Parkinsonism is the primary type of movement disorder in adults, encompassing a set of clinical symptoms, including rigidity, tremors, dystonia, bradykinesia, and postural instability. These symptoms are primarily caused by a deficiency in dopamine (DA), an essential neurotransmitter in the brain. Currently, the DA precursor levodopa (synthetic L-DOPA) is the standard medication to treat DA deficiency, but it only addresses symptoms rather than provides a cure. In this review, we provide an overview of disorders associated with DA dysregulation and deficiency, particularly Parkinson's disease and rare inherited disorders leading predominantly to dystonia and/or parkinsonism, even in childhood. Although levodopa is relatively effective for the management of motor dysfunctions, it is less effective for severe forms of parkinsonism and is also associated with side effects and a loss of efficacy over time. We present ongoing efforts to reinforce the effect of levodopa and to develop innovative therapies that target the underlying pathogenic mechanisms affecting DA synthesis and transport, increasing neurotransmission through disease-modifying approaches, such as cell-based therapies, nucleic acid- and protein-based biologics, and small molecules.

Parkinsonism refers to a constellation of clinical symptoms and manifestations primarily associated to motor dysfunction, such as rigidity, tremors, dystonia, bradykinesia and postural instability [1], and is considered as the predominant form of movement disorder in adults. The development of parkinsonism can be related to many factors such as traumatic brain injuries [2], inherited disorders [3], drugs [4] or prolonged exposure to toxins [5], but most cases are associated with Parkinson's disease (PD). In a molecular context, parkinsonism can result from oxidative stress or dysfunctions in the mitochondria, ubiquitin-proteasome system, endosomal trafficking, lysosomal autophagy or monoamine neurotransmission [3]. Regardless of the underlying molecular cause, patients with parkinsonism typically have a deficiency in dopamine (DA), due to direct effects from gene variants leading to deficiency in the function and/or levels of proteins involved in DA production, metabolism or signaling, or due to the indirect impacts of other dysfunctional cellular pathways that lead to the death of dopaminergic neurons, as occurs in PD.

DA is a monoamine neurotransmitter and hormone that is essential for signaling in the central and peripheral nervous system [6]. DA is responsible for memory, cognition, reward, learning and movement and thus, low DA levels contribute to parkinsonism. Due to the importance of DA for proper nervous system function, its synthesis, trafficking and metabolism must be tightly controlled. Monoamine neurotransmitters such as DA are synthesized from the amino acids L-Phe, L-Tyr and L-Trp in pathways involving the aromatic amino acid hydroxylases (AAAHs), a family of specialized enzymes comprised of phenylalanine hydroxylase (PAH), tyrosine hydroxylase (TH) and tryptophan hydroxylases 1 and 2 (TPH1 and TPH2), that require a catalytic non-heme ferrous iron and tetrahydrobiopterin (BH4) as cofactor [7]. The TPHs catalyze the rate-limiting step in serotonin biosynthesis and PAH hydroxylates L-Phe to L-Tyr, mainly in liver. In dopaminergic neurons, TH catalyzes the hydroxylation of L-Tyr to L-3,4-dihydroxyphenylalanine (L-DOPA), which is converted to DA by aromatic acid decarboxylase (AADC) (Figure 1).

Simplified overview of the pathways involved in the biosynthesis, metabolism and transport of DA, highlighting the targets of current therapeutic approaches designed to enhance DA signaling.

Figure 1.
Simplified overview of the pathways involved in the biosynthesis, metabolism and transport of DA, highlighting the targets of current therapeutic approaches designed to enhance DA signaling.

DA is synthesized by PAH, TH and AADC, and is then packed into synaptic vesicles by VMAT2 and released into the synapse for binding to receptors on the postsynaptic neuron. Residual DA in the synaptic cleft undergoes reuptake by DAT and is repackaged in synaptic vesicles or metabolized by monoamine oxidase (MAO) to 3,4-dihydroxyphenylacetic acid (DOPAC), which can be exported to astrocytes and microglia for further metabolism (see Meiser et al. [18] and Winner et al. [17]). See also Meiser et al. [18] for further reading on an alternative DA biosynthesis pathway. Current treatments for DA deficiencies that boost DA signaling involve administration of levodopa, inhibitors of AADC, COMT and/or MAO-B, BH4 (in BH4-deficiencies) and DA agonists. Abbreviations: 3-OMD, 3-O-methyldopa; 4a-OH-BH4, pterin-4-alpha-carbinolamine; 6-PTP, 6-pyruvoyl tetrahydropterin; AADC, aromatic acid decarboxylase; BH4, (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; DHNTP, 7,8-dihydroneopterin triphosphate; DHPR, dihydropteridine reductase; DOPAC, 3,4-dihydroxyphenylacetic acid; GTPCH1, GTP cyclohydrolase; GTP, guanosine triphosphate; L-DOPA, L-3,4-dihydroxyphenylalanine; MAO-B, monoamine oxidase B; PAH, phenylalanine hydroxylase; PCD, pterin-4-alpha-carbinolamine dehydratase; PTPS, 6-pyruvoyl tetrahydropterin synthase; qBH2, dihydrobiopterin quinoid; SR, sepiapterin reductase; TH, tyrosine hydroxylase; VMAT2, vesicular monoamine transporter 2.

Figure 1.
Simplified overview of the pathways involved in the biosynthesis, metabolism and transport of DA, highlighting the targets of current therapeutic approaches designed to enhance DA signaling.

DA is synthesized by PAH, TH and AADC, and is then packed into synaptic vesicles by VMAT2 and released into the synapse for binding to receptors on the postsynaptic neuron. Residual DA in the synaptic cleft undergoes reuptake by DAT and is repackaged in synaptic vesicles or metabolized by monoamine oxidase (MAO) to 3,4-dihydroxyphenylacetic acid (DOPAC), which can be exported to astrocytes and microglia for further metabolism (see Meiser et al. [18] and Winner et al. [17]). See also Meiser et al. [18] for further reading on an alternative DA biosynthesis pathway. Current treatments for DA deficiencies that boost DA signaling involve administration of levodopa, inhibitors of AADC, COMT and/or MAO-B, BH4 (in BH4-deficiencies) and DA agonists. Abbreviations: 3-OMD, 3-O-methyldopa; 4a-OH-BH4, pterin-4-alpha-carbinolamine; 6-PTP, 6-pyruvoyl tetrahydropterin; AADC, aromatic acid decarboxylase; BH4, (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin; COMT, catechol-O-methyltransferase; DAT, dopamine transporter; DHNTP, 7,8-dihydroneopterin triphosphate; DHPR, dihydropteridine reductase; DOPAC, 3,4-dihydroxyphenylacetic acid; GTPCH1, GTP cyclohydrolase; GTP, guanosine triphosphate; L-DOPA, L-3,4-dihydroxyphenylalanine; MAO-B, monoamine oxidase B; PAH, phenylalanine hydroxylase; PCD, pterin-4-alpha-carbinolamine dehydratase; PTPS, 6-pyruvoyl tetrahydropterin synthase; qBH2, dihydrobiopterin quinoid; SR, sepiapterin reductase; TH, tyrosine hydroxylase; VMAT2, vesicular monoamine transporter 2.

Close modal

The conversion of L-Tyr to L-DOPA is the rate-limiting step in DA synthesis [8]. Due to the cellular toxicity associated with excess DA, TH activity is tightly-regulated at both the transcriptional (e.g. regulation of mRNA levels, alternative splicing and modulation of mRNA stability) [9] and post-transcriptional levels (e.g. TH phosphorylation and dephosphorylation [10], protein-protein interactions, with GTPCH1, 14-3-3, and DNAJC12, among others [11–14] and DA feedback inhibition in interplay with phosphorylation at Ser40 [15,16]). Surplus of DA in the synapse is also taken back into the presynaptic terminal by the DA transporter (DAT) and is subsequently either metabolized by enzymes such as monoamine oxidase (MAO) or reloaded into vesicles by the monoamine transporter 2 (VMAT2) (Figure 1). Astrocytes and microglia have also been shown to be involved in further DA metabolism [17,18], and thus serve as disease modifiers in PD [19]. Furthermore, alternative pathways of DA synthesis from the precursor 3-methoxytyramine that result in the modulation of DA levels in vivo have been described for gut microbiota [20].

In the case of PD, the loss of DA-producing neurons is caused by a complex interplay between genetic and environmental factors with mechanisms that are still unclear. Variants in several genes have been associated with PD (GBA, LRRK2, PARK7, PINK1, PRKN, SNCA), but only ∼10% of reported cases have genetic etiology [21,22]. In the case of genetic errors in proteins directly involved in DA signaling, variants often lead to instability and misfolding, loss of function (loss of activity and/or interactions) and premature degradation, or to gain of function and aggregation, resulting to DA dysregulation and deficiency [23–25]. A list of rare deficiencies leading to decreased levels of DA and/or DA dysregulation that may underlie dystonia and parkinsonism since childhood is outlined in Table 1. Here, we focus on (i) defects in proteins involved in DA biosynthesis or transport; (ii) variants in enzymes involved in the synthesis and regeneration of the AAAH cofactor (BH4), and (iii) variants in molecular cochaperones that are involved in maintaining cellular homeostasis of the AAAHs, in particular the Hsp40 cochaperone DNAJC12. Except for the autosomal-dominant form of GTPCH1 (AD-GTPCH1) deficiency, all other deficiencies included in Table 1 are associated with biallelic variants, inherited in a recessive manner. For additional defects related to DA catabolism we refer to Brennenstuhl et al. [26] and Opladen and Hoffmann [27], and for a recent review on mouse models for inherited monoamine neurotransmitter disorders to Thöny et al. [28].

