Next Article in Journal
Hepatocyte Nuclear Factor 4α (HNF4α) Plays a Controlling Role in Expression of the Retinoic Acid Receptor β (RARβ) Gene in Hepatocytes
Next Article in Special Issue
HDAC6 Deficiency Has Moderate Effects on Behaviors and Parkinson’s Disease Pathology in Mice
Previous Article in Journal
Molecular Determinants of Species Specificity of α-Conotoxin TxIB towards Rat and Human α6/α3β4 Nicotinic Acetylcholine Receptors
Previous Article in Special Issue
Gut-to-Brain α-Synuclein Transmission in Parkinson’s Disease: Evidence for Prion-like Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Crucial Roles of Pitx3 in Midbrain Dopaminergic Neuron Development and Parkinson’s Disease-Associated Neurodegeneration

1
Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
2
Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 611731, China
3
Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(10), 8614; https://doi.org/10.3390/ijms24108614
Submission received: 29 March 2023 / Revised: 26 April 2023 / Accepted: 3 May 2023 / Published: 11 May 2023
(This article belongs to the Special Issue Development of Dopaminergic Neurons 2.0)

Abstract

:
The degeneration of midbrain dopaminergic (mDA) neurons, particularly in the substantia nigra pars compacta (SNc), is one of the most prominent pathological hallmarks of Parkinson’s disease (PD). To uncover the pathogenic mechanisms of mDA neuronal death during PD may provide therapeutic targets to prevent mDA neuronal loss and slow down the disease’s progression. Paired-like homeodomain transcription factor 3 (Pitx3) is selectively expressed in the mDA neurons as early as embryonic day 11.5 and plays a critical role in mDA neuron terminal differentiation and subset specification. Moreover, Pitx3-deficient mice exhibit some canonical PD-related features, including the profound loss of SNc mDA neurons, a dramatic decrease in striatal dopamine (DA) levels, and motor abnormalities. However, the precise role of Pitx3 in progressive PD and how this gene contributes to mDA neuronal specification during early stages remains unclear. In this review, we updated the latest findings on Pitx3 by summarizing the crosstalk between Pitx3 and its associated transcription factors in mDA neuron development. We further explored the potential benefits of Pitx3 as a therapeutic target for PD in the future. To better understand the transcriptional network of Pitx3 in mDA neuron development may provide insights into Pitx3-related clinical drug-targeting research and therapeutic approaches.

1. Background

Parkinson’s disease (PD) is the second most frequent age-dependent neurodegenerative disorder after Alzheimer’s disease and is estimated to affect more than 6 million people worldwide [1]. The typical clinical manifestations of PD include resting tremors, abnormal gait (shuffling and reduced step length), bradykinesia and rigidity, as well as nonmotor symptoms (hyposmia, constipation, and sleep behavioral disorder) [2,3]. These motor/non-motor symptoms could trigger widespread disability among PD patients [3]; therefore, how to slow down the disease progression to improve the quality of life in PD patients deserves more attention.
The main pathological feature of PD is the progressive loss of midbrain dopaminergic (mDA) neurons, which is possibly caused by mitochondrial dysfunction, the disruption of vesicle trafficking, autophagy impairment, or/and the defective proteasomal clearance of misfolded proteins [4,5]. These pathogenic effects, individually or collectively, lead to the apoptotic (programmed) cell death of mDA neurons in the end [6].
mDA neurons in the SNc and VTA of the midbrain are the primary DA source in the mammalian central nervous system [7]. During development, the precursors of DA neurons migrate from the neuroepithelium to the ventral midbrain. Sonic hedgehog (Shh) and fibroblast growth factor (Fgf8), two diffusible factors, collaborate to promote neuronal development. Lmx1a and Msx1 appear early, while precursor cells proliferate, and these are essential for the passage of the proliferating precursor cells into the first stages of differentiation into mDA neurons [8]. Lmx1b, the engrailed factors En1 and En2, Foxal, and other factors are also involved in the early stage of mDA-specific differentiation (at E9-E10 in mice) when the first gene for DA synthesis, amino acid decarboxylase (AADC), is induced. Between E9.5 and E13.5 in mice, midbrain DA progenitors exit the cell cycle and enter post-mitosis [9]. Then, at E10.5 and E11.5, the transcription factors for terminal differentiation are activated by Nurr1 and Pitx3, respectively. Nurr1 is required for the induction of tyrosine hydroxylase (TH) at E11.5, vesicular monoamine transporter 2 (Vmat2) at E12.5, and dopamine transporter (DAT) at E14 [10], and Pitx3 is necessary for some terminally differentiated SNc DA neurons to survive [7].
mDA neurons are generally divided into three main clusters, which are located in substantia nigra pars compacta (SNc), ventral tegmental area (VTA), and the retrorubral field (RRF). SNc mDA neurons primarily project their axons to the dorsolateral striatum, a component of the basal ganglia circuitry, and DA released from SNc mDA neurons mediates the activity of targeted striatal cells to regulate voluntary movements and motor learning [11]. In contrast, VTA mDA neurons mainly innervate the ventromedial striatum (nucleus accumbens, NAc), prefrontal cortex (PFC; mesocortical pathway), and limbic regions (mesolimbic network). DA release from VTA and RRF mDA neurons are involved in the modulation of cognitive, emotive/affective, motivational/salient, and rewarding behaviors [11]. All three types of mDA neurons undergo multiple developmental stages, including the patterning, induction, specification of mDA progenitors, and mDA neuron maturation, which are controlled by numerous genetic networks and cellular signaling pathways [12,13]. The distribution and projections of mDA neurons are detailed in Figure 1.
Currently, the main PD treatments still focus on increasing the DA level or regulating DA transmission via pharmacotherapy [14]. Moreover, patients who have noticeable tremors or uncontrollable motor fluctuations may also benefit from a deep brain stimulation operation [15]. However, instead of preventing or delaying the progression of the disease, these treatments can only partially mitigate the physical symptoms [16]. Meanwhile, substantial side effects, such as dyskinesia and impulse control difficulties, may appear after long-term pharmacological treatments [17]. Cell transplantation, a potential strategy used to replace impaired mDA neurons, has gained particular interest. It is known that some transcription factors are crucial for the growth, differentiation, specification, and transmitter synthesis of mDA neurons, including forkhead box A2 (Foxa2), LIM homeobox transcription factor1 A (Lmx1a), nuclear receptor-related factor1 (Nurr1), paired-like homeodomain3 (Pitx3), aldehyde Dehydrogenase 1 family member A1 (Ascl1), neurogenin2 (Ngn2), and neuronal differentiation 1 (NeuroD1) [16]. Pitx3 and Nurr1 are mdDA neuron-specific markers, whereas Ascl1, Ngn2, and NeuroD1 are markers for neural progenitor cells. Additionally, Neurotrophic factors, including glial-cell-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), also play a crucial part in the differentiation, development, and maintenance of mDA [16,18,19,20,21,22]. Thus, expanding the knowledge of these transcription factors and neurotrophic factors in neuronal formation will greatly benefit the generation of functional mDA neurons that are applied to cell replacement therapy for PD patients.
Among them, Pitx3 is mainly expressed in mDA neurons and plays an essential role in mDA neuronal development [23,24]. The expression of Pitx3 is known to initiate as early as embryonic day 11.5, and it is distributed in the lens, skeletal muscles, and mDA neurons during early development [23,25,26]. However, postnatally, its expression is preserved only in postmitotic mDA precursors and mature mDA neurons particularly in SN, suggesting a key role for Pitx3 in the maintenance of mDA neuronal function [23,24,27]. Pitx3-deficient Aphakia (ak) mice exhibit the preferential loss of SNc mdDA neurons during embryonic and postnatal development, reiterating that Pitx3 plays a critical role in early mdDA neuronal events. Additionally, the expression of Pitx3 was reported to be significantly downregulated in both PD patients’ peripheral blood lymphocytes and brain tissues [25,28]. Additionally, several Pitx3 gene variants have been identified to be linked to sporadic PD [29,30]. Thus, these data altogether establish the key role of Pitx3 not only in early mDA development, but also in retaining the normal physiological functions in adult neurons.
Pitx3 has the highest level of expression in mDA neurons among these transcription factors at the embryonic stages [31,32]. How this relatively abundant protein binds to other transcription factors to coordinate the mDA neuronal development is not clear yet. In this review, we focus on the discussion of Pitx3 and its crosstalk with other transcription factors during mDA neuronal specification and differentiation. Furthermore, the potential therapeutic role of Pitx3 is also summarized. Our study may help readers to further understand the Pitx3-centered neural transcription factor network and provide insights on how to improve future PD treatments.

2. Introduction of Pitx3

Pitx3, a member of the pituitary homeobox subfamily, is a transcription factor containing homeodomains located on chromosome 10q24 [33]. The first member of the pituitary homeobox family to be cloned was Pitxl (previously known as Ptx1). It was found to support pituitary development and function [34]. Shortly after a second family member, Pitx2 (called Ptx2 or Rieg1), was discovered to be involved with Rieger’s syndrome, causing a deformity of craniofacial characteristics [35] Then, Pitx3 (Plx3) was identified during embryogenesis. It is momentarily expressed in skeletal muscle and the eye lens [23], but after birth, the content of Pitx3 is highly restricted, and it is constitutively expressed in the SNc and VTA of the midbrain [23].

2.1. Pitx3 Structure

In humans, Pitx3 has four exons and is found on chromosome 10. The protein is 302 amino acids long, and the cDNA is 1407 bp in length [26]. The mouse Pitx3 gene also has four exons and is located on chromosome 19. cDNA has a length of 1379 bp, and the protein is also 302 amino acids long [25,36]. The pitx3 genes of humans and mice are highly homologous and nearly 90% identical at the nucleotide level throughout the coding region [36].

