Next Article in Journal
SIRT1: Genetic Variants and Serum Levels in Age-Related Macular Degeneration
Next Article in Special Issue
Differential Activation of pERK1/2 and c-Fos Following Injury to Different Regions of Primary Sensory Neuron
Previous Article in Journal
Nanoparticles: Synthesis and Their Role as Potential Drug Candidates for the Treatment of Parasitic Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Expression Analysis of Genes Involved in Transport Processes in Mice with MPTP-Induced Model of Parkinson’s Disease

by
Margarita M. Rudenok
1,*,
Maria I. Shadrina
1,
Elena V. Filatova
1,
Ivan N. Rybolovlev
1,
Maxim S. Nesterov
2,
Denis A. Abaimov
3,
Ruslan A. Ageldinov
2,
Anna A. Kolacheva
4,
Michael V. Ugrumov
4,
Petr A. Slominsky
1 and
Anelya Kh. Alieva
1
1
Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 2 Kurchatova Sq., 123182 Moscow, Russia
2
Scientific Center of Biomedical Technologies of the Federal Medical and Biological Agency of Russia, Svetlye Gory Village, Post Office Otradnoe, Krasnogorsk District, 143442 Moscow, Russia
3
The Research Centre of Neurology, 80, Volokolamskoye Shosse, 125367 Moscow, Russia
4
Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
*
Author to whom correspondence should be addressed.
Life 2022, 12(5), 751; https://doi.org/10.3390/life12050751
Submission received: 26 April 2022 / Revised: 13 May 2022 / Accepted: 17 May 2022 / Published: 19 May 2022
(This article belongs to the Special Issue Neurological Diseases: From Molecular Mechanisms to Therapy)

Abstract

:
Processes of intracellular and extracellular transport play one of the most important roles in the functioning of cells. Changes to transport mechanisms in a neuron can lead to the disruption of many cellular processes and even to cell death. It was shown that disruption of the processes of vesicular, axonal, and synaptic transport can lead to a number of diseases of the central nervous system, including Parkinson’s disease (PD). Here, we studied changes in the expression of genes whose protein products are involved in the transport processes (Snca, Drd2, Rab5a, Anxa2, and Nsf) in the brain tissues and peripheral blood of mice with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced models of PD. We detected changes in the expressions of Drd2, Anxa2, and Nsf at the earliest modeling stages. Additionally, we have identified conspicuous changes in the expression level of Anxa2 in the striatum and substantia nigra of mice with MPTP-induced models of PD in its early stages. These data clearly suggest the involvement of protein products in these genes in the earliest stages of the pathogenesis of PD.

1. Introduction

Among the most important processes in cell function are intracellular and extracellular transport [1,2,3]. These are carried out using transport proteins, cytoskeletal proteins, and transport vesicles [3,4].
Of particular importance is the cell transport in neurons. This is because of the structural features of these cell types, as in addition to the soma, they have very long processes (axons and dendrites) [3,5,6,7]. These require significant resources for the implementation of intracellular transport processes and provide one of the main functions of a neuron: transmission of a nerve impulse for synaptic transmission [8,9,10].
Nerve impulse transmission at the synapse is a complex physiological process that proceeds in several stages and includes multiple participants, such as synaptic vesicles, receptors, neurotransmitters, and transporters [11]. Vesicular transport is one of the most important components in synaptic transmission. In general, for a neuron, three main types of transport can be distinguished: axonal, vesicular, and synaptic [4,12]. Axonal transport ensures the integrity of the neuron and the connection of the soma with the synapse [12]. Vesicular transport is one of the most extensive types. Vesicles can be used for the transport of substances across the cell membrane by endocytosis and exocytosis. Numerous proteins and protein complexes participate in the formation, movement, and secretion of vesicle contents [9,10,13]. Synaptic transport is essential in the propagation of a nerve impulse and is also one of the most common modes of interaction between neurons [14,15,16].
Changes to transport mechanisms in a neuron can lead to the disruption of many cellular processes and even to cell death. Changes or disturbances in synaptic transmission can lead to a slowdown or interruption in the transmission of nerve impulses and to the disorganization of the brain and the organism as a whole [3,17].
Disruption of the processes of vesicular, axonal, and synaptic transport can lead to a number of diseases of the central nervous system [3,17,18,19,20,21,22,23,24,25,26,27,28], including Parkinson’s disease (PD) [4,29,30,31,32]. A key histopathology of PD is the presence of Lewy bodies (LBs) in the cell bodies and outgrowths of neurons in affected areas of the brain. They can have a toxic effect on the cell and isolate proteins vital for cell function [33]. There is evidence that the presence of LBs can be associated with disruptions to neuronal transport [28,34,35]. In addition, the presence of LBs complicates and changes transport in nerve cells [36,37,38]. This can lead to the accumulation of proteins in certain parts of the cell, obstruct synaptic signal transmission, and lead to slow neuronal death [39].
Previously, we performed a whole-transcriptome analysis of mouse brain tissues with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced models of the early stages of PD [40,41]. It was shown that changes in the expression of proteins associated with transport could affect the very early stages of pathogenesis. In addition, the use of bioinformatic approaches made it possible to analyze the data and identify a number of genes influencing changes to the transport processes in the early stages of neurodegeneration.
Here, we present an account of the data obtained as a result of whole-transcriptome analysis and a study of the changes in expression of selected genes in MPTP-induced models of the early stages of PD.

2. Materials and Methods

2.1. Parkinson’s Disease Models

We used models of the presymptomatic stage of PD with decapitation of mice 6 h after last injection of MPTP (6 h-PSS), the presymptomatic stage of PD with decapitation of mice 24 h after last injection of MPTP (24 h-PSS), and the advanced presymptomatic (AdvPSS) and early symptomatic (ESS) stages of PD. Models were created as previously described [42,43]. Detailed information is presented in the work performed by our team earlier [44]. A brief description is provided in Table 1.
All efforts were made to minimize the potential suffering and discomfort of animals and care was taken according to the 3R rule (replacement, reduction, and refinement).

2.2. RNA Isolation and Expression Analysis of Individual Candidate Genes

RNA isolation from tissue samples from each animal as well as analysis of mRNA levels using reverse transcription and real-time PCR (TaqMan technology, StepOnePlus™ System, Applied Biosystems, USA) was carried out in accordance with the protocols described previously [44].
Primer and Probe design was produced using Beacon designer 7.0 software (v. 7.0, Premier Biosoft International, Palo Alto, CA, USA) and the nucleotide sequences of the Snca, Drd2, Rab5a, Anxa2, and Nsf genes and the reference genes Bcat2 and Psmd7 [45,46,47,48]. The sequences of gene-specific primers and probes are presented in Table 2.

2.3. Statistical Processing and Bioinformatic Analysis

The protocol of statistical and bioinformatic analysis is described in detail in the work carried out in our laboratory earlier [44].