Table 1.
Overview of main proteins involved in DA synthesis and transport, and associated with inherited disorders that may cause DA deficiencies and underlie parkinsonism
Protein; geneFunctionPathophysiology
Phenylalanine hydroxylase (PAH); PAH PAH converts L-Phe to L-Tyr (Figure 1). High levels of L-Phe inhibit DA synthesis in the brain by inhibiting L-Tyr transport and TH activity [103,104]. PAH deficiency is associated with phenylketonuria (PKU), characterized by elevated L-Phe levels that can result in neurotoxicity and decreased DA levels. Untreated PKU patients present with intellectual disability, eczematous rash, autism, seizures, and motor deficits [80], although clear parkinsonism is rare in PKU [105]. Worldwide, PKU affects ∼0.45 million individuals [106]. 
Tyrosine hydroxylase (TH); TH TH catalyzes the conversion of L-Tyr to L-DOPA (Figure 1), the rate-limiting step in DA biosynthesis [107]. TH deficiency (THD) is a rare genetic disorder where patients present a phenotype ranging from L-DOPA responsive dystonia (DRD) to severe encephalopathy. Approximately 100 cases are reported so far [108]. 
GTP cyclohydrolase 1 (GTPCH1); GCH1 GTPCH1 is the first and rate-limiting enzyme in the de novo synthesis of BH4, responsible for the hydrolysis of GTP to DHNTP (Figure 1). Symptoms of GTPCH1 deficiency include hypertonia, DRD, gait difficulties, and hyperreflexia [109]. There are both autosomal dominant (AD) and autosomal recessive forms of GTPCH1 deficiency, the former being the most common cause of DRD. 
6-pyruvoyl tetrahydropterin synthase (PTPS); PTS PTPS catalyzes the second step in the synthesis of BH4, the conversion of DHNTP to 6-PTP (Figure 1). Individuals with PTPS deficiency present with impaired neurophysiological development, truncal hypotonia, dystonia, bradykinesia, swallowing difficulties and hyperthermia. This is the most common cause of BH4 deficiencies (54%) with ∼735 cases reported [110]. 
Sepiapterin reductase (SR or SPR); SPR SR catalyzes the final two-step reduction in 6-PTP to BH4 (Figure 1). SR deficiency (SRD) is characterized by motor and speech delay, axial hypotonia, dystonia and weakness [111]. 
Pterin-4-alpha-carbinolamine dehydratase (PCD); PCBD1 PCD acts on the regeneration of BH4, where it catalyzes the dehydration of the 4a-OH-BH4 to qBH2 (Figure 1). PCD deficiency is not associated with significant changes in L-Phe and neurotransmitter levels or severe clinical abnormalities, except for temporary fluctuations in tone [110]. 
Dihydropteridine reductase (DHPR); QDPR DHPR catalyzes the reduction in qBH2 to BH4 (Figure 1), using NADH as cofactor. DHPR deficiency results in progressive mental retardation, neuronal loss, abnormal vascular proliferation in the brain, basal ganglia calcification and sudden death [110]. It accounts for 33% of BH4 deficiencies, with ∼303 cases reported so far [110]. 
DnaJ homolog subfamily C, member 12 (DNAJC12); DNAJC12 DNAJC12 is an Hsp40 co-chaperone with a critical role in maintaining the protein homeostasis of the AAAHs, including PAH and TH [14]. DNAJC12 deficiency results in a multitude of neurological symptoms, including developmental delay, intellectual disability, infantile dystonia and parkinsonism [14,112,113]. So far, ∼52 patients have been described [114]. 
Aromatic L-amino acid decarboxylase (AADC or AAAD); DDC AADC decarboxylates L-DOPA to form DA (Figure 1). AADC deficiency is clinically characterized by infantile hypotonia, ophthalmic crisis and developmental delay [115]. 
Dopamine transporter (DAT); SLC6A3 DAT regulates DA homeostasis by transporting extracellular DA into the intracellular space (Figure 1). DAT is a key regulator of the amplitude and duration of dopaminergic transmission [116]. DAT deficiency leads to neuropsychiatric disorders and neurodegeneration [117], with a phenotype characterized by an infantile-onset hyperkinetic movement disorder [118,119]. Ng et al. reported 31 patients [119]. 
Vesicular monoamine transporter type 2 (VMAT2); SLC18A2 VMAT2 transports DA from the cytosol into synaptic vesicles, for their subsequent release from the neuron during neurotransmission (Figure 1). Moreover, the sequestration of cytosolic DA into vesicles prevents neuronal damage [120]. Clinical symptoms of VMAT2 deficiency include infantile-onset movement disorder, mood disturbance, autonomic instability, and developmental delay [121]. Twelve patients were reported by Padmakumar et al. [122]. 
Protein; geneFunctionPathophysiology
Phenylalanine hydroxylase (PAH); PAH PAH converts L-Phe to L-Tyr (Figure 1). High levels of L-Phe inhibit DA synthesis in the brain by inhibiting L-Tyr transport and TH activity [103,104]. PAH deficiency is associated with phenylketonuria (PKU), characterized by elevated L-Phe levels that can result in neurotoxicity and decreased DA levels. Untreated PKU patients present with intellectual disability, eczematous rash, autism, seizures, and motor deficits [80], although clear parkinsonism is rare in PKU [105]. Worldwide, PKU affects ∼0.45 million individuals [106]. 
Tyrosine hydroxylase (TH); TH TH catalyzes the conversion of L-Tyr to L-DOPA (Figure 1), the rate-limiting step in DA biosynthesis [107]. TH deficiency (THD) is a rare genetic disorder where patients present a phenotype ranging from L-DOPA responsive dystonia (DRD) to severe encephalopathy. Approximately 100 cases are reported so far [108]. 
GTP cyclohydrolase 1 (GTPCH1); GCH1 GTPCH1 is the first and rate-limiting enzyme in the de novo synthesis of BH4, responsible for the hydrolysis of GTP to DHNTP (Figure 1). Symptoms of GTPCH1 deficiency include hypertonia, DRD, gait difficulties, and hyperreflexia [109]. There are both autosomal dominant (AD) and autosomal recessive forms of GTPCH1 deficiency, the former being the most common cause of DRD. 
6-pyruvoyl tetrahydropterin synthase (PTPS); PTS PTPS catalyzes the second step in the synthesis of BH4, the conversion of DHNTP to 6-PTP (Figure 1). Individuals with PTPS deficiency present with impaired neurophysiological development, truncal hypotonia, dystonia, bradykinesia, swallowing difficulties and hyperthermia. This is the most common cause of BH4 deficiencies (54%) with ∼735 cases reported [110]. 
Sepiapterin reductase (SR or SPR); SPR SR catalyzes the final two-step reduction in 6-PTP to BH4 (Figure 1). SR deficiency (SRD) is characterized by motor and speech delay, axial hypotonia, dystonia and weakness [111]. 
Pterin-4-alpha-carbinolamine dehydratase (PCD); PCBD1 PCD acts on the regeneration of BH4, where it catalyzes the dehydration of the 4a-OH-BH4 to qBH2 (Figure 1). PCD deficiency is not associated with significant changes in L-Phe and neurotransmitter levels or severe clinical abnormalities, except for temporary fluctuations in tone [110]. 
Dihydropteridine reductase (DHPR); QDPR DHPR catalyzes the reduction in qBH2 to BH4 (Figure 1), using NADH as cofactor. DHPR deficiency results in progressive mental retardation, neuronal loss, abnormal vascular proliferation in the brain, basal ganglia calcification and sudden death [110]. It accounts for 33% of BH4 deficiencies, with ∼303 cases reported so far [110]. 
DnaJ homolog subfamily C, member 12 (DNAJC12); DNAJC12 DNAJC12 is an Hsp40 co-chaperone with a critical role in maintaining the protein homeostasis of the AAAHs, including PAH and TH [14]. DNAJC12 deficiency results in a multitude of neurological symptoms, including developmental delay, intellectual disability, infantile dystonia and parkinsonism [14,112,113]. So far, ∼52 patients have been described [114]. 
Aromatic L-amino acid decarboxylase (AADC or AAAD); DDC AADC decarboxylates L-DOPA to form DA (Figure 1). AADC deficiency is clinically characterized by infantile hypotonia, ophthalmic crisis and developmental delay [115]. 
Dopamine transporter (DAT); SLC6A3 DAT regulates DA homeostasis by transporting extracellular DA into the intracellular space (Figure 1). DAT is a key regulator of the amplitude and duration of dopaminergic transmission [116]. DAT deficiency leads to neuropsychiatric disorders and neurodegeneration [117], with a phenotype characterized by an infantile-onset hyperkinetic movement disorder [118,119]. Ng et al. reported 31 patients [119]. 
Vesicular monoamine transporter type 2 (VMAT2); SLC18A2 VMAT2 transports DA from the cytosol into synaptic vesicles, for their subsequent release from the neuron during neurotransmission (Figure 1). Moreover, the sequestration of cytosolic DA into vesicles prevents neuronal damage [120]. Clinical symptoms of VMAT2 deficiency include infantile-onset movement disorder, mood disturbance, autonomic instability, and developmental delay [121]. Twelve patients were reported by Padmakumar et al. [122]. 

While the increasing understanding of the pathogenic mechanisms behind parkinsonism has unveiled crucial pathways that can be targeted for the development of potential therapies, current treatments primarily aim at managing symptoms, but do not directly address the underlying molecular mechanisms that drive disease progression. Existing treatments, like levodopa, transiently elevate DA levels in the brain (Figure 1), since the conversion of the DA precursor L-DOPA to DA is not normally rate-limiting [29,30]. While levodopa is effective for the management of motor symptoms, its pharmacokinetics presents high interindividual variability and unpredictable plasma levels, leading to increased administration and unavoidable side effects, such as dyskinesia, after several years of treatment [31]. Thus, modifying the pharmacokinetics of levodopa by optimizing its delivery for prolonged release is a pursued strategy to alleviate the deterioration associated with prolonged treatments. This can be achieved by using innovative oral or non-oral delivery systems for extended-release, or by combining levodopa with other therapies [32]. So far, the most common approach is the co-administration with inhibitors of either AADC (i.e. carbidopa or benserazide) and/or peripheral COMT (i.e. tolcapone and entacapone). This prevents the peripheral degradation of levodopa before it reaches the brain, increasing its brain bioavailability and enhancing the effectiveness of the treatment [33]. Other approved approaches aiming to stabilize DA in the neuron, either independently or as an adjunct therapy to levodopa, are the use of MAO-B inhibitors (i.e. rasagiline and selegiline) [33]. Finally, DA receptor agonists like pramipexole and ropinirole bind with high-affinity to the D2 and D3 DA receptors, mimicking the actions of DA in the brain and effectively increases DA signaling [34] (Figure 1).

Numerous therapies are currently being developed that not only aim to alleviate symptoms but also target the correction of DA signaling (Figure 2). Decreased DA signaling is mainly caused by insufficient amounts of DA, inefficient signal transmission or death of dopaminergic neurons. In this chapter, we provide a brief overview of innovative therapeutic strategies that target these dysregulated processes.

Overview of novel therapies that aim to increase DA synthesis and signaling, focusing on cell-, nucleic acid-, and protein-based strategies, and small molecules.

Figure 2.
Overview of novel therapies that aim to increase DA synthesis and signaling, focusing on cell-, nucleic acid-, and protein-based strategies, and small molecules.

Cell-based strategies aim to increase DA by replenishing DA producing neurons in patients with neurodegeneration. These cells can either be derived from established cell lines or from the patients themselves. Nucleic acid-based strategies on the other hand, aim to increase wildtype proteins by delivering genes to compensate for deficient proteins, by using RNA to stabilize protein targets or by editing variants in parkinsonisms with genetic etiology. Protein-based strategies aim to increase the levels of deficient proteins, and peptides can also act as carriers to deliver DA. Lastly, small molecule compounds can impart general neuroprotective effects, or stabilize or modulate the activity of specific proteins involved in DA signaling.

Figure 2.
Overview of novel therapies that aim to increase DA synthesis and signaling, focusing on cell-, nucleic acid-, and protein-based strategies, and small molecules.

Cell-based strategies aim to increase DA by replenishing DA producing neurons in patients with neurodegeneration. These cells can either be derived from established cell lines or from the patients themselves. Nucleic acid-based strategies on the other hand, aim to increase wildtype proteins by delivering genes to compensate for deficient proteins, by using RNA to stabilize protein targets or by editing variants in parkinsonisms with genetic etiology. Protein-based strategies aim to increase the levels of deficient proteins, and peptides can also act as carriers to deliver DA. Lastly, small molecule compounds can impart general neuroprotective effects, or stabilize or modulate the activity of specific proteins involved in DA signaling.