2.2. Pitx3 Protein

Pitx3 coordinates gene activity that determines cell destiny during development in a variety of species, including yeasts and mammals [25]. The protein has an OAR domain at its C-terminus, which is present in several types of paired-like homeodomains [31]. Although the function of OAR in the Pitx3 protein is uncertain, OAR’s impact on DNA binding appears to limit the transcriptional activity of Cartl protein. The OAR domain and N-terminus of the Pitx and Prx OAR-related proteins are thought to intramolecularly interact, resulting in a protein conformation that is connected to a generally inactive state of these transcription factors [37].

2.3. Pitx3 in the Main Subgroups of mDA Neurons

Pitx3 is a transcription factor required for the development and survival of mdDA neurons [38]. In the midbrain, DA neurons have two primary subgroups: the SNc and the VTA [7]. Although most mDA neurons express Pitx3, the Pitx3 deficiency in mice only impacts a portion of the mDA neurons. These findings show that Pitx3 may play distinct roles in these two midbrain subregions [25]. Pitx3 and TH are both expressed in the VTA at approximately the same time, and TH is still expressed in VTA even in the absence of Pitx3. However, in SNc, Pitx3 is expressed before TH is, and the SNc precursors are unable to generate TH without Pitx3 [39]. Thus, the VTA DA neurons in Pitx3-deficient newborn mice remain unaffected, whereas SNc neurons have been significantly lost potentially due to decreased TH levels [40,41]. These findings imply that Pitx3 is crucial for maintaining normal function and preserving the specific molecular identities of postnatal mDA neurons [42].

3. Pitx3 and Pitx3-Related Transcriptional Networks in mDA Neuron Development

The mDA neuronal population is unevenly distributed during early developmental stages, where it divides into several subpopulations with distinct topographical and temporal expression patterns [43]. Pitx3 is a key player in subset specification and is required for the proper survival of an mDA neuron subset in prenatal and postnatal mouse brains [44]. Based on its crucial role in mDA neurons, the interaction network of Pitx3 with other important transcription factors is also elucidated, which may contribute to a better understanding of mDA neuronal differentiation and maturation.

3.1. Pitx3 and Lmx1 a/b

LIM homeodomain transcription factors 1a and 1b (Lmx1a and Lmx1b) play key roles in the developing midbrain, where their expressions start on embryonic days 9.5 and 7.5, respectively [45,46]. Lmx1a is initially expressed in the ventral midbrain, and then gradually spreads to dorsal regions, whereas Lmx1b expression is constrained to several specific areas, including the ventral midbrain, floor plate, mid-hindbrain boundary, and roof plate [47]. The knockdown of Lmx1a expression leads to the loss of mDA neurons, whereas the activation of this gene promotes the generation of mDA neurons [48]. Additionally, DAT-Lmx1a/bcKO adult mice, especially elderly mice, displayed motor, non-motor (olfaction), and cognitive impairments. Meanwhile, the model also showed lower striatal DA contents, altered synaptic structures at DA nerve terminals, and the progressive loss of mDA neurons and fibers [49].
Previous studies have reported that Lmx1a/b could directly regulate the expression of Nurr1 and Pitx3. Lmx1a binds directly to the murine Pitx3 promoter region in vivo, and retroviral Lmx1a expression increased the expression of Pitx3 in vitro. Moreover, Lmx1b is also bound to the promoters of Pitx3 and Nurr1, whereas in Lmx1b KO mice, the retained TH+ neurons all lack Pitx3 expression, indicating that Lmx1b is closely associated with Pitx3. Wnt1/β-catenin/LEF1 signaling plays an essential role in the development of the mouse’s ventral midbrain [50,51]. Lmx1a and Pitx3 are all potential direct targets of Wnt1/β-catenin/LEF1-mediated signaling [52], suggesting a cascade linking Wnt1/β-catenin/LEF1-Lmx1a-Pitx3 to mDA differentiation. Moreover, Lmx1b is also closely associated with Wnt1 signaling pathway, and the Wnt1-Lmx1b autoregulatory loop directly regulates Nurr1, En1, and Pitx3, all of which are crucial elements in mDA neuronal differentiation and survival [53,54]. Together, these observations suggest that Wnt1 signaling pathway is associated with Lmx1a/b and other important neural transcription factors (Nurr1, En1, and Pitx3) to participate in the early mDA developmental events.
Together, these observations suggest that Lmx1a/b and other important neural transcription factors (Nurr1, En1, and Pitx3) are associated with the Wnt1 signaling pathway to participate in the early mDA developmental events and Pitx3 could be directly regulated by Lmx1a/b.

3.2. Pitx3 and Nurr1

Orphan nuclear receptor Nurr1 (Nr4a2) is one of the main regulators for the development, survival, and functional maintenance of mDA neurons in vivo [10]. Nurr1 expression starts in the midbrain on embryonic day 10.5 and continues to be expressed in mDA neurons till adulthood [55]. Nurr1 is vital for establishing the dopaminergic phenotype in mDA neurons as it determines the expression of TH, the enzyme responsible for DA synthesis. In Nurr1-deficient mice, the expression of TH was completely absent during development, indicating that Nurr1 is essential for TH induction in mDA neurons [55,56]. Moreover, other important transcription factors (En1/2, FoxA2, Lmx1b, and Pitx3) were still retained in embryos; thus, no developmental abnormalities appeared in the mice. However, homozygous Nurr1 knock out mice died soon after birth, while heterozygous Nurr1+/− mice lived longer, but their mDA neurons were more vulnerable to neurotoxic damage caused by MPTP injection or proteasome inhibition. Meanwhile, they also exhibited severe age-related mDA neurodegeneration [57]. According to these findings, Nurr1 has a minor role in the early development of mDA neurons, but it is essential for the late survival and differentiation of mDA neurons.
Recent research has demonstrated the importance of the Lmx1a-miR-204/211-Nurr1 axis in the differentiation of mDA neurons [58]. Additionally, mice that are overexpressed with mutant alpha-synuclein and simultaneously lacking one Nurr1 allele showed the enhanced age-dependent reduction of Nurr1 protein levels, a decreased number of mDA neurons, as well as increased neuroinflammation and alpha-synuclein aggregation [59]. The possible mechanism underlying these pathological phenotypes is that alpha-synuclein promoted the GSK3B-mediated phosphorylation (a component of the Wnt1/β-catenin pathway) and the subsequent proteasomal degradation of Nurr1 [60], suggesting the presence of a Wnt1/β-catenin-lmx1a-Nurr1 pathway in the differentiation of mDA neurons.
Nurr1 and Pitx3 are both expressed during mDA neuron differentiation. Nurr1 is involved in DA synthesis and regulation, while Pitx3 is specifically engaged in the terminal differentiation and maintenance of mDA neurons. Moreover, Nurr1 is widely distributed in the cerebral cortex, hippocampus, thalamus, amygdala, and midbrain [61]. By contrast, Pitx3 is postnatally expressed only by mDA neurons and is constrained in the SNc and VTA regions. Clinical studies showed that the expression levels of Nurr1 and Pitx3 were significantly decreased in PD patients [62]. In addition, the presence of Pitx3 and Nurr1 is essential for triggering TH expressions in mDA neurons. A growing body of evidence indicates that Pitx3 and Nurr1 are functionally interconnected and cooperate to promote the differentiation of mouse and human ES cells toward the mDA phenotype [63]. Recent studies have shown that Lentiviral expression vectors that combined harbor Nurr1 and Pitx3 at the neural precursor stage dramatically and synergistically induced the expression of the late marker (DAT), and Pitx3 serves as a crucial Nurr1 potentiator in defining the dopaminergic phenotype [64]. Pitx3 triggers the release of SMRT/HDAC (co-repressor)-mediated repression from Nurr1 to regulate the Nurr1 transcriptional complex [64]. Specifically, Pitx3 and Nurr1 co-occupy promoters of mDA-related genes, and Pitx3 causes HDAC-mediated repression in the Nurr1 transcriptional complex. The loss of Pitx3 expression in vivo increases the interaction of co-repressor SMRT with Nurr1, causing Nurr1-mediated transcriptions of Vmat2, D2R, and TH to be repressed in an HDAC-dependent manner [64]. Furthermore, Pitx3 and Nurr1 interact with the promoter of Vmat2, a gene essential for the metabolism of DA [10].
These findings imply that Pitx3 could enhance the function of Nurr1 and they cooperatively induce the late maturation of midbrain DA neurons.