3. Results

Previously, the whole-transcriptome analysis of the murine striatum and substantia nigra in models of the presymptomatic stage of PD with the decapitation of mice 6 h after the last injection of MPTP (6 h-PSS), the presymptomatic stage of PD with the decapitation of mice 24 h after the last injection of MPTP (24 h-PSS), and the advanced presymptomatic (AdvPSS) and early symptomatic (ESS) stages of PD was carried out in our laboratory. As a result, 170 differentially expressed genes that belonged to the “transport” clusters were identified [41].
We performed a more detailed analysis of these genes using Pathway Studio® software 12.0 (Elsevier, The Netherlands) [49,50]. A network of interactions was built using the keywords “Parkinson’s disease”, “transport”, “neuron”, and “neurodegeneration” under the condition of direct interactions between network participants. As a result, a network of interactions of five genes, Snca, Drd2, Rab5a, Anxa2, and Nsf, was built, corresponding to all keywords as closely as possible (Figure 1).
Further, an analysis of the expression levels of selected genes was performed at the early stages of the development of PD in the brain tissues and peripheral blood of mice using MPTP-induced models of the early stages of the disease [43]. Earlier, we obtained data on the expression of the Snca and Drd2 genes in the brain tissues and peripheral blood using the AdvPSS and ESS models of PD [40]. This work presents data from previous [40] and current studies for building a holistic picture of the genes involved in this disease. The results of the expression analysis of the candidate genes are presented in Table 3.
The data presented in Table 3 demonstrate statistically significant changes in the expression of all the genes studied. However, the mRNA levels of Drd2 in the peripheral blood of mice in the 6 h-PSS and 24 h-PSS models of PD were under the detection level of the methods used.
Fewer genes changed their expression levels in the tissues of animals with the 6 h-PSS and 24 h-PSS models than in those using the models of the more advanced stages of PD (12 versus 19).
At the earliest stage modeled (6 h-PSS), there were changes in gene expression only in the substantia nigra and peripheral blood, whereas there were increases in the relative mRNA levels for all genes; the relative expression levels of Anxa2 and Drd2 increased in the substantia nigra while Snca, Rab5a, and Anxa2 transcript levels changed in the peripheral blood.
There was a change in the relative mRNA level of Anxa2 in all studied tissues at 24 h after MPTP administration. Moreover, a large increase in mRNA level—almost 7.5-fold—was observed in the striatum. There was a significant decrease in the relative mRNA level of Drd2 in the substantia nigra and an increase in the striatum by more than 1.5-fold. A change in expression at the mRNA level was also detected for Nsf in the striatum and peripheral blood.
There were significant changes in gene expression in the AdvPSS model of PD. There was an abrupt decrease in the relative mRNA level of Anxa2 by 3.5-fold and a 2.5-fold increase for Rab5a in the substantia nigra. Decreased expression levels of Rab5a mRNA were also detected in the striatum and peripheral blood.
For the ESS model of PD, we found increases in the relative mRNA levels of Nsf and Rab5a in the substantia nigra. In addition, there was an abrupt 9-fold decrease for Nsf and an almost 2-fold increase for Anxa2 in the striatum.
It is worth noting that the maximum number of significant data points on relative mRNA levels was obtained for Drd2 and Anxa2, with 7 of 12 analyzed samples for each.

4. Discussion

Proteins encoded by the human genes of the monogenic forms of PD, SNCA, VPS35, LRRK2, and ATP13A2 serve to maintain normal neuronal transport functions [36,51,52,53,54,55,56]. Mutations in these genes can contribute to the disruption of endo- and exocytosis and intracellular transport, which results in the incorrect redistribution of neurotransmitters, primarily dopamine (DA), in the synaptic gap and presynaptic terminals. These abnormalities can lead to impaired transport and neuronal death. However, despite the significance of these data [4], the entire spectrum of abnormal transport processes during the development of PD has not been fully described, and little is known about the roles of other genes involved in transport in the development of this disease.
The whole-transcriptome analysis of mouse brain tissues [40,41] and subsequent analysis of these data using Pathway Studio revealed potential significant participants in the transport processes in the early stages of the pathogenesis of PD (Figure 1). Therefore, the aim of this work was to study the expression levels of Snca, Drd2, Rab5a, Nsf, and Anxa2 in the brain tissues and peripheral blood of mice containing MPTP-induced models of PD.
In the present study, we used MPTP-induced models of the presymptomatic stages of PD, 6 h-PSS and 24 h-PSS, and of the advanced presymptomatic (AdvPSS) and early symptomatic (ESS) stages of PD (with decapitation 2 weeks after MPTP administration) [42,43]. The administration of MPTP makes it possible to simulate the death of substantia nigra DAergic neurons, DA deficiency in the striatum, and the manifestation of the classic signs of PD [57,58]. PD models based on MPTP injections are considered the most adequate experimental models of this disease. An important advantage of using MPTP is that it selectively penetrates into DAergic neurons due to its high affinity for the DA transporter DAT and selectively inhibits complex I of the mitochondrial electron transport chain, leading to oxidative stress and impaired calcium homeostasis. These events lead to the degeneration of DAergic neurons through necrosis or apoptosis [59,60,61]. The introduction of MPTP makes it possible to simulate the various stages of neurodegeneration in PD, including the earliest ones. This makes it possible to study the specific patterns and mechanisms underlying neurodegeneration in PD at different stages of the development of the pathological process as well as study the compensatory mechanisms that occur in nerve cells. Today, MPTP is the gold standard for the study of Parkinson’s disease, including at the earliest stages [62].
Other types of models have a number of their own features. In addition to MPTP, one of the most common toxins is 6-hydroxydopamine (6-OHDA). The introduction of this toxin promotes the degeneration of catecholamine neurons, including DAergic neurons, since 6-OHDA has a high affinity for DA transporters. The neurotoxic effect of 6-OHDA is mediated by a two-stage mechanism: at the first stage, the toxin accumulates in catecholamic neurons; at the second stage, a change in cellular homeostasis and neuron death occurs. Because of its similarity to endogenous catecholamines, 6-OHDA is taken up by the DAT and NAT transporters. The oxidation of 6-OHDA by monoamine oxidase (MAO-A) promotes the formation of hydrogen peroxide (H2O2), which is not only cytotoxic but also causes the formation of oxygen radicals [63]. An increase in the concentration of reactive oxygen species and other reactive substances caused by the entry of 6-OHDA into the cell leads to a rapid depletion of intracellular antioxidants, which, in turn, enhances neurotoxicity, leading to cell death [64].
A significant disadvantage of 6-OHDA is its inability to penetrate the BBB, which leads to its direct administration by stereotaxic method into the striatum and/or substantia nigra and less often into other parts of the brain [65,66,67,68]. Moreover, when using 6-OHDA, it is rather problematic to model the presymptomatic stages of PD since the very nature of toxin administration causes the rapid death of DA neurons [65,69].
Genetic models can mainly show only that variant of the pathogenesis of PD caused by a specific genetic defect. In addition, genetic models do not clearly reproduce the behavioral and neurodegenerative changes characteristic of PD. In addition, familial forms of PD account for only 5–15% of all cases of the disease, so this type of model cannot cover the entire spectrum of the causes of PD. Another disadvantage of such models is that they practically do not reproduce the presymptomatic stages of PD [70,71,72,73,74,75].