Close modal

Cell-based therapies

PD is the most common cause of parkinsonism, and the second most common progressive neurological disorder after Alzheimer's disease. PD is characterized by the loss of dopaminergic neurons in the substantia nigra and the consequential striatal DA deficiency [35]. It appears that 50–80% of the dopaminergic neurons are already lost by the time motor symptoms become apparent in PD patients [35,36]. Although the pathogenic mechanisms in PD are not clear, mitochondrial dysfunction, oxidative stress, neuroinflammation and loss of protein homeostasis are affected in idiopathic PD and appear involved in the dopaminergic neuronal death process [3]. Thus, in parallel with efforts to develop therapies that specifically target these pathways to prevent and delay PD progression [37,38], cell-based strategies are also being developed to increase DA signaling in patients that already have substantial loss of DA-producing neurons [39].

The transplantation of fetal mesencephalic tissue, a region rich in dopaminergic neuroblasts, to the DA-depleted striatum in patients with PD is one strategy to restore DA neurotransmission. While this approach increased DA signaling and ameliorated motor symptoms in patients with advanced PD, these improvements were not clear with respect to positive clinical outcome [40,41], but a sustained improvement in motor control and movement was however observed even 15–18 years post-operatively [42]. Nevertheless, ethical concerns regarding transplantation of fetal mesencephalic tissue from donors and issues in acquiring a reliable supply of cells have hampered the development and wide use of this treatment, thus alternative cell sources, such as stem cells, are also being explored.

Stem cells are self-renewing and can differentiate into a variety of cell types. In particular, the use of pluripotent stem cells (PSCs) poses less ethical concerns and risks as the cells are easier to acquire and are amenable for autologous transplantation in some situations. These PSCs can either be derived from developing blastocysts (embryonic stem cells; ESCs) or reprogrammed from somatic cells (induced PSCs; iPSCs). Transplantation of dopaminergic neuron progenitor cells derived from an established human ESC line to the putamen of mice has been shown to be efficacious [43], safe, well-tolerated and effective with respect to clinical outcome in the Phase 1 clinical trial 1 year post-operatively [44]. A study conducted in the monkey-model of PD showed improved motor and non-motor symptoms with extensive neuron axon growth and strong DA activity 2 years after autologous transplantation of rhesus iPSC-derived dopaminergic progenitor cells, which does not require immunosuppression [45]. The autologous co-transplantation of regulatory T-cells with iPSC-derived dopaminergic neurons has also been recently found to increase transplanted cell survival in mice, presenting a promising strategy to further improve clinical outcomes of cell-based therapies for PD [46].

Nucleic acid-based therapies

Nucleic acid-based therapeutic strategies aim to treat diseases by modulating gene expression or restoring the normal function of gene products through gene editing, or by delivering DNA or mRNA through viral or non-viral approaches. One strategy to enhance DA signaling and treat parkinsonism is gene therapy, where DNA encoding for enzymes that may restore DA synthesis is delivered to the brain, using both adeno-associated virus and lentivirus to deliver the genes. The initial gene therapy trials for PD focused on the introduction of the AADC gene into specific target neurons to enhance the conversion of administered L-DOPA to DA [47]. More recently, preclinical and clinical studies have investigated the enhancement of DA production and signaling by transducing the TH, AADC, GCH1 genes together into non-degenerating neurons. Although no therapy has yet been approved, on-going clinical trials support the potential of the approach [48,49]. The efficacy of gene therapy using viral vectors is also being proven in inherited disorders associated with DA deficiency and parkinsonism. Ongoing preclinical and clinical gene therapy studies for inherited disorders of neurotransmitter metabolism using viral vectors include successful preclinical studies targeting AADC-, DAT- and PTPS-deficiencies, as well as interventional clinical trials for AADC [50].

With respect to RNA-therapies, full-length PAH mRNA encapsulated in nanoparticles seems effective in restoring PAH levels and L-Phe metabolism in PAH-deficient mice, demonstrating the possibility of increasing protein expression [51]. Moreover, the long non-coding RNA HULC has been reported to modulate its activity by facilitating PAH-substrate and PAH-cofactor interactions, thus potentiating the enzyme for L-Phe metabolism [52]. Shorter HULC mimics also showed corrective effect of L-Phe levels in mice carrying a PAH disease variant [52].

CRISPR-Cas9-based gene editing is also emerging as an alternative approach to correct pathogenic variants in vivo. Moreover, the safety and therapeutic potential of combining gene editing ex vivo and stem cell therapy has been demonstrated by exagamglogene autotemcel (exa-cel or Casgevy), the first ex vivo gene therapy for sickle cell anemia and the first CRISPR-Cas9-based therapy approved in the US [53]. In the case of PD, as well as monogenic forms of parkinsonism, CRISPR-based genome editing also appears promising and research is underway to correct the predominant genetic variant in glucocerebrosidase (GBA), linked to an elevated risk of developing PD, in both Gaucher's patients and asymptomatic carriers [54]. The effectiveness of this approach was also investigated preclinically in a mouse model with classic PKU, where rescue of the disease phenotype was observed after a single base change in PAH [55].

Protein-based and enzyme replacement therapies

Protein-based therapeutic strategies traditionally involve the targeted delivery of proteins or peptides to replace or modulate the activity of their target. A potential approach to treat parkinsonism is the administration of functional proteins to compensate for their dysfunction (also known as enzyme replacement therapy or ERT). Functional full-length TH has been successfully delivered to dopaminergic cells in culture and to mouse brain using maltodextrin nanoparticles, leading to increased L-DOPA synthesis [56]. Interestingly, an artificial TH-mimicking Fe3O4 nanozyme that hydroxylates L-Tyr to L-DOPA and that also includes transferrin receptor aptamers to allow efficient crossing of the BBB and internalization into neurons has been prepared [57]. The device also includes a block strand with antisense oligonucleotides to α-synuclein (SNCA) mRNA to allow the specific targeting of degenerating dopaminergic neurons, as well as antisense oligonucleotide treatment and fluorescence imaging. Treatment of a PD mouse model with FNA-Fe3O4 resulted in recovery of L-DOPA and DA levels and amelioration of motor symptoms [57].

Small molecules

Small molecules still dominate the pharmaceutical industry at present despite the development of biologics (e.g. cell-, gene- or protein- based therapies) due to easier administration, amenability to be produced through chemical and bioengineering processes, cheaper costs, stability and ability to evade the immune response. There are numerous drug discovery initiatives underway, from screening of extensive compound libraries against predefined targets to the investigation of potent natural products for the identification of novel active agents. Below, we discuss ongoing efforts to identify small molecules to treat parkinsonism that focus on increasing DA levels and DA signaling in the brain. A selection of these compounds is presented in Table 2.