3.3. Pitx3 and Engrailed-1/Engrailed-2

Engrailed-1/Engrailed-2 (En1/2), a member of a family of transcription factors containing a DNA-binding homeodomain, started to express on embryonic day 8 in a sheet of cells in the anterior neuroepithelium of midbrain–hindbrain boundary. Later on, the expression of En1/2 was restricted to the midbrain (SNc and VTA regions) and hindbrain regions in adults [65,66,67]. The progressive loss of striatal neurons and SNc mDA neurons with associated motor abnormalities has been identified in the En1/2-deficient models [68]. These data indicated that En1/2 plays an important role in the process of terminal differentiation and is required for the acquisition and survival of mature neuronal identity during late embryonic life in a dose-dependent manner [13,69]. En1/2 also interacts with Wnt1 to control the induction of the midbrain–hindbrain boundary (MHB), and knockdown Wnt1 downregulates En1/2 expression in the MHB region [70].
Pitx3 and En1 were reported to be involved in the initiation of the expression of aldehyde dehydrogenase 1 family member A1 (Aldh1a1), a gene that is an important marker in rostro-lateral mDA neurons [13,71]. A previous study indicated a strong association between the Pitx3 promoter and PD (p = 0.0001), as well as between EN1 and PD (p = 0.046), after genotyping nine single nucleotide polymorphisms in the entire genomic region of Pitx3 and EN1 from 365 PD patients and 418 controls [72]. Thus, En1 and Pitx3 polymorphisms may function as genetic risk factors for sporadic PD.
En1 knock out mice showed a significant loss of mDA neurons and striatal innervation defects, which were similar to those observed in Pitx3 knock out mice [71]. Additionally, on embryonic day 14.5, a small number of mDA neurons expressed Nurr1, but not Pitx3 and En1. These neurons are known to lose their rostral or caudal subset identity, suggesting the necessity of Pitx3 and En1 in the rostral/caudal identity of mDA neuronal populations [13]. Moreover, Pitx3 regulates En1 activity through multiple Pitx3-driven modulatory proteins, and En1 expression is increased in the absence of Pitx3 [71]. Meanwhile, En1 may be also involved in the regulation of Pitx3 expression, and the levels of Pitx3 were reduced in En1-deleted animals [13,71]. On the other hand, Pitx3 plays an essential role in the survival of rostro-lateral mDA neurons, but not caudo-lateral ones. Accordingly, in a Pitx3 knock out model, most of the rostro-lateral mDA neurons were lost, but the caudo-lateral ones commonly referred to as VTA mDA neurons were retained [40,71], whereas in En1 knock out embryos, not only rostral mDA neuronal programming is affected, but also caudal mDA phenotypes [71], suggesting that Pitx3 and En1 contribute to the modulation and specification of different subtypes of mDA neurons.
These findings illustrate that Pitx3 and En1 are both genetic risk factors for sporadic Parkinson’s disease. Moreover, there is a close relationship between the two, with En1 probably increasing Pitx3 expression, while Pitx3 might be suppressed by En1 expression.

3.4. Pitx3 and Foxa1/2

Foxa1/2, members of the winged-helix/forkhead transcription factors [73], are essential for the generation, specification, differentiation, and transcription of mDA neurons during embryonic development [74,75]. Foxa1 was identified in the floorplate, notochord, and endoderm on embryonic day 7.5, and upon Foxa1-deficiency, the mice suffered from disrupted glucose homeostasis and died soon after birth [76,77,78]. Foxa2 is expressed in the posterior epiblast from the early stage onwards, and it is then confined to anterior definitive endoderm (ADE) and axial mesoderm, which consists of the head process, prechordal plate, notochord, and node [76]. Foxa2 expression is necessary for proper mesodermal and endodermal development among mice, and it starts to express as early as embryonic day 6.5. The consecutive Foxa2 loss results in embryonic lethality [76,77,78]. Together, both Foxa1 and Foxa2 are expressed in nearly all mDA neurons in adult mouse brains and are required for normal feeding and gait behavior, as well as for maintaining the expressions of mDA-specific markers during late embryonic/early postnatal and adult stages [75,79].
Consistent with haploinsufficiency of En1/2 or adult deletion of Nurr1, the haploinsufficiency of Foxa2 also results in the progressive loss of mDA neurons and decreased striatal DA, leading to the development of PD-related phenotypes in adult mice [16,80]. The co-expression and physical interaction of Foxa1/2 and Nurr1 have been identified in mDA neurons of adult mice, and the knockdown of Foxa1/2 could reduce the binding efficiency of Nurr1 to TH promoter regions [79,81]. Moreover, Foxa1/2 controls the expression of Lmx1a and Lmx1b (Lmx1a/b expression vanished in the Foxa1/2 knockout mutant) and is necessary for En1 and Nurr1 expression in developing mDA neurons [82]. It has been reported that Foxa1/2 could cooperate with Pitx3 and Nurr1 to enhance TH expression during late DA neuron development [64,82]. However, in Foxa1/2cKO mice, neither Pitx3 nor Nurr1 expression were impacted in the SNc and VTA mDA neurons, suggesting that Foxa1/2 may enhance the function of Pitx3 rather than its expression, and other redundant pathways may contribute to the expression of Pitx3 and Nurr1 during neuronal development [75].
The above findings suggest that among the neural transcription factors mentioned in this paper, Foxa1/2 is located upstream of them and could regulate their expression, and in addition to Pitx3, Foxa1/2 may enhance its function more.
Taken together, we summarized the expression time and distribution regions of important transcription factors (Pitx3, Lmx1a/b, Nurr1, Engrailed 1/2, and Foxa1/2) for mDA neuron development in Table 1. Additionally, the involvement of nuclear transcription factors (Pitx3, Nurr1, Eng 1/2, and Foxa 1/2) in PD pathogenesis is shown in Figure 1, and the genetic regulation of transcription factors mentioned above in the development of the mDA neurons is detailed in Figure 2.

4. The Broad Role of Pitx3 in PD: From Model to Patients

A variety of rodent models of PD have been used to study the pathogenesis of the disease, and they can currently be divided into two basic categories: neurotoxin-induced and genetic models [84,85]. The former models are usually treated with paraquat, rotenone, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), or 6-hydroxydopamine (6-OHDA) [86]; the latter models include those that overexpress α-synuclein or LRRK2 mutations, as well as DJ-1 knockout, PINK1 knockout, VMAT2 knockout, Nurr1 knockout, MitoPark knockout, Pitx3 knockout (aphakia) animal models, etc. [86,87]. These applied rodent models show highly suggestive human PD symptoms and are critical for understanding the motor/non-motor symptoms of PD [88].

4.1. Development of Pitx3-Deficient Mouse Models

Pitx3-deficiency was found in a natural mouse mutant (ophthalmic mice) that exhibited mDA neuron loss and motor abnormalities [89]. Unlike many genetic models, these mice selectively lost mDA neurons in the SNc, while mDA neurons in the ventral tegmental region and the olfactory bulb were largely retained, closely resembling the clinical aspects of PD [90], and the motor impairments of the mutants could be alleviated after levodopa injection [89]. Most intriguingly, these mice exhibited multiple non-motor deficits, such as an impaired spatial working memory, gastrointestinal dysfunction, and depressive behavior, which are analogous to the non-motor symptoms seen in PD patients. Meanwhile, inflammatory deficits were also identified in the Pitx3-deficient mice, including the morphologic alterations in astrocytes and microglia, and they potentially contributed to mDA neuron neurodegeneration as well [90]. Although the primary diagnostic criteria and most visible manifestations of PD are motor symptoms, non-motor symptoms are increasingly being acknowledged as an important component of PD dysfunctions and are frequently noticed before the onset of overt motor deficits [91]. A sleep disturbance, constipation, digestive dysfunction, and hyposmia have become important concerns for some PD patients [90]. Thus, Pitx3-deficient mice may provide some insight into the non-motor functional deficiencies of PD. Additionally, given that Pitx3 is an essential transcription factor for mDA development, Pitx3-deficient mice represent a useful tool that permits the study of early events in mDA formation. Moreover, studies have reported that Pitx3−/− mice produce decreased DA signaling rates and induce motor deficits [92] and can be also used to study the morphological and physiological modulation of striatal neurons [93] even after the administration of L-DOPA or other PD-related drugs [94].
Our laboratory recently generated a line of Pitx3fl/fl/DATCreERT2-conditional knockout mouse models that postnatally knocks out the Pitx3 gene in the cells expressing the dopamine transporter (DAT) protein after a tamoxifen (TAM) treatment. The mouse model showed canonical neuropathological features of PD, including age-dependent progressive motor deficits, a sharp decline in striatal DA content, and a significant loss of mDA neurons in the SNc, but not in the VTA [42], indicating the importance of the Pitx3 gene in adult neuronal survival. Moreover, Pitx3 deficiency also contributes to the pathology of the striatum. Our studies have shown that the contents of multiple neurotransmitters decreased in the Pitx3-deficient mice at early stages, including DA, GABA, and glutamate. Meanwhile, the morphologies of medium spiny neurons (MSNs) were altered due to Pitx3 deficiency, including nuclear, soma, and dendritic atrophy, as well as an increased number of nuclear invaginations. Additionally, more nuclear DNA damages were observed in MSNs during aging, while Pitx3-deficiency aggravated this phenomenon, together with alterations of the DNA methylation profiling associated with lipoprotein and nucleus pathways at late stages [42,93], suggesting that early perturbations of neurotransmitters within MSNs may potentially contribute to the alterations of metabolism, morphology, and epigenetics within the striatum at late stages.

4.2. The Role of Pitx3 in Cell Therapy

Cell transplantation, a potential strategy used to replace impaired mDA neurons, has gained particular interest since the 21st century. Stem cells have unique qualities, with the capacity to self-renew and differentiate into various cell types [95]. An increasing body of evidence supports that a series of transcription factors essential for mDA neuron development, differentiation, and specification also contribute to cell reprogramming: the generation of functional mDA neurons from stem cells or other non-neuronal cells [96].
Human teratocarcinoma cell line Ntera2 (NT2), which is derived from a human testicular cancer, is a potential source of cell therapy for neurological diseases [97]. NT2-derived neuron-like cells can give rise to the growth of axons and dendrites [98]. Thus, NT2 cells have long been thought of as a replacement source of mDA neurons for PD cell treatment [99]. Recent studies showed that after overexpressing Pitx3 and being exposed to GDNF, NT2 cells could release DA, indicating that Pitx3-GDNF interactions in DA signaling may promote the dopaminergic neuronal properties of NT2 cells, making it clinically applicable for cell replacement therapy in PD [99].
Mesenchymal stem cells (MSCs) derived from adult human bone marrow have varying capacities for brain development depending on the tissue of origin [100] and have shown lots of potential for immediate clinical applications due to their low immunogenicity [101]. However, a limited degree of mDA differentiation was seen in MSCs with a poor graft survival after transplantation [102]. Especially, adult bone marrow MSCs obtained from an older donor exhibited a significantly reduced neuroectodermal differentiation capability due to aging [103]. Pitx3 has been known to be a selective marker for transplantable embryonic stem cells [104,105]. The overexpression of Pitx3 in MSCs promotes the induction of mDA phenotype [25], suggesting that the administration of Pitx3 in MSCs during transplantation may be an effective cell therapy in PD. However, more research may be required to find the most suitable source of MSCs and to standardize the production process for ensuring the uniform neural differentiation of MSCs.
Research has shown that only 5–10% of neural stem cells survive after transplantation due to the toxic effects of the inflammatory state. Meanwhile, the majority of neural stem cells transplanted in vivo differentiate into glial cells rather than neurons [106,107]. Thus, it is crucial to find a way to prevent neuroinflammation in mDA neurons and promote their survival and development. Applying developmental transcription factors to therapeutics has become one of the most popular and appealing strategies [16]. Exogenous Pitx3 in ESCs-derived progenitor cells promotes the development of DA neurons in vitro by controlling the expression of genes for TH, Ngn2, and tubulin III [108], and an increased Pitx3 expression level was substantially linked with high TH expression [109] and benefits the formation of functional mDA neurons.

