Skip to main content

REVIEW article

Front. Cell. Neurosci., 04 March 2022
Sec. Cellular Neurophysiology
Volume 16 - 2022 | https://doi.org/10.3389/fncel.2022.826193

Evidence That Substantia Nigra Pars Compacta Dopaminergic Neurons Are Selectively Vulnerable to Oxidative Stress Because They Are Highly Metabolically Active

  • Department of Human Genetics, McGill University, Montreal, QC, Canada

There are 400–500 thousand dopaminergic cells within each side of the human substantia nigra pars compacta (SNpc) making them a minuscule portion of total brain mass. These tiny clusters of cells have an outsized impact on motor output and behavior as seen in disorders such as Parkinson’s disease (PD). SNpc dopaminergic neurons are more vulnerable to oxidative stress compared to other brain cell types, but the reasons for this are not precisely known. Here we provide evidence to support the hypothesis that this selective vulnerability is because SNpc neurons sustain high metabolic rates compared to other neurons. A higher baseline requirement for ATP production may lead to a selective vulnerability to impairments in oxidative phosphorylation (OXPHOS) or genetic insults that impair Complex I of the electron transport chain. We suggest that the energy demands of the unique morphological and electrophysiological properties of SNpc neurons may be one reason these cells produce more ATP than other cells. We further provide evidence to support the hypothesis that transcription factors (TFs) required to drive induction, differentiation, and maintenance of midbrain dopaminergic neural progenitor cells which give rise to terminally differentiated SNpc neurons are uniquely involved in both developmental patterning and metabolism, a dual function unlike other TFs that program neurons in other brain regions. The use of these TFs during induction and differentiation may program ventral midbrain progenitor cells metabolically to higher ATP levels, allowing for the development of those specialized cell processes seen in terminally differentiated cells. This paper provides a cellular and developmental framework for understanding the selective vulnerability of SNpc dopaminergic cells to oxidative stress.

Introduction

Dopaminergic cells of the ventral midbrain that project to the striatum play an essential role in governing motor behavior in mammals. The small cluster of cells in the substantia nigra pars compacta (SNpc) release the neurotransmitter dopamine to neurons of the striatum which project to the basal ganglia. The basal ganglia is connected to the thalamus and motor cortex which imparts control over motor output. The loss of the majority of SNpc cells is the underlying cause of Parkinson’s disease (PD), a progressive movement disorder characterized by resting tremor, bradykinesia, rigidity and non-motor symptoms such as cognitive decline and sleep disturbance (Hayes, 2019). The causes of PD highlight the critical importance of mitochondria and cellular respiration because toxins that inhibit mitochondrial function or mutations in genes that support mitochondrial function associate with cell loss in animal models and humans (Park et al., 2018).

Inhibitors of the electron transport chain, the group of enzymes in the inner mitochondrial matrix that replenishes adenosine triphosphate (ATP) using oxygen, lead to selective destruction of SNpc dopaminergic cells (Langston et al., 1983; Sayre et al., 1991; Betarbet et al., 2000; Morais et al., 2014), suggesting they are particularly sensitive to inhibition of oxidative phosphorylation (OXPHOS) or reactive oxygen species (ROS) created in this process (Jenner, 1993), collectively termed oxidative stress (Haddad and Nakamura, 2015). This idea is supported by the discovery that many genetic mutations associated with PD appear to be associated in some way with mitochondrial dynamics or oxidative stress (e.g., PARK7, PARKIN, and PINK1) (Shen and Cookson, 2004). Furthermore, genetic insults involving other energy pathways also appear to be basal ganglia-related in that they can affect motor function; examples include deficiency of glucose transporter SLC2A1 (Leen et al., 2010) (OMIM 606777) or GCH1 (Trender-Gerhard et al., 2009) (OMIM 128230). The links between oxidative stress and basal ganglia-related movement disorders lead to the fundamental question of what drives the underlying cause of disease. There are many potential explanations for this, but here we will evaluate the evidence that SNpc dopaminergic cells have higher ATP requirements than other brain cell types making them more susceptible to genetic insults or toxins which impair OXPHOS. The motivation for this piece came from our recent serendipitous discovery showing that the dopaminergic midbrain progenitors were operating at full OXPHOS capacity compared to isogenic glutamatergic forebrain progenitor cells (Bell et al., 2021). While this was not the primary concern of that paper, the effect size in healthy, unbranched, and non-dopamine producing cells was striking. Might SNpc cells then be programmed to high energy states? We wanted to pursue this idea more in depth, hence the current article. Here, we will argue that selective vulnerability is because SNpc neurons operate at a higher baseline levels of ATP synthesis/oxygen consumption both because they are intrinsically programmed that way, and must sustain highly energy consuming processes due to particular cell properties. While this idea is not mutually exclusive from other ideas such as the selective vulnerability to ROS in SNpc, a formal review of this idea is warranted.

We first define the concept of selective vulnerability in SNpc DA cells, then review evidence in two parts. In the first part we focus on morphological and electrophysiological properties of terminally differentiated SNpc neurons. We review findings showing that SNpc neurons are more vulnerable to OXPHOS insults than other dopaminergic cell types, then discuss the morphological and electrophysiological properties of SNpc dopaminergic neurons that contribute to increasing ATP demands. In the second part, we focus on transcription factors (TFs) required for the development of midbrain SNpc progenitor cells. We review the role of these TFs in both SNpc development and metabolism, and suggest that the dual feature of these TFs is particular to SNpc cells. SNpc cells may be programmed to a higher energetic state before the differentiation of morphological and electrophysiological cell properties emerges. This might suggest that developmental programming of midbrain progenitors allows for the terminal differentiation of highly ATP consuming processes, rather than the other way around.

Dopaminergic Cells of the Substantia Nigra Pars Compacta and the Concept of Selective Vulnerability

Dopamine is part of the catecholamine family of monoamines, defined by the presence of an amine and a catechol group, derived from phenylalanine or dietary tyrosine. It is synthesized in several areas in mammals including chromaffin cells of the adrenal medulla and in at least 11 clusters in brain from the olfactory bulb to the midbrain (Felten and Sladek, 1983). One such cluster resides in the ventral tegmental area (VTA) of the midbrain (A10 area in mouse), where dopaminergic neurons project to the nucleus accumbens. Another such cluster lies in the SNpc (A9 in mouse), which is adjacent to the VTA and comprises neurons that project to the striatum. The million or so cells that make up the bilateral substantia nigra have an outsize impact on functional output. Destruction of tens of millions of cortical cells after a stroke for example, can lead to hemi-paralysis which in many cases can be partially recoverable (Murphy and Corbett, 2009), while loss of a majority of the dopamine producing cells of the midbrain SNpc can lead to significant motor impairment. While dopamine replacement therapy can alleviate some PD symptoms in most people, cell destruction and degeneration are not slowed.