Table 2.
Names, chemical structures, targets and detailed activities of small molecules aimed at increasing DA levels and DA signaling in the brain, offering potential as treatments for parkinsonism
Small molecules activating/stabilizing enzymes involved in DA signaling
NameStructureTargetActivity
Compound III (3-amino-2-benzyl-7-nitro-4-(2-quinolyl)-1,2-dihydroisoquinolin-1-one)   PAH Compound III stabilizes PAH in vitro and increases activity and steady-state protein levels in cells and mice [63]. 
  TH Compound III stabilizes TH in vitro and increases total TH activity in mouse brains [68]. 
Compound IV (5,6-dimethyl-3-(4-methyl-2-pyridinyl)-2-thioxo-2,3-dihydrothieno[2,3-d] pyrimidin-4(1H)-one)   PAH Compound IV stabilizes PAH in vitro and increases activity and steady-state protein levels in cells and mice [63,69]. 
Compound 2 ((2E)-3-(2-chlorophenyl)-N-(4-[(3,4-dimethyl-1,2-oxazol-5-yl) sulfamoyl]phenyl) acrylamide)   TH Compound 2 stabilizes TH in vitro and increases the activity and steady-state protein levels in cells [64]. 
Compound 4 (N-allyl-2-(4-oxo-3,4,5,6,7,8-hexahydro[1] benzothieno[2,3-d]pyrimidin-2-yl) hydrazinecarbothio amide)   TH Compound 4 increases the half-life of the TH activity in vitro [64]. 
Compound 5 ((7S)-2,7-dimethyl-5,6,7,8-tetrahydro[1] benzothieno[2,3-d]pyrimidin-4(3H)-one)   TH Compound 5 stabilizes TH in vitro and increases activity and steady-state protein levels in cells [64]. 
Levalbuterol   TH Levalbuterol is an FDA-approved drug that stabilizes TH, lowers the feedback inhibition by DA and delays misfolding and aggregation of TH in vitro [65]. 
Ibogaine   DAT Ibogaine restores the folding of DAT, increases its surface expression and enhances DA transport in cellular and in vivo models [70,71]. 
Noribogaine   DAT Noribogaine restores the folding of DAT and enhances DA transport in cellular and in vivo models [70]. 
Bupropion   DAT Bupropion is an FDA-approved drug that increases the surface expression of DAT and enhances DA uptake activity in cells [71]. 
Sapropterin dihydrochloride (orally active synthetic form of BH4  GTPCH1, PTPS, DHPR, PCD, SPR, PAH, TH BH4 supplementation therapy (sapropterin dihydrochloride, Kuvan®) is used for BH4 deficiencies [79] and PAH deficiency [80]. For TH deficiencies, BH4 has demonstrated to stabilize TH in vitro and in vivo, increasing TH levels and activity [81] and improves phenotype in human iPSCs and in the THD mouse model [82]. 
Small molecules activating/stabilizing enzymes involved in DA signaling
NameStructureTargetActivity
Compound III (3-amino-2-benzyl-7-nitro-4-(2-quinolyl)-1,2-dihydroisoquinolin-1-one)   PAH Compound III stabilizes PAH in vitro and increases activity and steady-state protein levels in cells and mice [63]. 
  TH Compound III stabilizes TH in vitro and increases total TH activity in mouse brains [68]. 
Compound IV (5,6-dimethyl-3-(4-methyl-2-pyridinyl)-2-thioxo-2,3-dihydrothieno[2,3-d] pyrimidin-4(1H)-one)   PAH Compound IV stabilizes PAH in vitro and increases activity and steady-state protein levels in cells and mice [63,69]. 
Compound 2 ((2E)-3-(2-chlorophenyl)-N-(4-[(3,4-dimethyl-1,2-oxazol-5-yl) sulfamoyl]phenyl) acrylamide)   TH Compound 2 stabilizes TH in vitro and increases the activity and steady-state protein levels in cells [64]. 
Compound 4 (N-allyl-2-(4-oxo-3,4,5,6,7,8-hexahydro[1] benzothieno[2,3-d]pyrimidin-2-yl) hydrazinecarbothio amide)   TH Compound 4 increases the half-life of the TH activity in vitro [64]. 
Compound 5 ((7S)-2,7-dimethyl-5,6,7,8-tetrahydro[1] benzothieno[2,3-d]pyrimidin-4(3H)-one)   TH Compound 5 stabilizes TH in vitro and increases activity and steady-state protein levels in cells [64]. 
Levalbuterol   TH Levalbuterol is an FDA-approved drug that stabilizes TH, lowers the feedback inhibition by DA and delays misfolding and aggregation of TH in vitro [65]. 
Ibogaine   DAT Ibogaine restores the folding of DAT, increases its surface expression and enhances DA transport in cellular and in vivo models [70,71]. 
Noribogaine   DAT Noribogaine restores the folding of DAT and enhances DA transport in cellular and in vivo models [70]. 
Bupropion   DAT Bupropion is an FDA-approved drug that increases the surface expression of DAT and enhances DA uptake activity in cells [71]. 
Sapropterin dihydrochloride (orally active synthetic form of BH4  GTPCH1, PTPS, DHPR, PCD, SPR, PAH, TH BH4 supplementation therapy (sapropterin dihydrochloride, Kuvan®) is used for BH4 deficiencies [79] and PAH deficiency [80]. For TH deficiencies, BH4 has demonstrated to stabilize TH in vitro and in vivo, increasing TH levels and activity [81] and improves phenotype in human iPSCs and in the THD mouse model [82]. 
Small molecules with neuroprotective activity
Name Structure Activity 
Epoxide 4   Epoxide 4 promotes the survival of cultured dopaminergic neurons and protect them against degeneration. In the PD mouse model, Epoxide 4 increases the density of DA neuron fibers in the striatum [83].  
MCC950 (N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl) carbamoyl)-4-(2-hydroxypropan-2-yl) furan-2-sulfonamide)   MCC950 mitigates motor deficits and nigrostriatal dopaminergic degeneration in multiple rodent PD models [84].  
BRF110 (4-[Methyl[2-phenyl-5-(2-propen-1-yl)-6-(trifluoromethyl)-4-pyrimidinyl]amino] benzoic acid)   BRF110 prevents dopaminergic neuron demise and striatal dopaminergic denervation in vivo. Remarkably, besides neuroprotection, BRF110 up-regulates TH, DDC and GCH1 transcription and increases striatal DA in the PD mouse model [85]. 
IRX4204 ((2E,4E)-3-Methyl-5-[(1S,2S)-2-methyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthalenyl) cyclopropyl]-2,4-pentadienoic acid)   IRX4204 promotes the survival and maintenance of nigral DA neurons in primary mesencephalic cultures, and attenuates motor deficits in a rat model of PD [86].  
Fasudil   Fasudil, an FDA-approved drug, reduces dopaminergic cell loss and improves motor function in a mouse model of PD [87]. 
Lapatinib   Administration of Lapatinib in a rat model of PD improves motor function and restores dopaminergic neurons by increasing TH expression, leading to elevated DA levels [88]. 
Pedicularioside A   Pedicularioside, a plant-derived terpene, has been shown to prevent the loss of DA neurons in a PD neuron model [90]. 
Celastrol   Celastrol, a plant-derived terpene, provides neuroprotection in PD cell models and also improves motor symptoms in PD mouse model [91]. 
Curcumin   Curcumin is a potent antioxidant from turmeric plant, with protects dopaminergic neurons and improves motor behavioral performance in PD animal models [92,93]. 
Epigallocatechin-3-gallate   A polyphenolic component extracted from green tea, which has shown neuroprotective effects in cellular and animal models of PD, through different molecular mechanisms such as the modulation of DA production [94]. 
Resveratrol   Resveratrol, a polyphenol contained in blueberries or grapes, exhibits potent neuroprotective activity in cellular and animal models of PD [95]. 
Small molecules with neuroprotective activity
Name Structure Activity 
Epoxide 4   Epoxide 4 promotes the survival of cultured dopaminergic neurons and protect them against degeneration. In the PD mouse model, Epoxide 4 increases the density of DA neuron fibers in the striatum [83].  
MCC950 (N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl) carbamoyl)-4-(2-hydroxypropan-2-yl) furan-2-sulfonamide)   MCC950 mitigates motor deficits and nigrostriatal dopaminergic degeneration in multiple rodent PD models [84].  
BRF110 (4-[Methyl[2-phenyl-5-(2-propen-1-yl)-6-(trifluoromethyl)-4-pyrimidinyl]amino] benzoic acid)   BRF110 prevents dopaminergic neuron demise and striatal dopaminergic denervation in vivo. Remarkably, besides neuroprotection, BRF110 up-regulates TH, DDC and GCH1 transcription and increases striatal DA in the PD mouse model [85]. 
IRX4204 ((2E,4E)-3-Methyl-5-[(1S,2S)-2-methyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-2-naphthalenyl) cyclopropyl]-2,4-pentadienoic acid)   IRX4204 promotes the survival and maintenance of nigral DA neurons in primary mesencephalic cultures, and attenuates motor deficits in a rat model of PD [86].  
Fasudil   Fasudil, an FDA-approved drug, reduces dopaminergic cell loss and improves motor function in a mouse model of PD [87]. 
Lapatinib   Administration of Lapatinib in a rat model of PD improves motor function and restores dopaminergic neurons by increasing TH expression, leading to elevated DA levels [88]. 
Pedicularioside A   Pedicularioside, a plant-derived terpene, has been shown to prevent the loss of DA neurons in a PD neuron model [90]. 
Celastrol   Celastrol, a plant-derived terpene, provides neuroprotection in PD cell models and also improves motor symptoms in PD mouse model [91]. 
Curcumin   Curcumin is a potent antioxidant from turmeric plant, with protects dopaminergic neurons and improves motor behavioral performance in PD animal models [92,93]. 
Epigallocatechin-3-gallate   A polyphenolic component extracted from green tea, which has shown neuroprotective effects in cellular and animal models of PD, through different molecular mechanisms such as the modulation of DA production [94]. 
Resveratrol   Resveratrol, a polyphenol contained in blueberries or grapes, exhibits potent neuroprotective activity in cellular and animal models of PD [95]. 
Small molecules inhibiting VMAT2 
Name Structure Target Activity 
Tetrabenazine   VMAT2 Tetrabenazine is an approved inhibitor of VMAT2 for the treatment of chorea associated to Huntington's disease [99]. Salmeterol and ziprasidone are FDA-approved compounds able to inhibit VMAT2, which could potentially be repurposed for the symptomatic treatment of tardive dyskinesia and other hyperkinetic movements [102]. 
Salmeterol    
Ziprasidone    
Small molecules inhibiting VMAT2 
Name Structure Target Activity 
Tetrabenazine   VMAT2 Tetrabenazine is an approved inhibitor of VMAT2 for the treatment of chorea associated to Huntington's disease [99]. Salmeterol and ziprasidone are FDA-approved compounds able to inhibit VMAT2, which could potentially be repurposed for the symptomatic treatment of tardive dyskinesia and other hyperkinetic movements [102]. 
Salmeterol    
Ziprasidone    

The small molecules are classified into three groups according to their functions: activators and stabilizers of enzymes involved in DA synthesis and signaling, small molecules with neuroprotective activity and inhibitors of VMAT2.

Small molecules are low-molecular mass organic compounds that selectively bind to their target proteins, resulting in modulation of conformation and/or activity. The interest in the development of compounds that restore function by correcting the folding and/or increasing stability to protect the targets from accelerated degradation (pharmacological chaperones, PCs) for the treatment of inborn error of metabolism and other misfolding diseases is increasing [58–60]. Successful therapies for Fabry's disease (Migalastat), transthyretin amyloidosis (Tafamidis), cystic fibrosis (Elexacafor and Tezakafor) have strengthened the PC-field. One of the most common strategies for identifying PCs is by high-throughput screening (HTS), which allows rapid testing of large compound libraries against a pre-selected target. HTS can be performed with biophysical screens, using purified proteins as targets, or cellular screens either monitoring the cellular phenotype or changes in protein levels [61]. This methodology is normally applied using large collections of diverse drug-like compounds or FDA-approved drugs, in this latter case leading to the possibility of drug repurposing, which may accelerate the drug development timeline and reduce costs related to hit development and safety testing [62].

HTS using differential scanning fluorimetry (DSF) to monitor the binding of compound hits to specific targets has been effective to identify compounds with PC capability for DA deficiencies, especially for PAH and TH [63–65]. Other screening methods relying on affinity-immobilization, immunoprecipitation and measurement of PAH activity by tandem mass spectrometry have also been developed [66]. In the case of PAH, HTS of 1000 compounds randomly selected from a larger diversity library identified two PCs (compounds III and IV) that stabilized PAH in vitro, increased activity and steady-state protein levels of wildtype and variant PAH in cells and mice [63]. Later, a new generation of compound IV-analogues producing higher affinity and stabilization of PAH were developed by alchemical design [67]. Compounds III and IV (Table 2) were also investigated for their potential activity on the other BH4 dependent AAAHs, TH and TPH2 [68], as these hydroxylases are highly-conserved in sequence, structure and catalytic mechanism. Compound III stabilized TH in vitro and increased total TH activity in mouse brains [68], representing a valuable proof of concept for the potential of PCs as therapies for the treatment of THD and other parkinsonisms. However, as this compound is not specific for TH, additional HTS runs were performed to identify more effective and specific compounds. HTS-DSF of 10 000 compounds identified three additional hits (compounds 2, 4 and 5; Table 2) that stabilized TH in vitro, with compound 5 additionally increasing TH activity when studied in cells transiently transfected with THD variants [64]. EPR, X-ray crystallization and docking revealed that compounds 4, 5 and previous compound IV constitute a group of binders that weakly coordinate via a pyrimidine nitrogen to the catalytic iron of TH without significant inhibition, and seem to protect the iron from rapid inactivation and formation of reactive oxygen species [64,69]. Levalbuterol (Table 2), a TH stabilizer obtained from HTS-DSF of the Prestwick chemical library of FDA-approved drugs, also seems to exert a similar protective mechanism by coordinating to the iron [65]. Despite their stabilizing effect on TH in vitro, the low affinity of these PCs implies too high doses to obtain in vivo proof of concept, and the search and design of more effective PCs, binding outside the active site to stabilize TH and variants specifically and effectively, continues. Another dopaminergic target for which PCs has been investigated is DAT. The impact of ibogaine and noribogaine (Table 2), PCs for the closely related serotonin transporter, was studied for the folding of DAT. These compounds demonstrated the ability to bind to the inward-facing DAT conformation, effectively restoring its folding and thereby enhancing DA transport in disease-related variants in both cellular and in vivo models [70]. Ibogaine and bupropion were also identified as compounds that increased the surface expression of DAT, enhanced DA uptake activity, and promoted the maturation of DAT protein in some disease-mutants in cells [71]. More recently, a series of analogues of ibogaine have been generated to find more effective PCs for DAT deficiencies [72,73]. A novel strategy to address DA deficiencies includes not only focusing on the enzymes responsible for DA synthesis or metabolism, but also addressing the molecular partners crucial for their homeostasis. For example, TH interacts with regulatory proteins such as 14-3-3 [74,75], GTPCH1 [76] and DNAJC12 [14] (Figure 1), which play essential roles in maintaining TH stability. The search for compounds that can modulate these protein–protein interactions is also emerging as a promising avenue for the development of alternative therapeutic approaches [25,77]. Likewise, enzyme cofactors are also considered to have PC potential for their target enzymes [78]. BH4 supplementation therapy (sapropterin dihydrochloride, Kuvan®) (Table 2) is used not only for BH4 deficiencies [79], but also is one of the option treatments for BH4 responsive PAH deficiency [80]. For TH deficiencies, BH4 has been found to stabilize TH, wildtype and variants, in vitro and in vivo, increasing TH levels and activity [81]. Recently it has also been shown that BH4 treatment improves THD phenotype in human iPSCs and in the THD mouse model [82].

Other small molecules have shown potential to develop and maintain dopaminergic neurons, such as Epoxide 4 [83], MCC950 [84], BRF110 [85] and IRX4204 [86] (Table 2). These molecules have been effective in increasing the survival of DA neurons, raising DA content in the brain, and alleviating motor symptoms in experimental PD animals. Furthermore, two FDA drugs approved for other applications, i.e. Fasudil [87] and Lapatinib (Table 2) [88], have been shown to boost striatal DA levels, enhance dopaminergic neuronal survival and improve the motor function in PD animal models. Fasudil is being evaluated in a clinical trial in PD patients (EudraCT: 2021-003879-34).

A large fraction of small molecules is extracted from natural compounds. Some natural products display well-known anti-oxidative and anti-inflammatory capabilities, inhibit protein misfolding, and boost mitochondrial homeostasis and other neuroprotective processes [89]. Notably, terpenes such as pedicularioside A [90] and celastrol [91] (Table 2), have shown efficacy in preventing the loss of dopaminergic neurons in both in vitro and in vivo studies. Additionally, polyphenols such as curcumin [92,93], epigallocatechin-3-gallate [94] or resveratrol [95] (Table 2) have also been demonstrated to provide neuroprotection through different pathways, exhibiting positive effects on motor and behavioral function by promoting the survival of dopaminergic neurons and increasing DA levels in different PD models [96,97].

Finally, it is worth mentioning that the general pathomechanisms in parkinsonisms rather infer the need of therapies that increase function of dysregulated or dysfunctional targets (both in PD and in the inherited disorders in Table 1), and most of the therapies discussed above thus refer to activating/stabilizing approaches. However, there are some instances with need of novel inhibitors. For example, in the treatment of tardive dyskinesia (TD) and other hyperkinetic movements, it may be beneficial to inhibit VMAT2 activity to reduce DA storage and release from pre-synaptic neurons into the synaptic cleft, reducing stimulation of post-synaptic DA receptors and subsequently decreasing dyskinetic movements. Reserpine is a known VMAT2 inhibitor that is used as an antipsychotic and for the symptomatic treatment of chorea associated to Huntington's disease [98], but it is no longer used today due to adverse side effects. Tetrabenazine [99] (Table 2), and its variants deutetrabenazine and valbenazine are also VMAT2 inhibitors that have been approved by the FDA for treating TD [100,101], but efforts continue to develop better VMAT2 inhibitors. Interestingly, target based HTS using DSF to monitor ligand binding can also be successfully applied to the discovery of inhibitors, after the hit binders are further evaluated in specific functional assays. This strategy was used for the identification of several approved drugs as novel VMAT2 inhibitors, such as salmeterol or ziprasidone (Table 2), with great potential to be repurposed for the symptomatic treatment of TD in PD and other conditions [102].

DA is a neurotransmitter that plays an essential role in many important body functions such as movement, memory, and pleasure. Insufficient levels of DA can lead to a spectrum of mental and physical health issues and contribute to the development of parkinsonisms. The current treatment to address DA deficiencies involves supplementation with levodopa, which is converted to DA in the brain. While levodopa effectively manages symptoms, it does not offer a cure for the underlying condition. Consequently, it is essential to investigate alternative treatment strategies to target the fundamental pathomechanisms of DA deficiencies. Numerous molecular mechanisms can disrupt DA levels in the brain, therefore several strategies and targets should be considered. Advances in understanding the molecular basis of different DA deficiencies have identified new therapeutic targets and promising disease-modifying strategies. However, translating these research findings into tangible improvements in patient care remains a significant challenge. Many of the promising strategies have yet to reach human clinical trials, primarily due to challenges related to pharmacokinetics, pharmacodynamics, safety and toxicity. Future research should address these aspects to facilitate the translation of scientific discoveries into practical and effective treatments for patients.

  • DA is an essential neurotransmitter involved in behavioral, cognitive, and motor functions. Deficiencies in DA lead to debilitating motor disorders, such as dystonia and parkinsonism. Pediatric inherited forms of these disorders often include neurodevelopmental and cognitive impairments.

  • The currently available treatments for DA deficiencies are primarily based on levodopa and exhibit variable efficacy, only addressing symptoms. Alternative treatment strategies that target the fundamental pathomechanisms of DA deficiencies are highly needed.

  • Novel disease-modifying therapeutic approaches, rooted in advancing insights into the underlying pathogenic mechanisms affecting DA synthesis and transport, hold great promise. These approaches aim not only to address parkinsonism but also to mitigate neurodevelopmental and cognitive impairments.

The authors declare that there are no potential conflicts of interest or competing financial interest associated with this article.

We acknowledge funds from Stiftelsen K.G. Jebsen (Center for Translational Research in Parkinson's Disease), La Fundació la Marató de TV3 (Project 202012-31-32-33), The Neuro-SysMed Center (RCN, Project No. 288164) (to A.M.), Fundación Ramon Areces (to G.G.-A.), and The Meltzer Research Fund (Project 103578) (to M.D.S.T.).

We thank current and past members of the Biorecognition research group for valuable discussions and feedback.

AAAH

aromatic amino acid hydroxylase

AADC

aromatic acid decarboxylase

AD

autosomal dominant

DA

dopamine

DAT

dopamine transporter

DHPR

dihydropteridine reductase

DSF

differential scanning fluorimetry

ESC

embryonic stem cell

HTS

high-throughput screening

iPSC

induced PSC

MAO

monoamine oxidase

PAH

phenylalanine hydroxylase

PC

pharmacological chaperone

PD

Parkinson's disease

PSC

pluripotent stem cell

TD

tardive dyskinesia

TH

tyrosine hydroxylase

1
Daniel
,
S.E.
and
Lees
,
A.J.
(
1993
)
Parkinson's Disease Society Brain Bank, London: overview and research
.
J. Neural Transm. Suppl.
39
,
165
172
2
Padmakumar
,
S.
,
Kulkarni
,
P.
,
Ferris
,
C.F.
,
Bleier
,
B.S.
and
Amiji
,
M.M.
(
2022
)
Traumatic brain injury and the development of parkinsonism: understanding pathophysiology, animal models, and therapeutic targets
.
Biomed. Pharmacother.
149
,
112812
3
Von Scheibler
,
E.N.M.M.
,
Van Eeghen
,
A.M.
,
De Koning
,
T.J.
,
Kuijf
,
M.L.
,
Zinkstok
,
J.R.
,
Müller
,
A.R.
et al. (
2023
)
Parkinsonism in genetic neurodevelopmental disorders: a systematic review
.
Mov. Disord. Clin. Pract.
10
,
17
31
4
Shin
,
H.W.
and
Chung
,
S.J.
(
2012
)
Drug-induced parkinsonism
.
J. Clin. Neurol.
8
,
15
21
5
Dalton
,
K.R.
,
Kidd
,
C.J.
and
Hack
,
N.
(
2023
)
Toxin induced parkinsonism and hospitalization related adverse outcome mitigation for Parkinson's disease: a comprehensive review
.
J. Clin. Med.
12
,
1074
6
Klein
,
M.O.
,
Battagello
,
D.S.
,
Cardoso
,
A.R.
,
Hauser
,
D.N.
,
Bittencourt
,
J.C.
and
Correa
,
R.G.
(
2019
)
Dopamine: functions, signaling, and association with neurological diseases
.
Cell. Mol. Neurobiol.
39
,
31
59
7
Fitzpatrick
,
P.F.
(
2023
)
The aromatic amino acid hydroxylases: structures, catalysis, and regulation of phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase
.
Arch. Biochem. Biophys.
735
,
109518
8
Nagatsu
,
T.
,
Levitt
,
M.
and
Udenfriend
,
S.
(
1964
)
Tyrosine hydroxylase. The initial step in norepinephrine biosynthesis
.
J. Biol. Chem.
239
,
2910
2917
9
Tekin
,
I.
,
Roskoski
, Jr,
R.
,
Carkaci-Salli
,
N.
and
Vrana
,
K.E.
(
2014
)
Complex molecular regulation of tyrosine hydroxylase
.
J. Neural. Transm.
121
,
1451
1481
10
Dunkley
,
P.R.
and
Dickson
,
P.W.
(
2019
)
Tyrosine hydroxylase phosphorylation in vivo
.
J. Neurochem.
149
,
706
728
11
Bowling
,
K.M.
,
Huang
,
Z.
,
Xu
,
D.
,
Ferdousy
,
F.
,
Funderburk
,
C.D.
,
Karnik
,
N.
et al. (
2008
)
Direct binding of GTP cyclohydrolase and tyrosine hydroxylase: regulatory interactions between key enzymes in dopamine biosynthesis
.
J. Biol. Chem.
283
,
31449
31459
12
Ichimura
,
T.
,
Isobe
,
T.
,
Okuyama
,
T.
,
Yamauchi
,
T.
and
Fujisawa
,
H.
(
1987
)
Brain 14-3-3 protein is an activator protein that activates tryptophan 5-monooxygenase and tyrosine 3-monooxygenase in the presence of Ca2+,calmodulin-dependent protein kinase II
.
FEBS Lett.
219
,
79
82
13
Leong
,
S.L.
,
Cappai
,
R.
,
Barnham
,
K.J.
and
Pham
,
C.L.
(
2009
)
Modulation of alpha-synuclein aggregation by dopamine: a review
.
Neurochem. Res.
34
,
1838
1846
14
Anikster
,
Y.
,
Haack
,
T.B.
,
Vilboux
,
T.
,
Pode-Shakked
,
B.
,
Thony
,
B.
,
Shen
,
N.
et al. (
2017
)
Biallelic mutations in DNAJC12 cause hyperphenylalaninemia, dystonia, and intellectual disability
.
Am. J. Hum. Genet.
100
,
257
266
15
Dickson
,
P.W.
and
Briggs
,
G.D.
(
2013
)
Tyrosine hydroxylase: regulation by feedback inhibition and phosphorylation
.
Adv. Pharmacol.
68
,
13
21
16
Bueno-Carrasco
,
M.T.
,
Cuellar
,
J.
,
Flydal
,
M.I.
,
Santiago
,
C.
,
Krakenes
,
T.A.
,
Kleppe
,
R.
et al. (
2022
)
Structural mechanism for tyrosine hydroxylase inhibition by dopamine and reactivation by Ser40 phosphorylation
.
Nat. Commun.
13
,
74
17
Winner
,
B.M.
,
Zhang
,
H.
,
Farthing
,
M.M.
,
Karchalla
,
L.M.
,
Lookingland
,
K.J.
and
Goudreau
,
J.L.
(
2017
)
Metabolism of dopamine in nucleus accumbens astrocytes is preserved in aged mice exposed to MPTP
.
Front. Aging Neurosci.
9
,
410
18
Meiser
,
J.
,
Weindl
,
D.
and
Hiller
,
K.
(
2013
)
Complexity of dopamine metabolism
.
Cell Commun. Signal.
11
,
34
19
Takahashi
,
S.
and
Mashima
,
K.
(
2022
)
Neuroprotection and disease modification by astrocytes and microglia in Parkinson disease
.
Antioxidants (Basel)
11
,
170
20
Bock
,
D.
,
Rothgangl
,
T.
,
Villiger
,
L.
,
Schmidheini
,
L.
,
Matsushita
,
M.
,
Mathis
,
N.
et al. (
2022
)
In vivo prime editing of a metabolic liver disease in mice
.
Sci. Transl. Med.
14
,
eabl9238
21
Day
,
J.O.
and
Mullin
,
S.
(
2021
)
The genetics of Parkinson's disease and implications for clinical practice
.
Genes (Basel)
12
,
1006
22
Lanore
,
A.
,
Casse
,
F.
,
Tesson
,
C.
,
Courtin
,
T.
,
Menon
,
P.J.
,
Sambin
,
S.
et al. (
2023
)
Differences in survival across monogenic forms of Parkinson's disease
.
Ann. Neurol.
94
,
123
132
23
Bross
,
P.
,
Corydon
,
T.J.
,
Andresen
,
B.S.
,
Jorgensen
,
M.M.
,
Bolund
,
L.
and
Gregersen
,
N.
(
1999
)
Protein misfolding and degradation in genetic diseases
.
Hum. Mutat.
14
,
186
198
24
Fossbakk
,
A.
,
Kleppe
,
R.
,
Knappskog
,
P.M.
,
Martinez
,
A.
and
Haavik
,
J.
(
2014
)
Functional studies of tyrosine hydroxylase missense variants reveal distinct patterns of molecular defects in dopa-responsive dystonia
.
Hum. Mutat.
35
,
880
890
25
Jung-Klawitter
,
S.
,
Richter
,
P.
,
Yuan
,
Y.
,
Welzel
,
K.
,
Kube
,
M.
,
Bahr
,
S.
et al. (
2024
)
Tyrosine hydroxylase variants influence protein expression, cellular localization, stability, enzymatic activity and the physical interaction between tyrosine hydroxylase and GTP cyclohydrolase 1
.
J. Inherit. Metab. Dis.
47
,
517
532
26
Brennenstuhl
,
H.
,
Jung-Klawitter
,
S.
,
Assmann
,
B.
and
Opladen
,
T.
(
2019
)
Inherited disorders of neurotransmitters: classification and practical approaches for diagnosis and treatment
.
Neuropediatrics
50
,
2
14
27
Opladen
,
T.
and
Hoffmann
,
G.F.
(
2022
) Disorders of monoamine metabolism. In
Physician's Guide to the Diagnosis, Treatment, and Follow-Up of Inherited Metabolic Diseases
(
Blau
,
N.
, et al., eds), pp.
313
329
,
Springer International Publishing
,
Cham
28
Thony
,
B.
,
Ng
,
J.
,
Kurian
,
M.A.
,
Mills
,
P.
and
Martinez
,
A.
(
2024
)
Mouse models for inherited monoamine neurotransmitter disorders
.
J. Inherit. Metab. Dis.
47
,
533
550
29
Carlsson
,
A.
,
Lindqvist
,
M.
and
Magnusson
,
T.
(
1957
)
3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists
.
Nature
180
,
1200
30
Liu
,
C.
,
Goel
,
P.
and
Kaeser
,
P.S.
(
2021
)
Spatial and temporal scales of dopamine transmission
.
Nat. Rev. Neurosci.
22
,
345
358
31
Willemsen
,
M.A.
,
Verbeek
,
M.M.
,
Kamsteeg
,
E.J.
,
de Rijk-van Andel
,
J.F.
,
Aeby
,
A.
,
Blau
,
N.
et al. (
2010
)
Tyrosine hydroxylase deficiency: a treatable disorder of brain catecholamine biosynthesis
.
Brain
133
,
1810
1822
32
Lees
,
A.
,
Tolosa
,
E.
,
Stocchi
,
F.
,
Ferreira
,
J.J.
,
Rascol
,
O.
,
Antonini
,
A.
et al. (
2023
)
Optimizing levodopa therapy, when and how? Perspectives on the importance of delivery and the potential for an early combination approach
.
Expert Rev. Neurother.
23
,
15
24
33
Deleu
,
D.
,
Northway
,
M.G.
and
Hanssens
,
Y.
(
2002
)
Clinical pharmacokinetic and pharmacodynamic properties of drugs used in the treatment of Parkinson's disease
.
Clin. Pharmacokinet.
41
,
261
309
34
Reavill
,
C.
,
Boyfield
,
I.
,
Coldwell
,
M.
and
Nelson
,
P.
(
2000
)
Comparative pharmacological study of ropinirole (SKF-101468) and its metabolites in rats
.
J. Pharm. Pharmacol.
52
,
1129
1135
35
Surmeier
,
D.J.
,
Obeso
,
J.A.
and
Halliday
,
G.M.
(
2017
)
Selective neuronal vulnerability in Parkinson disease
.
Nat. Rev. Neurosci.
18
,
101
113
36
Sulzer
,
D.
and
Surmeier
,
D.J.
(
2013
)
Neuronal vulnerability, pathogenesis, and Parkinson's disease
.
Mov. Disord.
28
,
41
50
37
Pardo-Moreno
,
T.
,
Garcia-Morales
,
V.
,
Suleiman-Martos
,
S.
,
Rivas-Dominguez
,
A.
,
Mohamed-Mohamed
,
H.
,
Ramos-Rodriguez
,
J.J.
et al. (
2023
)
Current treatments and new, tentative therapies for Parkinson's disease
.
Pharmaceutics
15
,
770
38
Ahmad
,
J.
,
Haider
,
N.
,
Khan
,
M.A.
,
Md
,
S.
,
Alhakamy
,
N.A.
,
Ghoneim
,
M.M.
et al. (
2022
)
Novel therapeutic interventions for combating Parkinson's disease and prospects of Nose-to-Brain drug delivery
.
Biochem. Pharmacol.
195
,
114849
39
Cha
,
Y.
,
Park
,
T.Y.
,
Leblanc
,
P.
and
Kim
,
K.S.
(
2023
)
Current status and future perspectives on stem cell-based therapies for Parkinson's disease
.
J. Mov. Disord.
16
,
22
41
40
Freed
,
C.R.
,
Greene
,
P.E.
,
Breeze
,
R.E.
,
Tsai
,
W.Y.
,
DuMouchel
,
W.
,
Kao
,
R.
et al. (
2001
)
Transplantation of embryonic dopamine neurons for severe Parkinson's disease
.
N. Engl. J. Med.
344
,
710
719
41
Olanow
,
C.W.
,
Goetz
,
C.G.
,
Kordower
,
J.H.
,
Stoessl
,
A.J.
,
Sossi
,
V.
,
Brin
,
M.F.
et al. (
2003
)
A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease
.
Ann. Neurol.
54
,
403
414
42
Kefalopoulou
,
Z.
,
Politis
,
M.
,
Piccini
,
P.
,
Mencacci
,
N.
,
Bhatia
,
K.
,
Jahanshahi
,
M.
et al. (
2014
)
Long-term clinical outcome of fetal cell transplantation for Parkinson disease: two case reports
.
JAMA Neurol.
71
,
83
87
43
Piao
,
J.
,
Zabierowski
,
S.
,
Dubose
,
B.N.
,
Hill
,
E.J.
,
Navare
,
M.
,
Claros
,
N.
et al. (
2021
)
Preclinical efficacy and safety of a human embryonic stem cell-derived midbrain dopamine progenitor product, MSK-DA01
.
Cell Stem Cell
28
,
217
229 e7
44
BlueRock Therapeutics
. (
2023
)
BlueRock's neuronal stem cell therapy for Parkinson's disease is first to show positive results in Phase I clinical study. Available from:
https://www.bluerocktx.com/bluerocks-neuronal-stem-cell-therapy-for-parkinsons-disease-is-first-to-show-positive-results-in-phase-i-clinical-study/
45
Tao
,
Y.
,
Vermilyea
,
S.C.
,
Zammit
,
M.
,
Lu
,
J.
,
Olsen
,
M.
,
Metzger
,
J.M.
et al. (
2021
)
Autologous transplant therapy alleviates motor and depressive behaviors in parkinsonian monkeys
.
Nat. Med.
27
,
632
639
46
Park
,
T.Y.
,
Jeon
,
J.
,
Lee
,
N.
,
Kim
,
J.
,
Song
,
B.
,
Kim
,
J.H.
et al. (
2023
)
Co-transplantation of autologous T(reg) cells in a cell therapy for Parkinson's disease
.
Nature
619
,
606
615
47
Eberling
,
J.L.
,
Jagust
,
W.J.
,
Christine
,
C.W.
,
Starr
,
P.
,
Larson
,
P.
,
Bankiewicz
,
K.S.
et al. (
2008
)
Results from a phase I safety trial of hAADC gene therapy for Parkinson disease
.
Neurology
70
,
1980
1983
48
Van Laar
,
A.D.
,
Van Laar
,
V.S.
,
San Sebastian
,
W.
,
Merola
,
A.
,
Elder
,
J.B.
,
Lonser
,
R.R.
et al. (
2021
)
An update on gene therapy approaches for Parkinson's disease: restoration of dopaminergic function
.
J. Parkinsons Dis.
11
,
S173
S182
49
Morizane
,
A.
(
2023
)
Cell therapy for Parkinson's disease with induced pluripotent stem cells
.
Inflamm Regen.
43
,
16
50
Chu
,
W.S.
,
Ng
,
J.
,
Waddington
,
S.N.
and
Kurian
,
M.A.
(
2024
)
Gene therapy for neurotransmitter-related disorders
.
J. Inherit. Metab. Dis.
47
,
176
191
51
Perez-Garcia
,
C.G.
,
Diaz-Trelles
,
R.
,
Vega
,
J.B.
,
Bao
,
Y.
,
Sablad
,
M.
,
Limphong
,
P.
et al. (
2022
)
Development of an mRNA replacement therapy for phenylketonuria
.
Mol. Ther. Nucleic Acids.
28
,
87
98
52
Abrahams
,
S.
,
Miller
,
H.C.
,
Lombard
,
C.
,
van der Westhuizen
,
F.H.
and
Bardien
,
S.
(
2021
)
Curcumin pre-treatment may protect against mitochondrial damage in LRRK2-mutant Parkinson's disease and healthy control fibroblasts
.
Biochem. Biophys. Rep.
27
,
101035
53
Rostami
,
T.
,
Rad
,
S.
,
Rostami
,
M.R.
,
Mirhosseini
,
S.A.
,
Alemi
,
H.
,
Khavandgar
,
N.
et al. (
2024
)
Hematopoietic stem cell transplantation in sickle cell disease: A multidimentional review
.
Cell Transplant.
33
,
1
13
54
ASPEN
(
2023
)
ASPEN'S clinical pipeline. Available from:
https://aspenneuroscience.com/technology/pipeline/
55
Rich
,
B.E.
,
Jackson
,
J.C.
,
de Ora
,
L.O.
,
Long
,
Z.G.
,
Uyeda
,
K.S.
and
Bess
,
E.N.
(
2022
)
Alternative pathway for dopamine production by acetogenic gut bacteria that O-Demethylate 3-Methoxytyramine, a metabolite of catechol O-Methyltransferase
.
J. Appl. Microbiol.
133
,
1697
1708
56
Bezem
,
M.T.
,
Johannessen
,
F.G.
,
Jung-Kc
,
K.
,
Gundersen
,
E.T.
,
Jorge-Finnigan
,
A.
,
Ying
,
M.
et al. (
2018
)
Stabilization of human tyrosine hydroxylase in maltodextrin nanoparticles for delivery to neuronal cells and tissue
.
Bioconjug. Chem.
29
,
493
502
57
Fang
,
X.
,
Yuan
,
M.
,
Zhao
,
F.
,
Yu
,
A.
,
Lin
,
Q.
,
Li
,
S.
et al. (
2023
)
In situ continuous Dopa supply by responsive artificial enzyme for the treatment of Parkinson's disease
.
Nat. Commun.
14
,
2661
58
Bustad
,
H.J.
,
Kallio
,
J.P.
,
Vorland
,
M.
,
Fiorentino
,
V.
,
Sandberg
,
S.
,
Schmitt
,
C.
et al. (
2021
)
Acute intermittent porphyria: an overview of therapy developments and future perspectives focusing on stabilisation of HMBS and proteostasis regulators
.
Int. J. Mol. Sci.
22
,
675
59
Liguori
,
L.
,
Monticelli
,
M.
,
Allocca
,
M.
,
Hay Mele
,
B.
,
Lukas
,
J.
,
Cubellis
,
M.V.
et al. (
2020
)
Pharmacological chaperones: a therapeutic approach for diseases caused by destabilizing missense mutations
.
Int. J. Mol. Sci.
21
,
489
60
Tao
,
Y.X.
and
Conn
,
P.M.
(
2018
)
Pharmacoperones as novel therapeutics for diverse protein conformational diseases
.
Physiol. Rev.
98
,
697
725
61
Brennecke
,
P.
,
Rasina
,
D.
,
Aubi
,
O.
,
Herzog
,
K.
,
Landskron
,
J.
,
Cautain
,
B.
et al. (
2019
)
EU-OPENSCREEN: a novel collaborative approach to facilitate chemical biology
.
SLAS Discov.
24
,
398
413
62
Kulkarni
,
V.S.
,
Alagarsamy
,
V.
,
Solomon
,
V.R.
,
Jose
,
P.A.
and
Murugesan
,
S.
(
2023
)
Drug repurposing: an effective tool in modern drug discovery
.
Russ. J. Bioorg. Chem.
49
,
157
166
63
Pey
,
A.L.
,
Ying
,
M.
,
Cremades
,
N.
,
Velazquez-Campoy
,
A.
,
Scherer
,
T.
,
Thony
,
B.
et al. (
2008
)
Identification of pharmacological chaperones as potential therapeutic agents to treat phenylketonuria
.
J. Clin. Invest.
118
,
2858
2867
64
Hole
,
M.
,
Underhaug
,
J.
,
Diez
,
H.
,
Ying
,
M.
,
Rohr
,
A.K.
,
Jorge-Finnigan
,
A.
et al. (
2015
)
Discovery of compounds that protect tyrosine hydroxylase activity through different mechanisms
.
Biochim. Biophys. Acta
1854
,
1078
1089
65
Flydal
,
M.I.
,
Krakenes
,
T.A.
,
Tai
,
M.D.S.
,
Tran
,
M.P.A.
,
Teigen
,
K.
and
Martinez
,
A.
(
2021
)
Levalbuterol lowers the feedback inhibition by dopamine and delays misfolding and aggregation in tyrosine hydroxylase
.
Biochimie
183
,
126
132
66
Zhang
,
Z.
,
Ma
,
D.
,
Wang
,
X.
,
Wang
,
Y.
,
Li
,
Y.
,
Yang
,
P.
et al. (
2021
)
A joint method for the screening of pharmacological chaperones for phenylalanine hydroxylase
.
Org. Biomol. Chem.
19
,
5827
5835
67
Conde-Gimenez
,
M.
,
Galano-Frutos
,
J.J.
,
Galiana-Cameo
,
M.
,
Mahia
,
A.
,
Victor
,
B.L.
,
Salillas
,
S.
et al. (
2022
)
Alchemical design of pharmacological chaperones with higher affinity for phenylalanine hydroxylase
.
Int. J. Mol. Sci.
23
,
4502
68
Calvo
,
A.C.
,
Scherer
,
T.
,
Pey
,
A.L.
,
Ying
,
M.
,
Winge
,
I.
,
McKinney
,
J.
et al. (
2010
)
Effect of pharmacological chaperones on brain tyrosine hydroxylase and tryptophan hydroxylase 2
.
J. Neurochem.
114
,
853
863
69
Torreblanca
,
R.
,
Lira-Navarrete
,
E.
,
Sancho
,
J.
and
Hurtado-Guerrero
,
R.
(
2012
)
Structural and mechanistic basis of the interaction between a pharmacological chaperone and human phenylalanine hydroxylase
.
Chembiochem
13
,
1266
1269
70
Kasture
,
A.
,
El-Kasaby
,
A.
,
Szollosi
,
D.
,
Asjad
,
H.M.M.
,
Grimm
,
A.
,
Stockner
,
T.
et al. (
2016
)
Functional rescue of a misfolded Drosophila melanogaster dopamine transporter mutant associated with a sleepless phenotype by pharmacological chaperones
.
J. Biol. Chem.
291
,
20876
20890
71
Beerepoot
,
P.
,
Lam
,
V.M.
and
Salahpour
,
A.
(
2016
)
Pharmacological chaperones of the dopamine transporter rescue dopamine transporter deficiency syndrome mutations in heterologous cells
.
J. Biol. Chem.
291
,
22053
22062
72
Bhat
,
S.
,
Guthrie
,
D.A.
,
Kasture
,
A.
,
El-Kasaby
,
A.
,
Cao
,
J.
,
Bonifazi
,
A.
et al. (
2021
)
Tropane-based ibogaine analog rescues folding-deficient serotonin and dopamine transporters
.
ACS Pharmacol. Transl. Sci.
4
,
503
516
73
Sutton
,
C.
,
Williams
,
E.Q.
,
Homsi
,
H.
,
Beerepoot
,
P.
,
Nazari
,
R.
,
Han
,
D.
et al. (
2022
)
Structure-activity relationships of dopamine transporter pharmacological chaperones
.
Front. Cell. Neurosci.
16
,
832536
74
Itagaki
,
C.
,
Isobe
,
T.
,
Taoka
,
M.
,
Natsume
,
T.
,
Nomura
,
N.
,
Horigome
,
T.
et al. (
1999
)
Stimulus-coupled interaction of tyrosine hydroxylase with 14-3-3 proteins
.
Biochemistry
38
,
15673
15680
75
Ghorbani
,
S.
,
Fossbakk
,
A.
,
Jorge-Finnigan
,
A.
,
Flydal
,
M.I.
,
Haavik
,
J.
and
Kleppe
,
R.
(
2016
)
Regulation of tyrosine hydroxylase is preserved across different homo- and heterodimeric 14-3-3 proteins
.
Amino Acids
48
,
1221
1229
76
Larbalestier
,
H.
,
Keatinge
,
M.
,
Watson
,
L.
,
White
,
E.
,
Gowda
,
S.
,
Wei
,
W.
et al. (
2022
)
GCH1 deficiency activates brain innate immune response and impairs tyrosine hydroxylase homeostasis
.
J. Neurosci.
42
,
702
716
77
Waloen
,
K.
,
Kleppe
,
R.
,
Martinez
,
A.
and
Haavik
,
J.
(
2017
)
Tyrosine and tryptophan hydroxylases as therapeutic targets in human disease
.
Expert Opin. Ther. Targets
21
,
167
180
78
Rodrigues
,
J.V.
,
Henriques
,
B.J.
,
Lucas
,
T.G.
and
Gomes
,
C.M.
(
2012
)
Cofactors and metabolites as protein folding helpers in metabolic diseases
.
Curr. Top. Med. Chem.
12
,
2546
2559
79
Opladen
,
T.
,
Lopez-Laso
,
E.
,
Cortes-Saladelafont
,
E.
,
Pearson
,
T.S.
,
Sivri
,
H.S.
,
Yildiz
,
Y.
et al. (
2020
)
Consensus guideline for the diagnosis and treatment of tetrahydrobiopterin (BH(4)) deficiencies
.
Orphanet J. Rare Dis.
15
,
126
80
Blau
,
N.
,
van Spronsen
,
F.J.
and
Levy
,
H.L.
(
2010
)
Phenylketonuria
.
Lancet
376
,
1417
1427
81
Thony
,
B.
,
Calvo
,
A.C.
,
Scherer
,
T.
,
Svebak
,
R.M.
,
Haavik
,
J.
,
Blau
,
N.
et al. (
2008
)
Tetrahydrobiopterin shows chaperone activity for tyrosine hydroxylase
.
J. Neurochem.
106
,
672
681
82
Jung-Kc
,
K.
,
Tristan-Noguero
,