4.3. Pitx3 and PD

Polymorphisms of the Pitx3 gene are associated with the sporadic and early onset of PD [25,26,29]. One genetic association study from a screening sample of 340 PD patients and 680 controls and a large replication sample of 669 PD patients and 669 controls found the C allele of the Pitx3 promoter SNP rs3758549: C>T (p = 0.004) in PD patients appeared to be recessive with an estimated population frequency of 83%, suggesting an allele-dependent dysregulation of Pitx3 expression might contribute to the susceptibility to PD [110]. In contrast to abovementioned study, another study indicated that based on their clinical results, SNP is not confirmed to be associated with PD [111]. Additionally, an allele of rs4919621 SNP (chr10:103988621) in the intron of the Pitx3 gene and the C allele of rs2281983 SNP appeared significantly more often in PD patients with an early age of onset than they did in PD patients who experienced late onset (p = 0.001) and the controls (p = 0.002) [111]. Moreover, a significantly lower expression level of Pitx3 was recognized in the peripheral blood lymphocytes of PD patients compared to that of the controls. Importantly, this reduction was remarkably associated with an increased risk of developing PD in male and elderly subjects [28]. The data indicate that variations in transcription factors, which are crucial for the growth and maintenance of mDA neurons, merits our attention as they may be directly associated with PD vulnerability. To increase the precision and reproducibility of experimental outcomes, the onset of PD (early or late onset) and whether there is a PD family history in PD patients should also be taken into account.
At present, the primary therapeutic strategy used to alleviate or control motor symptoms in PD patients is to generally increase the bioavailability of DA by supplementing DA precursors or inhibiting DA breakdown [112]. However, as the disease progresses and the continuous loss of SNc mDA neurons occurs, the treatment efficiency decreases, and some of the common side effects of a long-term levodopa treatment occur (motor fluctuations, “on/off” phenomena, and dyskinesias). These levodopa-related complications and disabilities have become a treatment challenge for patients with advanced PD [14]. It has been reported that the delivery of pro-survival/anti-apoptotic factors to PD patients either locally (e.g., into the brain/VM or directly into mDA neurons) or systemically (e.g., into the bloodstream) is not sufficient to create efficient PD prevention and neuroprotective therapies. The possible reasons may be the timing and manner of administration, as well as potential concentration–dependent interactions, i.e., side effects in the body or brain, must be taken into account [113]. In our Pitx3cKO mouse model, a profound loss of mDA neurons in SNc was identified; however, the proximal neurons within VTA are less vulnerable to degeneration and are largely spared [42]. This means that different mDA neuron subtypes own their distinct vulnerability to the PD pathology. Therefore, our model provides a useful tool for elucidating how neuronal specification is maintained in subtypes of adult mDA neurons, which contributes to the development of timing and manner of drug administration in PD.
Furthermore, although Pitx3 is not expressed in astrocytes, the transfection of Pitx3 in astrocytes increased the expression levels of BDNF and GDNF, resulting in a protective effect on mDA neurons. Specifically, Pitx3-transfected astrocytes greatly mitigate rotenone-induced damage to mDA neurons, whereas this protection can be significantly reversed by preincubation with antibodies against BDNF or GDNF [114]. These results suggested that Pitx3 may protect mDA neurons by being overexpressed in non-mDA neurons. Thus, the co-transplantation of non-neuronal cells overexpressing Pitx3 with MSCs may increase graft survival and integration, but this still requires further study in the future. Studies have shown that genetic factors affect patients more during early-onset PD than they do during late-onset PD. Likely, Pitx3 is also critical for early-onset PD [115]. Therefore, the stage of PD onset is a key factor that needs to be considered when performing PD treatments associated with Pitx3. Moreover, further unraveling the complexity of PD pathogenesis and expanding the knowledge of the synergistic effects between transcription factors in mDA neuron development will contribute to promoting the development of alternative therapeutic strategies for PD.

5. Conclusions and Perspective

A better understanding of neural networks of mDA-related transcription factors could help to enhance the prevention or improve the treatments of PD. Pitx3 is a crucial transcription factor for the growth of mDA neurons and promotes the differentiation and survival of SNc mDA neurons. It has been reported that Pitx3 associates with multiple transcription factors (Lmx1 a/b, Nurr1, En1/2, and Foxa 1/2) and cooperate with them to participate in the differentiation and expansion of mDA neurons. Therefore, Pitx3 may be an entry point to regulate pathways essential for the prevention and treatment of PD. We believe that increasing the knowledge of Pitx3 will enhance the mechanistic understating, drug development, and clinical evaluation of PD.

Author Contributions

W.L. and H.C. designed and conceived the manuscript. X.W., X.C. and G.L. wrote the manuscript, created pictures and tables, and edited it. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by funding from the National Natural Science Foundation of China (Key Program) (32220103006), the Key Research and Development Program of Sichuan (2021YFS0382), the National Natural Science Foundation of China (General Program) (82271524), and the intramural program of National Institute on Aging, USA (ZIA AG000944, AG000928).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declared no conflicts of interest.

Abbreviations

mDA: Midbrain dopaminergic; PD: Parkinson’s disease; Pitx3: paired-like homeodomain transcription factor 3; RRF: retrorubral field; SNc: substantia nigra pars compacta; VTA: ventral tegmental area; DA: dopamine; AADC: aldehyde dehydrogenase 2; Dat: dopamine transporter; Vmat2: vesicular monoamine transporter; ADH2: aldehyde dehydrogenase 2; Th: tyrosine hydroxylase; En1/2: Engrailed-1/Engrailed-2; Ahd2/Aldh1a1: aldehyde dehydrogenase family 1; Foxa1/2: Foxa1 and Foxa2; MSCs: mesenchymal stem cells; BM: bone marrow; L-DOPA: L-3,4-dihydroxyphenylalanine; BBB: blood–brain barrier; MHB: midbrain–hindbrain boundary; MSCs: mesenchymal stem cells.

