To phenotype eye morphology, flies were collected after eclosion at 18 °C and frozen to death at − 80 °C for one night. The eyes were photographed using a Zeiss imager A1 stereomicroscope. More than six flies were scored per genotype, and each experiment was repeated three times. Scanning electron microscopy analysis of fly eyes was performed as described previously with FEI Quanta 200 [70].

Alkaline phosphatase(ALP) activity assay

ALP activity assays were performed as described previously with some modifications [71]. For yeast samples, yeast was cultured in synthetic dextrose media (SD) at 30 °C to OD = 0.4, collected by centrifugation at 3000g for 5 min, and then transferred to the induction medium (synthetic glucose and raffinose (SGR) media). The yeast was cultured in SGR media at 30 °C to OD = 1, collected by centrifugation, and washed three times with ice-cold PBS. The collected yeast was homogenized in ALP lysis buffer (1.0 mM Tris–HCl pH 7.4, 0.5 mM MgCl2, and 0.1% Triton X-100), and the protein concentration was measured by the BCA kit (Thermal). Approximately 2 μg protein was added to 90 μL solution A (1.0 M diethanolamine, 0.5 mM MgCl2 pH 9.8) and 10 μL solution B (150 mM p-nitrophenyl phosphate). After incubation for 30 min at 30 °C, ALP activity was measured based on the p-nitrophenol release by its absorbance at 405 nm.

For fly samples, 80 adult heads were lysed in the ALP lysis buffer, then 2 μg protein was added to 90 μL solution A and 10 μL solution B. The absorbance at 405 nm was measured after incubation for 30 min at 25 °C. The experiments were repeated three times.

Luciferase activity assay

Yeast was cultured in SD media at 30 °C to OD = 0.4, collected by centrifugation at 3000g for 5 min, and then transferred to SGR media. The yeast was cultured in SGR media at 30 °C to OD = 1, collected by centrifugation, and washed three times with ice-cold PBS. The collected yeast was homogenized in cold PBS, and the protein concentration was measured by the BCA kit (Thermal). Approximately 10 μg protein was used to test the luciferase activity by Firefly Luciferase Reporter Gene Assay Kit (Beyotime, #RG027); fluorescence intensity was measured for 10 s by the fluorescence microplate reader. In order to avoid the error caused by the difference in the amount of samples, the reporter gene of Renilla luciferase was used as an internal reference.

Western blot analysis

SH-SY5Y cells were used to test the effect of metals on mammalian TH phosphorylation at Ser40. Antibodies of anti-tyrosine hydroxylase (AB152) and anti-tyrosine hydroxylase, phosphoSer 40 (AB5935) were purchased from Sigma-Aldrich (Shanghai, China). The secondary antibody HRP-conjugated goat anti-rabbit IgG was purchased from Zhongshan Goldenbridge Biotechnology (Beijing, China). For Western blot analysis, cells were homogenized in the buffer containing 1% Triton X-100 plus 10% proteinase inhibitor cocktail (Sigma), centrifuged, separated on 10% SDS-PAGE, and transferred to nitrocellulose membranes (Millipore, Watford, UK). Signals were developed with the ECL detection kit (Vigorous Biotechnology, Beijing, China). The experiments were repeated three times.

Metal sensitivity/resistance assay in yeast

Catsup was cloned into pYES2 (Invitrogen) and transformed into ZHY3. The vector pYES2 was also transformed into yeast and used as the control. For growth testing on agar plates (spotting assay), yeast grown in the culture medium (synthetic dextrose media, SD) was 10-fold serially diluted with sterile ddH2O and then spotted on SD (the transgene could not be induced on this medium) or synthetic galactose and raffinose media (SGR, the transgene could be induced on this medium) plates with or without metal or chelators. Incubation was all at 30 °C; 3 mM ZnCl2 or 20 μΜ TPEN was added in the medium, n = 6.

Metal ions binding assays of TH

A double-tagged Drosophila TH protein (dTH) expression plasmid containing N-terminal His6 and SUMO tag (pET28a-His6-SUMO-dTH) was constructed. The ensuing plasmid was transformed into E. coli BL21(DE3). Protein expression was induced by 250 μM β-D-1-thiogalactopyranoside (IPTG) with 100 μM FeSO4 supplied in LB medium for enzyme stability. Induction proceeded at 16 °C for 14 h. The His6-SUMO-dTH protein was purified with Ni-NTA resin, and the tags were removed by ULP1 digestion. Protein was then loaded onto a Superdex 200 gel filtration column for size separation. The protein composition of each fraction was determined by SDS-PAGE analysis. Fractions containing dTH were collected, and EDTA was added to a final concentration of 1 mM to remove divalent metal ions. After incubation at 4 °C for 30 min, the purified protein was concentrated by Amicon® Ultra Centrifugal Filters (type 30,000 NMWL). Then, the protein was diluted 10-fold with EDTA-free 50 mM MES buffer (pH 6.5) and applied to Ultra filters again to be concentrated to its original volume. The procedure was repeated for five times to remove EDTA in the protein buffer.