There are also models employing the use of amphetamine psychostimulants and reserpine. Currently, they are used very rarely to model the presymptomatic stages of PD due to their high nonspecificity. The principle of action is very different from the compounds described above and is associated with the release of DA from the terminals of neurons into the cytosol, leading to a lack of DA in the striatum [76,77,78,79]. These models do not show clear symptoms of parkinsonism and can be used to study the dysfunction of the catecholaminergic system as a whole.
In this work, the tissues of the striatum, substantia nigra, and peripheral blood of mice with MPTP-induced PD models were used. The striatum and substantia nigra are mainly subject to neurodegeneration in PD in the earliest stages. Blood was examined to further explore candidate genes as potential transcriptional peripheral biomarkers for this disease.
Table 3 demonstrates a greater number of significant changes in the tissues of the brain and peripheral blood of mice with models of AdvPSS and ESS of PD compared with those with 6 h-PSS and 24 h-PSS models. We observed more pronounced changes at the transcriptome level at the advanced stages of the development of the pathological process in PD. These results correlate with our previous data that showed that the number of differentially expressed genes increased with the progression of the disease [41,44].
In our previous study, we obtained data on the expression of Snca and Drd2 in the brain tissues and peripheral blood of animals using AdvPSS and ESS models [40] (Table 3). Mutations in the human DRD2 gene encoding the DA uptake receptor and release inhibitor D2 might be associated with some psychiatric and neurological diseases [80]. Alpha-synuclein (SNCA), encoded by the SNCA gene, is a presynaptic protein and is actively involved in the functioning of various types of neurons [81]. It is worth noting that SNCA can be involved in the development of both familial and sporadic forms of PD [82]. Alieva et al. (2017) demonstrated that these genes changed their expression levels in the brain tissues of mice with MPTP-induced models of AdvPSS and ESS, and the most pronounced and significant changes were observed in the substantia nigra [40]. These data indicated the involvement of the Snca and Drd2 genes in the neurodegenerative processes of PD. At the same time, an analysis of earlier time points (6 h-PSS and 24 h-PSS models of PD) suggested that changes in the Snca mRNA levels did not occur at the early stages of neurodegeneration. Here, we observed an increase in the relative level of Drd2 mRNA in the striatum and a decrease in the substantia nigra 24 h after the last injection of MPTP. Thus, this gene might be involved in pathogenesis at the very early stages of PD.
There were significant changes in the expression of Nsf mRNA in the striatum and substantia nigra in the PD models studied. Nsf, encoding the N-ethylmaleimide-sensitive factor, is required at all stages of the endosome pathway, from the endoplasmic reticulum to the Golgi apparatus and from the Golgi apparatus to the plasma membrane [83]. Several studies have demonstrated that the dysfunction of this protein can lead to the disruption of intracellular transport, which includes not only a decrease in vesicular traffic but also damage to the cytoskeleton [84,85,86]. An increase in the expression of Nsf mRNA in the substantia nigra in the ESS model of PD indicates that, at this stage of neurodegeneration, the mechanisms of intracellular transport are enhanced precisely in the nerve cell body. However, there are insufficient resources to support these processes in nerve cell bodies and neuronal outgrowths. This is reflected in a decrease in the Nsf mRNA levels in the striatum at the same simulated stage.
Rab5a, encoded by a member of the RAS oncogene family, as well as Nsf, is required for the fusion of endosomes with the plasma membrane [87,88]. This protein is involved in the conversion of early endosomes to late ones [89]. In 2004, Delprato et al. showed that motor neurons degenerate during Rab5a dysfunction [90]. The Rab5a gene showed increased expression in the substantia nigra in the AdvPSS and ESS models of PD. Moreover, the mRNA level of this gene decreased only in the striatum in the AdvPSS model but increased in the control samples in the ESS model. It is likely that an increase in the mRNA level of this gene at these stages of neurodegeneration in the substantia nigra might lead to the development of the compensatory mechanisms observed in the striatum in the ESS model.
In the present study, Anxa2 showed the maximum number of significant results at the mRNA level (Table 3). The protein annexin A2 encoded by this gene performs a number of biological functions such as the regulation of the organization of organelle membranes [91], participation in clathrin-mediated endocytosis, and other types of vesicular transport [92]. Annexin A2 is also involved in the formation of the actin cytoskeleton [93]. Babbin et al. (2007) showed that inhibition of Anxa2 expression in cell cultures can lead to the destabilization of actin filaments and their subsequent depolymerization [94].
Although reports on the dysfunction of annexin A2 appear primarily in the field of cancer research, a role for this protein in diseases of the nervous system, such as Alzheimer’s disease [95,96] and Niemann–Pick type C disease, has also been demonstrated [97]. However, there is very little evidence that the Anxa2 gene is directly involved in the pathogenesis of PD. It was shown that astrocytes expressing pathogenic Lrrk2 showed a significant decrease in the expression of Anxa2 at the protein level and a subsequent decrease in intracellular α-synuclein [98].
To maintain proper synaptic functioning, a neuron needs to concentrate all its resources precisely in the near-synaptic region, including the enhanced expression of proteins involved in vesicular transport. Our data support this assumption (Table 3 and Figure 2). We observed a significant increase in the mRNA level of the Anxa2 gene in the substantia nigra at the earliest modeled stage (6 h-PSS). In this case, it is likely that the cells were trying to transport RNA to the neuronal terminal, which is reflected in the striatum in the 24 h-PSS model of PD. This indicates that neurons are trying to compensate for the lack of resources caused by the development of neurodegeneration.
The study of gene expression in the blood opens up broad prospects in the search for genes that change expression at different stages of the disease, which will make it possible to better understand the mechanisms of PD pathogenesis. Blood cells, in particular lymphocytes, have some characteristics typical of a dopaminergic neuron: they contain both enzymes for dopamine synthesis, a dopamine membrane transporter, and a number of dopamine receptors are expressed. It has been assumed that the changes found in the peripheral blood of patients with PD might reflect the processes occurring in the DAergic neurons of the substantia nigra [99,100,101] and that blood can be taken for analysis to further study candidate genes as transcriptional peripheral markers of this disease. In our work, we also analyzed the changes in gene expression in the peripheral blood of mice with MPTP-induced PD. Previously, a correlation of changes in the relative levels of the Snca and Drd2 mRNAs was revealed in the substantia nigra and peripheral blood [42]. Our work also revealed statistically significant changes in the peripheral blood for all the studied genes. At the same time, one can see a trend toward a decrease in the mRNA levels in the models of the more advanced stages of PD. Such changes may reflect an increase in the neurodegenerative process through a decrease in the expression of genes involved in cell transport processes.