References

  1. GBD 2016 Neurology Collaborators. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef] [PubMed]
  2. Balestrino, R.; Schapira, A.H.V. Parkinson disease. Eur. J. Neurol. 2020, 27, 27–42. [Google Scholar] [CrossRef] [PubMed]
  3. Blesa, J.; Foffani, G.; Dehay, B.; Bezard, E.; Obeso, J.A. Motor and non-motor circuit disturbances in early Parkinson disease: Which happens first? Nat. Rev. Neurosci. 2022, 23, 115–128. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, M.E.; Stecher, B.; Labrie, V.; Brundin, L.; Brundin, P. Triggers, Facilitators, and Aggravators: Redefining Parkinson’s Disease Pathogenesis. Trends Neurosci. 2019, 42, 4–13. [Google Scholar] [CrossRef]
  5. Panicker, N.; Ge, P.; Dawson, V.L.; Dawson, T.M. The cell biology of Parkinson’s disease. J. Cell Biol. 2021, 220, e202012095. [Google Scholar] [CrossRef]
  6. Stoker, T.B.; Greenland, J.C. (Eds.) Parkinson’s Disease: Pathogenesis and Clinical Aspects; Codon Publications: Brisbane, Australian, 2018. [Google Scholar]
  7. Smidt, M.P.; Burbach, J.P. How to make a mesodiencephalic dopaminergic neuron. Nat. Rev. Neurosci. 2007, 8, 21–32. [Google Scholar] [CrossRef]
  8. Burbach, J.P.; Smidt, M.P. Molecular programming of stem cells into mesodiencephalic dopaminergic neurons. Trends Neurosci. 2006, 29, 601–603. [Google Scholar] [CrossRef]
  9. Bayer, H.M.; Glimcher, P.W. Midbrain dopamine neurons encode a quantitative reward prediction error signal. Neuron 2005, 47, 129–141. [Google Scholar] [CrossRef]
  10. Smits, S.M.; Ponnio, T.; Conneely, O.M.; Burbach, J.P.; Smidt, M.P. Involvement of Nurr1 in specifying the neurotransmitter identity of ventral midbrain dopaminergic neurons. Eur. J. Neurosci. 2003, 18, 1731–1738. [Google Scholar] [CrossRef]
  11. Arber, S.; Costa, R.M. Networking brainstem and basal ganglia circuits for movement. Nat. Rev. Neurosci. 2022, 23, 342–360. [Google Scholar] [CrossRef]
  12. Cox, J.; Witten, I.B. Striatal circuits for reward learning and decision-making. Nat. Rev. Neurosci. 2019, 20, 482–494. [Google Scholar] [CrossRef]
  13. Kouwenhoven, W.M.; von Oerthel, L.; Smidt, M.P. Pitx3 and En1 determine the size and molecular programming of the dopaminergic neuronal pool. PLoS ONE 2017, 12, e0182421. [Google Scholar] [CrossRef]
  14. Gerlach, M.; Reichmann, H.; Riederer, P. Levodopa in the treatment of Parkinson’s disease: Current controversies. Mov. Disord. 2005, 20, 643. [Google Scholar] [CrossRef]
  15. Nemade, D.; Subramanian, T.; Shivkumar, V. An Update on Medical and Surgical Treatments of Parkinson’s Disease. Aging Dis. 2021, 12, 1021–1035. [Google Scholar] [CrossRef]
  16. Tian, L.; Al-Nusaif, M.; Chen, X.; Li, S.; Le, W. Roles of Transcription Factors in the Development and Reprogramming of the Dopaminergic Neurons. Int. J. Mol. Sci. 2022, 23, 845. [Google Scholar] [CrossRef]
  17. Carmichael, K.; Sullivan, B.; Lopez, E.; Sun, L.; Cai, H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson’s disease. Ageing Neurodegener Dis. 2021, 1. [Google Scholar] [CrossRef]
  18. Canals, I.; Quist, E.; Ahlenius, H. Transcription Factor-Based Strategies to Generate Neural Cell Types from Human Pluripotent Stem Cells. Cell Reprogram. 2021, 23, 206–220. [Google Scholar] [CrossRef]
  19. Mesman, S.; Smidt, M.P. Acquisition of the Midbrain Dopaminergic Neuronal Identity. Int. J. Mol. Sci. 2020, 21, 4638. [Google Scholar] [CrossRef]
  20. Azimi, S.M.; Sheridan, S.D.; Ghannad-Rezaie, M.; Eimon, P.M.; Yanik, M.F. Combinatorial programming of human neuronal progenitors using magnetically-guided stoichiometric mRNA delivery. Elife 2018, 7, e31922. [Google Scholar] [CrossRef]
  21. Di Val Cervo, P.R.; Romanov, R.A.; Spigolon, G.; Masini, D.; Martin-Montanez, E.; Toledo, E.M.; La Manno, G.; Feyder, M.; Pifl, C.; Ng, Y.-H.; et al. Induction of functional dopamine neurons from human astrocytes in vitro and mouse astrocytes in a Parkinson’s disease model. Nat. Biotechnol. 2017, 35, 444–452. [Google Scholar] [CrossRef]
  22. Ng, Y.H.; Chanda, S.; Janas, J.A.; Yang, N.; Kokubu, Y.; Südhof, T.C.; Wernig, M. Efficient generation of dopaminergic induced neuronal cells with midbrain characteristics. Stem Cell Reports 2021, 16, 1763–1776. [Google Scholar] [CrossRef] [PubMed]
  23. Smidt, M.P.; van Schaick, H.S.A.; Lanctôt, C.; Tremblay, J.J.; Cox, J.J.; van der Kleij, A.A.M.; Wolterink, G.; Drouin, J.; Burbach, J.P.H. A homeodomain gene Ptx3 has highly restricted brain expression in mesencephalic dopaminergic neurons. Proc. Natl. Acad. Sci. USA 1997, 94, 13305–13310. [Google Scholar] [CrossRef] [PubMed]
  24. Smidt, M.P.; Smits, S.M.; Burbach, J.P. Homeobox gene Pitx3 and its role in the development of dopamine neurons of the substantia nigra. Cell Tissue Res. 2004, 318, 35–43. [Google Scholar] [CrossRef] [PubMed]
  25. Li, J.; Dani, J.A.; Le, W. The role of transcription factor Pitx3 in dopamine neuron development and Parkinson’s disease. Curr. Top. Med. Chem. 2009, 9, 855–859. [Google Scholar] [PubMed]
  26. Guo, Y.; Le, W.-D.; Jankovic, J.; Yang, H.-R.; Xu, H.-B.; Xie, W.-J.; Song, Z.; Deng, H. Systematic genetic analysis of the PITX3 gene in patients with Parkinson disease. Mov. Disord. 2011, 26, 1729–1732. [Google Scholar] [CrossRef]
  27. Kim, H.; Quan, X.; Seong, Y.; Kim, J. Impaired motor coordination in Pitx3 overexpression mice. Biochem. Biophys. Res. Commun. 2014, 446, 1211–1218. [Google Scholar] [CrossRef]
  28. Liu, H.; Wei, L.; Tao, Q.; Deng, H.; Ming, M.; Xu, P.; Le, W. Decreased NURR1 and PITX3 gene expression in Chinese patients with Parkinson’s disease. Eur. J. Neurol. 2012, 19, 870–875. [Google Scholar] [CrossRef]
  29. Le, W.; Nguyen, D.; Lin, X.W.; Rawal, P.; Huang, M.; Ding, Y.; Xie, W.; Deng, H.; Jankovic, J. Transcription factor PITX3 gene in Parkinson’s disease. Neurobiol. Aging 2011, 32, 750–753. [Google Scholar] [CrossRef]
  30. Anand, D.; Agrawal, S.A.; Slavotinek, A.; Lachke, S.A. Mutation update of transcription factor genes FOXE3, HSF4, MAF, and PITX3 causing cataracts and other developmental ocular defects. Hum. Mutat. 2018, 39, 471–494. [Google Scholar] [CrossRef]
  31. Semina, E.V.; Reiter, R.S.; Murray, J.C. Isolation of a new homeobox gene belonging to the Pitx/Rieg family: Expression during lens development and mapping to the aphakia region on mouse chromosome 19. Hum. Mol. Genet. 1997, 6, 2109–2116. [Google Scholar] [CrossRef]
  32. Messmer, K.; Remington, M.P.; Skidmore, F.; Fishman, P.S. Induction of tyrosine hydroxylase expression by the transcription factor Pitx3. Int. J. Dev. Neurosci. 2007, 25, 29–37. [Google Scholar] [CrossRef]
  33. Chen, H.; Song, Z.; Yuan, L.; Xiong, W.; Yang, Z.; Gong, L.; Deng, H. Genetic analysis of PITX3 variants in patients with essential tremor. Acta Neurol. Scand. 2017, 135, 373–376. [Google Scholar] [CrossRef]
  34. Lamonerie, T.; Tremblay, J.J.; Lanctôt, C.; Therrien, M.; Gauthier, Y.; Drouin, J. Ptx1, a bicoid-related homeo box transcription factor involved in transcription of the pro-opiomelanocortin gene. Genes Dev. 1996, 10, 1284–1295. [Google Scholar] [CrossRef]
  35. Gage, P.J.; Camper, S.A. Pituitary homeobox 2, a novel member of the bicoid-related family of homeobox genes, is a potential regulator of anterior structure formation. Hum. Mol. Genet. 1997, 6, 457–464. [Google Scholar] [CrossRef]
  36. Semina, E.V.; Ferrell, R.E.; Mintz-Hittner, H.A.; Bitoun, P.; Alward, W.L.M.; Reiter, R.S.; Funkhauser, C.; Daack-Hirsch, S.; Murray, J.C. A novel homeobox gene PITX3 is mutated in families with autosomal-dominant cataracts and ASMD. Nat. Genet. 1998, 19, 167–170. [Google Scholar] [CrossRef]
  37. Brouwer, A.; Berge, D.T.; Wiegerinck, R.; Meijlink, F. The OAR/aristaless domain of the homeodomain protein Cart1 has an attenuating role in vivo. Mech. Dev. 2003, 120, 241–252. [Google Scholar] [CrossRef]
  38. Bäckström, D.; Domellöf, M.E.; Granåsen, G.; Linder, J.; Mayans, S.; Elgh, E.; Mo, S.J.; Forsgren, L. PITX3 genotype and risk of dementia in Parkinson’s disease: A population-based study. J. Neurol. Sci. 2017, 381, 278–284. [Google Scholar] [CrossRef]