Competitive binding assays were performed as described previously with some modification [52]. Briefly, purified dTH (2.5 μM subunit) was incubated with 15 μM FeCl2, 0-25 μM ZnCl2, and 0.5 mg/mL catalase in 50 mM MES Potassium (pH 6.5) for 20 min at 4 °C. After incubation, the mixture was applied to Amicon Ultra filter devices (Merck Millipore). Free and bound metal ions were separated by centrifugation (5 min at 5000 rpm) at 4 °C. The filtrate was collected and contents of iron ion or zinc ion were determined by BPS-based colorimetric assays or ICP-MS. All the experiments were repeated at least three times.

Determination of L-dopa

For L-dopa assay in S. cerevisiae, Catsup and dTH were cloned into pYES2 (Invitrogen) and pYES3 (Invitrogen), respectively. The plasmids were transformed into ZHY3 or wild-type yeast INVSc1. The vectors pYES3 and pYES2 were also transformed into yeast and used as the controls. For L-dopa produced in yeast, a single colony was picked and grown in SD medium at 30 °C to OD600 = 1. Then, the yeast cells were collected by centrifugation at 3000g for 5 min, washed with sterile water, and transferred to SGR medium supplemented with 0.4 mg/mL tyrosine, 2.0 mg/mL ascorbic acid, and 1 mM DMPH4. In the experiments for testing metal effects, 400 mM BPS along with FeSO4 or ZnCl2 was added in the medium. The yeast was shaken at 30 °C for 18 h, and the supernatant was collected for L-dopa level determination.

For L-dopa assays in E.coli, pET28a-His6-SUMO-dTH mentioned above was transformed into BL21 (DE3). The vector pET28a was transformed into BL21(DE3) as control. Protein expression was induced by 250 μM IPTG at 37 °C. The LB medium was supplied with 400 μg/mL tyrosine, 2.0 mg/mL ascorbic acid, and 1 mM DMPH4 for L-dopa production. FeSO4 or ZnCl2 was added as indicated in each experiment. L-dopa level was determined after 8 h growth at 37 °C. L-dopa level was determined as previously described [72] with some modifications. Briefly, the colorimetric method for estimation of L-DOPA was performed in 96-well microtiter plates. Fifty microliters each of 0.5 M HCl, nitrite-molybdate reagent (composed of 10% w/v sodium nitrite and 10% w/v sodium molybdate), and 1.0 M NaOH was sequentially added to 50 μL of the sample. The absorbance was measured at 530 nm using a Thermo Multiskan GO reader. A calibration curve with different amounts of L-dopa was constructed. L-dopa level in each sample was calculated based on the standard curve or normalized to the control. All experiments were repeated at least three times.

Dopamine level determination

For sample preparation, 3rd larvae were collected and sonicated in 0.6 mL of ice-cold 0.4 M perchloric acid. After centrifugation at 13000g at 4 °C for 10 min, the supernatant was transferred into a new tube and mixed with a half supernatant volume of 20 mM potassium citrate, 300 mM K2HPO4, and 2 mM EDTA. The mixture was kept on ice in the dark for 20 min. Then, the chilled mixture was filtered through a low-binding Durapore (0.22 μm) PVDF membrane. The amounts of dopamine were measured by an HPLC-ECD system. Chromatographic separation was performed on a Diamonsil C18 column (5 μm, 250 × 4.6 mm). The mobile phase contained 60 mM KH2PO4, 350 μM C8H19NaO4S, 100 μM EDTA, and 8% formaldehyde. Dopamine levels were monitored using a HPLC system (Water e2695) equipped with an electrochemical detector (Water 2645). The experiments were repeated three times.

TH activity assay

TH activity was determined as previously described with some modifications [73, 74]. For the TH activity assay in Drosophila heads, samples were collected and homogenized in 20 mM Tris-HCl buffer, pH 7.5 by ultrasonic disintegration over ice. Protein concentrations were determined with BCA kit and adjusted for each group. Fifty microliters homogenate was further diluted with 300 μL 30 mM Tris-acetic acid containing 0.1% Triton X-100 and 25 μCi of radioactive tyrosine HCl (0.4 mCi/nMol of L-[ring-3,5-3H]tyrosine), 50 nMol of the cofactor 6, 7-dimethyl 5, 6, 7, 8-tetrahydropterin (DMPH4, Sigma, St. Louis, MO, USA), 5000 units of catalase, and 5 nM DTT in 100 mM potassium phosphate, pH 6.0 for incubation at 37 °C for 20 min. Then, aliquots were taken and placed in 5% (v/v) perchloric acid to stop the reaction. Unreacted tyrosine and the product DOPA were absorbed with 1.0 mL of an aqueous slurry of activated charcoal (in 0.1 M HCl) and the released [3H]H20 was analyzed by liquid scintillation counting. TH utilizes O2 to produce DOPA, and [3H]H20 is derived from [3H]tyrosine. The TH activity was expressed as radiation value of [3H]H20 produced per group.