5. Conclusions

Here, we studied changes in the expression of genes whose protein products are involved in transport processes (Snca, Drd2, Rab5a, Anxa2, and Nsf) in the brain tissues and peripheral blood of mice with MPTP-induced models of PD. We detected changes in the expressions of Drd2, Anxa2, and Nsf at the earliest modeling stages. These findings suggest that changes in transport already occur in the early stages of neurodegeneration.
Moreover, we are the first to report conspicuous changes in the expression levels of Anxa2 in the striatum and substantia nigra of mice with MPTP-induced models of the early stages of PD. These data clearly suggest the involvement of this gene in the compensatory mechanisms in the early stages of the pathogenesis of PD.

Author Contributions

Conceptualization, M.M.R., A.K.A. and M.I.S.; methodology, M.M.R., E.V.F., I.N.R. and M.S.N.; software, M.M.R., A.K.A. and I.N.R.; validation, M.S.N., D.A.A., R.A.A. and A.A.K.; formal analysis, M.M.R., E.V.F., M.S.N., D.A.A., R.A.A. and A.A.K.; investigation, M.M.R., A.K.A., and M.I.S.; resources, M.I.S., P.A.S. and M.V.U.; data curation, A.K.A. and M.I.S.; writing—original draft preparation, M.M.R.; writing—review and editing, A.K.A., M.I.S. and P.A.S.; visualization, M.M.R.; supervision, A.K.A., M.I.S. and P.A.S.; project administration, A.K.A., M.I.S. and P.A.S.; funding acquisition, M.I.S. and P.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Russian Science Foundation (RSF) (project no. 20-15-00262). The work of Kolacheva A.A., Ugrumov M.V. was conducted under the IDB RAS Government basic research program in 2022 No. 0088-2021-0018. This work of Petr Slominsky was supported by the Ministry of Science and Higher Education of the Russian Federation, agreement 075-15-2021-1357.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the Research Center of Neurology (protocol 18/4, 3 March 2021) and the Ethics Committee of the Institute of Molecular Genetics of National Research Centre “Kurchatov Institute” (protocol 3/21, 17 February 2021).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was carried out on the basis of the center for collective use of IMG RAS “Center of Cell and Gene Technologies”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wilson, D.B. Cellular transport mechanisms. Annu. Rev. Biochem. 1978, 47, 933–965. [Google Scholar] [CrossRef] [PubMed]
  2. Berger, F.; Keller, C.; Muller, M.J.; Klumpp, S.; Lipowsky, R. Co-operative transport by molecular motors. Biochem. Soc. Trans. 2011, 39, 1211–1215. [Google Scholar] [CrossRef] [PubMed]
  3. Benarroch, E.E. Membrane trafficking and transport: Overview and neurologic implications. Neurology 2012, 79, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  4. Abeliovich, A.; Gitler, A.D. Defects in trafficking bridge Parkinson’s disease pathology and genetics. Nature 2016, 539, 207–216. [Google Scholar] [CrossRef] [PubMed]
  5. Goldstein, L.S.; Yang, Z. Microtubule-based transport systems in neurons: The roles of kinesins and dyneins. Annu. Rev. Neurosci. 2000, 23, 39–71. [Google Scholar] [CrossRef]
  6. Maeder, C.I.; Shen, K.; Hoogenraad, C.C. Axon and dendritic trafficking. Curr. Opin. Neurobiol. 2014, 27, 165–170. [Google Scholar] [CrossRef]
  7. Bridgman, P.C. Myosin-dependent transport in neurons. J. Neurobiol. 2004, 58, 164–174. [Google Scholar] [CrossRef]
  8. Isola, A.L.; Chen, S. Exosomes: The Messengers of Health and Disease. Curr. Neuropharmacol. 2017, 15, 157–165. [Google Scholar] [CrossRef]
  9. Rizo, J.; Sudhof, T.C. The membrane fusion enigma: SNAREs, Sec1/Munc18 proteins, and their accomplices—Guilty as charged? Annu. Rev. Cell Dev. Biol. 2012, 28, 279–308. [Google Scholar] [CrossRef]
  10. Kononenko, N.L.; Haucke, V. Molecular mechanisms of presynaptic membrane retrieval and synaptic vesicle reformation. Neuron 2015, 85, 484–496. [Google Scholar] [CrossRef]
  11. Gondre-Lewis, M.C.; Park, J.J.; Loh, Y.P. Cellular mechanisms for the biogenesis and transport of synaptic and dense-core vesicles. Int. Rev. Cell Mol. Biol. 2012, 299, 27–115. [Google Scholar] [CrossRef] [PubMed]
  12. Grafstein, B.; Forman, D.S. Intracellular transport in neurons. Physiol. Rev. 1980, 60, 1167–1283. [Google Scholar] [CrossRef] [PubMed]
  13. Hannah, M.J.; Schmidt, A.A.; Huttner, W.B. Synaptic vesicle biogenesis. Annu. Rev. Cell Dev. Biol. 1999, 15, 733–798. [Google Scholar] [CrossRef] [PubMed]
  14. Eccles, J.C. The Mechanism of Synaptic Transmission. In Ergebnisse der Physiologie Biologischen Chemie und Experimentellen Pharmakologie; Kramer, K., Krayer, O., Lehnartz, E., Muralt, A.V., Weber, H.H., Eds.; Springer: Berlin/Heidelberg, Germany, 1961; pp. 299–430. [Google Scholar]
  15. Huettner, J.E. Kainate receptors and synaptic transmission. Prog. Neurobiol. 2003, 70, 387–407. [Google Scholar] [CrossRef]
  16. Katz, B.; Miledi, R. A study of synaptic transmission in the absence of nerve impulses. J. Physiol. 1967, 192, 407–436. [Google Scholar] [CrossRef]
  17. Bronfman, F.C.; Escudero, C.A.; Weis, J.; Kruttgen, A. Endosomal transport of neurotrophins: Roles in signaling and neurodegenerative diseases. Dev. Neurobiol. 2007, 67, 1183–1203. [Google Scholar] [CrossRef]
  18. Halim, N.D.; Weickert, C.S.; McClintock, B.W.; Hyde, T.M.; Weinberger, D.R.; Kleinman, J.E.; Lipska, B.K. Presynaptic proteins in the prefrontal cortex of patients with schizophrenia and rats with abnormal prefrontal development. Mol. Psychiatry 2003, 8, 797–810. [Google Scholar] [CrossRef]
  19. Gabriel, S.M.; Davidson, M.; Haroutunian, V.; Powchik, P.; Bierer, L.M.; Purohit, D.P.; Perl, D.P.; Davis, K.L. Neuropeptide deficits in schizophrenia vs. Alzheimer’s disease cerebral cortex. Biol. Psychiatry 1996, 39, 82–91. [Google Scholar] [CrossRef]
  20. Shankar, G.M.; Walsh, D.M. Alzheimer′s disease: Synaptic dysfunction and Abeta. Mol. Neurodegener. 2009, 4, 48. [Google Scholar] [CrossRef]
  21. Waites, C.L.; Garner, C.C. Presynaptic function in health and disease. Trends Neurosci. 2011, 34, 326–337. [Google Scholar] [CrossRef]
  22. Willis, M.; Leitner, I.; Jellinger, K.A.; Marksteiner, J. Chromogranin peptides in brain diseases. J. Neural Transm. 2011, 118, 727–735. [Google Scholar] [CrossRef] [PubMed]
  23. Lauterborn, J.C.; Rex, C.S.; Kramar, E.; Chen, L.Y.; Pandyarajan, V.; Lynch, G.; Gall, C.M. Brain-derived neurotrophic factor rescues synaptic plasticity in a mouse model of fragile X syndrome. J. Neurosci. Off. J. Soc. Neurosci. 2007, 27, 10685–10694. [Google Scholar] [CrossRef] [PubMed]