  39. Maxwell, S.L.; Ho, H.Y.; Kuehner, E.; Zhao, S.; Li, M. Pitx3 regulates tyrosine hydroxylase expression in the substantia nigra and identifies a subgroup of mesencephalic dopaminergic progenitor neurons during mouse development. Dev. Biol. 2005, 282, 467–479. [Google Scholar] [CrossRef]
  40. Smidt, M.P.; Smits, S.M.; Bouwmeester, H.; Hamers, F.P.T.; van der Linden, A.J.A.; Hellemons, A.J.C.G.M.; Graw, J.; Burbach, J.P.H. Early developmental failure of substantia nigra dopamine neurons in mice lacking the homeodomain gene Pitx3. Development 2004, 131, 1145–1155. [Google Scholar] [CrossRef]
  41. Nunes, I.; Tovmasian, L.T.; Silva, R.M.; Burke, R.E.; Goff, S.P. Pitx3 is required for development of substantia nigra dopaminergic neurons. Proc. Natl. Acad. Sci. USA 2003, 100, 4245–4250. [Google Scholar] [CrossRef]
  42. Wang, Y.; Chen, X.; Wang, Y.; Li, S.; Cai, H.; Le, W. The essential role of transcription factor Pitx3 in preventing mesodiencephalic dopaminergic neurodegeneration and maintaining neuronal subtype identities during aging. Cell Death Dis. 2021, 12, 1008. [Google Scholar] [CrossRef] [PubMed]
  43. Smits, S.M.; Burbach, J.P.; Smidt, M.P. Developmental origin and fate of meso-diencephalic dopamine neurons. Prog. Neurobiol. 2006, 78, 1–16. [Google Scholar] [CrossRef] [PubMed]
  44. Tran, T.Q.; Kioussi, C. Pitx genes in development and disease. Cell Mol. Life Sci. 2021, 78, 4921–4938. [Google Scholar] [CrossRef] [PubMed]
  45. Ang, S.L. Transcriptional control of midbrain dopaminergic neuron development. Development 2006, 133, 3499–3506. [Google Scholar] [CrossRef] [PubMed]
  46. Smidt, M.; Asbreuk, C.H.J.; Cox, J.J.; Chen, H.; Johnson, R.L.; Burbach, J.P.H. A second independent pathway for development of mesencephalic dopaminergic neurons requires Lmx1b. Nat. Neurosci. 2000, 3, 337–341. [Google Scholar] [CrossRef]
  47. Andersson, E.; Tryggvason, U.; Deng, Q.; Friling, S.; Alekseenko, Z.; Robert, B.; Perlmann, T.; Ericson, J. Identification of intrinsic determinants of midbrain dopamine neurons. Cell 2006, 124, 393–405. [Google Scholar] [CrossRef]
  48. Alavian, K.N.; Scholz, C.; Simon, H.H. Transcriptional regulation of mesencephalic dopaminergic neurons: The full circle of life and death. Mov. Disord. 2008, 23, 319–328. [Google Scholar] [CrossRef]
  49. Doucet-Beaupré, H.; Gilbert, C.; Profes, M.S.; Chabrat, A.; Pacelli, C.; Giguère, N.; Rioux, V.; Charest, J.; Deng, Q.; Laguna, A.; et al. Lmx1a and Lmx1b regulate mitochondrial functions and survival of adult midbrain dopaminergic neurons. Proc. Natl. Acad. Sci. USA 2016, 113, E4387–E4396. [Google Scholar] [CrossRef]
  50. Chung, S.; Leung, A.; Han, B.S.; Chang, M.Y.; Moon, J.I.; Kim, C.H.; Hong, S.; Pruszak, J.; Isacson, O.; Kim, K.S. Wnt1-lmx1a forms a novel autoregulatory loop and controls midbrain dopaminergic differentiation synergistically with the SHH-FoxA2 pathway. Cell Stem Cell 2009, 5, 646–658. [Google Scholar] [CrossRef]
  51. Villaescusa, J.C.; Li, B.; Toledo, E.M.; Rivetti di Val Cervo, P.; Yang, S.; Stott, S.R.; Kaiser, K.; Islam, S.; Gyllborg, D.; Laguna-Goya, R.; et al. A PBX1 transcriptional network controls dopaminergic neuron development and is impaired in Parkinson’s disease. EMBO J. 2016, 35, 1963–1978. [Google Scholar] [CrossRef]
  52. Nouri, P.; Götz, S.; Rauser, B.; Irmler, M.; Peng, C.; Trümbach, D.; Kempny, C.; Lechermeier, C.G.; Bryniok, A.; Dlugos, A.; et al. Dose-Dependent and Subset-Specific Regulation of Midbrain Dopaminergic Neuron Differentiation by LEF1-Mediated WNT1/b-Catenin Signaling. Front. Cell Dev. Biol. 2020, 8, 587778. [Google Scholar] [CrossRef]
  53. Andersson, E.R.; Saltó, C.; Villaescusa, J.C.; Cajanek, L.; Yang, S.; Bryjova, L.; Nagy, I.I.; Vainio, S.J.; Ramirez, C.; Bryja, V.; et al. Wnt5a cooperates with canonical Wnts to generate midbrain dopaminergic neurons in vivo and in stem cells. Proc. Natl. Acad. Sci. USA 2013, 110, E602–E610. [Google Scholar] [CrossRef]
  54. Prakash, N.; Brodski, C.; Naserke, T.; Puelles, E.; Gogoi, R.; Hall, A.; Panhuysen, M.; Echevarria, D.; Sussel, L.; Weisenhorn, D.M.V.; et al. A Wnt1-regulated genetic network controls the identity and fate of midbrain-dopaminergic progenitors in vivo. Development 2006, 133, 89–98. [Google Scholar] [CrossRef]
  55. Zetterstrom, R.H.; Solomin, L.; Jansson, L.; Hoffer, B.J.; Olson, L.; Perlmann, T. Dopamine neuron agenesis in Nurr1-deficient mice. Science 1997, 276, 248–250. [Google Scholar] [CrossRef]
  56. Saucedo-Cardenas, O.; Quintana-Hau, J.D.; Le, W.-D.; Smidt, M.P.; Cox, J.J.; De Mayo, F.; Burbach, J.P.H.; Conneely, O.M. Nurr1 is essential for the induction of the dopaminergic phenotype and the survival of ventral mesencephalic late dopaminergic precursor neurons. Proc. Natl. Acad. Sci. USA 1998, 95, 4013–4018. [Google Scholar] [CrossRef]
  57. Jiang, C.; Wan, X.; He, Y.; Pan, T.; Jankovic, J.; Le, W. Age-dependent dopaminergic dysfunction in Nurr1 knockout mice. Exp. Neurol. 2005, 191, 154–162. [Google Scholar] [CrossRef]
  58. Pulcrano, S.; De Gregorio, R.; De Sanctis, C.; Lahti, L.; Perrone-Capano, C.; Ponti, D.; di Porzio, U.; Perlmann, T.; Caiazzo, M.; Volpicelli, F.; et al. Lmx1a-Dependent Activation of miR-204/211 Controls the Timing of Nurr1-Mediated Dopaminergic Differentiation. Int. J. Mol. Sci. 2022, 23, 6961. [Google Scholar] [CrossRef]
  59. Argyrofthalmidou, M.; Spathis, A.D.; Maniati, M.; Poula, A.; Katsianou, M.A.; Sotiriou, E.; Manousaki, M.; Perier, C.; Papapanagiotou, I.; Papadopoulou-Daifoti, Z.; et al. Nurr1 repression mediates cardinal features of Parkinson’s disease in alpha-synuclein transgenic mice. Hum. Mol. Genet. 2021, 30, 1469–1483. [Google Scholar] [CrossRef]
  60. García-Yagüe, J.; Lastres-Becker, I.; Stefanis, L.; Vassilatis, D.K.; Cuadrado, A. alpha-Synuclein Induces the GSK-3-Mediated Phosphorylation and Degradation of NURR1 and Loss of Dopaminergic Hallmarks. Mol. Neurobiol. 2021, 58, 6697–6711. [Google Scholar] [CrossRef]
  61. Katunar, M.R.; Saez, T.; Brusco, A.; Antonelli, M.C. Immunocytochemical expression of dopamine-related transcription factors Pitx3 and Nurr1 in prenatally stressed adult rats. J. Neurosci. Res. 2009, 87, 1014–1022. [Google Scholar] [CrossRef]
  62. Le, W.; Pan, T.; Huang, M.; Xu, P.; Xie, W.; Zhu, W.; Zhang, X.; Deng, H.; Jankovic, J. Decreased NURR1 gene expression in patients with Parkinson’s disease. J. Neurol. Sci. 2008, 273, 29–33. [Google Scholar] [CrossRef] [PubMed]
  63. Martinat, C.; Bacci, J.-J.; Leete, T.; Kim, J.; Vanti, W.B.; Newman, A.H.; Cha, J.H.; Gether, U.; Wang, H.; Abeliovich, A. Cooperative transcription activation by Nurr1 and Pitx3 induces embryonic stem cell maturation to the midbrain dopamine neuron phenotype. Proc. Natl. Acad. Sci. USA 2006, 103, 2874–2879. [Google Scholar] [CrossRef] [PubMed]
  64. Jacobs, F.M.J.; van Erp, S.; van der Linden, A.J.A.; von Oerthel, L.; Burbach, J.P.H.; Smidt, M.P. Pitx3 potentiates Nurr1 in dopamine neuron terminal differentiation through release of SMRT-mediated repression. Development 2009, 136, 531–540. [Google Scholar] [CrossRef] [PubMed]
  65. Rekaik, H.; de Thé, F.-X.B.; Prochiantz, A.; Fuchs, J.; Joshi, R.L. Dissecting the role of Engrailed in adult dopaminergic neurons--Insights into Parkinson disease pathogenesis. FEBS Lett. 2015, 589 Pt A, 3786–3794. [Google Scholar] [CrossRef]
  66. Prochiantz, A.; Di Nardo, A.A. Homeoprotein signaling in the developing and adult nervous system. Neuron 2015, 85, 911–925. [Google Scholar] [CrossRef]
  67. Simon, H.H.; Saueressig, H.; Wurst, W.; Goulding, M.D.; O'Leary, D.D.M. Fate of midbrain dopaminergic neurons controlled by the engrailed genes. J. Neurosci. 2001, 21, 3126–3134. [Google Scholar] [CrossRef]
  68. Nordströma, U.; Beauvais, G.; Ghosh, A.; Pulikkaparambil Sasidharan, B.C.; Lundblad, M.; Fuchs, J.; Joshi, R.L.; Lipton, J.W.; Roholt, A.; Medicetty, S.; et al. Progressive nigrostriatal terminal dysfunction and degeneration in the engrailed1 heterozygous mouse model of Parkinson’s disease. Neurobiol. Dis. 2015, 73, 70–82. [Google Scholar] [CrossRef]
  69. Blaudin de The, F.X.; Rekaik, H.; Prochiantz, A.; Fuchs, J.; Joshi, R.L. Neuroprotective Transcription Factors in Animal Models of Parkinson Disease. Neural. Plast. 2016, 2016, 6097107. [Google Scholar] [CrossRef]