A.
,
Altankhuyag
,
A.
,
Pinol Belenguer
,
D.
,
Prestegard
,
K.S.
,
Fernandez-Carasa
,
I.
et al. (
2024
)
Tetrahydrobiopterin (BH(4)) treatment stabilizes tyrosine hydroxylase: rescue of tyrosine hydroxylase deficiency phenotypes in human neurons and in a knock-in mouse model
.
J. Inherit. Metab. Dis.
47
,
494
508
83
Ardashov
,
O.V.
,
Pavlova
,
A.V.
,
Mahato
,
A.K.
,
Sidorova
,
Y.
,
Morozova
,
E.A.
,
Korchagina
,
D.V.
et al. (
2019
)
A novel small molecule supports the survival of cultured dopamine neurons and may restore the dopaminergic innervation of the brain in the MPTP mouse model of Parkinson's disease
.
ACS Chem. Neurosci.
10
,
4337
4349
84
Gordon
,
R.
,
Albornoz
,
E.A.
,
Christie
,
D.C.
,
Langley
,
M.R.
,
Kumar
,
V.
,
Mantovani
,
S.
et al. (
2018
)
Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice
.
Sci. Transl. Med.
10
,
eaah4066
85
Spathis
,
A.D.
,
Asvos
,
X.
,
Ziavra
,
D.
,
Karampelas
,
T.
,
Topouzis
,
S.
,
Cournia
,
Z.
et al. (
2017
)
Nurr1:RXRalpha heterodimer activation as monotherapy for Parkinson's disease
.
Proc. Natl Acad. Sci. U.S.A.
114
,
3999
4004
86
Wang
,
J.
,
Bi
,
W.
,
Zhao
,
W.
,
Varghese
,
M.
,
Koch
,
R.J.
,
Walker
,
R.H.
et al. (
2016
)
Selective brain penetrable Nurr1 transactivator for treating Parkinson's disease
.
Oncotarget
7
,
7469
7479
87
Zhao
,
Y.F.
,
Zhang
,
Q.
,
Xi
,
J.Y.
,
Li
,
Y.H.
,
Ma
,
C.G.
and
Xiao
,
B.G.
(
2015
)
Multitarget intervention of Fasudil in the neuroprotection of dopaminergic neurons in MPTP-mouse model of Parkinson's disease
.
J. Neurol. Sci.
353
,
28
37
88
Mansour
,
H.M.
,
Mohamed
,
A.F.
,
Khattab
,
M.M.
and
El-Khatib
,
A.S.
(
2023
)
Lapatinib ditosylate rescues motor deficits in rotenone-intoxicated rats: potential repurposing of anti-cancer drug as a disease-modifying agent in Parkinson's disease
.
Eur. J. Pharmacol.
954
,
175875
89
Rahman
,
M.M.
,
Wang
,
X.
,
Islam
,
M.R.
,
Akash
,
S.
,
Supti
,
F.A.
,
Mitu
,
M.I.
et al. (
2022
)
Multifunctional role of natural products for the treatment of Parkinson's disease: at a glance
.
Front. Pharmacol.
13
,
976385
90
Li
,
Y.Y.
,
Lu
,
J.H.
,
Li
,
Q.
,
Zhao
,
Y.Y.
and
Pu
,
X.P.
(
2008
)
Pedicularioside A from Buddleia lindleyana inhibits cell death induced by 1-methyl-4-phenylpyridinium ions (MPP+) in primary cultures of rat mesencephalic neurons
.
Eur. J. Pharmacol.
579
,
134
140
91
Lin
,
M.W.
,
Lin
,
C.C.
,
Chen
,
Y.H.
,
Yang
,
H.B.
and
Hung
,
S.Y.
(
2019
)
Celastrol inhibits dopaminergic neuronal death of Parkinson's disease through activating mitophagy
.
Antioxidants (Basel)
9
,
37
92
Nebrisi
,
E.E.
(
2021
)
Neuroprotective activities of curcumin in Parkinson's disease: a review of the literature
.
Int. J. Mol. Sci.
22
,
11248
93
Patel
,
A.
,
Olang
,
C.A.
,
Lewis
,
G.
,
Mandalaneni
,
K.
,
Anand
,
N.
and
Gorantla
,
V.R.
(
2022
)
An overview of Parkinson's disease: curcumin as a possible alternative treatment
.
Cureus
14
,
e25032
94
Wang
,
Y.
,
Wu
,
S.
,
Li
,
Q.
,
Lang
,
W.
,
Li
,
W.
,
Jiang
,
X.
et al. (
2022
)
Epigallocatechin-3-gallate: a phytochemical as a promising drug candidate for the treatment of Parkinson's disease
.
Front. Pharmacol.
13
,
977521
95
Yadav
,
E.
,
Yadav
,
P.
,
Khan
,
M.M.U.
,
Singh
,
H.
and
Verma
,
A.
(
2022
)
Resveratrol: a potential therapeutic natural polyphenol for neurodegenerative diseases associated with mitochondrial dysfunction
.
Front. Pharmacol.
13
,
922232
96
Arias-Sanchez
,
R.A.
,
Torner
,
L.
and
Fenton Navarro
,
B.
(
2023
)
Polyphenols and neurodegenerative diseases: potential effects and mechanisms of neuroprotection
.
Molecules
28
,
5415
97
Shah
,
M.A.
,
Faheem
,
H.I.
,
Hamid
,
A.
,
Yousaf
,
R.
,
Haris
,
M.
,
Saleem
,
U.
et al. (
2024
)
The entrancing role of dietary polyphenols against the most frequent aging-associated diseases
.
Med. Res. Rev.
44
,
235
274
98
Chandler
,
J.H.
(
1955
)
Reserpine in the treatment of Huntington's chorea
.
Med. Bull.
21
,
95
100
ISSN 0196-5336
99
Guay
,
D.R.
(
2010
)
Tetrabenazine, a monoamine-depleting drug used in the treatment of hyperkinetic movement disorders
.
Am. J. Geriatr. Pharmacother.
8
,
331
373
100
Peckham
,
A.M.
and
Nicewonder
,
J.A.
(
2019
)
VMAT2 inhibitors for tardive dyskinesia-practice implications
.
J. Pharm. Pract.
32
,
450
457
101
Golsorkhi
,
M.
,
Koch
,
J.
,
Pedouim
,
F.
,
Frei
,
K.
,
Bondariyan
,
N.
and
Dashtipour
,
K.
(
2024
)
Comparative analysis of deutetrabenazine and valbenazine as VMAT2 inhibitors for tardive dyskinesia: a systematic review
.
Tremor Other Hyperkinet. Mov.
14
,
13
102
Stove
,
S.I.
,
Skjevik
,
A.A.
,
Teigen
,
K.
and
Martinez
,
A.
(
2022
)
Inhibition of VMAT2 by beta2-adrenergic agonists, antagonists, and the atypical antipsychotic ziprasidone
.
Commun. Biol.
5
,
1283
103
Curtius
,
H.C.
,
Niederwieser
,
A.
,
Viscontini
,
M.
,
Leimbacher
,
W.
,
Wegmann
,
H.
,
Blehova
,
B.
et al. (
1981
)
Serotonin and dopamine synthesis in phenylketonuria
.
Adv. Exp. Med. Biol.
133
,
277
291
104
Boot
,
E.
,
Hollak
,
C.E.M.
,
Huijbregts
,
S.C.J.
,
Jahja
,
R.
,
van Vliet
,
D.
,
Nederveen
,
A.J.
et al. (
2017
)
Cerebral dopamine deficiency, plasma monoamine alterations and neurocognitive deficits in adults with phenylketonuria
.
Psychol. Med.
47
,
2854
2865
105
Velema
,
M.
,
Boot
,
E.
,
Engelen
,
M.
and
Hollak
,
C.
(
2015
)
Parkinsonism in phenylketonuria: a consequence of dopamine depletion?
JIMD Rep.
20
,
35
38
106
Hillert
,
A.
,
Anikster
,
Y.
,
Belanger-Quintana
,
A.
,
Burlina
,
A.
,
Burton
,
B.K.
,
Carducci
,
C.
et al. (
2020
)
The genetic landscape and epidemiology of phenylketonuria
.
Am. J. Hum. Genet.
107
,
234
250
107
Nagatsu
,
T.
and
Nagatsu
,
I.
(
2016
)
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.
123
,
1255
1278
108
Nygaard
,
G.
,
Szigetvari
,
P.D.
,
Grindheim
,
A.K.
,
Ruoff
,
P.
,
Martinez
,
A.
,
Haavik
,
J.
et al. (
2021
)
Personalized medicine to improve treatment of dopa-responsive dystonia-a focus on tyrosine hydroxylase deficiency
.
J. Pers. Med.
11
,
1186
109
Clot
,
F.
,
Grabli
,
D.
,
Cazeneuve
,
C.
,
Roze
,
E.
,
Castelnau
,
P.
,
Chabrol
,
B.
et al. (
2009
)
Exhaustive analysis of BH4 and dopamine biosynthesis genes in patients with Dopa-responsive dystonia
.
Brain
132
,
1753
1763
110
Eichwald
,
T.
,
da Silva
,
L.B.
,
Staats Pires
,
A.C.
,
Niero
,
L.
,
Schnorrenberger
,
E.
,
Filho
,
C.C.
et al. (
2023
)
Tetrahydrobiopterin: beyond its traditional role as a cofactor
.
Antioxidants (Basel)
12
,
1037
111
Wu
,
Y.
,
Chen
,
P.
,
Sun
,
L.
,
Yuan
,
S.
,
Cheng
,
Z.
,
Lu
,
L.
et al. (
2020
)
Sepiapterin reductase: characteristics and role in diseases
.
J. Cell. Mol. Med.
24
,
9495
9506
112
van Spronsen
,
F.J.
,
Himmelreich
,
N.
,
Rufenacht
,
V.
,
Shen
,
N.
,
Vliet
,
D.V.
,
Al-Owain
,
M.
et al. (
2018
)
Heterogeneous clinical spectrum of DNAJC12-deficient hyperphenylalaninemia: from attention deficit to severe dystonia and intellectual disability
.
J. Med. Genet.
55
,
249
253
113
Straniero
,
L.
,
Guella
,
I.
,
Cilia
,
R.
,
Parkkinen
,
L.
,
Rimoldi
,
V.
,
Young
,
A.
et al. (
2017
)
DNAJC12 and dopa-responsive nonprogressive parkinsonism
.
Ann. Neurol.
82
,
640
646
114
Deng
,
I.B.
,
Follett
,
J.
,
Bu
,
M.
and
Farrer
,
M.J.
(
2024
)
DNAJC12 in monoamine metabolism, neurodevelopment, and neurodegeneration
.
Mov. Disord.
39
,
249
258
115
Brun
,
L.
,
Ngu
,
L.H.
,
Keng
,
W.T.
,
Ch'ng
,
G.S.
,
Choy
,
Y.S.
,
Hwu
,
W.L.
et al. (
2010
)
Clinical and biochemical features of aromatic L-amino acid decarboxylase deficiency
.
Neurology
75
,
64
71
116
Torres
,
G.E.
,
Gainetdinov
,
R.R.
and
Caron
,
M.G.
(
2003
)
Plasma membrane monoamine transporters: structure, regulation and function
.
Nat. Rev. Neurosci.
4
,
13
25
117
Ng
,
J.
,
Barral
,
S.
,
De La Fuente Barrigon
,
C.
,
Lignani
,
G.
,
Erdem
,
F.A.
,
Wallings
,
R.
et al. (
2021
)
Gene therapy restores dopamine transporter expression and ameliorates pathology in iPSC and mouse models of infantile parkinsonism
.
Sci. Transl. Med.
13
,
eaaw1564
118
Kurian
,
M.A.
,
Li
,
Y.
,
Zhen
,
J.
,
Meyer
,
E.
,
Hai
,
N.
,
Christen
,
H.J.
et al. (
2011
)
Clinical and molecular characterisation of hereditary dopamine transporter deficiency syndrome: an observational cohort and experimental study
.
Lancet Neurol.
10
,
54
62
119
Ng
,
J.
,
Barral
,
S.
,
Waddington
,
S.N.
and
Kurian
,
M.A.
(
2023
)
Dopamine transporter deficiency syndrome (DTDS): expanding the clinical phenotype and precision medicine approaches
.
Cells
12
,
1737
120
Sulzer
,
D.
and
Zecca
,
L.
(
2000
)
Intraneuronal dopamine-quinone synthesis: a review
.
Neurotox. Res.
1
,
181
195
121
Rilstone
,
J.J.
,
Alkhater
,
R.A.
and
Minassian
,
B.A.
(
2013
)
Brain dopamine-serotonin vesicular transport disease and its treatment
.
N. Engl. J. Med.
368
,
543
550
122
Padmakumar
,
M.
,
Jaeken
,
J.
,
Ramaekers
,
V.
,
Lagae
,
L.
,
Greene
,
D.
,
Thys
,
C.
, et al. (
2019
)
A novel missense variant in SLC18A2 causes recessive brain monoamine vesicular transport disease and absent serotonin in platelets
.
JIMD Rep.
47
,
9
16
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).