  24. Annangudi, S.P.; Luszpak, A.E.; Kim, S.H.; Ren, S.; Hatcher, N.G.; Weiler, I.J.; Thornley, K.T.; Kile, B.M.; Wightman, R.M.; Greenough, W.T.; et al. Neuropeptide Release is Impaired in a Mouse Model of Fragile X Mental Retardation Syndrome. ACS Chem. Neurosci. 2010, 1, 306–314. [Google Scholar] [CrossRef] [PubMed]
  25. Verhoeven, K.; De Jonghe, P.; Coen, K.; Verpoorten, N.; Auer-Grumbach, M.; Kwon, J.M.; FitzPatrick, D.; Schmedding, E.; De Vriendt, E.; Jacobs, A.; et al. Mutations in the small GTP-ase late endosomal protein RAB7 cause Charcot-Marie-Tooth type 2B neuropathy. Am. J. Hum. Genet. 2003, 72, 722–727. [Google Scholar] [CrossRef] [PubMed]
  26. Henderson, L.P.; Lin, L.; Prasad, A.; Paul, C.A.; Chang, T.Y.; Maue, R.A. Embryonic striatal neurons from niemann-pick type C mice exhibit defects in cholesterol metabolism and neurotrophin responsiveness. J. Biol. Chem. 2000, 275, 20179–20187. [Google Scholar] [CrossRef] [PubMed]
  27. Chevalier-Larsen, E.; Holzbaur, E.L. Axonal transport and neurodegenerative disease. Biochim. Biophys. Acta 2006, 1762, 1094–1108. [Google Scholar] [CrossRef]
  28. Volpicelli-Daley, L.A. Effects of alpha-synuclein on axonal transport. Neurobiol. Dis. 2017, 105, 321–327. [Google Scholar] [CrossRef]
  29. Bossers, K.; Meerhoff, G.; Balesar, R.; Van Dongen, J.W.; Kruse, C.G.; Swaab, D.F.; Verhaagen, J. Analysis of Gene Expression in Parkinson’s Disease: Possible Involvement of Neurotrophic Support and Axon Guidance in Dopaminergic Cell Death. Brain Pathol. 2009, 19, 91–107. [Google Scholar] [CrossRef]
  30. Hauser, M.A.; Li, Y.-J.; Xu, H.; Noureddine, M.A.; Shao, Y.S.; Gullans, S.R.; Scherzer, C.R.; Jensen, R.V.; McLaurin, A.C.; Gibson, J.R.; et al. Expression Profiling of Substantia Nigra in Parkinson Disease, Progressive Supranuclear Palsy, and Frontotemporal Dementia With Parkinsonism. Arch Neurol. 2005, 62, 917–921. [Google Scholar] [CrossRef]
  31. Miller, R.M.; Kiser, G.L.; Kaysser-Kranich, T.M.; Lockner, R.J.; Palaniappan, C.; Federoff, H.J. Robust dysregulation of gene expression in substantia nigra and striatum in Parkinson’s disease. Neurobiol. Dis. 2006, 21, 305–313. [Google Scholar] [CrossRef]
  32. Bieri, G.; Gitler, A.D.; Brahic, M. Internalization, axonal transport and release of fibrillar forms of alpha-synuclein. Neurobiol. Dis. 2018, 109, 219–225. [Google Scholar] [CrossRef] [PubMed]
  33. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
  34. Yamamoto, S.; Fukae, J.; Mori, H.; Mizuno, Y.; Hattori, N. Positive immunoreactivity for vesicular monoamine transporter 2 in Lewy bodies and Lewy neurites in substantia nigra. Neurosci. Lett. 2006, 396, 187–191. [Google Scholar] [CrossRef] [PubMed]
  35. Power, J.H.; Barnes, O.L.; Chegini, F. Lewy Bodies and the Mechanisms of Neuronal Cell Death in Parkinson’s Disease and Dementia with Lewy Bodies. Brain Pathol. 2017, 27, 3–12. [Google Scholar] [CrossRef]
  36. Lin, M.K.; Farrer, M.J. Genetics and genomics of Parkinson’s disease. Genome Med. 2014, 6, 48. [Google Scholar] [CrossRef]
  37. Tsika, E.; Glauser, L.; Moser, R.; Fiser, A.; Daniel, G.; Sheerin, U.M.; Lees, A.; Troncoso, J.C.; Lewis, P.A.; Bandopadhyay, R.; et al. Parkinson’s disease-linked mutations in VPS35 induce dopaminergic neurodegeneration. Hum. Mol. Genet. 2014, 23, 4621–4638. [Google Scholar] [CrossRef]
  38. Alter, S.P.; Lenzi, G.M.; Bernstein, A.I.; Miller, G.W. Vesicular integrity in Parkinson’s disease. Curr. Neurol. Neurosci. Rep. 2013, 13, 362. [Google Scholar] [CrossRef]
  39. Mukherjee, A.; Biswas, A.; Das, S.K. Gut dysfunction in Parkinson’s disease. World J. Gastroenterol. 2016, 22, 5742–5752. [Google Scholar] [CrossRef]
  40. Alieva, A.K.; Filatova, E.V.; Kolacheva, A.A.; Rudenok, M.M.; Slominsky, P.A.; Ugrumov, M.V.; Shadrina, M.I. Transcriptome Profile Changes in Mice with MPTP-Induced Early Stages of Parkinson’s Disease. Mol. Neurobiol. 2017, 54, 6775–6784. [Google Scholar] [CrossRef]
  41. Alieva, A.K.; Zyrin, V.S.; Rudenok, M.M.; Kolacheva, A.A.; Shulskaya, M.V.; Ugryumov, M.V.; Slominsky, P.A.; Shadrina, M.I. Whole-Transcriptome Analysis of Mouse Models with MPTP-Induced Early Stages of Parkinson’s Disease Reveals Stage-Specific Response of Transcriptome and a Possible Role of Myelin-Linked Genes in Neurodegeneration. Mol. Neurobiol. 2018, 55, 7229–7241. [Google Scholar] [CrossRef]
  42. Ugrumov, M.V.; Khaindrava, V.G.; Kozina, E.A.; Kucheryanu, V.G.; Bocharov, E.V.; Kryzhanovsky, G.N.; Kudrin, V.S.; Narkevich, V.B.; Klodt, P.M.; Rayevsky, K.S.; et al. Modeling of presymptomatic and symptomatic stages of parkinsonism in mice. Neuroscience 2011, 181, 175–188. [Google Scholar] [CrossRef] [PubMed]
  43. Kolacheva, A.A.; Kozina, E.A.; Volina, E.V.; Ugryumov, M.V. Time course of degeneration of dopaminergic neurons and respective compensatory processes in the nigrostriatal system in mice. Dokl. Biol.Sci. Proc. Acad. Sci. USSR Biol. Sci. Sect. Transl. Russ. 2014, 456, 160–164. [Google Scholar] [CrossRef] [PubMed]
  44. Rudenok, M.M.; Alieva, A.K.; Starovatykh, J.S.; Nesterov, M.S.; Stanishevskaya, V.A.; Kolacheva, A.A.; Ugryumov, M.V.; Slominsky, P.A.; Shadrina, M.I. Expression analysis of genes involved in mitochondrial biogenesis in mice with MPTP-induced model of Parkinson’s disease. Mol. Genet. Metab. Rep. 2020, 23, 100584. [Google Scholar] [CrossRef] [PubMed]
  45. Chang, Y.C.; Ding, Y.; Dong, L.; Zhu, L.J.; Jensen, R.V.; Hsiao, L.L. Differential expression patterns of housekeeping genes increase diagnostic and prognostic value in lung cancer. PeerJ 2018, 6, e4719. [Google Scholar] [CrossRef]
  46. Haslinger, D.; Waltes, R.; Yousaf, A.; Lindlar, S.; Schneider, I.; Lim, C.K.; Tsai, M.M.; Garvalov, B.K.; Acker-Palmer, A.; Krezdorn, N.; et al. Loss of the Chr16p11.2 ASD candidate gene QPRT leads to aberrant neuronal differentiation in the SH-SY5Y neuronal cell model. Mol. Autism 2018, 9, 56. [Google Scholar] [CrossRef]
  47. Hoerndli, F.J.; Toigo, M.; Schild, A.; Gotz, J.; Day, P.J. Reference genes identified in SH-SY5Y cells using custom-made gene arrays with validation by quantitative polymerase chain reaction. Anal. Biochem. 2004, 335, 30–41. [Google Scholar] [CrossRef]
  48. Warrington, J.A.; Nair, A.; Mahadevappa, M.; Tsyganskaya, M. Comparison of human adult and fetal expression and identification of 535 housekeeping/maintenance genes. Physiol. Genom. 2000, 2, 143–147. [Google Scholar] [CrossRef]
  49. Hettne, K.M.; Thompson, M.; van Haagen, H.H.; van der Horst, E.; Kaliyaperumal, R.; Mina, E.; Tatum, Z.; Laros, J.F.; van Mulligen, E.M.; Schuemie, M.; et al. The Implicitome: A Resource for Rationalizing Gene-Disease Associations. PLoS ONE 2016, 11, e0149621. [Google Scholar] [CrossRef]