  70. Danielian, P.S.; McMahon, A.P. Engrailed-1 as a target of the Wnt-1 signalling pathway in vertebrate midbrain development. Nature 1996, 383, 332–334. [Google Scholar] [CrossRef]
  71. Veenvliet, J.V.; dos Santos, M.T.M.A.; Kouwenhoven, W.M.; von Oerthel, L.; Lim, J.L.; van der Linden, A.J.A.; Koerkamp, M.J.A.G.; Holstege, F.C.P.; Smidt, M.P. Specification of dopaminergic subsets involves interplay of En1 and Pitx3. Development 2013, 140, 3373–3384. [Google Scholar] [CrossRef]
  72. Haubenberger, D.; Reinthaler, E.; Mueller, J.C.; Pirker, W.; Katzenschlager, R.; Froehlich, R.; Bruecke, T.; Daniel, G.; Auff, E.; Zimprich, A. Association of transcription factor polymorphisms PITX3 and EN1 with Parkinson’s disease. Neurobiol. Aging 2011, 32, 302–307. [Google Scholar] [CrossRef]
  73. Jackson, B.C.; Carpenter, C.; Nebert, D.W.; Vasiliou, V. Update of human and mouse forkhead box (FOX) gene families. Hum. Genomics. 2010, 4, 345–352. [Google Scholar] [CrossRef]
  74. Ang, S.L. Foxa1 and Foxa2 transcription factors regulate differentiation of midbrain dopaminergic neurons. Adv. Exp. Med. Biol. 2009, 651, 58–65. [Google Scholar]
  75. Pristerà, A.; Lin, W.; Kaufmann, A.-K.; Brimblecombe, K.R.; Threlfell, S.; Dodson, P.D.; Magill, P.J.; Fernandes, C.; Cragg, S.J.; Ang, S.-L. Transcription factors FOXA1 and FOXA2 maintain dopaminergic neuronal properties and control feeding behavior in adult mice. Proc. Natl. Acad. Sci. USA 2015, 112, E4929–E4938. [Google Scholar] [CrossRef]
  76. Sasaki, H.; Hogan, B.L. Differential expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo. Development 1993, 118, 47–59. [Google Scholar] [CrossRef]
  77. Burtscher, I.; Lickert, H. Foxa2 regulates polarity and epithelialization in the endoderm germ layer of the mouse embryo. Development 2009, 136, 1029–1038. [Google Scholar] [CrossRef]
  78. Shih, D.Q.; Navas, M.A.; Kuwajima, S.; Duncan, S.A.; Stoffel, M. Impaired glucose homeostasis and neonatal mortality in hepatocyte nuclear factor 3alpha-deficient mice. Proc. Natl. Acad. Sci. USA 1999, 96, 10152–10157. [Google Scholar] [CrossRef]
  79. Stott, S.R.W.; Metzakopian, E.; Lin, W.; Kaestner, K.H.; Hen, R.; Ang, S.-L. Foxa1 and foxa2 are required for the maintenance of dopaminergic properties in ventral midbrain neurons at late embryonic stages. J. Neurosci. 2013, 33, 8022–8034. [Google Scholar] [CrossRef]
  80. Kittappa, R.; Chang, W.W.; Awatramani, R.B.; McKay, R.D.G. The foxa2 gene controls the birth and spontaneous degeneration of dopamine neurons in old age. PLoS. Biol. 2007, 5, e325. [Google Scholar] [CrossRef]
  81. Oh, S.-M.; Chang, M.-Y.; Song, J.-J.; Rhee, Y.-H.; Joe, E.-H.; Lee, H.-S.; Yi, S.-H.; Lee, S.-H. Combined Nurr1 and Foxa2 roles in the therapy of Parkinson’s disease. EMBO Mol. Med. 2015, 7, 510–525. [Google Scholar] [CrossRef]
  82. Lin, W.; Metzakopian, E.; Mavromatakis, Y.E.; Gao, N.; Balaskas, N.; Sasaki, H.; Briscoe, J.; Whitsett, J.A.; Goulding, M.; Kaestner, K.H.; et al. Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development. Dev. Biol. 2009, 333, 386–396. [Google Scholar] [CrossRef] [PubMed]
  83. Wallen, A.; Perlmann, T. Transcriptional control of dopamine neuron development. Ann. N. Y. Acad. Sci. 2003, 991, 48–60. [Google Scholar] [CrossRef] [PubMed]
  84. Blandini, F.; Armentero, M.T. Animal models of Parkinson’s disease. FEBS J. 2012, 279, 1156–1166. [Google Scholar] [CrossRef] [PubMed]
  85. Vingill, S.; Connor-Robson, N.; Wade-Martins, R. Are rodent models of Parkinson’s disease behaving as they should? Behav. Brain. Res. 2018, 352, 133–141. [Google Scholar] [CrossRef]
  86. Dawson, T.M.; Ko, H.S.; Dawson, V.L. Genetic animal models of Parkinson’s disease. Neuron 2010, 66, 646–661. [Google Scholar] [CrossRef]
  87. Chesselet, M.F.; Richter, F. Modelling of Parkinson’s disease in mice. Lancet Neurol. 2011, 10, 1108–1118. [Google Scholar] [CrossRef]
  88. Langley, M.R.; Ghaisas, S.; Palanisamy, B.N.; Ay, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Characterization of nonmotor behavioral impairments and their neurochemical mechanisms in the MitoPark mouse model of progressive neurodegeneration in Parkinson’s disease. Exp. Neurol. 2021, 341, 113716. [Google Scholar] [CrossRef]
  89. van den Munckhof, P.; Gilbert, F.; Chamberland, M.; Levesque, D.; Drouin, J. Striatal neuroadaptation and rescue of locomotor deficit by L-dopa in aphakia mice, a model of Parkinson’s disease. J. Neurochem. 2006, 96, 160–170. [Google Scholar] [CrossRef]
  90. Song, B.; Feldmann, J.W.; Cao, S.; Feitosa, M.; Kong, Y.; Kim, W.; Schweitzer, A.; Leblanc, P.; Schweitzer, J.S.; Kim, K.-S. A Pitx3-deficient developmental mouse model for fine motor, olfactory, and gastrointestinal symptoms of Parkinson’s disease. Neurobiol. Dis. 2022, 170, 105777. [Google Scholar] [CrossRef]
  91. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  92. Le, W.; Zhang, L.; Xie, W.; Li, S.; Dani, J.A. Pitx3 deficiency produces decreased dopamine signaling and induces motor deficits in Pitx3(-/-) mice. Neurobiol. Aging 2015, 36, 3314–3320. [Google Scholar] [CrossRef]
  93. Chen, X.; Yang, Z.; Shao, Y.; Kim, K.; Wang, Y.; Wang, Y.; Wu, H.; Xu, X.; Le, W. Pitx3 deficiency promotes age-dependent alterations in striatal medium spiny neurons. Front. Aging Neurosci. 2022, 14, 960479. [Google Scholar] [CrossRef]
  94. Suarez, L.M.; Alberquilla, S.; García-Montes, J.R.; Moratalla, R. Differential Synaptic Remodeling by Dopamine in Direct and Indirect Striatal Projection Neurons in Pitx3(-/-) Mice, a Genetic Model of Parkinson’s Disease. J. Neurosci. 2018, 38, 3619–3630. [Google Scholar] [CrossRef]
  95. Mitalipov, S.; Wolf, D. Totipotency, pluripotency and nuclear reprogramming. Adv. Biochem. Eng. Biotechnol. 2009, 114, 185–199. [Google Scholar]
  96. Wang, M.; Ling, K.-H.; Tan, J.J.; Lu, C.-B. Development and Differentiation of Midbrain Dopaminergic Neuron: From Bench to Bedside. Cells 2020, 9, 1489. [Google Scholar] [CrossRef]
  97. Paquet-Durand, F.; Bicker, G. Human model neurons in studies of brain cell damage and neural repair. Curr. Mol. Med. 2007, 7, 541–554. [Google Scholar] [CrossRef]
  98. Pleasure, S.J.; Lee, V.M. NTera 2 cells: A human cell line which displays characteristics expected of a human committed neuronal progenitor cell. J. Neurosci. Res. 1993, 35, 585–602. [Google Scholar] [CrossRef]
  99. Eskandarian Boroujeni, M.; Aliaghaei, A.; Maghsoudi, N.; Gardaneh, M. Complementation of dopaminergic signaling by Pitx3-GDNF synergy induces dopamine secretion by multipotent Ntera2 cells. J. Cell Biochem. 2020, 121, 200–212. [Google Scholar] [CrossRef]
  100. Wegmeyer, H.; Bröske, A.-M.; Leddin, M.; Kuentzer, K.; Nisslbeck, A.K.; Hupfeld, J.; Wiechmann, K.; Kuhlen, J.; Von Schwerin, C.; Stein, C.; et al. Mesenchymal stromal cell characteristics vary depending on their origin. Stem Cells Dev. 2013, 22, 2606–2618. [Google Scholar] [CrossRef]
  101. Trzaska, K.A.; Kuzhikandathil, E.V.; Rameshwar, P. Specification of a dopaminergic phenotype from adult human mesenchymal stem cells. Stem Cells 2007, 25, 2797–2808. [Google Scholar] [CrossRef]
  102. Suon, S.; Yang, M.; Iacovitti, L. Adult human bone marrow stromal spheres express neuronal traits in vitro and in a rat model of Parkinson’s disease. Brain Res. 2006, 1106, 46–51. [Google Scholar] [CrossRef] [PubMed]
  103. Hermann, A.; List, C.; Habisch, H.-J.; Vukicevic, V.; Ehrhart-Bornstein, M.; Brenner, R.; Bernstein, P.; Fickert, S.; Storch, A. Age-dependent neuroectodermal differentiation capacity of human mesenchymal stromal cells: Limitations for autologous cell replacement strategies. Cytotherapy 2010, 12, 17–30. [Google Scholar] [CrossRef] [PubMed]
  104. Chung, S.; Hedlund, E.; Hwang, M.; Kim, D.; Shin, B.-S.; Hwang, D.-Y.; Kang, U.J.; Isacson, O.; Kim, K.-S. The homeodomain transcription factor Pitx3 facilitates differentiation of mouse embryonic stem cells into AHD2-expressing dopaminergic neurons. Mol. Cell Neurosci. 2005, 28, 241–252. [Google Scholar] [CrossRef] [PubMed]
  105. Hedlund, E.; Pruszak, J.; Lardaro, T.; Ludwig, W.; Viñuela, A.; Kim, K.-S.; Isacson, O. Embryonic stem cell-derived Pitx3-enhanced green fluorescent protein midbrain dopamine neurons survive enrichment by fluorescence-activated cell sorting and function in an animal model of Parkinson’s disease. Stem Cells 2008, 26, 1526–3156. [Google Scholar] [CrossRef]