Selective vulnerability refers to the idea that a given insult affects a given cell type more negatively than another cell type. For example, the expanded CAG repeat in the HUNTINGTIN gene causes Huntington’s disease which primarily affects medium spiny neurons of the striatum (Ehrlich, 2012). Similarly, there are germline mutations that lead to very specific cancer types, despite expression of a mutated gene in all or most areas of the body; one example is that of RUNX1 germline mutations predisposing to blood cancer (Avagyan and Brown, 2021). The different molecules that define a cell state are likely also the same factors that can lead to selective vulnerability. Since tissue types are different by definition, it becomes easier to understand how some cell types can be vulnerable to specific insults (whether genetic or environmental) than others. In the case of SNpc cells, they are much more vulnerable to inhibitors of OXPHOS (reviewed below). The depletion of ATP might force cells into cell death programs if they cannot sustain their basic energy needs (Kushnareva and Newmeyer, 2010).

Part A: Morphological and Electrophysiological Properties of Terminally Differentiated SNpc Dopaminergic Cells

Substantia Nigra Pars Compacta Dopaminergic Cells Are More Vulnerable to Electron Transport Inhibitors Than Other Dopaminergic Cell Types

Mitochondria provide about 93% of the ATP in the brain (Sokoloff, 1960) and most of the ATP required for neuronal activity (Hall et al., 2012; Harris et al., 2012) making OXPHOS critical for global brain function. In rodent studies testing exposure to MPP+ (5–10 μM) or rotenone (25–50 nM), both of which inhibit mitochondrial complex 1, loss of dopaminergic neurons in the SNpc is significantly higher than in dopaminergic neurons from the VTA, the midbrain area adjacent to the SNpc (Pacelli et al., 2015; Jaumotte et al., 2016), a result supported by in vitro human findings (Oosterveen et al., 2021). The VTA is a favored comparator region (Chung et al., 2005) because these cells are dopamine-producing, they are adjacent to SNpc (so presumably follow at least similar developmental trajectories), but are often preserved in PD (Hirsch et al., 1988; Maingay et al., 2006). Under low doses of 6-OHDA, a toxin that also impairs mitochondrial complex 1 and a commonly used approach to model PD in animals, SNpc dopaminergic neurons show a loss of 40% while VTA dopaminergic neurons do not show significant loss (Giguere et al., 2019; Tanguay et al., 2021). SNpc neurons seem to be more susceptible to impairment of OXPHOS compared to dopaminergic cells in the VTA (Subramaniam and Chesselet, 2013). A complete comparison of VTA and SNpc cells in the respect to selective vulnerability has been described (Brichta and Greengard, 2014).

Substantia Nigra Pars Compacta Dopaminergic Cells Have More Energy Consuming Synaptic Contacts Compared to Other Dopaminergic Cell Types

A single SNpc neuron has a very large number of projections into the striatum (Anden et al., 1966), and it has been possible to trace these large structures in a dissected brain over long distances and branch points. Matsuda et al. (2009) showed that one single SNpc neuron can form connections with, on average, 2.7 ± 1.5% of striatal neurons (Matsuda et al., 2009; Bolam and Pissadaki, 2012). Comparing subtypes of dopaminergic cells in rat shows that SNpc TH-positive neurons are 69 and 326% larger than that of VTA and OB TH-positive neurons (Pacelli et al., 2015) and that a single SNpc neuron forms from 100,00 to 250,000 connections in the rat striatum (Bolam and Pissadaki, 2012) whereas a single VTA DA neuron makes 12,000 to 30,000 connections. Based on these numbers and the known differences between rat and human brains, it is estimated that one single human SNpc neuron could have 1 million to 2.5 million striatal synaptic sites (Bolam and Pissadaki, 2012). This work is supported by diffusion tensor imaging studies from human brain suggesting significantly more connections from SN than VTA (Kwon and Jang, 2014). SNpc DA neurons are thus not only highly branched, but they are also highly active given the number of synaptic sites.

Why might extensive branching and connections cause increased energy burden, as defined by ATP utilization? The number of ATP molecules required to restore membrane potential after the propagation of an action potential exponentially increases as a function of the size and complexity of arborization (Pissadaki and Bolam, 2013). The high energy cost associated with sustaining such activity is known to translate into higher mitochondrial biogenesis, density, and respiration in cultured primary neurons (Pacelli et al., 2015). Compared to VTA neurons, SNpc neurons show significantly higher levels of PGC-1α, a regulator of mitochondrial biogenesis, and higher mitoDSRed signal, a proxy for mitochondrial network expression. Furthermore, basal oxygen consumption rates (OCR) of SNpc neurons are threefold higher than that of VTA or OB neurons (Pacelli et al., 2015). SNpc neurons do not significantly differ from the two other DA cell types in terms of maximal OCR, so the results suggest that SNpc DA neurons function at maximal capacity under baseline conditions whereas VTA and OB dopaminergic cell types have the potential to achieve this high energy state only if required by metabolic pressures.

Electrophysiological Activity of Substantia Nigra Pars Compacta Neurons Require More ATP Than Other Dopaminergic Cell Types

Substantia nigra pars compacta DA neurons have unique electrophysiological properties. They are autonomous pacemakers (Clark and Chiodo, 1988; Grace and Onn, 1989; Hyland et al., 2002), transiently firing broad and slow action potentials in addition to burst firing, in a process that is dependent on Ca2 + influx (Puopolo et al., 2007). SNpc neurons not only have to ensure that their electrochemical environment allows firing thresholds to be reached, but must also do so with high frequency, to maintain tonic rhythm. Ca2 + entry via Ca2 + ATPase channels is essential in sustaining such electrochemical gradients, but is relatively expensive in energy as the channels consume one molecule of ATP to pump one molecule of Ca2 + (Surmeier et al., 2012). In comparison, one molecule of ATP can be used to pump three Na+ ions and two K+ ions via Na+/K+ -ATPase channels, which are required to maintain electrochemical gradients for action potentials in, for example, cortical pyramidal neurons. Further, SNpc neurons’ activity involves L-type Cav1.3 Ca2 + channels (Chan et al., 2007; Guzman et al., 2009), a type of channel that is rarely opened or lowly expressed in other brain regions and that opens at steep subthreshold membrane potentials (Wilson and Callaway, 2000; Puopolo et al., 2007). This causes large amounts of Ca2 + to flow in the SNpc neurons, which increases the cells’ metabolic load (Guzman et al., 2010; Goldberg et al., 2012). Support for this idea can be seen by blocking L-type Cav1.3 Ca2 + channels in rodent SNpc brain slices which leads to decreased oxygen consumption and lower levels of ATP (Guzman et al., 2010).

The influx/efflux of calcium is costly, but required in SNpc cells. In VTA DA neurons, Ca2 + can be sequestered by Ca2 + -buffering proteins which are not ATP-dependent and thus “energetically cheap” to maintain (Surmeier et al., 2012), so SNpc cells have a particular need for readily moveable Ca2 + across the membrane and cannot benefit as much from cheap Ca2 + buffering used in VTA neurons. SNpc neurons do buffer calcium but this is low relative to their Ca2 + influx during pacemaking activity (Foehring et al., 2009), so SNpc cells may rely on a more costly mechanism to regulate Ca2 + levels. An example of such mechanism is Ca2 + sequestration by the ER network, which also requires ATP-dependent transporters and mitochondrial Ca2 + uniporter [reviewed in (Surmeier et al., 2012)].

Part B: Developmental Programming of Ventral Midbrain DA Cells

Substantia Nigra Pars Compacta Cells Are Induced via a Series of Transcription Factors With Known Roles in Metabolism

Several TFs are required to induce, differentiate, or maintain SNpc neurons throughout development, and many of these are not specific to SNpc but rather are specific to dorsal-ventral or anterior-posterior patterning. What defines an SNpc neuron is not any one TF but rather the combination of TFs at a given time and 3D position on these axes. In this way, an organism uses the same TFs, in for example, ventral patterning, such as LMX1A to contribute to specification of both motor neurons and SNpc neurons. Some of the TFs that contribute to SNpc fate specification are very well known for their roles in metabolism, a function presumably unrelated to their developmental patterning roles, although it is possible that their metabolic function is in fact patterning these cells as well. Here, we describe several TFs important in SNpc neurons at different developmental stages and described both their role in SNpc fate specification and metabolism. We note that studies investigating the metabolic roles of genes described here are mostly conducted in liver, kidney, pancreas, and muscle. Studies showing the direct relationship between midbrain patterning TFs and energy expenditure/storage in the brain have not yet, to our knowledge, been investigated.

Recently, we found strong evidence that undifferentiated but committed cells that can give rise to SNpc cells have much higher baseline OXPHOS and ATP levels than forebrain cells that were developmentally comparable and isogenic (Bell et al., 2021). This suggested to us that SNpc cells may be programmed to a high energy state prior to the differentiation of morphological or electrophysiological properties characteristic of SNpc neurons described in the first section of this paper. If this were true, it may imply two things: (1) That the TFs that induce cells to become committed progenitors of SNpc fate are programming the cells very early on to high ATP consumption and high OXPHOS levels, and (2) The high ATP-requiring processes of SNpc cells may have been able to develop only because of this programming step; that is, they are a consequence of the developmental programming, rather than high ATP consumption being a consequence of particular cell morphology and electrophysiological characteristics.

LMX1A/B, NR4A2, EN1/2, OTX1/2, CORIN, PITX3, and FOXA1/2 are probably the best described TFs that have a role in SNpc fate specification (Asgrimsdottir and Arenas, 2020), and some of these have a role in metabolism, which are the TFs we focus on here. Our purpose in this section is to highlight that ventral midbrain programming uses TFs that have both a metabolic role and a cell patterning role. While we cannot rule out that other TFs that drive cell fate for other neuronal cell types also have roles in metabolism that have yet to be discovered (e.g., PAX6 or FOXG1 in forebrain), the well-known role of the ventral midbrain neuronal factors in metabolism is possibly unique to TFs that drive SNpc neuron development.

FOXA1 and FOXA2 are part of the FoxA subfamily of the forkhead/winged-helix family. They are key regulators of neural tube patterning (Monaghan et al., 1993; Sasaki and Hogan, 1993) and are expressed both in ventral midbrain progenitors early in development (E8.5 onward in mouse) (Ang et al., 1993) and in terminally differentiated neurons (Ferri et al., 2007), so may have both an inductive and maintenance function. Homozygous deletion of Foxa2 in mice results in the absence of a notochord, and thus the absence of a floor plate and abnormality in the dorsal-ventral patterning of the developing neural tube (Ang and Rossant, 1994), but brain specific knock-outs have better refined their role in the ventral midbrain. Specifically, Foxa1/2 regulate the extent of neurogenesis and thus the final number of SNpc neurons, as well as the activation of other important TFs in midbrain and described here (Ferri et al., 2007). In later stages of development, brain specific knock-out of both Foxa1/2 results in less tyrosine hydroxylase and less burst firing specifically in SNpc neurons (Pristera et al., 2015).

With respect to metabolic function, Foxa1 and Foxa2 are required for regulating glucose homeostasis (Kaestner et al., 1999; Lantz et al., 2004; Friedman and Kaestner, 2006). Foxa1-null mice, despite being fed, show glucose levels similar to that of starved animals, which results in death of the animals due to hypoglycemia (Kaestner et al., 1999). This is due to inefficient glucagon signaling likely due to poor Gcg gene expression in response to hypoglycemic conditions, a result supported by in vitro studies (Philippe et al., 1994). Under fasting conditions, mice hepatocytes deficient in Foxa2 show significantly lower cAMP and glucocorticoid signaling and blunted efficiency of a cAMP target protein (CREB) binding to its target sites (Zhang et al., 2005). In pancreatic beta cells, cell-specific ablation of Foxa2 results in the complete loss of K-ATP channels, which normally couple glucose metabolism with insulin secretion (Lantz et al., 2004). Specifically, Foxa2 mutant mice (Sund et al., 2001; Lantz et al., 2004) show significant reductions of both K-ATP channel subunits, Abcc8 and Kcnj11 (Reis and Velho, 2002; Vaxillaire et al., 2004), genes that are also markers of midbrain dopaminergic cells (Liss et al., 1999; Schiemann et al., 2012). FOXA1/2 may program both pancreatic beta cells and ventral midbrain NPCs and use the same molecular tools (e.g., K-ATP subunit genes) in different tissue types to achieve different ends (presumably insulin secretion and regulation of calcium transients, respectively). Finally, Foxa2 is regulated by insulin itself (Wolfrum et al., 2003) and several single nucleotide polymorphisms (SNPs) in the gene are significantly associated with different metabolic indicators, such as fasting blood glucose levels [e.g., rs6048205 (Manning et al., 2012; Wojcik et al., 2019; Sinnott-Armstrong et al., 2021), rs3833331 (Chen et al., 2021; Sakaue et al., 2021), rs1209523 (Xing et al., 2010; Wojcik et al., 2019), rs72470563 (Chung et al., 2021), rs1337918 (Chen et al., 2021)].

NR4A2 is a nuclear receptor that belongs to the nuclear receptor subfamily 4 group A (NR4A) and is also known as NURR1. Knockout of Nurr1 in mice results in complete loss of midbrain dopaminergic cells, implying that Nurr1 is required for induction of ventral midbrain cell fate (Zetterstrom et al., 1997) and the survival of SNpc neurons (Zetterstrom et al., 1997; Saucedo-Cardenas et al., 1998). In metabolism, Nr4a2 is a key regulator in glucose metabolism, lipid metabolism, and the TCA cycle [reviewed in Herring et al. (2019)]. In isolated mouse primary hepatocytes, gluconeogenesis stimulates the expression of Nurr1, and fasting mice show increased Nr4a2 expression in extracted livers. NR4A family members can bind the promoters of the gluconeogenesis genes and a glucose transporter gene Glut2 (Pei et al., 2006). Outside of the liver, Nr4a2 is upregulated in skeletal muscle following endurance exercise (Mahoney et al., 2005; Catoire et al., 2012) and has a critical role in glucose uptake (Amoasii et al., 2016, 2019).

LIMX1A/B are part of the LIM homeodomain family of genes and knockout in mice strongly links them the SNpc dopaminergic cell development (Yan et al., 2011). During brain development, at around embryonic day 9.5 in mice, Lmx1a is expressed in the roof plate of the neural tube and is responsible for neural tube specification (Millonig et al., 2000; Chizhikov and Millen, 2004). Lmx1a specifically mediates the differentiation of floor plate dopaminergic progenitors (Deng et al., 2011), whereas Lmx1b is involved in the establishment of the midbrain-hindbrain boundary and mediates the differentiation of lateral, non-floor plate, dopaminergic progenitors (Guo et al., 2007; Deng et al., 2011). Recent evidence shows that Lmx1b is required for the autophagic-lysosomal pathway and neuroprotection of post-mitotic dopaminergic neurons (Laguna et al., 2015; Doucet-Beaupre et al., 2016).

In pancreatic beta islet cells, Lmx1a regulates the expression of insulin via a synergistic interaction with the basic helix-loop-helix protein E47/Pan-1 at the promoter of the insulin gene (German et al., 1992, 1994). Lmx1b might also regulate genes that respond to a diet switch (from normal food diet to a restricted food diet) in drosophila (Whitaker et al., 2014). In humans, several polymorphisms in LMX1B have been associated with metabolic diseases. The polymorphism rs10733682 is associated with BMI (Locke et al., 2015) and the AA genotype of rs10733682 interacts with intake of total energy, fat, and carbohydrate in determining risk of developing elevated triglycerides levels, which is associated with higher obesity incidence (Zhu et al., 2020). Numerous other LMX1B variants have been associated with BMI or fat body mass in genome-wide association studies, such as rs867559 (Speliotes et al., 2010), rs7027304 (Tachmazidou et al., 2017; Sakaue et al., 2021), rs3829849 (Costa-Urrutia et al., 2020), rs2235056 (Akiyama et al., 2017), rs1336980 (Vogelezang et al., 2020), rs7030609 and rs2275241 (Richardson et al., 2020).

Conclusion

We provide several lines of evidence that SNpc cells have higher metabolic activity than other cell types including other dopaminergic cell types outside the SNpc. In part A of this work, we looked at the cellular properties of these cells. The evidence reviewed might suggest that SNpc cells require more ATP and thus more OXPHOS to sustain and maintain these particular functions than other cells and so are selectively vulnerable to genetic or metabolic insults, such as mutations in genes that support mitophagy or chemicals that interfere with Complex I of the electron transport chain. We do not necessarily consider this idea exclusive from other hypotheses of SNpc selective vulnerability, namely the creation of ROS from OXPHOS or the oxidation of dopamine itself.

In Part B, we proposed a new idea for the association between SNpc cells and a heightened energy state. We suggest that SNpc neurons may be programmed to this state prior to the development of potentially energy intensive cellular activities. Committed ventral midbrain progenitor cells are unbranched, have no synaptic contact, and do not fire action potentials, yet we previously found that OXPHOS capacity is much higher than comparator forebrain progenitor cells (Bell et al., 2021). This could be direct evidence that high levels of OXPHOS are already present in progenitor cells prior to the development of electrophysiological or cellular properties. We suggest that this state may come about due to the unique assortment of TFs required to induce SNpc development. Specifically, we highlight TFs that are also known actors in metabolic programming with essential roles in insulin and glucose maintenance. It may be that the metabolic role of these TFs is in fact the programming step, and it is not that these TFs have independent and dual roles in metabolism and SNpc development.

Author Contributions

Both authors wrote the manuscript, contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Akiyama, M., Okada, Y., Kanai, M., Takahashi, A., Momozawa, Y., Ikeda, M., et al. (2017). Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat. Genet. 49, 1458–1467. doi: 10.1038/ng.3951

PubMed Abstract | CrossRef Full Text | Google Scholar

Amoasii, L., Holland, W., Sanchez-Ortiz, E., Baskin, K. K., Pearson, M., Burgess, S. C., et al. (2016). A MED13-dependent skeletal muscle gene program controls systemic glucose homeostasis and hepatic metabolism. Genes Dev. 30, 434–446. doi: 10.1101/gad.273128.115

PubMed Abstract | CrossRef Full Text | Google Scholar

Amoasii, L., Sanchez-Ortiz, E., Fujikawa, T., Elmquist, J. K., Bassel-Duby, R., and Olson, E. N. (2019). NURR1 activation in skeletal muscle controls systemic energy homeostasis. Proc. Natl. Acad. Sci. U.S.A. 116, 11299–11308. doi: 10.1073/pnas.1902490116

PubMed Abstract | CrossRef Full Text | Google Scholar

Anden, N. E., Dahlstrom, A., Fuxe, K., Olson, L., and Ungerstedt, U. (1966). Ascending noradrenaline neurons from the pons and the medulla oblongata. Experientia 22, 44–45. doi: 10.1007/BF01897761

PubMed Abstract | CrossRef Full Text | Google Scholar

Ang, S. L., and Rossant, J. (1994). HNF-3 beta is essential for node and notochord formation in mouse development. Cell 78, 561–574. doi: 10.1016/0092-8674(94)90522-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Ang, S. L., Wierda, A., Wong, D., Stevens, K. A., Cascio, S., Rossant, J., et al. (1993). The formation and maintenance of the definitive endoderm lineage in the mouse: involvement of HNF3/forkhead proteins. Development 119, 1301–1315.

PubMed Abstract | Google Scholar

Asgrimsdottir, E. S., and Arenas, E. (2020). Midbrain dopaminergic neuron development at the single cell level: in vivo and in stem cells. Front. Cell Dev. Biol. 8:463. doi: 10.3389/fcell.2020.00463

PubMed Abstract | CrossRef Full Text | Google Scholar

Avagyan, S., and Brown, A. L. (2021). To t or not to b: germline runx1 mutation preferences in pediatric all predisposition. J. Clin. Invest. 131:e15246. doi: 10.1172/JCI152464

PubMed Abstract | CrossRef Full Text | Google Scholar

Bell, S., McCarty, V., Peng, H., Jefri, M., Hettige, N., Antonyan, L., et al. (2021). Lesch-Nyhan disease causes impaired energy metabolism and reduced developmental potential in midbrain dopaminergic cells. Stem Cell Rep. 16, 1749–1762. doi: 10.1016/j.stemcr.2021.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Betarbet, R., Sherer, T. B., MacKenzie, G., Garcia-Osuna, M., Panov, A. V., and Greenamyre, J. T. (2000). Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat. Neurosci. 3, 1301–1306.

Google Scholar

Bolam, J. P., and Pissadaki, E. K. (2012). Living on the edge with too many mouths to feed: why dopamine neurons die. Mov. Disord. 27, 1478–1483. doi: 10.1002/mds.25135

PubMed Abstract | CrossRef Full Text | Google Scholar

Brichta, L., and Greengard, P. (2014). Molecular determinants of selective dopaminergic vulnerability in Parkinson’s disease: an update. Front. Neuroanat. 8:152. doi: 10.3389/fnana.2014.00152

PubMed Abstract | CrossRef Full Text | Google Scholar

Catoire, M., Mensink, M., Boekschoten, M. V., Hangelbroek, R., Muller, M., Schrauwen, P., et al. (2012). Pronounced effects of acute endurance exercise on gene expression in resting and exercising human skeletal muscle. PLoS One 7:e51066. doi: 10.1371/journal.pone.0051066

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, C. S., Guzman, J. N., Ilijic, E., Mercer, J. N., Rick, C., Tkatch, T., et al. (2007). ‘Rejuvenation’ protects neurons in mouse models of Parkinson’s disease. Nature 447, 1081–1086. doi: 10.1038/nature05865

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Spracklen, C. N., Marenne, G., Varshney, A., Corbin, L. J., Luan, J., et al. (2021). The trans-ancestral genomic architecture of glycemic traits. Nat. Genet. 53, 840–860. doi: 10.1038/s41588-021-00852-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Chizhikov, V. V., and Millen, K. J. (2004). Control of roof plate formation by Lmx1a in the developing spinal cord. Development 131, 2693–2705. doi: 10.1242/dev.01139

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung, C. Y., Seo, H., Sonntag, K. C., Brooks, A., Lin, L., and Isacson, O. (2005). Cell type-specific gene expression of midbrain dopaminergic neurons reveals molecules involved in their vulnerability and protection. Hum. Mol. Genet. 14, 1709–1725. doi: 10.1093/hmg/ddi178

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung, R. H., Chiu, Y. F., Wang, W. C., Hwu, C. M., Hung, Y. J., Lee, I. T., et al. (2021). Hsiung, multi-omics analysis identifies CpGs near G6PC2 mediating the effects of genetic variants on fasting glucose. Diabetologia 64, 1613–1625. doi: 10.1007/s00125-021-05449-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Clark, D., and Chiodo, L. A. (1988). Electrophysiological and pharmacological characterization of identified nigrostriatal and mesoaccumbens dopamine neurons in the rat. Synapse 2, 474–485. doi: 10.1002/syn.890020503

PubMed Abstract | CrossRef Full Text | Google Scholar

Costa-Urrutia, P., Abud, C., Franco-Trecu, V., Colistro, V., Rodríguez-Arellano, M. E., Alvarez-Fariña, R., et al. (2020). Effect of 15 BMI-associated polymorphisms, reported for Europeans, across ethnicities and degrees of amerindian ancestry in Mexican children. Int. J. Mol. Sci. 21, 374. doi: 10.3390/ijms21020374

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, Q., Andersson, E., Hedlund, E., Alekseenko, Z., Coppola, E., Panman, L., et al. (2011). Specific and integrated roles of Lmx1a, Lmx1b and Phox2a in ventral midbrain development. Development 138, 3399–3408. doi: 10.1242/dev.065482

PubMed Abstract | CrossRef Full Text | Google Scholar

Doucet-Beaupre, H., Gilbert, C., Profes, M. S., Chabrat, A., Pacelli, C., Giguere, N., et al. (2016). Lmx1a and Lmx1b regulate mitochondrial functions and survival of adult midbrain dopaminergic neurons. Proc. Natl. Acad. Sci. U.S.A. 113, E4387–E4396. doi: 10.1073/pnas.1520387113

PubMed Abstract | CrossRef Full Text | Google Scholar

Ehrlich, M. E. (2012). Huntington’s disease and the striatal medium spiny neuron: cell-autonomous and non-cell-autonomous mechanisms of disease. Neurotherapeutics 9, 270–284. doi: 10.1007/s13311-012-0112-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Felten, D. L., and Sladek, J. R. (1983). Monoamine distribution in primate brain V. Monoaminergic nuclei: anatomy, pathways and local organization. Brain Res. Bull. 10, 171–284. doi: 10.1016/0361-9230(83)90045-x

CrossRef Full Text | Google Scholar

Ferri, A. L., Lin, W., Mavromatakis, Y. E., Wang, J. C., Sasaki, H., Whitsett, J. A., et al. (2007). Foxa1 and Foxa2 regulate multiple phases of midbrain dopaminergic neuron development in a dosage-dependent manner. Development 134, 2761–2769. doi: 10.1242/dev.000141

PubMed Abstract | CrossRef Full Text | Google Scholar

Foehring, R. C., Zhang, X. F., Lee, J. C., and Callaway, J. C. (2009). Endogenous calcium buffering capacity of substantia nigral dopamine neurons. J. Neurophysiol. 102, 2326–2333. doi: 10.1152/jn.00038.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Friedman, J. R., and Kaestner, K. H. (2006). The Foxa family of transcription factors in development and metabolism. Cell Mol. Life Sci. 63, 2317–2328. doi: 10.1007/s00018-006-6095-6

PubMed Abstract | CrossRef Full Text | Google Scholar

German, M. S., Wang, J., Chadwick, R. B., and Rutter, W. J. (1992). Synergistic activation of the insulin gene by a LIM-homeo domain protein and a basic helix-loop-helix protein: building a functional insulin minienhancer complex. Genes Dev. 6, 2165–2176. doi: 10.1101/gad.6.11.2165

PubMed Abstract | CrossRef Full Text | Google Scholar

German, M. S., Wang, J., Fernald, A. A., Espinosa, R. III, Le Beau, M. M., and Bell, G. I. (1994). Localization of the genes encoding two transcription factors, LMX1 and CDX3, regulating insulin gene expression to human chromosomes 1 and 13. Genomics 24, 403–404. doi: 10.1006/geno.1994.1639

PubMed Abstract | CrossRef Full Text | Google Scholar

Giguere, N., Delignat-Lavaud, B., Herborg, F., Voisin, A., Li, Y., Jacquemet, V., et al. (2019). Increased vulnerability of nigral dopamine neurons after expansion of their axonal arborization size through D2 dopamine receptor conditional knockout. PLoS Genet. 15:e1008352. doi: 10.1371/journal.pgen.1008352

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldberg, J. A., Guzman, J. N., Estep, C. M., Ilijic, E., Kondapalli, J., Sanchez-Padilla, J., et al. (2012). Calcium entry induces mitochondrial oxidant stress in vagal neurons at risk in Parkinson’s disease. Nat. Neurosci. 15, 1414–1421. doi: 10.1038/nn.3209

PubMed Abstract | CrossRef Full Text | Google Scholar

Grace, A. A., and Onn, S. P. (1989). Morphology and electrophysiological properties of immunocytochemically identified rat dopamine neurons recorded in vitro. J. Neurosci. 9, 3463–3481. doi: 10.1523/JNEUROSCI.09-10-03463.1989

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, C., Qiu, H. Y., Huang, Y., Chen, H., Yang, R. Q., Chen, S. D., et al. (2007). Lmx1b is essential for Fgf8 and Wnt1 expression in the isthmic organizer during tectum and cerebellum development in mice. Development 134, 317–325. doi: 10.1242/dev.02745

PubMed Abstract | CrossRef Full Text | Google Scholar

Guzman, J. N., Sanchez-Padilla, J., Wokosin, D., Kondapalli, J., Ilijic, E., Schumacker, P. T., et al. (2010). Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature 468, 696–700.

Google Scholar

Guzman, J. N., ánchez-Padilla, J. S., Chan, C. S., and Surmeier, D. J. (2009). Robust pacemaking in substantia nigra dopaminergic neurons. J. Neurosci. 29, 11011–11019. doi: 10.1523/JNEUROSCI.2519-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Haddad, D., and Nakamura, K. (2015). Understanding the susceptibility of dopamine neurons to mitochondrial stressors in Parkinson’s disease. FEBS Lett. 589, 3702–3713. doi: 10.1016/j.febslet.2015.10.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, C. N., Klein-Flügge, M. C., Howarth, C., and Attwell, D. (2012). Oxidative phosphorylation, not glycolysis, powers presynaptic and postsynaptic mechanisms underlying brain information processing. J. Neurosci. 32, 8940–8951. doi: 10.1523/JNEUROSCI.0026-12.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Harris, J. J., Jolivet, R., and Attwell, D. (2012). Synaptic energy use and supply. Neuron 75, 762–777. doi: 10.1016/j.neuron.2012.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Hayes, M. T. (2019). Parkinson’s disease and parkinsonism. Am. J. Med. 132, 802–807.

Google Scholar

Herring, J. A., Elison, W. S., and Tessem, J. S. (2019). Function of Nr4a orphan nuclear receptors in proliferation, apoptosis and fuel utilization across tissues. Cells 8:1373. doi: 10.3390/cells8111373

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirsch, E., Graybiel, A. M., and Agid, Y. A. (1988). Melanized dopaminergic neurons are differentially susceptible to degeneration in Parkinson’s disease. Nature 334, 345–348. doi: 10.1038/334345a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Hyland, B. I., Reynolds, J. N., Hay, J., Perk, C. G., and Miller, R. (2002). Firing modes of midbrain dopamine cells in the freely moving rat. Neuroscience 114, 475–492. doi: 10.1016/s0306-4522(02)00267-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Jaumotte, J. D., Wyrostek, S. L., and Zigmond, M. J. (2016). Protection of cultured dopamine neurons from MPP(+) requires a combination of neurotrophic factors. Eur. J. Neurosci. 44, 1691–1699. doi: 10.1111/ejn.13252

PubMed Abstract | CrossRef Full Text | Google Scholar

Jenner, P. (1993). Altered mitochondrial function, iron metabolism and glutathione levels in Parkinson’s disease. Acta Neurol. Scand. Suppl. 146, 6–13.

PubMed Abstract | Google Scholar

Kaestner, K. H., Katz, J., Liu, Y., Drucker, D. J., and Schütz, G. (1999). Inactivation of the winged helix transcription factor HNF3alpha affects glucose homeostasis and islet glucagon gene expression in vivo. Genes Dev. 13, 495–504. doi: 10.1101/gad.13.4.495

PubMed Abstract | CrossRef Full Text | Google Scholar

Kushnareva, Y., and Newmeyer, D. D. (2010). Bioenergetics and cell death. Ann. N.Y. Acad. Sci. 1201, 50–57.

Google Scholar

Kwon, H. G., and Jang, S. H. (2014). Differences in neural connectivity between the substantia nigra and ventral tegmental area in the human brain. Front. Hum. Neurosci. 8:41. doi: 10.3389/fnhum.2014.00041

PubMed Abstract | CrossRef Full Text | Google Scholar

Laguna, A., Schintu, N., Nobre, A., Alvarsson, A., Volakakis, N., Jacobsen, J. K., et al. (2015). Dopaminergic control of autophagic-lysosomal function implicates Lmx1b in Parkinson’s disease. Nat. Neurosci. 18, 826–835. doi: 10.1038/nn.4004

PubMed Abstract | CrossRef Full Text | Google Scholar

Langston, J. W., Ballard, P., Tetrud, J. W., and Irwin, I. (1983). Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science 219, 979–980. doi: 10.1126/science.6823561

PubMed Abstract | CrossRef Full Text | Google Scholar

Lantz, K. A., Vatamaniuk, M. Z., Brestelli, J. E., Friedman, J. R., Matschinsky, F. M., and Kaestner, K. H. (2004). Foxa2 regulates multiple pathways of insulin secretion. J. Clin. Invest. 114, 512–520. doi: 10.1172/JCI21149

PubMed Abstract | CrossRef Full Text | Google Scholar

Leen, W. G., Klepper, J., Verbeek, M. M., Leferink, M., Hofste, T., van Engelen, B. G., et al. (2010). Glucose transporter-1 deficiency syndrome: the expanding clinical and genetic spectrum of a treatable disorder. Brain 133, 655–670. doi: 10.1093/brain/awp336

PubMed Abstract | CrossRef Full Text | Google Scholar

Liss, B., Bruns, R., and Roeper, J. (1999). Alternative sulfonylurea receptor expression defines metabolic sensitivity of K-ATP channels in dopaminergic midbrain neurons. EMBO J. 18, 833–846. doi: 10.1093/emboj/18.4.833

PubMed Abstract | CrossRef Full Text | Google Scholar

Locke, A. E., Kahali, B., Berndt, S. I., Justice, A. E., Pers, T. H., Day, F. R., et al. (2015). Genetic studies of body mass index yield new insights for obesity biology. Nature 518, 197–206. doi: 10.1038/nature14177

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahoney, D. J., Parise, G., Melov, S., Safdar, A., and Tarnopolsky, M. A. (2005). Analysis of global mRNA expression in human skeletal muscle during recovery from endurance exercise. FASEB J. 19, 1498–1500. doi: 10.1096/fj.04-3149fje

PubMed Abstract | CrossRef Full Text | Google Scholar

Maingay, M., Romero-Ramos, M., Carta, M., and Kirik, D. (2006). Ventral tegmental area dopamine neurons are resistant to human mutant alpha-synuclein overexpression. Neurobiol. Dis. 23, 522–532. doi: 10.1016/j.nbd.2006.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Manning, A. K., Hivert, M. F., Scott, R. A., Grimsby, J. L., Bouatia-Naji, N., Chen, H., et al. (2012). A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat. Genet. 44, 659–669. doi: 10.1038/ng.2274

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsuda, W., Furuta, T., Nakamura, K. C., Hioki, H., Fujiyama, F., Arai, R., et al. (2009). Single nigrostriatal dopaminergic neurons form widely spread and highly dense axonal arborizations in the neostriatum. J. Neurosci. 29, 444–453. doi: 10.1523/JNEUROSCI.4029-08.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Millonig, J. H., Millen, K. J., and Hatten, M. E. (2000). The mouse Dreher gene Lmx1a controls formation of the roof plate in the vertebrate CNS. Nature 403, 764–769. doi: 10.1038/35001573

PubMed Abstract | CrossRef Full Text | Google Scholar

Monaghan, A. P., Kaestner, K. H., Grau, E., and Schütz, G. (1993). Postimplantation expression patterns indicate a role for the mouse forkhead/HNF-3 alpha, beta and gamma genes in determination of the definitive endoderm, chordamesoderm and neuroectoderm. Development 119, 567–578.

PubMed Abstract | Google Scholar

Morais, V. A., Haddad, D., Craessaerts, K., De Bock, P. J., Swerts, J., Vilain, S., et al. (2014). PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science 344, 203–207. doi: 10.1126/science.1249161

PubMed Abstract | CrossRef Full Text | Google Scholar

Murphy, T. H., and Corbett, D. (2009). Plasticity during stroke recovery: from synapse to behaviour. Nat. Rev. Neurosci. 10, 861–872. doi: 10.1038/nrn2735

PubMed Abstract | CrossRef Full Text | Google Scholar

Oosterveen, T., Garcao, P., Moles-Garcia, E., Soleilhavoup, C., Travaglio, M., Sheraz, S., et al. (2021). Pluripotent stem cell derived dopaminergic subpopulations model the selective neuron degeneration in Parkinson’s disease. Stem Cell Rep. 16, 2718–2735. doi: 10.1016/j.stemcr.2021.09.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Pacelli, C., Giguere, N., Bourque, M. J., Levesque, M., Slack, R. S., and Trudeau, L. E. (2015). Elevated mitochondrial bioenergetics and axonal arborization size are key contributors to the vulnerability of dopamine neurons. Curr. Biol. 25, 2349–2360. doi: 10.1016/j.cub.2015.07.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, J. S., Davis, R. L., and Sue, C. M. (2018). Mitochondrial dysfunction in parkinson’s disease: new mechanistic insights and therapeutic perspectives. Curr. Neurol. Neurosci. Rep. 18:21. doi: 10.1007/s11910-018-0829-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Pei, L., Waki, H., Vaitheesvaran, B., Wilpitz, D. C., Kurland, I. J., and Tontonoz, P. (2006). NR4A orphan nuclear receptors are transcriptional regulators of hepatic glucose metabolism. Nat. Med. 12, 1048–1055. doi: 10.1038/nm1471

PubMed Abstract | CrossRef Full Text | Google Scholar

Philippe, J., Morel, C., and Prezioso, V. R. (1994). Glucagon gene expression is negatively regulated by hepatocyte nuclear factor 3 beta. Mol. Cell Biol. 14, 3514–3523. doi: 10.1128/mcb.14.5.3514-3523.1994

PubMed Abstract | CrossRef Full Text | Google Scholar

Pissadaki, E. K., and Bolam, J. P. (2013). The energy cost of action potential propagation in dopamine neurons: clues to susceptibility in Parkinson’s disease. Front. Comput. Neurosci. 7:13. doi: 10.3389/fncom.2013.00013

PubMed Abstract | CrossRef Full Text | Google Scholar

Pristera, A., Lin, W., Kaufmann, A. K., Brimblecombe, K. R., Threlfell, S., Dodson, P. D., et al. (2015). Transcription factors FOXA1 and FOXA2 maintain dopaminergic neuronal properties and control feeding behavior in adult mice. Proc. Natl. Acad. Sci. U.S.A. 112, E4929–E4938. doi: 10.1073/pnas.1503911112

PubMed Abstract | CrossRef Full Text | Google Scholar

Puopolo, M., Raviola, E., and Bean, B. P. (2007). Roles of subthreshold calcium current and sodium current in spontaneous firing of mouse midbrain dopamine neurons. J. Neurosci. 27, 645–656. doi: 10.1523/JNEUROSCI.4341-06.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

Reis, A. F., and Velho, G. (2002). Sulfonylurea receptor -1 (SUR1): genetic and metabolic evidences for a role in the susceptibility to type 2 diabetes mellitus. Diabetes Metab. 28, 14–19.

PubMed Abstract | Google Scholar

Richardson, T. G., Sanderson, E., Elsworth, B., Tilling, K., and Davey Smith, G. (2020). Use of genetic variation to separate the effects of early and later life adiposity on disease risk: mendelian randomisation study. BMJ 369:m1203. doi: 10.1136/bmj.m1203

PubMed Abstract | CrossRef Full Text | Google Scholar

Sakaue, S., Kanai, M., Tanigawa, Y., Karjalainen, J., Kurki, M., Koshiba, S., et al. (2021). A cross-population atlas of genetic associations for 220 human phenotypes. Nat. Genet. 53, 1415–1424. doi: 10.1038/s41588-021-00931-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sasaki, H., and Hogan, B. L. (1993). Differential expression of multiple fork head related genes during gastrulation and axial pattern formation in the mouse embryo. Development 118, 47–59. doi: 10.1242/dev.118.1.47

PubMed Abstract | CrossRef Full Text | Google Scholar

Saucedo-Cardenas, O., Quintana-Hau, J. D., Le, W. D., Smidt, M. P., Cox, J. J., De Mayo, F., et al. (1998). Nurr1 is essential for the induction of the dopaminergic phenotype and the survival of ventral mesencephalic late dopaminergic precursor neurons. Proc. Natl. Acad. Sci. U.S.A. 95, 4013–4018. doi: 10.1073/pnas.95.7.4013

PubMed Abstract | CrossRef Full Text | Google Scholar

Sayre, L. M., Wang, F. J., Arora, P. K., Riachi, N. J., Harik, S. I., and Hoppel, C. L. (1991). Dopaminergic neurotoxicity in vivo and inhibition of mitochondrial respiration in vitro by possible endogenous pyridinium-like substances. J. Neurochem. 57, 2106–2115. doi: 10.1111/j.1471-4159.1991.tb06429.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Schiemann, J., Schlaudraff, F., Klose, V., Bingmer, M., Seino, S., Magill, P. J., et al. (2012). K-ATP channels in dopamine substantia nigra neurons control bursting and novelty-induced exploration. Nat. Neurosci. 15, 1272–1280. doi: 10.1038/nn.3185

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, J., and Cookson, M. R. (2004). Mitochondria and dopamine: new insights into recessive parkinsonism. Neuron 43, 301–304. doi: 10.1016/j.neuron.2004.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Sinnott-Armstrong, N., Tanigawa, Y., Amar, D., Mars, N., Benner, C., Aguirre, M., et al. (2021). Genetics of 35 blood and urine biomarkers in the UK Biobank. Nat. Genet. 53, 185–194.

Google Scholar

Sokoloff, L. (1960). “The metabolism of the central nervous system in vivo,” in Handbook of Physiology–Neurophysiology, vol III. Washington, DC: American Physiological Society, 1843–1864.

Google Scholar

Speliotes, E. K., Willer, C. J., Berndt, S. I., Monda, K. L., Thorleifsson, G., Jackson, A. U., et al. (2010). Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 42, 937–948. doi: 10.1038/ng.686

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramaniam, S. R., and Chesselet, M. F. (2013). Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Prog. Neurobiol. 106, 17–32.

Google Scholar

Sund, N. J., Vatamaniuk, M. Z., Casey, M., Ang, S. L., Magnuson, M. A., Stoffers, D. A., et al. (2001). Tissue-specific deletion of Foxa2 in pancreatic beta cells results in hyperinsulinemic hypoglycemia. Genes Dev. 15, 1706–1715. doi: 10.1101/gad.901601

PubMed Abstract | CrossRef Full Text | Google Scholar

Surmeier, D. J., Guzman, J. N., Sanchez, J., and Schumacker, P. T. (2012). Physiological phenotype and vulnerability in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2, a009290. doi: 10.1101/cshperspect.a009290

PubMed Abstract | CrossRef Full Text | Google Scholar

Tachmazidou, I., Süveges, D., Min, J. L., Ritchie, G. R. S., Steinberg, J., Walter, K., et al. (2017). Whole-genome sequencing coupled to imputation discovers genetic signals for anthropometric traits. Am. J. Hum. Genet. 100, 865–884. doi: 10.1016/j.ajhg.2017.04.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanguay, W., Ducrot, C., Giguere, N., Bourque, M. J., and Trudeau, L. E. (2021). Neonatal 6-OHDA lesion of the SNc induces striatal compensatory sprouting from surviving SNc dopaminergic neurons without VTA contribution. Eur. J. Neurosci. 54, 6618–6632. doi: 10.1111/ejn.15437

PubMed Abstract | CrossRef Full Text | Google Scholar

Trender-Gerhard, I., Sweeney, M. G., Schwingenschuh, P., Mir, P., Edwards, M. J., and Gerhard, A. (2009). Autosomal-dominant GTPCH1-deficient DRD: clinical characteristics and long-term outcome of 34 patients. J. Neurol. Neurosurg. Psychiatry 80, 839–845. doi: 10.1136/jnnp.2008.155861

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaxillaire, M., Populaire, C., Busiah, K., Cave, H., Gloyn, A. L., Hattersley, A. T., et al. (2004). Kir6.2 mutations are a common cause of permanent neonatal diabetes in a large cohort of French patients. Diabetes 53, 2719–2722. doi: 10.2337/diabetes.53.10.2719

PubMed Abstract | CrossRef Full Text | Google Scholar

Vogelezang, S., Bradfield, J. P., Ahluwalia, T. S., Curtin, J. A., Lakka, T. A., Grarup, N., et al. (2020). Novel loci for childhood body mass index and shared heritability with adult cardiometabolic traits. PLoS Genet. 16:e1008718. doi: 10.1371/journal.pgen.1008718

PubMed Abstract | CrossRef Full Text | Google Scholar

Whitaker, R., Gil, M. P., Ding, F., Tatar, M., Helfand, S. L., and Neretti, N. (2014). Dietary switch reveals fast coordinated gene expression changes in Drosophila melanogaster. Aging 6, 355–368. doi: 10.18632/aging.100662

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, C. J., and Callaway, J. C. (2000). Coupled oscillator model of the dopaminergic neuron of the substantia nigra. J. Neurophysiol. 83, 3084–3100. doi: 10.1152/jn.2000.83.5.3084

PubMed Abstract | CrossRef Full Text | Google Scholar

Wojcik, G. L., Graff, M., Nishimura, K. K., Tao, R., Haessler, J., Gignoux, C. R., et al. (2019). Genetic analyses of diverse populations improves discovery for complex traits. Nature 570, 514–518. doi: 10.1038/s41586-019-1310-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolfrum, C., Besser, D., Luca, E., and Stoffel, M. (2003). Insulin regulates the activity of forkhead transcription factor Hnf-3beta/Foxa-2 by Akt-mediated phosphorylation and nuclear/cytosolic localization. Proc. Natl. Acad. Sci. U.S.A. 100, 11624–11629. doi: 10.1073/pnas.1931483100

PubMed Abstract | CrossRef Full Text | Google Scholar

Xing, C., Cohen, J. C., and Boerwinkle, E. (2010). A weighted false discovery rate control procedure reveals alleles at FOXA2 that influence fasting glucose levels. Am. J. Hum. Genet. 86, 440–446. doi: 10.1016/j.ajhg.2010.01.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, C. H., Levesque, M., Claxton, S., Johnson, R. L., and Ang, S. L. (2011). Lmx1a and lmx1b function cooperatively to regulate proliferation, specification, and differentiation of midbrain dopaminergic progenitors. J. Neurosci. 31, 12413–12425. doi: 10.1523/JNEUROSCI.1077-11.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

Zetterstrom, R. H., Solomin, L., Jansson, L., Hoffer, B. J., Olson, L., and Perlmann, T. (1997). Dopamine neuron agenesis in Nurr1-deficient mice. Science 276, 248–250. doi: 10.1126/science.276.5310.248

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Rubins, N. E., Ahima, R. S., Greenbaum, L. E., and Kaestner, K. H. (2005). Foxa2 integrates the transcriptional response of the hepatocyte to fasting. Cell Metab. 2, 141–148. doi: 10.1016/j.cmet.2005.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Q., Xue, K., Guo, H. W., and Yang, Y. H. (2020). LMX1B rs10733682 polymorphism interacts with macronutrients, dietary patterns on the risk of obesity in han chinese girls. Nutrients 12:1227. doi: 10.3390/nu12051227

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: dopamine, midbrain development, FOXA, NR4A2, LMX1

Citation: Ni A and Ernst C (2022) Evidence That Substantia Nigra Pars Compacta Dopaminergic Neurons Are Selectively Vulnerable to Oxidative Stress Because They Are Highly Metabolically Active. Front. Cell. Neurosci. 16:826193. doi: 10.3389/fncel.2022.826193

Received: 30 November 2021; Accepted: 28 January 2022;
Published: 04 March 2022.

Edited by:

Alison Axtman, University of North Carolina at Chapel Hill, United States

Reviewed by:

Martin Darvas, University of Washington, United States
Matthew Wanat, University of Texas at San Antonio, United States

Copyright © 2022 Ni and Ernst. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Carl Ernst, carl.ernst@mcgill.ca

Download