For the TH activity assay in yeast, yeast was cultured in SD media at 30 °C for 24 h to OD = 1. Yeast was collected by centrifugation at 3000g for 5 min and then transferred to SGR media supplemented with metals or metal chelators, as indicated in each experiment; 500 nMol DMPH4 (Sigma, St. Louis, MO, USA) and 10 μL L-[ring-3,5-3H]tyrosine were added in the media and then the yeast was further cultured at 30 °C for 24 h. Cells were removed after centrifugation at 12,000g for 15 min. The supernatant was transferred to a new container. Unreacted tyrosine and the product DOPA were absorbed with 1.0 mL of an aqueous slurry of activated charcoal (in 0.1 M HCl) and the released [3H]H20 was analyzed by liquid scintillation counting. The TH activity was expressed as radiation value of [3H]H20 produced per group. All experiments were repeated at least three times.

Statistical analysis

All data were recorded and analyzed with Microsoft Excel and GraphPad Prism (version 6.00; La Jolla, CA, USA). Data were analyzed by Student’s t-test between the groups, and ANOVA was used for multiple comparison. Statistical results were presented as means ± SEM. Asterisks indicate critical levels of significance (*p < 0.05, **p < 0.01, ***p < 0.001).

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Abbreviations

DA:

Dopamine

PD:

Parkinson’s disease

TH:

Tyrosine hydroxylase

Catsup:

Catecholamines up

CNS:

Central nervous system

OE:

Overexpression

ALP:

Alkaline phosphatase

TPEN:

N,N,N′,N′-tetrakis(2-pyridylmethyl)ethylenediamine

BPS:

Bathophenanthrolinedisulfonic acid

BH4:

Tetrahydrobiopterin

DMPH4:

6, 7-Dimethyl 5, 6, 7, 8-tetrahydropterin

GTPCH:

GTP cyclohydrolase I

ICP-MS:

Inductively coupled plasma mass spectrometry

References

  1. Massano J, Bhatia KP. Clinical approach to Parkinson’s disease: features, diagnosis, and principles of management. Cold Spring Harb Perspect Med. 2012;2(6):a008870. https://doi.org/10.1101/cshperspect.a008870.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Neckameyer WS. Multiple roles for dopamine in Drosophila development. Dev Biol. 1996;176(2):209–19. https://doi.org/10.1006/dbio.1996.0128.

    Article  CAS  PubMed  Google Scholar 

  3. Berridge CW, Devilbiss DM. Psychostimulants as cognitive enhancers: the prefrontal cortex, catecholamines, and attention-deficit/hyperactivity disorder. Biol Psychiatry. 2011;69(12):e101–11. https://doi.org/10.1016/j.biopsych.2010.06.023.

    Article  CAS  PubMed  Google Scholar 

  4. Haenisch B, Bonisch H. Depression and antidepressants: insights from knockout of dopamine, serotonin or noradrenaline re-uptake transporters. Pharmacol Ther. 2011;129(3):352–68. https://doi.org/10.1016/j.pharmthera.2010.12.002.

    Article  CAS  PubMed  Google Scholar 

  5. Berke JD. What does dopamine mean? Nat Neurosci. 2018;21(6):787–93. https://doi.org/10.1038/s41593-018-0152-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Daubner SC, Le T, Wang S. Tyrosine hydroxylase and regulation of dopamine synthesis. Arch Biochem Biophys. 2011;508(1):1–12. https://doi.org/10.1016/j.abb.2010.12.017.

    Article  CAS  PubMed  Google Scholar 

  7. Tekin I, Roskoski R Jr, Carkaci-Salli N, Vrana KE. Complex molecular regulation of tyrosine hydroxylase. J Neural Transm (Vienna). 2014;121(12):1451–81. https://doi.org/10.1007/s00702-014-1238-7.

    Article  CAS  Google Scholar 

  8. Memmert S, Damanaki A, Nogueira AVB, Nokhbehsaim M, Götz W, Cirelli JA, et al. Regulation of tyrosine hydroxylase in periodontal fibroblasts and tissues by obesity-associated stimuli. Cell Tissue Res. 2019;375(3):619–28. https://doi.org/10.1007/s00441-018-2941-8.

    Article  CAS  PubMed  Google Scholar 

  9. Nagatsu T, Nagatsu I. Tyrosine hydroxylase (TH), its cofactor tetrahydrobiopterin (BH4), other catecholamine-related enzymes, and their human genes in relation to the drug and gene therapies of Parkinson’s disease (PD): historical overview and future prospects. J Neural Transm (Vienna). 2016;123(11):1255–78. https://doi.org/10.1007/s00702-016-1596-4.

    Article  CAS  Google Scholar 

  10. Zhu Y, Zhang J, Zeng Y. Overview of tyrosine hydroxylase in Parkinson’s disease. CNS Neurol Disord Drug Targets. 2012;11(4):350–8. https://doi.org/10.2174/187152712800792901.

    Article  CAS  PubMed  Google Scholar 

  11. Woimant F, Trocello JM. Disorders of heavy metals. Handb Clin Neurol. 2014;120:851–64. https://doi.org/10.1016/B978-0-7020-4087-0.00057-7.

    Article  PubMed  Google Scholar 

  12. Montgomery EB Jr. Heavy metals and the etiology of Parkinson’s disease and other movement disorders. Toxicology. 1995;97(1-3):3–9. https://doi.org/10.1016/0300-483X(94)02962-T.

    Article  CAS  PubMed  Google Scholar 

  13. Sikk K, et al. Manganese-induced parkinsonism due to ephedrone abuse. Parkinsons Dis. 2011;2011:865319.

    PubMed  PubMed Central  Google Scholar 

  14. Singh N, Haldar S, Tripathi AK, McElwee MK, Horback K, Beserra A. Iron in neurodegenerative disorders of protein misfolding: a case of prion disorders and Parkinson’s disease. Antioxid Redox Signal. 2014;21(3):471–84. https://doi.org/10.1089/ars.2014.5874.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dexter DT, Wells FR, Lee AJ, Agid F, Agid Y, Jenner P, et al. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson’s disease. J Neurochem. 1989;52(6):1830–6. https://doi.org/10.1111/j.1471-4159.1989.tb07264.x.

    Article  CAS  PubMed  Google Scholar 

  16. Kumar V, Singh BK, Chauhan AK, Singh D, Patel DK, Singh C. Minocycline rescues from zinc-induced nigrostriatal dopaminergic neurodegeneration: biochemical and molecular interventions. Mol Neurobiol. 2016;53(5):2761–77. https://doi.org/10.1007/s12035-015-9137-y.

    Article  CAS  PubMed  Google Scholar 

  17. Kumar V, Singh D, Singh BK, Singh S, Mittra N, Jha RR, et al. Alpha-synuclein aggregation, ubiquitin proteasome system impairment, and L-Dopa response in zinc-induced parkinsonism: resemblance to sporadic Parkinson’s disease. Mol Cell Biochem. 2018;444(1-2):149–60. https://doi.org/10.1007/s11010-017-3239-y.

    Article  CAS  PubMed  Google Scholar 

  18. Dos Santos AB, et al. Hair in Parkinson’s disease patients exhibits differences in calcium, iron and zinc concentrations measured by flame atomic absorption spectrometry - FAAS. J Trace Elem Med Biol. 2018;47:134–9. https://doi.org/10.1016/j.jtemb.2018.02.003.

    Article  CAS  PubMed  Google Scholar 

  19. Stathakis DG, Burton DY, McIvor WE, Krishnakumar S, Wright TRF, O'Donnell JM. The catecholamines up (Catsup) protein of Drosophila melanogaster functions as a negative regulator of tyrosine hydroxylase activity. Genetics. 1999;153(1):361–82. https://doi.org/10.1093/genetics/153.1.361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chaudhuri A, Bowling K, Funderburk C, Lawal H, Inamdar A, Wang Z, et al. Interaction of genetic and environmental factors in a Drosophila parkinsonism model. J Neurosci. 2007;27(10):2457–67. https://doi.org/10.1523/JNEUROSCI.4239-06.2007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang Z, Ferdousy F, Lawal H, Huang Z, Daigle JG, Izevbaye I, et al. Catecholamines up integrates dopamine synthesis and synaptic trafficking. J Neurochem. 2011;119(6):1294–305. https://doi.org/10.1111/j.1471-4159.2011.07517.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hsouna A, Lawal HO, Izevbaye I, Hsu T, O'Donnell JM. Drosophila dopamine synthesis pathway genes regulate tracheal morphogenesis. Dev Biol. 2007;308(1):30–43. https://doi.org/10.1016/j.ydbio.2007.04.047.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Harbison ST, Carbone MA, Ayroles JF, Stone EA, Lyman RF, Mackay TFC. Co-regulated transcriptional networks contribute to natural genetic variation in Drosophila sleep. Nat Genet. 2009;41(3):371–5. https://doi.org/10.1038/ng.330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Groth C, Sasamura T, Khanna MR, Whitley M, Fortini ME. Protein trafficking abnormalities in Drosophila tissues with impaired activity of the ZIP7 zinc transporter Catsup. Development. 2013;140(14):3018–27. https://doi.org/10.1242/dev.088336.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Begum NA, Kobayashi M, Moriwaki Y, Matsumoto M, Toyoshima K, Seya T. Mycobacterium bovis BCG cell wall and lipopolysaccharide induce a novel gene, BIGM103, encoding a 7-TM protein: identification of a new protein family having Zn-transporter and Zn-metalloprotease signatures. Genomics. 2002;80(6):630–45. https://doi.org/10.1006/geno.2002.7000.