  106. Brundin, P.; Karlsson, J.; Emgård, M.; Schierle, G.S.K.; Hansson, O.; Petersén, A.; Castilho, R.F. Improving the survival of grafted dopaminergic neurons: A review over current approaches. Cell Transplant. 2000, 9, 179–195. [Google Scholar] [CrossRef]
  107. Haas, S.J.-P.; Beckmann, S.; Petrov, S.; Andressen, C.; Wree, A.; Schmitt, O. Transplantation of immortalized mesencephalic progenitors (CSM14.1 cells) into the neonatal parkinsonian rat caudate putamen. J. Neurosci. Res. 2007, 85, 778–786. [Google Scholar] [CrossRef]
  108. Hong, S.; Chung, S.; Leung, K.; Hwang, I.; Moon, J.; Kim, K.-S. Functional roles of Nurr1, Pitx3, and Lmx1a in neurogenesis and phenotype specification of dopamine neurons during in vitro differentiation of embryonic stem cells. Stem Cells Dev. 2014, 23, 477–487. [Google Scholar] [CrossRef]
  109. Tiklová, K.; Björklund, Å.K.; Lahti, L.; Fiorenzano, A.; Nolbrant, S.; Gillberg, L.; Volakakis, N.; Yokota, C.; Hilscher, M.M.; Hauling, T.; et al. Single-cell RNA sequencing reveals midbrain dopamine neuron diversity emerging during mouse brain development. Nat. Commun. 2019, 10, 581. [Google Scholar] [CrossRef]
  110. Fuchs, J.; Mueller, J.C.; Lichtner, P.; Schulte, C.; Munz, M.; Berg, D.; Wüllner, U.; Illig, T.; Sharma, M.; Gasser, T. The transcription factor PITX3 is associated with sporadic Parkinson’s disease. Neurobiol. Aging 2009, 30, 731–738. [Google Scholar] [CrossRef]
  111. Bergman, O.; Håkansson, A.; Westberg, L.; Nordenström, K.; Belin, A.C.; Sydow, O.; Olson, L.; Holmberg, B.; Eriksson, E.; Nissbrandt, H. PITX3 polymorphism is associated with early onset Parkinson’s disease. Neurobiol. Aging 2010, 31, 114–117. [Google Scholar] [CrossRef]
  112. Postuma, R.B.; Berg, D.; Stern, M.; Poewe, W.; Olanow, C.W.; Oertel, W.; Obeso, J.; Marek, K.; Litvan, I.; Lang, A.E.; et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov. Disord. 2015, 30, 1591–1601. [Google Scholar] [CrossRef]
  113. Prakash, N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front. Mol. Neurosci. 2022, 15, 1071731. [Google Scholar] [CrossRef]
  114. Yang, D.; Peng, C.; Li, X.; Fan, X.; Li, L.; Ming, M.; Chen, S.; Le, W. Pitx3-transfected astrocytes secrete brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor and protect dopamine neurons in mesencephalon cultures. J. Neurosci. Res. 2008, 86, 3393–3400. [Google Scholar] [CrossRef]
  115. Håkansson, A.; Westberg, L.; Nilsson, S.; Buervenich, S.; Carmine, A.; Holmberg, B.; Sydow, O.; Olson, L.; Johnels, B.; Eriksson, E.; et al. Interaction of polymorphisms in the genes encoding interleukin-6 and estrogen receptor beta on the susceptibility to Parkinson’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2005, 133, 88–92. [Google Scholar] [CrossRef]
Figure 1. Projections of mDA neurons and involvement of nuclear transcription factors (Pitx3, Nurr1, Eng 1/2, and Foxa 1/2) in PD pathogenesis. A sagittal section of the adult brain shows the location of the cell bodies (ovals) of the RRF (pink), SNc (green), and VTA (blue) mDA clusters in the brain stem and their projection (indicated by arrows) into the limbic region (midbrain pathway), striatum (substantia nigra pathway), and PFC (midcortical pathway). Under physiological conditions, these neurotranscription factors (Pitx3, Nurr1, Eng 1/2, and Foxa 1/2) function normally. However, the dysfunction of these neurotranscription factors is accompanied by a series of abnormal indicators in the brain of PD patients, including abnormal transcription and regulation, an impaired mTOR pathway/autophagic pathway, the CREB survival pathway, and DNA repair under various stimuli (neurotoxin, aging, oxidative stress, etc., as indicated by the red lightning bolt icon), which ultimately leads to the death of mDA neurons.
Figure 1. Projections of mDA neurons and involvement of nuclear transcription factors (Pitx3, Nurr1, Eng 1/2, and Foxa 1/2) in PD pathogenesis. A sagittal section of the adult brain shows the location of the cell bodies (ovals) of the RRF (pink), SNc (green), and VTA (blue) mDA clusters in the brain stem and their projection (indicated by arrows) into the limbic region (midbrain pathway), striatum (substantia nigra pathway), and PFC (midcortical pathway). Under physiological conditions, these neurotranscription factors (Pitx3, Nurr1, Eng 1/2, and Foxa 1/2) function normally. However, the dysfunction of these neurotranscription factors is accompanied by a series of abnormal indicators in the brain of PD patients, including abnormal transcription and regulation, an impaired mTOR pathway/autophagic pathway, the CREB survival pathway, and DNA repair under various stimuli (neurotoxin, aging, oxidative stress, etc., as indicated by the red lightning bolt icon), which ultimately leads to the death of mDA neurons.
Ijms 24 08614 g001
Figure 2. Genetic regulation of transcription factors in the development of the mDA neurons. Wnt1 interacts with Lmx1a/b, forming a Wnt-Lmx1a/b loop. The specific signaling pathways include WNT/β-catenin/LEF1-Lmx1a/b-Pitx3. Lmx1a/b is also regulated by Foxa1/2, which could control the expression of En1/2. En1/2 regulates Pitx3 expression and Pitx3 suppress En1/2. Pitx3 plays a critical role in SMRT-Sin3-HDAC-mediated repression of mDA-related genes by recruitment of the Nurr1-PSF transcriptional complex and releasing SMRT-HDAC complexes to activate Nurr1 transcription.
Figure 2. Genetic regulation of transcription factors in the development of the mDA neurons. Wnt1 interacts with Lmx1a/b, forming a Wnt-Lmx1a/b loop. The specific signaling pathways include WNT/β-catenin/LEF1-Lmx1a/b-Pitx3. Lmx1a/b is also regulated by Foxa1/2, which could control the expression of En1/2. En1/2 regulates Pitx3 expression and Pitx3 suppress En1/2. Pitx3 plays a critical role in SMRT-Sin3-HDAC-mediated repression of mDA-related genes by recruitment of the Nurr1-PSF transcriptional complex and releasing SMRT-HDAC complexes to activate Nurr1 transcription.
Ijms 24 08614 g002
Table 1. The expression time and distribution region of important transcription factors for mDA neuron development.
Table 1. The expression time and distribution region of important transcription factors for mDA neuron development.
Transcription FactorThe Expression Time
Embryonic Day (E)
The Cells It Acts onThe Distribution Region Reference
Pitx3E11.5Mature mDA neuronsSubstantia nigra and ventral tegmental region[23,61]
Lmx1aE9Mitotic ventral mesencephalic precursor cellsVentral midbrain and dorsally[46,47]
Lmx1bE7.5Neural progenitor cells, Mitotic ventral mesencephalic precursor cellsDerive from the midbrain, then restricted to the ventral midbrain, the floor plate, mid-hindbrain boundary, and roof plate[46,47]
Nurr1E10.5Immature/ mature mDA neuronsCerebral cortex, hippocampus, thalamus, amygdala, and midbrain[61,83]
En1/2E8Ventral mDA precursorMidbrain and hindbrain[65,66,67]
Foxa 1E7.5mDA progenitors, immature/ mature neuronsFloorplate, notochord, and endoderm[76,77,79]
Foxa 2E6.5mDA progenitors, immature/ mature neuronsPosterior epiblast, anterior definitive endoderm, axial mesoderm[76,77,79]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, X.; Chen, X.; Liu, G.; Cai, H.; Le, W. The Crucial Roles of Pitx3 in Midbrain Dopaminergic Neuron Development and Parkinson’s Disease-Associated Neurodegeneration. Int. J. Mol. Sci. 2023, 24, 8614. https://doi.org/10.3390/ijms24108614

AMA Style

Wang X, Chen X, Liu G, Cai H, Le W. The Crucial Roles of Pitx3 in Midbrain Dopaminergic Neuron Development and Parkinson’s Disease-Associated Neurodegeneration. International Journal of Molecular Sciences. 2023; 24(10):8614. https://doi.org/10.3390/ijms24108614

Chicago/Turabian Style

Wang, Xin, Xi Chen, Guangdong Liu, Huaibin Cai, and Weidong Le. 2023. "The Crucial Roles of Pitx3 in Midbrain Dopaminergic Neuron Development and Parkinson’s Disease-Associated Neurodegeneration" International Journal of Molecular Sciences 24, no. 10: 8614. https://doi.org/10.3390/ijms24108614

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop