Open access peer-reviewed chapter

Mitochondrial KATP Channel and Dopaminergic Vulnerability Neurons in Parkinson’s Disease

Written By

Gesivaldo Santos, Julita Maria Pereira Borges, Marcos Avilla-Rodriguez, Érika Pereira Rubio, Cattiúscia Batista Bromochenkel, Djalma Menezes Oliveira, Jane Lima dos Santos, Rosane Moura Aguiar, Milena Mascarenhas Ferraz, Silvana Batista Gaino, Francisco Capani and George E. Barreto

Submitted: 08 June 2018 Reviewed: 04 October 2018 Published: 12 March 2019

DOI: 10.5772/intechopen.81862

From the Edited Volume

Parkinson's Disease and Beyond - A Neurocognitive Approach

Edited by Sara Palermo, Mario Stanziano and Rosalba Morese

Chapter metrics overview

1,341 Chapter Downloads

View Full Metrics

Abstract

The motor deficiency control commonly characterizes Parkinson’s disease (PD), resulting in impairment of neuromuscular command, because of basal ganglia nuclei degeneration and late formation of Lewy’s bodies in the remaining dopaminergic (DA) neurons. Motor signals are triggered in high cortical motor areas and go toward the midbrain regions, where the final tuning movement takes place. PD is characterized primarily by the death of dopaminergic neurons in the regions known as substantia nigra compacta (STNc). Mutations in a couple of genes, such as Parkin1 and DJ1, correspond to the usual familial form of the disease, due to its association with oxidative stress and depolarization of mitochondrial membrane. However, this form does not explain the selective pattern of apoptosis between the neuronal dopaminergic areas of midbrain regions. In this chapter, we are putting forward the hypothesis of oxidative stress and mitochondrial changes as the apparent most relevant cause in PD, as well as the neuroprotective role played by Kir6.2, a potassium-ATP channel and calcium voltage-gated v1.3.

Keywords

  • Parkinson’s disease
  • potassium-ATP channel
  • calcium v1.3 channel
  • mitochondria ETC impairments
  • oxidative stress

Glossary:

  1. PD nonhuman animal model: simulation of PD may be acquired using rotenone and MPTP, two pesticides whose use leads to impairments in mitochondrial complex-I and in consequence an outburst of oxidative species and free radicals.

  2. Kir6.2/SUR1: during oxidative stress, potassium-ATP channel may act as a neuroprotector factor, by inducing GABA release in subcortical areas, thereby preventing neuronal glutamatergic overexcitation.

Advertisement

1. Introduction

Parkinson’s disease (PD) is related to the selective loss of neurons, which contain dopamine (DA) in substantia nigra compacta (STNc) and late formation of Lewy’s bodies in the remaining dopaminergic (DA) neurons [1]. PD is one of the most conditions, falling behind only Alzheimer’s disease. Parkinson’s disease prevalence is progressive, ranging from 1% in people >60 years old until 2–4% in people >70 years old.

PD is commonly known by the disease of the motor deficiency process [2]. Due to its association with impairments in basal ganglia, the presence of resting tremor, muscular rigidity, bradykinesia, sleep disturbances, gait impairment, and difficulties with balance [3, 4, 5] became the most prominent footprint in PD characterization (see Figure 1) [4].

Figure 1.

The basal ganglia and a series of subcortical nuclei responsible for the motor fine-tuning in health brain (right) and in PD (left). Direct pathway provides the disinhibition of the thalamus (D1 signaling in blue). Indirect pathway, in red, will stop the movement, previously initiated. D1, dopamine receptor 1; D2, dopamine receptor 2; GPi, globus pallidus internus; GPe, globus pallidus external.

In humans, movements are coordinated by a series of high-precision steps that begin in the regions of the prefrontal cortex and go toward the areas directly related to movement coordination, such as supplementary area and primary motor area [6]. From the start point, the signal follows pathways from inside the brain into the subcortical regions and reaches the spinal cord and finally the skeletal muscle [7]. However, it is not enough to simply generate the motor signal and deliver it to the muscle; it is necessary to coordinate and control the accuracy of the process generated in high cortical regions. That is the point where basal ganglia take place on this process as a stakeholder (Figure 1) [8].

Basal ganglia are composed of a series of subcortical nuclei scattered for the midbrain regions, whose role extends from the motor fine-tuning, initialization, and finalization of the movement (processes supported by the substantia nigra pars compacta (STNc)), as well as the development of cognitive functions such as learning, reward, and emotions, mostly supported by the ventral tegmental area (VTA) and nucleus accumbens (NC). Together, they are functionally known as the ventral and dorsal striatum, respectively [9].

As one can see in Figure 1, in parkinsonism, the striatum plays an important role in the pathophysiology of the disease, and the substantia nigra compacta (STNc) emerges as the main nucleus responsible for the core mechanism related to the initialization of the movement, assigned to the direct pathway [10]. Direct pathway provides the disinhibition of the thalamus by the dopamine D1 signaling, performed by STNc [11]. In opposite side, the indirect pathway will stop the movement, previously initiated by the release of GABA from the external globus pallidus (GPe) and substantia nigra pars reticulata (STNr) [12].

James Parkinson did the first description of the disease in 1817 in his book: An Essay on the Shaking Palsy [13]. PD is characterized primarily by the death of dopaminergic neurons in the STNc resulting in defective control of the movements in the basal ganglia (Figure 1) [14]. However, in addition to dopamine metabolism impairment, another pathological process implicated in the etiology of the disease is the aggregation of alpha-synuclein (a-syn), resulting in Lewy’s body formation in the remaining dopaminergic neurons, characterizing the non-motor symptoms of the disease (Figure 2) [4, 5, 15].

Figure 2.

Dopaminergic neurons undergo several mechanisms that may explain in part the selective cell death in substantia nigra. Mutations associated with major PD related genes such as α-synuclein, DJ-1 and Parkin are well characterized. It is possible that the mutated genes induced a protein imbalance that lead to the aggregation of proteins to form the so-called Lewy’s bodies. Another key mechanism studied is the ROS imbalance in mitochondria resulting in a diminished ATP that affect the ability of dopaminergic neuron to reach its main functions. A plausible mechanism to be explored is the role of K-ATP channel that is sensitive to ATP/ADP changes to govern the depolarization in GABAergic neuron.

Despite the dominant prevalence of sporadic form (90%), mutations in genes, such as a-synuclein, DJ1, Parkin1, and others, have been implicated in the familial pattern of PD, mainly due to their associations with oxidative stress responses and depolarization of mitochondrial membrane. However, gene alterations in the familial form of PD are ambiguous because they do not explain the selective pattern of apoptosis between STNc and VTA, both formed predominantly by dopaminergic neurons (Figure 2) [14, 16]. Another hypothesis associated to the oxidative stress and mitochondrial changes is related to Ca+2 imbalance homeostasis in dopaminergic neurons of the STNc, as well as the presence of KATP channels acting as a metabolic sensor by coupling glucose metabolism to mitochondrial membrane potential (Mit-ψ) (see Figures 3 and 4) [17]. In other words, understanding the mechanism of specific vulnerability, by which only dopaminergic neurons in specific areas are associated with PD, remains an impressive challenge; however, some evidences, that will be discussed here, may shed light in an intricate neurobiology of Parkinson’s condition [14, 18].

Figure 3.

K-ATP and mitochondrial functions may be a potential relationship. K-ATP sensor is subjected to regulation via several mechanisms as glucose metabolism, ATP/ADP ratio, mitochondrial decoupling, and gene expression. K-ATP may lead the neuron to depolarize via potassium and calcium fluxes. It is reported that K-ATP has other subtypes of receptors, for example, the mito-KATP. It is possible that the pathogenic production of ROS may alter the function of K-ATP with protective functions. So, it is plausible that the modulation of K-ATP receptors can be used to protect dopaminergic neurons.

Figure 4.

Glucose and energetic metabolism may regulate the function of K-ATP channel. The change in the ATP/ADP ratio by glucose metabolism induces a response in K-ATP that blockade the potassium outward with the subsequent loss of membrane potential. It may lead to depolarization with several functions, for example, the release of GABA.

Advertisement

2. Substantia nigra compacta and ROS formation

The use of rotenone and MPTP, two pesticides, which act on the mitochondrial complex-I, brought mitochondrial damage induced by oxidative stress to the center of PD etiology. The uplift of reactive oxygen species (ROS), caused by a defective complex-I, such as observed in PD patients, makes mitochondria the main source of ROS in the intracellular environment (Figure 3) [19, 20]. Superoxide (O2) and peroxide of hydrogen (H2O2), the two main species of ROS derived from electron transport chain (ETC) activity, seem to govern the internal state of the cell between proliferative and apoptotic. According to this model, high levels of O2 tend to favor cell proliferation due to increased transcription of oncogenes, such as the Rac1-Bcl2 pathway, whose overexpression increases mitochondrial respiration. On the other hand, prevailing levels of H2O2 promote apoptosis, and finally, very high rates of H2O2 promote cellular necrosis due to irreparable damage to cell physiology (Figure 3) [21].

The hypothesis of oxidative stress and mitochondrial changes is apparently the most relevant cause in the sporadic PD (Figure 3). However, they alone do not explain selective dopaminergic neuron vulnerability. The key element for this may lie in the differential expression of KATP channels (Kir6.2) between STNc neurons and ventral tegmental area (VTA) [22]. In in vitro studies, the total decoupling of the mitochondrial respiratory chain leads to the activation of KATP channels in all dopaminergic neurons. However, partial decoupling of complex-I strongly affects neurons from STNc, showing an opposite effect on KATP channels in dopaminergic neurons of VTA, whose process increases the neuronal activity, reducing ROS formation due to the closure of the KATP channels (Figure 4). It has been identified that D2-autoreceptor (D2-AR) acts as an inhibitor of STNc-DA neurons in response to local high DA release [23]. All the processes have been achieved by activation of Kir6.2, an inward rectifier potassium channel coupled to G-protein [23]. Although ventral tegmental area DA neurons (VTA-DA) do not present the same response in such like condition [24], studies account for the presence of Cav+21.3 channel as the main responsible for this selectivity in STN-DA neurons, whose function would be associated with the downregulation of STN-DA neuronal cell activity in response to dangerous transient of local DA release [25].

2.1 The starting point of oxidative stress and KATP channel disposition

In PD, pathogenesis predominates the selective loss of dopaminergic neurons. These neurons are in the STNc, with projections up to the striatum zone. The neurochemistry of degeneration involves several molecular events triggered by mitochondrial dysfunction with increased oxidative stress and excitotoxicity caused by extracellular Ca+2 overflow. These events promote important changes in protein conformation, e.g., alpha-synuclein, responsible for Lewy’s body formation, a defective protein aggregation resulting in mitophagies and apoptosis [26, 27, 28].

Dopamine is a catecholamine synthesized from the L-dihydroxyphenylalanine (L-dopa) of the amino acid tyrosine by the enzyme tyrosine hydroxylase (TH). In the next step, L-dopa undergoes decarboxylation by the aromatic amino acid decarboxylase (AADC) to generate dopamine and CO2. TH and AADC form a complex with the vesicular monoaminergic carrier-2 (VMAT-2), facilitating the uptake of dopamine in the monoaminergic synaptic vesicles [29]. The TH, the AADC complex, and the VMAT-2 transporter facilitate the absorption of dopamine in the monoaminergic synaptic vesicle, thus avoiding the oxidation of dopamine to o-quinones by the dissociation of the hydroxyls of the catechol present in the molecule when at p. 7.4 of the cytosol [30]. In the monoaminergic vesicle, the stability of the molecule in the protonated form is conferred by an estimated pH of approximately 5 [30].

Dopamine, which is not absorbed by VMAT-2, is transported freely by the cytosol and may undergo oxidation by monoamine oxidase to give rise to dihydroxyphenylacetic acid, methylation by ortho-methyltransferase, and structure oxidation of catechol to o-quinone aminochrome [31]. 0-Quinones derived from dopamine to form aminochrome are rapidly mopped by cysteines (or other thiols present) generating forms that are oxidized to form melanotic pigments [32, 33, 34]. The action of flavenzyme FADH, such as DT-diaphorase induce the formation of hydroquinone and ROS from reduction of o-quinones derived of freely dopamine in the cytosol [35, 36]. Semiquinone is a highly reactive radical, and under aerobic conditions, it catalyzes the reduction reaction of oxygen to the superoxide that activates the redox cycle between the leucoaminochrome o-semiquinone radical and the aminochrome [37, 38].

Aminochrome leads to the formation of species with proteins of complexes I and III of the mitochondrial electron transport chain associated with reduced flavin adenine dinucleotide (FADH2) [39]. It forms compounds with the isocitrate dehydrogenase, leading to mitochondrial dysfunction due to a decreased ATP production [40]. In addition, aminochrome forms adducts with alpha-synuclein protofibrils and with Parkin leading to proteasome dysfunction, with actin leading to dysfunction in axonal and cytoskeletal transport, aggregation of α-β tubulins leading to autophagy dysfunction [41, 42]. In PD models, the active metabolite of 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), MPP+, and rotenone are mitochondrial toxins that inhibit the NADH-quinone oxidoreductase complex respiratory [37, 43, 44].

2.2 Mitochondrial KATP channels

Mitochondrial dysfunction seems to play a crucial role in PD [44, 45, 46, 47]. Mitochondria is directly implicated in ROS generation and consequently in neuronal cell death in vulnerable population. Mitochondrial KATP channels (mito-KATP) act as a gate control linking metabolism with cell survival and neurotransmitter releasing (Figures 2 and 4) [48]. Kir6.2 mito-KATP channels are encoded by KCNJ11 gene family, whereas SUR1, the KATP channel sensor subunit, belongs to the class of ABCC8, a subfamily of membranes transporters [49, 50].

KATP channels are triggered by the ATP/ADP rate, opening in response to reduced levels of ATP and, closing, otherwise, linking KATP channels in a direct relationship with the neuronal sugar metabolism and the action potential [51]. In physiological conditions, in dopaminergic neurons, these channels are probably closed. On the other hand, in PD model induced by MPTP administration in mice, the ATP depletion and mitochondrial dysfunction are observed, thus triggering the opening of the mito-KATP channels (Figure 4) [52].

Activation of KATP channels upon damage may play a neuroprotective role by decreasing the cellular metabolic demand, reducing activation rate of the action potential, thus leading to hyperpolarization of the dopaminergic neurons and loss of its normal pacemaker activity [53].

KATP channels play an important role in signal transduction in the central nervous system (CNS). For example, these channels are implicated in rest potential of most neuronal cell controlling the duration of action potential, firing frequency, and nonspecific intervals, thus regulating pacemaker time [54]. These ionic channels stabilize the membrane potential and the mitochondrial matrix volume during the ATP decline in order to increase the firing in dopaminergic neurons, as well as activation of metabolic pathways to provide cell energy (Figures 3 and 4) [25].

During mitochondrial dysfunction caused by oxidative stress or in the presence of neurotoxins (MPTP or rotenone), high calcium concentration and the hyperpolarization of membrane potential may be involved in the reduction of cellular activity in adult rats [52]. However, the vulnerability of dopaminergic neurons may be related to the differential expression of KATP (Kir6.2) between STNc and VTA neurons. Acute activation of rotenone-induced KATP channels in rat brain slices in responsive dopaminergic neurons increases the expression of the KATP channel subunits, the SUR1 and Kir6.2 [55]. However, the chronic effect of ATP depletion and consequent opening of KATP channels (Kir6.2/SUR1) due to its metabolic sensitivity in the vulnerable STNc and VTA-DA neurons decrease the expression of the decoupling protein (UCP2) due to the lower degree of mitochondrial decoupling conducted by the metabolic stress in PD [25].

The opening of the KATP channels may result in the hyperpolarization of the neurotransmitters modulating the release of glutamate and g-aminobutyric acid (GABA) in the substantia nigra reticulata (StNr) and in the striatum, reducing glutamatergic transmission into the brain (Figure 4) [56]. This fact suggests the significance of oxidative stress and mitochondrial alterations as a common remarkable cause in the development of PD (Figures 3 and 4).

During PD development, Cav1.2 in favor of Cav1.3 is differentially expressed in brain areas, thus resulting in an increase of neuronal susceptibility to events associated with oxidative stress [1, 24]. The hypothesis of differential expression of Cav1 subtypes to explain neuronal selective cell death remains inconclusive, but the fact that there is a change in its prior expression throughout the brain in early stages of the disease’s development reinforces our previous proposition that calcium imbalance is a fundamental requirement to understand PD pathogenesis.

Advertisement

3. Conclusions

In this review, we suggest that both mitochondrial KATP channel and calcium1.3-voltage-gated contribute to maintain a balance between cell proliferation and apoptosis, acting as a metabolic sensor by coupling ROS and glucose metabolism to mitochondrial membrane potential in dopaminergic neurons. Finally, in conclusion, it may lead to a new pathway of drug development and treatment of PD.

Advertisement

List of abbreviations

AADC

aromatic amino acid decarboxylase

ABCC8

ATP-binding cassette subfamily C member 8

ADP

adenosine diphosphate

a-syn

alpha-synuclein

ATP

adenosine triphosphate

Cav1.2

voltage-dependent calcium channel subunit alpha 1C

Cav1.3

voltage-dependent calcium channel subunit alpha 1D

CNS

central nervous system

D2-AR

dopaminergic D2-autoreceptor

DA

dopaminergic

DJ1

protein deglycase 1

PD

Parkinson’s disease

ETC

electron transport chain

FADH2

Flavin adenine dinucleotide

GABA

g-aminobutyric acid

GPe

external globus pallidus

KATP

potassium channels

KCNJ11

potassium voltage-gated channel subfamily J member 11

Kir6.2

subunit ATP-sensitive K+ channel

L-dopa

L-3,4-dihydroxyphenylalanine

Mito-KATP

mitochondrial ATP-dependent K+ channel

mit-psi

mitochondrial membrane potential

MPP+

metabolite of 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine

MPTP

1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine

NADH

nicotinamide adenine dinucleotide

NC

nucleus accumbens

ROS

reactive oxygen species

STNc

substantia nigra compacta

STNr

substantia nigra pars reticulata

SUR1

sulfonylurea receptor

TH

tyrosine hydroxylase

UCP2

decoupling protein 2

VMAT-2

vesicular monoaminergic carrier-2

VTA

ventral tegmental area

VTA-DA

ventral tegmental area DA neurons

References

  1. 1. Hurley MJ, Gentleman SM, Dexter DT. Calcium CaV1 channel subtype mRNA expression in Parkinson’s disease examined by in situ hybridization. Journal of Molecular Neuroscience. 2015;55(3):715-724. ISSN: 0895-8696
  2. 2. Mazzoni P, Shabbott B, Cortes JC. Motor control abnormalities in Parkinson’s disease. Cold Spring Harbor Perspectives in Medicine. 2012;2(6):a009282. ISSN: 2157-1422
  3. 3. Rizek P, Kumar N, Jog MS. An update on the diagnosis and treatment of Parkinson disease. CMAJ. 2016;188(16):1157-1165. ISSN: 0820-3946
  4. 4. Kalia LV et al. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurology. 2015;72(1):100-105. ISSN: 2168-6149
  5. 5. Aarsland D, Creese B, Chaudhuri KR. A new tool to identify patients with Parkinson’s disease at increased risk of dementia. Lancet Neurology. 2017;16(8):576-578. ISSN: 1474-4422
  6. 6. Masumoto J, Inui N. Motor control hierarchy in joint action that involves bimanual force production. Journal of Neurophysiology. 2015;113(10):3736-3743. ISSN: 0022-3077
  7. 7. Nakajima T et al. Reassessment of non-monosynaptic excitation from the motor cortex to motoneurons in single motor units of the human biceps brachii. Frontiers in Human Neuroscience. 2017;11:19. ISSN: 1662-5161 (Print)1662-5161
  8. 8. Nambu A. A new dynamic model of the cortico-basal ganglia loop. Progress in Brain Research. 2004;143:461-466. ISSN: 0079-6123 (Print)0079-6123
  9. 9. Mannella F, Baldassarre G. Selection of cortical dynamics for motor behaviour by the basal ganglia. Biological Cybernetics. 2015;109(6):575-595. ISSN: 0340-1200
  10. 10. Bolam JP et al. Synaptic organisation of the basal ganglia. Journal of Anatomy. 2000;196(Pt 4):527-542. ISSN: 0021-8782 (Print)0021-8782
  11. 11. Wu J et al. Iptakalim modulates ATP-sensitive K(+) channels in dopamine neurons from rat substantia nigra pars compacta. Journal of Pharmacology and Experimental Therapeutics. 2006;319(1):155-164. ISSN: 0022-3565 (Print)0022-3565
  12. 12. Nikolaus S et al. GABAergic control of nigrostriatal and mesolimbic dopamine in the rat brain. Frontires in Behavioral Neuroscience. 2018;12:38. ISSN: 1662-5153 (Print)1662-5153
  13. 13. Goetz CG. The history of Parkinson’s disease: Early clinical descriptions and neurological therapies. Cold Spring Harbor Perspectives in Medicine. 2011;1(1):a008862. ISSN: 2157-1422
  14. 14. Bose A, Beal MF. Mitochondrial dysfunction in Parkinson’s disease. Journal of Neurochemistry. 2016;139(Supp. 1):216-231. ISSN: 0022-3042
  15. 15. Soukup SF, Vanhauwaert R, Verstreken P. Parkinson’s disease: Convergence on synaptic homeostasis. The EMBO Journal. 2018;37:e98960. ISSN: 0261-4189
  16. 16. Tan EK, Skipper LM. Pathogenic mutations in Parkinson disease. Human Mutation. 2007;28(7):641-653. ISSN: 1059-7794
  17. 17. Yee AG et al. Paradoxical lower sensitivity of locus coeruleus than substantia nigra pars compacta neurons to acute actions of rotenone. Experimental Neurology. 2017;287(Pt 1):34-43. ISSN: 0014-4886
  18. 18. Conradsson D, Paquette C, Franzen E. Turning stability in individuals with Parkinson disease. Journal of Neurologic Physical Therapy. 2018;42(4):241?247. ISSN: 1557-0576
  19. 19. Dos Santos AB, Kohlmeier KA, Barreto GE. Are sleep disturbances preclinical markers of Parkinson’s disease? Neurochemical Research. 2015;40(3):421-427. ISSN: 0364-3190
  20. 20. Dos Santos AB, Barreto GE, Kohlmeier KA. Treatment of sleeping disorders should be considered in clinical management of Parkinson’s disease. Frontiers in Aging Neuroscience. 2014;6:273. ISSN: 1663-4365 (Print)1663-4365
  21. 21. Zhang L et al. Interception of benzo[a]pyrene-7,8-dione by UDP glucuronosyltransferases (UGTs) in human lung cells. Chemical Research in Toxicology. 2013;26(10):1570-1578. ISSN: 0893-228x
  22. 22. Han SS et al. The expression of KATP channel subunits in alpha-synuclein-transfected MES23.5 cells. Annals of Translational Medicine. 2018;6(10):170. ISSN 2305-5839 (Print)2305-5839
  23. 23. Koyrakh L et al. Molecular and cellular diversity of neuronal G-protein-gated potassium channels. The Journal of Neuroscience. 2005;25(49):11468-11478. ISSN: 0270-6474
  24. 24. Dragicevic E et al. Cav1.3 channels control D2-autoreceptor responses via NCS-1 in substantia nigra dopamine neurons. Brain. 2014;137(Pt 8):2287-2302. ISSN: 0006-8950
  25. 25. Dragicevic E, Schiemann J, Liss B. Dopamine midbrain neurons in health and Parkinson’s disease: Emerging roles of voltage-gated calcium channels and ATP-sensitive potassium channels. Neuroscience. 2015;284:798-814. ISSN: 0306-4522
  26. 26. Jenner P. Dopamine agonists, receptor selectivity and dyskinesia induction in Parkinson’s disease. Current Opinion in Neurology. 2003;16(Supp. 1):S3-S7. ISSN: 1350-7540 (Print)1350-7540
  27. 27. Halliday GM, Stevens CH. Glia: Initiators and progressors of pathology in Parkinson’s disease. Movement Disorders. 2011;26(1):6-17. ISSN: 0885-3185
  28. 28. Vives-Bauza C, Przedborski S. Mitophagy: The latest problem for Parkinson’s disease. Trends in Molecular Medicine. 2011;17(3):158-165. ISSN: 1471-4914
  29. 29. Cartier EA et al. A biochemical and functional protein complex involving dopamine synthesis and transport into synaptic vesicles. Journal of Biological Chemistry. 2010;285(3):1957-1966. ISSN: 0021-9258
  30. 30. Guillot TS, Miller GW. Protective actions of the vesicular monoamine transporter 2 (VMAT2) in monoaminergic neurons. Molecular Neurobiology. 2009;39(2):149-170. ISSN 0893-7648 (Print)0893-7648
  31. 31. Segura-Aguilar J, Kostrzewa RM. Neurotoxins and neurotoxicity mechanisms. An overview. Neurotoxicity Research. 2006;10(3-4):263-287. ISSN 1029-8428 (Print)1029-8428
  32. 32. Eisenhofer G, Kopin IJ, Goldstein DS. Catecholamine metabolism: A contemporary view with implications for physiology and medicine. Pharmacological Reviews. 2004;56(3):331-349. ISSN 0031-6997 (Print)0031-6997
  33. 33. Zafar KS, Siegel D, Ross D. A potential role for cyclized quinones derived from dopamine, DOPA, and 3,4-dihydroxyphenylacetic acid in proteasomal inhibition. Molecular Pharmacology. 2006;70(3):1079-1086. ISSN: 0026-895X (Print)0026-895x
  34. 34. Bisaglia M et al. Dopamine quinones interact with alpha-synuclein to form unstructured adducts. Biochemical and Biophysical Research Communications. 2010;394(2):424-428. ISSN: 0006-291x
  35. 35. Pereira MRG, de Oliveira ES, de Villar FAGA, Grangeiro MS, Fonseca J, Silva AR, et al. Cytotoxicity of catechol towards human glioblastoma cells via superoxide and reactive quinones generation. Brazilian Journal of Pathology and Laboratory Medicine 2004;40:80-85. ISSN 1678-4774
  36. 36. Ito S et al. The metabolic fate of ortho-quinones derived from catecholamine metabolites. International Journal of Molecular Sciences. 2016;17(2). ISSN: 1422-0067
  37. 37. Esteves AR et al. Mitochondrial dysfunction: The road to alpha-synuclein oligomerization in PD. Parkinsons Disease. 2011;2011:693761. ISSN: 2042-0080
  38. 38. Biosa A et al. Dopamine oxidation products as mitochondrial endotoxins, a potential molecular mechanism for preferential neurodegeneration in Parkinson’s Disease. ACS Chemical Neuroscience. 2018. ISSN: 1948-7193
  39. 39. Barreto GE et al. Catechol inhibits FADH2-linked respiration in rat liver mitochondrial fraction. Acta Cirúrgica Brasileira. 2005;20(Supp. 1):72-77. ISSN 0102-8650 (Print)0102-8650
  40. 40. Van Laar VS, Berman SB. Mitochondrial dynamics in Parkinson’s disease. Experimental Neurology. 2009;218(2):247-256. ISSN: 0014-4886
  41. 41. Norris EH, Giasson BI. Role of oxidative damage in protein aggregation associated with Parkinson’s disease and related disorders. Antioxidants & Redox Signaling. 2005;7(5-6):672-684. ISSN: 1523-0864 (Print)1523-0864
  42. 42. Segura-Aguilar J, Huenchuguala S. Aminochrome induces irreversible mitochondrial dysfunction by inducing autophagy dysfunction in Parkinson’s disease. Frontiers in Neuroscience. 2018;12:106. ISSN 1662-4548 (Print)1662-453x
  43. 43. Barreto RA et al. Monocrotaline pyrrol is cytotoxic and alters the patterns of GFAP expression on astrocyte primary cultures. Toxicology In Vitro. 2008;22(5):1191-1197. ISSN 0887-2333 (Print)0887-2333
  44. 44. Cabezas R et al. Mitochondrial functions in astrocytes: Neuroprotective implications from oxidative damage by rotenone. Neuroscience Research. 2012;74(2):80-90. ISSN: 0168-0102
  45. 45. Cabezas R, Baez-Jurado E, Hidalgo- Lanussa O, Echeverria V, Ashrad GM, Sahebkar A, Barreto GE. Growth factors and Neuroglobin in astrocyte protection against neurodegeneration and oxidative stress. Molecular Neurobiology. Jul 7, 2018:1-3. ISSN: 0893-7648
  46. 46. Cabezas R, Ávila M, Gonzalez J, et al. Astrocytic modulation of blood brain barrier: Perspectives on Parkinson’s disease. Frontiers in Cellular Neuroscience. 2014;8:211. ISSN: 1662-5102
  47. 47. Sutachan JJ et al. Cellular and molecular mechanisms of antioxidants in Parkinson’s disease. Nutritional Neuroscience. 2012;15(3):120-126. ISSN: 1028-415x
  48. 48. Lutas A, Birnbaumer L, Yellen G. Metabolism regulates the spontaneous firing of substantia nigra pars reticulata neurons via KATP and nonselective cation channels. The Journal of Neuroscience. 2014;34(49):16336-16347. ISSN: 0270-6474
  49. 49. Vajapey R et al. The impact of age-related dysregulation of the angiotensin system on mitochondrial redox balance. Frontiers in Physiology. 2014;5:439. ISSN: 1664-042X (Print)1664-042x
  50. 50. Lee KPK, Chen J, Mackinnon R. Molecular structure of human KATP in complex with ATP and ADP. eLife. 2017;6. ISSN: 2050-084x
  51. 51. Minami K et al. Roles of ATP-sensitive K+ channels as metabolic sensors: Studies of Kir6.x null mice. Diabetes. 2004;53(Supp. 3):S176-S180. ISSN: 0012-1797 (Print)0012-1797
  52. 52. Liss B et al. K-ATP channels promote the differential degeneration of dopaminergic midbrain neurons. Nature Neuroscience. 2005;8(12):1742-1751. ISSN: 1097-6256 (Print)1097-6256
  53. 53. Piri H et al. The anti-Parkinsonism effects of KATP channel blockade in the 6-hydroxydopamine-induced animal model: The role of oxidative stress. Basic and Clinical Neuroscience. 2017;8(3):183-192. ISSN: 2008-126X (Print)2008-126x
  54. 54. Kou J, Klorig DC, Bloomquist JR. Potentiating effect of the ATP-sensitive potassium channel blocker glibenclamide on complex I inhibitor neurotoxicity in vitro and in vivo. Neurotoxicology. 2006;27(5):826-834. ISSN: 0161-813X (Print)0161-813x
  55. 55. Avshalumov MV et al. H2O2 signaling in the nigrostriatal dopamine pathway via ATP-sensitive potassium channels: Issues and answers. Antioxidants & Redox Signaling. 2007;9(2):219-231. ISSN 1523-0864 (Print)1523-0864
  56. 56. Milton SL, Lutz PL. Adenosine and ATP-sensitive potassium channels modulate dopamine release in the anoxic turtle (Trachemys scripta) striatum. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2005;289(1):R77-R83. ISSN: 0363-6119 (Print)0363-6119

Written By

Gesivaldo Santos, Julita Maria Pereira Borges, Marcos Avilla-Rodriguez, Érika Pereira Rubio, Cattiúscia Batista Bromochenkel, Djalma Menezes Oliveira, Jane Lima dos Santos, Rosane Moura Aguiar, Milena Mascarenhas Ferraz, Silvana Batista Gaino, Francisco Capani and George E. Barreto

Submitted: 08 June 2018 Reviewed: 04 October 2018 Published: 12 March 2019