    Article  CAS  PubMed  Google Scholar 

  26. Huang L, Kirschke CP, Zhang Y, Yu YY. The ZIP7 gene (Slc39a7) encodes a zinc transporter involved in zinc homeostasis of the Golgi apparatus. J Biol Chem. 2005;280(15):15456–63. https://doi.org/10.1074/jbc.M412188200.

    Article  CAS  PubMed  Google Scholar 

  27. Lee MG, Bin BH. Different Actions of Intracellular Zinc Transporters ZIP7 and ZIP13 Are Essential for Dermal Development. Int J Mol Sci. 2019;20(16). https://doi.org/10.3390/ijms20163941.

  28. Tuncay E, Bitirim VC, Durak A, Carrat GRJ, Taylor KM, Rutter GA, et al. Hyperglycemia-induced changes in ZIP7 and ZnT7 expression cause Zn(2+) release from the sarco(endo)plasmic reticulum and mediate ER stress in the heart. Diabetes. 2017;66(5):1346–58. https://doi.org/10.2337/db16-1099.

    Article  CAS  PubMed  Google Scholar 

  29. Lye JC, Richards CD, Dechen K, Paterson D, de Jonge MD, Howard DL, et al. Systematic functional characterization of putative zinc transport genes and identification of zinc toxicosis phenotypes in Drosophila melanogaster. J Exp Biol. 2012;215(Pt 18):3254–65. https://doi.org/10.1242/jeb.069260.

    Article  CAS  PubMed  Google Scholar 

  30. Xiao G, Wan Z, Fan Q, Tang X, Zhou B. The metal transporter ZIP13 supplies iron into the secretory pathway in Drosophila melanogaster. Elife. 2014;3:e03191. https://doi.org/10.7554/eLife.03191.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Richards CD, Burke R. A fly’s eye view of zinc homeostasis: novel insights into the genetic control of zinc metabolism from Drosophila. Arch Biochem Biophys. 2016;611:142–9. https://doi.org/10.1016/j.abb.2016.07.015.

    Article  CAS  PubMed  Google Scholar 

  32. Lye JC, Richards CD, Dechen K, Warr CG, Burke R. In vivo zinc toxicity phenotypes provide a sensitized background that suggests zinc transport activities for most of the Drosophila Zip and ZnT genes. J Biol Inorg Chem. 2013;18(3):323–32. https://doi.org/10.1007/s00775-013-0976-6.

    Article  CAS  PubMed  Google Scholar 

  33. Dechen K, Richards CD, Lye JC, Hwang JEC, Burke R. Compartmentalized zinc deficiency and toxicities caused by ZnT and Zip gene over expression result in specific phenotypes in Drosophila. Int J Biochem Cell Biol. 2015;60:23–33. https://doi.org/10.1016/j.biocel.2014.12.017.

    Article  CAS  PubMed  Google Scholar 

  34. Xiao G, Fan Q, Wang X, Zhou B. Huntington disease arises from a combinatory toxicity of polyglutamine and copper binding. Proc Natl Acad Sci U S A. 2013;110(37):14995–5000. https://doi.org/10.1073/pnas.1308535110.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Le Du MH, et al. Crystal structure of alkaline phosphatase from human placenta at 1.8 A resolution. Implication for a substrate specificity. J Biol Chem. 2001;276(12):9158–65. https://doi.org/10.1074/jbc.M009250200.

    Article  PubMed  Google Scholar 

  36. Suzuki T, Ishihara K, Migaki H, Matsuura W, Kohda A, Okumura K, et al. Zinc transporters, ZnT5 and ZnT7, are required for the activation of alkaline phosphatases, zinc-requiring enzymes that are glycosylphosphatidylinositol-anchored to the cytoplasmic membrane. J Biol Chem. 2005;280(1):637–43. https://doi.org/10.1074/jbc.M411247200.

    Article  CAS  PubMed  Google Scholar 

  37. Zhao M, Zhou B. A distinctive sequence motif in the fourth transmembrane domain confers ZIP13 iron function in Drosophila melanogaster. Biochim Biophys Acta Mol Cell Res. 1867;2020(2):118607. https://doi.org/10.1016/j.bbamcr.2019.118607.

    Article  CAS  Google Scholar 

  38. Friggi-Grelin F, Coulom H, Meller M, Gomez D, Hirsh J, Birman S. Targeted gene expression in Drosophila dopaminergic cells using regulatory sequences from tyrosine hydroxylase. J Neurobiol. 2003;54(4):618–27. https://doi.org/10.1002/neu.10185.

    Article  CAS  PubMed  Google Scholar 

  39. Wang X, Wu Y, Zhou B. Dietary zinc absorption is mediated by ZnT1 in Drosophila melanogaster. FASEB J. 2009;23(8):2650–61. https://doi.org/10.1096/fj.08-126649.

    Article  CAS  PubMed  Google Scholar 

  40. Wang Z, Li X, Zhou B. Drosophila ZnT1 is essential in the intestine for dietary zinc absorption. Biochem Biophys Res Commun. 2020;533(4):1004–11. https://doi.org/10.1016/j.bbrc.2020.09.077.

    Article  CAS  PubMed  Google Scholar 

  41. Ohashi W, Kimura S, Iwanaga T, Furusawa Y, Irié T, Izumi H, et al. Zinc transporter SLC39A7/ZIP7 promotes intestinal epithelial self-renewal by resolving ER stress. PLoS Genet. 2016;12(10):e1006349. https://doi.org/10.1371/journal.pgen.1006349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Qin Q, Wang X, Zhou B. Functional studies of Drosophila zinc transporters reveal the mechanism for dietary zinc absorption and regulation. BMC Biol. 2013;11(1):101. https://doi.org/10.1186/1741-7007-11-101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dunkley PR, Bobrovskaya L, Graham ME, von Nagy-Felsobuki EI, Dickson PW. Tyrosine hydroxylase phosphorylation: regulation and consequences. J Neurochem. 2004;91(5):1025–43. https://doi.org/10.1111/j.1471-4159.2004.02797.x.

    Article  CAS  PubMed  Google Scholar 

  44. Neckameyer WS, Quinn WG. Isolation and characterization of the gene for Drosophila tyrosine hydroxylase. Neuron. 1989;2(2):1167–75. https://doi.org/10.1016/0896-6273(89)90183-9.

    Article  CAS  PubMed  Google Scholar 

  45. Neckameyer WS, White K. Drosophila tyrosine hydroxylase is encoded by the pale locus. J Neurogenet. 1993;8(4):189–99. https://doi.org/10.3109/01677069309083448.

    Article  CAS  PubMed  Google Scholar 

  46. Neckameyer WS, Holt B, Paradowski TJ. Biochemical conservation of recombinant Drosophila tyrosine hydroxylase with its mammalian cognates. Biochem Genet. 2005;43(7-8):425–43. https://doi.org/10.1007/s10528-005-6781-3.

    Article  CAS  PubMed  Google Scholar 

  47. Vie A, et al. Differential regulation of Drosophila tyrosine hydroxylase isoforms by dopamine binding and cAMP-dependent phosphorylation. J Biol Chem. 1999;274(24):16788–95. https://doi.org/10.1074/jbc.274.24.16788.

    Article  CAS  PubMed  Google Scholar 

  48. Krishnakumar S, Burton D, Rasco J, Chen X, O'donnell J. Functional interactions between GTP cyclohydrolase I and tyrosine hydroxylase in Drosophila. J Neurogenet. 2000;14(1):1–23. https://doi.org/10.3109/01677060009083474.

    Article  CAS  PubMed  Google Scholar 

  49. O’Donnell JM, McLean JR, Reynolds ER. Molecular and developmental genetics of the Punch locus, a pterin biosynthesis gene in Drosophila melanogaster. Dev Genet. 1989;10(3):273–86. https://doi.org/10.1002/dvg.1020100316.

    Article  PubMed  Google Scholar 

  50. Yamamoto K, Kobayashi N, Yoshitama K, Teramoto S, Komamine A. Isolation and purification of tyrosine hydroxylase from callus cultures of Portulaca grandiflora. Plant Cell Physiol. 2001;42(9):969–75. https://doi.org/10.1093/pcp/pce125.

    Article  CAS  Google Scholar 

  51. Kim DH, Shin M, Jung SH, Kim YJ, Jones WD. A fat-derived metabolite regulates a peptidergic feeding circuit in Drosophila. PLoS Biol. 2017;15(3):e2000532. https://doi.org/10.1371/journal.pbio.2000532.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Haavik J, et al. Recombinant human tyrosine hydroxylase isozymes. Reconstitution with iron and inhibitory effect of other metal ions. Eur J Biochem. 1991;199(2):371–8. https://doi.org/10.1111/j.1432-1033.1991.tb16133.x.

    Article  CAS  PubMed  Google Scholar 

  53. Haavik J, et al. The incorporation of divalent metal ions into recombinant human tyrosine hydroxylase apoenzymes studied by intrinsic fluorescence and 1H-NMR spectroscopy. Eur J Biochem. 1992;210(1):23–31. https://doi.org/10.1111/j.1432-1033.1992.tb17386.x.

    Article  CAS  PubMed  Google Scholar 

  54. Yuan M, Li X, Xiao J, Wang S. Molecular and functional analyses of COPT/Ctr-type copper transporter-like gene family in rice. BMC Plant Biol. 2011;11(1):69. https://doi.org/10.1186/1471-2229-11-69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Qiao W, Ellis C, Steffen J, Wu CY, Eide DJ. Zinc status and vacuolar zinc transporters control alkaline phosphatase accumulation and activity in Saccharomyces cerevisiae. Mol Microbiol. 2009;72(2):320–34. https://doi.org/10.1111/j.1365-2958.2009.06644.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ramsey AJ, Daubner SC, Ehrlich JI, Fitzpatrick PF. Identification of iron ligands in tyrosine hydroxylase by mutagenesis of conserved histidinyl residues. Protein Sci. 1995;4(10):2082–6. https://doi.org/10.1002/pro.5560041013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Braak H, Ghebremedhin E, Rüb U, Bratzke H, del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res. 2004;318(1):121–34. https://doi.org/10.1007/s00441-004-0956-9.

    Article  PubMed  Google Scholar 

  58. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3(12):1301–6. https://doi.org/10.1038/81834.

    Article  CAS  PubMed  Google Scholar 

  59. Sherer TB, Kim JH, Betarbet R, Greenamyre JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol. 2003;179(1):9–16. https://doi.org/10.1006/exnr.2002.8072.

    Article  CAS  PubMed  Google Scholar 

  60. Calap-Quintana P, González-Fernández J, Sebastiá-Ortega N, Llorens J, Moltó M. Drosophila melanogaster Models of Metal-Related Human Diseases and Metal Toxicity. Int J Mol Sci. 2017;18(7). https://doi.org/10.3390/ijms18071456.

  61. Xiao G, Zhou B. ZIP13: a study of Drosophila offers an alternative explanation for the corresponding human disease. Front Genet. 2017;8:234.

    Article  Google Scholar 

  62. Djuric A, Begic A, Gobeljic B, Stanojevic I, Ninkovic M, Vojvodic D, et al. Oxidative stress, bioelements and androgen status in testes of rats subacutely exposed to cadmium. Food Chem Toxicol. 2015;86:25–33. https://doi.org/10.1016/j.fct.2015.09.004.

    Article  CAS  PubMed  Google Scholar 

  63. Kuznetsova SS, Azarkina NV, Vygodina TV, Siletsky SA, Konstantinov AA. Zinc ions as cytochrome C oxidase inhibitors: two sites of action. Biochemistry (Mosc). 2005;70(2):128–36. https://doi.org/10.1007/s10541-005-0091-6.

    Article  CAS  Google Scholar 

  64. Maret W. The redox biology of redox-inert zinc ions. Free Radic Biol Med. 2019;134:311–26. https://doi.org/10.1016/j.freeradbiomed.2019.01.006.

    Article  CAS  PubMed  Google Scholar 

  65. Maret W. Zinc in Cellular Regulation: The Nature and Significance of “Zinc Signals”. Int J Mol Sci. 2017;18(11). https://doi.org/10.3390/ijms18112285.

  66. Ramsey AJ, Hillas PJ, Fitzpatrick PF. Characterization of the active site iron in tyrosine hydroxylase. Redox states of the iron. J Biol Chem. 1996;271(40):24395–400. https://doi.org/10.1074/jbc.271.40.24395.

    Article  CAS  PubMed  Google Scholar 

  67. Bjorklund G, Stejskal V, Urbina MA, Dadar M, Chirumbolo S, Mutter J. Metals and Parkinson’s disease: mechanisms and biochemical processes. Curr Med Chem. 2018;25(19):2198–214. https://doi.org/10.2174/0929867325666171129124616.

    Article  CAS  PubMed  Google Scholar 

  68. Tosato M, Di Marco V. Metal Chelation Therapy and Parkinson’s Disease: A Critical Review on the Thermodynamics of Complex Formation between Relevant Metal Ions and Promising or Established Drugs. Biomolecules. 2019;9(7). https://doi.org/10.3390/biom9070269.

  69. Thingholm TE, Ronnstrand L, Rosenberg PA. Why and how to investigate the role of protein phosphorylation in ZIP and ZnT zinc transporter activity and regulation. Cell Mol Life Sci. 2020;77(16):3085–102. https://doi.org/10.1007/s00018-020-03473-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chatterjee S, Sang TK, Lawless GM, Jackson GR. Dissociation of tau toxicity and phosphorylation: role of GSK-3beta, MARK and Cdk5 in a Drosophila model. Hum Mol Genet. 2009;18(1):164–77. https://doi.org/10.1093/hmg/ddn326.

    Article  CAS  PubMed  Google Scholar 

  71. Bergerman J, Blethen S. Determination of alkaline phosphatase isoenzymes. Clin Chim Acta. 1972;36(2):389–96. https://doi.org/10.1016/0009-8981(72)90013-7.

    Article  CAS  PubMed  Google Scholar 

  72. EarleArnow L. Colorimetric determination of the components of 3,4 dihydroxyphenylalanine-tyrosine mixtures. J Biol Chem. 1937;118(2):531–7. https://doi.org/10.1016/S0021-9258(18)74509-2.

    Article  Google Scholar 

  73. Rodriguez-Gomez JA, et al. Increased activity and expression of tyrosine hydroxylase in the rat substantia nigra after chronic treatment with nomifensine. Mol Pharmacol. 1997;52(4):641–7. https://doi.org/10.1124/mol.52.4.641.

    Article  CAS  PubMed  Google Scholar 

  74. Borelli MI, Rubio M, García ME, Flores LE, Gagliardino JJ. Tyrosine hydroxylase activity in the endocrine pancreas: changes induced by short-term dietary manipulation. BMC Endocr Disord. 2003;3(1):2. https://doi.org/10.1186/1472-6823-3-2.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Yeast strains were originally kind gifts from David Eide, PhD. Fly stocks were from the Bloomington Stock Center (Bloomington, IN, USA) and the Vienna Drosophila RNAi Center. We are thankful for the technical help from Junxuan Xu, Zhiqing Wang, Xuekai Ma, Xiaowen Ji, and other lab members. We also thank Zhiwu Zhu, PhD, and Miao Liu for reading and commenting on this manuscript.

Funding

This study was supported by the National Key Research and Development Program of China (2018YFA0900100) and the National Natural Science Foundation of China (31971087, 31671284).

Author information

Authors and Affiliations

Authors

Contributions

GX contributed to the methodology, investigation, data curation, formal analysis, conceptualization, and writing—original draft and writing—review. MZ contributed to the methodology, investigation, data curation, formal analysis, conceptualization, and writing—review. ZL and FD contributed to the methodology and data curation. BZ contributed to the conceptualization, writing—review and editing, project administration, supervision, and funding acquisition. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bing Zhou.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

Quantification of the signal intensity in Figure 4B. No obvious and consistent changes were observed. All values are presented as mean ± SEM and included in Additional file 2: table S13; n=4. Figure S2. L-dopa production is promoted by iron and suppressed by zinc in INVSc1. (A) The generation of L-dopa in INVSc1 was elevated by iron and inhibited by zinc added in the iron deficient medium. All values are presented as mean ± SEM and included in Additional file 2: table S14; n=3. *p<0.05, **p<0.01,***p<0.001. (B) L-dopa production in INVSc1 was inhibited by Catsup expression in the iron abundant medium. All values are presented as mean ± SEM and included in Additional file 2: table S15; n=3. *p<0.05. Figure S3. TH protein levels were not much changed under metal treatments. A coomassie blue staining of protein gel was performed to test the effect of iron and zinc on the expression of TH in E. coli. Figure S4. ICP-MS indicated that zinc competes with iron in binding with Drosophila TH. Zinc binding sites could exceed 1 at high zinc concentrations, possibly due to additional non-specific bindings. All values presented in this graph are included in Additional file 2: table S12. Figure S5. Effects of modulating zinc and iron homeostasis on the mobility and survival of flies in the absence of rotenone. (A) The mobility of flies of various genotypes raised on normal food. n=6. ***p<0.001. (B) The lifespan of flies of various genotypes in the absence of rotenone. Genotypes in (A) and (B) of the flies used were TH-Gal4>w1118 (control), TH-Gal4>Catsup RNAi, TH-Gal4>Catsup OE, TH-Gal4>dZnT7 OE, TH-Gal4>Mvl OE. All values are presented as mean ± SEM; n= 6. Figure S6. TH phosphorylation in SH-SY5Y cells under different metal or metal chelator treatments. Gel blot was cut around the potential target region as indicated by the markers and then hybridized to TH and P-TH respectively. These are the original gel exposure pictures. Related to Figure 4B.

Additional file 2.

The individual raw data values of figures and Additional files for number of replicates ≤ 6. All the data are cited in the figure legend.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiao, G., Zhao, M., Liu, Z. et al. Zinc antagonizes iron-regulation of tyrosine hydroxylase activity and dopamine production in Drosophila melanogaster. BMC Biol 19, 236 (2021). https://doi.org/10.1186/s12915-021-01168-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12915-021-01168-0

Keywords