  50. Zoetmulder, M.; Biernat, H.B.; Nikolic, M.; Korbo, L.; Friberg, L.; Jennum, P.J. Prepulse inhibition is associated with attention, processing speed, and 123I-FP-CIT SPECT in Parkinson’s disease. J. Parkinson’s Dis. 2014, 4, 77–87. [Google Scholar] [CrossRef]
  51. Brunger, A.T. Structural insights into the molecular mechanism of Ca(2+)-dependent exocytosis. Curr. Opin. Neurobiol. 2000, 10, 293–302. [Google Scholar] [CrossRef]
  52. Steger, M.; Tonelli, F.; Ito, G.; Davies, P.; Trost, M.; Vetter, M.; Wachter, S.; Lorentzen, E.; Duddy, G.; Wilson, S.; et al. Phosphoproteomics reveals that Parkinson′s disease kinase LRRK2 regulates a subset of Rab GTPases. eLife 2016, 5, e12813. [Google Scholar] [CrossRef] [PubMed]
  53. Inoshita, T.; Arano, T.; Hosaka, Y.; Meng, H.; Umezaki, Y.; Kosugi, S.; Morimoto, T.; Koike, M.; Chang, H.Y.; Imai, Y.; et al. Vps35 in cooperation with LRRK2 regulates synaptic vesicle endocytosis through the endosomal pathway in Drosophila. Hum. Mol. Genet. 2017, 26, 2933–2948. [Google Scholar] [CrossRef]
  54. Esteves, A.R.; Swerdlow, R.H.; Cardoso, S.M. LRRK2, a puzzling protein: Insights into Parkinson′s disease pathogenesis. Exp. Neurol. 2014, 261, 206–216. [Google Scholar] [CrossRef] [PubMed]
  55. Demirsoy, S.; Martin, S.; Motamedi, S.; van Veen, S.; Holemans, T.; Van den Haute, C.; Jordanova, A.; Baekelandt, V.; Vangheluwe, P.; Agostinis, P. ATP13A2/PARK9 regulates endo-/lysosomal cargo sorting and proteostasis through a novel PI(3, 5)P2-mediated scaffolding function. Hum. Mol. Genet. 2017, 26, 1656–1669. [Google Scholar] [CrossRef] [PubMed]
  56. Park, J.S.; Blair, N.F.; Sue, C.M. The role of ATP13A2 in Parkinson’s disease: Clinical phenotypes and molecular mechanisms. Mov. Disord. 2015, 30, 770–779. [Google Scholar] [CrossRef] [PubMed]
  57. German, D.C.; Nelson, E.L.; Liang, C.L.; Speciale, S.G.; Sinton, C.M.; Sonsalla, P.K. The neurotoxin MPTP causes degeneration of specific nucleus A8, A9 and A10 dopaminergic neurons in the mouse. Neurodegeneration 1996, 5, 299–312. [Google Scholar] [CrossRef]
  58. Meredith, G.E.; Totterdell, S.; Potashkin, J.A.; Surmeier, D.J. Modeling PD pathogenesis in mice: Advantages of a chronic MPTP protocol. Parkinsonism. Relat. Disord. 2008, 14 (Suppl. 2), S112–S115. [Google Scholar] [CrossRef]
  59. Gerlach, M.; Riederer, P. Animal models of Parkinson′s disease: An empirical comparison with the phenomenology of the disease in man. J. Neural. Transm. 1996, 103, 987–1041. [Google Scholar] [CrossRef]
  60. Langston, J.W.; Ballard, P.; Tetrud, J.W.; Irwin, I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science 1983, 219, 979–980. [Google Scholar] [CrossRef]
  61. Przedborski, S.; Jackson-Lewis, V.; Djaldetti, R.; Liberatore, G.; Vila, M.; Vukosavic, S.; Almer, G. The parkinsonian toxin MPTP: Action and mechanism. Restor. Neurol. Neurosci. 2000, 16, 135–142. [Google Scholar]
  62. Salari, S.; Bagheri, M. In vivo, in vitro and pharmacologic models of Parkinson’s disease. Physiol. Res. 2019, 68, 17–24. [Google Scholar] [CrossRef] [PubMed]
  63. Cohen, G.; Heikkila, R.E.; Allis, B.; Cabbat, F.; Dembiec, D.; MacNamee, D.; Mytilineou, C.; Winston, B. Destruction of sympathetic nerve terminals by 6-hydroxydopamine: Protection by 1-phenyl-3-(2-thiazolyl)-2-thiourea, diethyldithiocarbamate, methimazole, cysteamine, ethanol and n-butanol. J. Pharmacol. Exp. Ther. 1976, 199, 336–352. [Google Scholar]
  64. Blum, D.; Torch, S.; Lambeng, N.; Nissou, M.; Benabid, A.L.; Sadoul, R.; Verna, J.M. Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: Contribution to the apoptotic theory in Parkinson′s disease. Prog. Neurobiol. 2001, 65, 135–172. [Google Scholar] [CrossRef]
  65. Deumens, R.; Blokland, A.; Prickaerts, J. Modeling Parkinson’s Disease in Rats: An Evaluation of 6-OHDA Lesions of the Nigrostriatal Pathway. Exp. Neurol. 2002, 175, 303–317. [Google Scholar] [CrossRef] [PubMed]
  66. Lorigados, P.L.; Fuentes, N.P.; Alvarez, G.L.; McRae, A.; Serrano, S.T.; Blanco, L.L.; Macías, G.R. Nerve growth factor levels in Parkinson disease and experimental parkinsonian rats. Brain Res. 2002, 952, 122–127. [Google Scholar] [CrossRef]
  67. Dimatelis, J.J.; Hendricks, S.; Hsieh, J.; Vlok, N.M.; Bugarith, K.; Daniels, W.M.; Russell, V.A. Exercise partly reverses the effect of maternal separation on hippocampal proteins in 6-hydroxydopamine-lesioned rat brain. Exp. Physiol. 2013, 98, 233–244. [Google Scholar] [CrossRef] [PubMed]
  68. Sun, W.; Sugiyama, K.; Asakawa, T.; Yamaguchi, H.; Akamine, S.; Ouchi, Y.; Magata, Y.; Namba, H. Dynamic changes of striatal dopamine D2 receptor binding at later stages after unilateral lesions of the medial forebrain bundle in Parkinsonian rat models. Neurosci. Lett. 2011, 496, 157–162. [Google Scholar] [CrossRef]
  69. Carman, L.S.; Gage, F.H.; Shults, C.W. Partial lesion of the substantia nigra relation between extent of lesion. Brain Res. 1991, 553, 275–283. [Google Scholar] [CrossRef]
  70. Gonzalez, S.; Mena, M.A.; Lastres-Becker, I.; Serrano, A.; de Yebenes, J.G.; Ramos, J.A.; Fernandez-Ruiz, J. Cannabinoid CB(1) receptors in the basal ganglia and motor response to activation or blockade of these receptors in parkin-null mice. Brain Res. 2005, 1046, 195–206. [Google Scholar] [CrossRef]
  71. Rial, D.; Castro, A.A.; Machado, N.; Garcao, P.; Goncalves, F.Q.; Silva, H.B.; Tome, A.R.; Kofalvi, A.; Corti, O.; Raisman-Vozari, R.; et al. Behavioral phenotyping of Parkin-deficient mice: Looking for early preclinical features of Parkinson’s disease. PLoS ONE 2014, 9, e114216. [Google Scholar] [CrossRef]
  72. Kelm-Nelson, C.A.; Yang, K.M.; Ciucci, M.R. Exercise Effects on Early Vocal Ultrasonic Communication Dysfunction in a PINK1 Knockout Model of Parkinson’s Disease. J. Parkinson’s Dis. 2015, 5, 749–763. [Google Scholar] [CrossRef] [PubMed]
  73. Bishop, M.W.; Chakraborty, S.; Matthews, G.A.; Dougalis, A.; Wood, N.W.; Festenstein, R.; Ungless, M.A. Hyperexcitable substantia nigra dopamine neurons in PINK1- and HtrA2/Omi-deficient mice. J. Neurophysiol. 2010, 104, 3009–3020. [Google Scholar] [CrossRef] [PubMed]
  74. Rousseaux, M.W.; Qu, D.; Hewitt, S.J.; Seang, S.; Kim, R.H.; Slack, R.S.; Schlossmacher, M.G.; Lagace, D.C.; Mak, T.W.; Park, D.S. Progressive dopaminergic cell loss with unilateral-to-bilateral progression in a genetic model of Parkinson disease. Proc. Natl. Acad. Sci. USA 2012, 25, 15918–15923. [Google Scholar] [CrossRef] [PubMed]
  75. Yue, Z.; Lachenmayer, M.L. Genetic LRRK2 models of Parkinson’s disease: Dissecting the pathogenic pathway and exploring clinical applications. Mov. Disord. Off. J. Mov. Disord. Soc. 2011, 26, 1386–1397. [Google Scholar] [CrossRef] [PubMed]
  76. Bernheimer, H.; Birkmayer, W.; Hornykiewicz, O.; Jellinger, K.; Seitelberger, F. Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J. Neurol. Sci. 1973, 20, 415–455. [Google Scholar] [CrossRef]
  77. Betarbet, R.; Sherer, T.B.; Greenamyre, J.T. Animal models of Parkinson’s disease. BioEssays News Rev.Mol. Cell. Dev. Biol. 2002, 24, 308–318. [Google Scholar] [CrossRef]
  78. Fibiger, H.C.; Mogeer, E.G. Effect of acute and chronic methamphetamine treatment on tyrosine hydroxylase activity in brain and adrenal medulla. Eur. J. Pharmacol. 1971, 16, 176–180. [Google Scholar] [CrossRef]
  79. Sonsalla, P.K.; Nicklas, W.J.; Heikkila, R.E. Role for excitatory amino acids in methamphetamine-induced nigrostriatal dopaminergic toxicity. Science 1989, 243, 398–400. [Google Scholar] [CrossRef]
  80. Noble, E.P. D2 dopamine receptor gene in psychiatric and neurologic disorders and its phenotypes. Am. J. Med. Genet. Part B Neuropsychiatr.Genet. Off. Publ. Int. Soc. Psychiatr. Genet. 2003, 116b, 103–125. [Google Scholar] [CrossRef]
  81. Burre, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Sudhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef]
  82. Ozansoy, M.; Basak, A.N. The central theme of Parkinson’s disease: Alpha-synuclein. Mol. Neurobiol. 2013, 47, 460–465. [Google Scholar] [CrossRef] [PubMed]
  83. Whiteheart, S.W.; Schraw, T.; Matveeva, E.A. N-ethylmaleimide sensitive factor (NSF) structure and function. Int. Rev. Cytol. 2001, 207, 71–112. [Google Scholar] [CrossRef] [PubMed]
  84. Hayashi, M.; Taniguchi, S.; Ishizuka, Y.; Kim, H.S.; Wataya, Y.; Yamamoto, A.; Moriyama, Y. A homologue of N-ethylmaleimide-sensitive factor in the malaria parasite Plasmodium falciparum is exported and localized in vesicular structures in the cytoplasm of infected erythrocytes in the brefeldin A-sensitive pathway. J. Biol. Chem. 2001, 276, 15249–15255. [Google Scholar] [CrossRef] [PubMed]
  85. Nunes, P.; Haines, N.; Kuppuswamy, V.; Fleet, D.J.; Stewart, B.A. Synaptic vesicle mobility and presynaptic F-actin are disrupted in a N-ethylmaleimide-sensitive factor allele of Drosophila. Mol. Biol. Cell 2006, 17, 4709–4719. [Google Scholar] [CrossRef]
  86. Faugaret, D.; Chouinard, F.C.; Harbour, D.; El Azreq, M.-A.; Bourgoin, S.G. An essential role for phospholipase D in the recruitment of vesicle amine transport protein-1 to membranes in human neutrophils. Biochem. Pharm. 2011, 81, 144–156. [Google Scholar] [CrossRef]
  87. Woodman, P. Vesicle transport: More work for the Rabs? Curr. Biol. 1998, 8, R199–R201. [Google Scholar] [CrossRef]
  88. Tanmay, B.; Kumar, R.J. Rab proteins: The key regulators of intracellular vesicle transport. Exp. Cell Res. 2014, 328, 1–19. [Google Scholar] [CrossRef]
  89. Roth, B.L.; Willins, D.L.; Kroeze, W.K. G protein-coupled receptor (GPCR) trafficking in the central nervous system: Relevance for drugs of abuse. Drug Alcohol. Depend. 1998, 51, 73–85. [Google Scholar] [CrossRef]
  90. Delprato, A.; Merithew, E.; Lambright, D.G. Structure, exchange determinants, and family-wide rab specificity of the tandem helical bundle and Vps9 domains of Rabex-5. Cell 2004, 118, 607–617. [Google Scholar] [CrossRef]
  91. Luo, M.; Hajjar, K.A. Annexin A2 system in human biology: Cell surface and beyond. Semin. Thromb. Hemost. 2013, 39, 338–346. [Google Scholar] [CrossRef]
  92. Bharadwaj, A.; Bydoun, M.; Holloway, R.; Waisman, D. Annexin A2 heterotetramer: Structure and function. Int. J. Mol. Sci. 2013, 14, 6259–6305. [Google Scholar] [CrossRef] [PubMed]
  93. Filipenko, N.R.; Waisman, D.M. The C Terminus of Annexin II Mediates Binding to F-actin. J. Biol. Chem. 2001, 276, 5310–5315. [Google Scholar] [CrossRef] [PubMed]
  94. Babbin, B.A.; Parkos, C.A.; Mandell, K.J.; Winfree, L.M.; Laur, O.; Ivanov, A.I.; Nusrat, A. Annexin 2 regulates intestinal epithelial cell spreading and wound closure through Rho-related signaling. Am. J. Pathol. 2007, 170, 951–966. [Google Scholar] [CrossRef] [PubMed]
  95. Joanna, B.-P. Annexins in the Central Nervous System: Are they Neuroprotective or Proapoptotic Agents? Med. Chem. Rev. Online 2004, 1, 233–252. [Google Scholar] [CrossRef]
  96. Brandt, R.; Bakota, L. Microtubule dynamics and the neurodegenerative triad of Alzheimer′s disease: The hidden connection. J. Neurochem. 2017, 143, 409–417. [Google Scholar] [CrossRef] [PubMed]
  97. Domon, M.M.; Besson, F.; Bandorowicz-Pikula, J.; Pikula, S. Annexin A6 is recruited into lipid rafts of Niemann-Pick type C disease fibroblasts in a Ca2+-dependent manner. Biochem. Biophys. Res. Commun. 2011, 405, 192–196. [Google Scholar] [CrossRef]
  98. Streubel-Gallasch, L.; Giusti, V.; Sandre, M.; Tessari, I.; Plotegher, N.; Giusto, E.; Masato, A.; Iovino, L.; Battisti, I.; Arrigoni, G.; et al. Parkinson’s Disease-Associated LRRK2 Interferes with Astrocyte-Mediated Alpha-Synuclein Clearance. Mol. Neurobiol. 2021, 58, 3119–3140. [Google Scholar] [CrossRef]
  99. Barbanti, P.; Fabbrini, G.; Ricci, A.; Cerbo, R.; Bronzetti, E.; Caronti, B.; Calderaro, C.; Felici, L.; Stocchi, F.; Meco, G.; et al. Increased expression of dopamine receptors on lymphocytes in Parkinson’s disease. Mov. Disord. 1999, 14, 764–771. [Google Scholar] [CrossRef]
  100. Buttarelli, F.R.; Capriotti, G.; Pellicano, C.; Prosperi, D.; Circella, A.; Festa, A.; Giovannelli, M.; Tofani, A.; Pontieri, F.E.; Scopinaro, F. Central and peripheral dopamine transporter reduction in Parkinson′s disease. Neurol. Res. 2009, 31, 687–691. [Google Scholar] [CrossRef]
  101. Nagai, Y.; Ueno, S.; Saeki, Y.; Soga, F.; Hirano, M.; Yanagihara, T. Decrease of the D3 dopamine receptor mRNA expression in lymphocytes from patients with Parkinson’s disease. Neurology 1996, 46, 791–795. [Google Scholar] [CrossRef]
Figure 1. A network of genes that are somehow involved in the functioning of transport. Pathway Studio was used to build this network. Cellular processes are marked with a yellow box, genes and protein structures under study are marked with a red ellipse, disease (PD) and related processes are marked with a purple box, and a cell (neuron) is marked with light blue. Interactions between the gene and the object are indicated by arrows.
Figure 1. A network of genes that are somehow involved in the functioning of transport. Pathway Studio was used to build this network. Cellular processes are marked with a yellow box, genes and protein structures under study are marked with a red ellipse, disease (PD) and related processes are marked with a purple box, and a cell (neuron) is marked with light blue. Interactions between the gene and the object are indicated by arrows.
Life 12 00751 g001
Figure 2. Relative mRNA and protein levels of Anxa2 in striatum and substantia nigra of mice with MPTP-induced models of PD. The expression level studied in the control was taken as 1. Life 12 00751 i001 p-Value ≤ 0.05.
Figure 2. Relative mRNA and protein levels of Anxa2 in striatum and substantia nigra of mice with MPTP-induced models of PD. The expression level studied in the control was taken as 1. Life 12 00751 i001 p-Value ≤ 0.05.
Life 12 00751 g002
Table 1. Description of used models.
Table 1. Description of used models.
6 h-PSS24 h-PSSAdvPSSESS
Dose of MPTP12 mg/kg12 mg/kg12 mg/kg12 mg/kg
Number and frequency of injections4 injections at 2-h intervals4 injections at 2-h intervals2 injections at 2-h intervals4 injections at 2-h intervals
Withdrawal time after the last injection of MPTP6 h24 h2 weeks2 weeks
Change in DA levels in the striatum89% reduction72% increase56% reduction75% reduction
Death of DAergic
neurons in substantia nigra
33% reductionunchanged26% reduction43% reduction
Changes in animal
behavior
unchangedunchangedunchangeda decrease in motor activity was detected
6 h-PSS—model of the presymptomatic stage of PD with decapitation of mice 6 h after last injection of MPTP; 24 h-PSS—model of the presymptomatic stage of PD with decapitation of mice 24 h after last injection of MPTP; AdvPSS—model of the advanced presymptomatic stage of PD; ESS—early symptomatic stage of PD. MPTP—1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine.
Table 2. Nucleotide sequences of gene-specific primers and probes.
Table 2. Nucleotide sequences of gene-specific primers and probes.
GeneNucleotide Sequence
Snca
(Synuclein Alpha)
NM_001042451.2 *
Probe: 5′-VIC-CCTCCTCACCCTTGCCCATCTGGTCC-BHQ2-3′
Forward primer: 5′-AGTGGAGGGAGCTGGGAATATAG-3′
Reverse primer: 5′-GCATGTCTTCCAGGATTCCTTCC-3′
Drd2
(Dopamine Receptor D2)
NM_010077.2 *
Probe: 5′-ROX-CAGCCAGCAGATGATGAACACACCAAGA-BHQ2-3′
Forward primer: 5′-TCCCAGCAGAAGGAGAAGAAAG-3′
Reverse primer: 5′-TGTATATTCAGGATGTGCGTGATG-3′
Rab5a
(RAB5A, Member RAS Oncogene Family)
NM_025887.4 *
Probe: 5′-VIC-CATCTGCATAGGACTGTGCTTCC-BHQ2-3′
Forward primer: 5′-GCAAGTCCTAATATTGTGATA-3′
Reverse primer: 5′-CTCCATAAATAATAAGCTGTTG-3′
Anxa2
(Annexin A2)
NM_007585.3 *
Probe: 5′-VIC-AGAAGGACATCATCTCTGACACATCTG-BHQ2-3′
Forward primer: 5′-GAGTGTACAAGGAAATGTAC-3′
Reverse primer: 5′-CGTAGTCAATAACTGAGC-3′
Nsf
(N-ethylmaleimide sensitive fusion protein)
NM_008740.4 *
Probe: 5′-VIC-CGTCCTCACCATCACATGCTG-BHQ2-3′
Forward primer: 5′-CCCTACTGATGAATTATCTTTA-3′
Reverse primer: 5′-TCCTCAGCGTAAATATGTA-3′
Bcat2
(Branched chain aminotransferase 2)
NM_001243053.1 *
Probe: 5′-FAM-CGGATACACTCCAACAGCTCCTGCTTGT-BHQ1-3′
Forward primer: 5′-TCAACATGGACAGGATGCTACG-3′
Reverse primer: 5′-CCAGTCTTTGTCTACTTCAATGAGC-3′
Psmd7
(Proteasome 26S Subunit, Non-ATPase 7)
NM_010817.2 *
Probe: 5′-FAM-AGTCCTAGGTCCTTTGGCTTCACGTCGA-BHQ1-3′
Forward primer: 5′-CTGCACAAGAATGATATCGCCATC-3′
Reverse primer: 5′-CTCCACTGAGATGTAGGCTTCG-3′
* Numbers in the database GenBank (Accession numbers). FAM, ROX and VIC—fluorescent dyes; BHQ1 and BHQ2—fluorescence quenchers.
Table 3. Relative mRNA levels of the genes studied in brain and peripheral blood of mice with MPTP-induced models of early stages of PD.
Table 3. Relative mRNA levels of the genes studied in brain and peripheral blood of mice with MPTP-induced models of early stages of PD.
GenesStriatumSubstantia NigraPeripheral Blood
6 h-PSS
(n = 10)
24 h-PSS
(n = 10)
Adv-PSS
(n = 10)
ESS
(n = 10)
6 h-PSS
(n = 10)
24 h-PSS
(n = 10)
Adv-PSS
(n = 10)
ESS
(n = 10)
6 h-PSS
(n = 10)
24 h-PSS
(n = 10)
Adv-PSS
(n = 10)
ESS
(n = 10)
Drd20.94 11.760.90 *0.42 *1.340.573.55 *0.27 *0.90 *0.38 *
0.77–1.18 21.25–2.390.17–1.090.33–0.861.24–1.460.46–0.813.14–5.800.13–0.59 0.75–1.520.27–0.44
Snca1.100.870.93 *0.73 *0.870.497.52 *0.17 *3.370.740.69 *0.20 *
1.01–1.330.78–1.410.12–1.000.61–1.300.81–1.130.33–1.066.42–8.360.16–0.362.12–4.290.60–0.930.21–1.820.14–0.36
Rab5a1.031.010.571.120.931.012.682.431.810.610.360.96
0.83–1.520.82–1.110.47–0.850.39–1.590.72–1.080.88–1.441.79–4.892.01–3.381.56–2.720.39–1.300.22–0.470.73–1.82
Anxa20.987.410.931.926.381.210.291.242.880.711.301.05
0.84–1.225.80–9.600.68–2.201.37–2.755.38–6.470.72–3.830.19–1.010.99–1.561.12–5.110.36–0.870.83–1.960.73–1.70
Nsf0.981.292.690.111.131.031.323.501.140.580.721.07
0.88–1.091.00–1.591.67–3.270.07–0.331.12–1.250.82–1.310.71–3.132.47–5.211.06–3.18 0.39–1.670.46–0.840.79–1.13
1 Median. 2 25–75—quartiles. n = 10 for each group. The levels of the mRNA of the genes studied in the control group were taken as 1. The data with p < 0.05 and fold change ≥1.5 are bold and highlighted. The data with p < 0.05 and fold change ˂1.5 are bold but not highlighted. Dashes indicate samples in which analysis was not performed. * The results obtained in previous studies [40]. 6 h-PSS—model of the presymptomatic stage of PD with decapitation of mice 6 h after last injection of MPTP; 24 h-PSS—model of the presymptomatic stage of PD with decapitation of mice 24 h after last injection of MPTP; AdvPSS—model of the advanced presymptomatic stage of PD; ESS—early symptomatic stage of PD.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rudenok, M.M.; Shadrina, M.I.; Filatova, E.V.; Rybolovlev, I.N.; Nesterov, M.S.; Abaimov, D.A.; Ageldinov, R.A.; Kolacheva, A.A.; Ugrumov, M.V.; Slominsky, P.A.; et al. Expression Analysis of Genes Involved in Transport Processes in Mice with MPTP-Induced Model of Parkinson’s Disease. Life 2022, 12, 751. https://doi.org/10.3390/life12050751

AMA Style

Rudenok MM, Shadrina MI, Filatova EV, Rybolovlev IN, Nesterov MS, Abaimov DA, Ageldinov RA, Kolacheva AA, Ugrumov MV, Slominsky PA, et al. Expression Analysis of Genes Involved in Transport Processes in Mice with MPTP-Induced Model of Parkinson’s Disease. Life. 2022; 12(5):751. https://doi.org/10.3390/life12050751

Chicago/Turabian Style

Rudenok, Margarita M., Maria I. Shadrina, Elena V. Filatova, Ivan N. Rybolovlev, Maxim S. Nesterov, Denis A. Abaimov, Ruslan A. Ageldinov, Anna A. Kolacheva, Michael V. Ugrumov, Petr A. Slominsky, and et al. 2022. "Expression Analysis of Genes Involved in Transport Processes in Mice with MPTP-Induced Model of Parkinson’s Disease" Life 12, no. 5: 751. https://doi.org/10.3390/life12050751

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