• No results found

The midbrain dopaminergic system -development and heterogeneity in animalmodels of Parkinson's diseaseEllinor Betnér

N/A
N/A
Protected

Academic year: 2021

Share "The midbrain dopaminergic system -development and heterogeneity in animalmodels of Parkinson's diseaseEllinor Betnér"

Copied!
28
0
0

Loading.... (view fulltext now)

Full text

(1)

The midbrain dopaminergic system - development and heterogeneity in animal models of Parkinson's disease

Ellinor Betnér

Degree project inbiology, Master ofscience (2years), 2019 Examensarbete ibiologi 45 hp tillmasterexamen, 2019

Biology Education Centre and Comparative Physiology, Uppsala University Supervisors: Åsa Mackenzie and Maria Papathanou

(2)

Abstract

Dopamine (DA) is a neurotransmitter related to motor, limbic and cognitive functions. The development of DA is regulated by a cascade of transcription factors working together in a temporal and spatial manner, with its origin in the floor plate (FP) and midbrain-hindbrain boundary (MHB). One of the transcription factors involved in the maintenance of the FP is Developing Brain Homeobox 1 (Dbx1). Dysfunctions in this dopaminergic system can result in many diseases, for example Parkinson’s disease (PD). PD is caused by a degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc), resulting in tremors, inability to initiate movement, but also limbic and cognitive effects. Not only the SNc is affected in PD, but also the ventral tegmental area (VTA). The VTA is involved in limbic and cognitive functions such as memory and reward, and has been shown to degenerate in PD. Since PD also results in non-motor symptoms, the effect on the VTA in PD needs more research. Studies have shown that some subpopulations within the SNc and VTA seem to survive in PD, while others do not. Research to investigate factors that characterize these surviving subpopulations are of interest to understand potential neuroprotective effects from these factors. Gastrin Releasing Peptide (Grp) is one gene that has been shown to be spared in human PD patients, which raises the question whether animal models for PD also show spared Grp mRNA positive neurons.

To investigate the function of Dbx1, a CRISPR/Cas9 mediated knockout of Dbx1 was performed in zebrafish, which resulted in a more anterior located MHB. To investigate how Grp and other markers are affected in PD animal models, two of the most common animal models of PD were used: the genetic model Thy1-a-Syn and the toxin-based model 6-OHDA.

Thy-a-Syn showed no difference in the expression pattern of DA markers. The 6-OHDA showed that Aldh1a1 and Grp mRNA positive neurons survive in 6-OHDA mice.

Keywords: development, midbrain, dopamine, substantia nigra, ventral tegmental area, Parkinson’s disease

(3)

Introduction

The basal ganglia are structures involved in motoric and limbic functions of the brain. The function of the basal ganglia is highly dependent on dopamine (DA), and a consistent dopaminergic input. The two main dopaminergic input regions of the basal ganglia are the substantia nigra pars compacta (SNc), mainly involved in motor functions, and the ventral tegmental area (VTA), that mainly is involved in the limbic functions.

Dopamine neurons are formed

The process of making a midbrain dopaminergic (mDA) neuron involves a cascade of transcription factors (TFs) interacting in a temporal and spatial matter during development.

Firstly, the neural tube, which is the early version of central nervous system, is formed from the ectoderm after gastrulation, which occurs around embryonic day (E) 6.0 in mice (Theiler 1989). Early in the formation of the neural tube, at E7.5 in mice, two organizing centres are formed: the isthmic organizer (IsO) and the floor plate (FP). The IsO marks the boundary between the midbrain and the hindbrain neuromeres and is therefore also called the midbrain- hindbrain boundary (MHB). mDA neurons are formed from the FP and MHB (Arenas et al.

2015). The MHB acts both as a physical separator between divisions of the developing brain, but also acts as a signalling and patterning centre (Gibbs et al. 2017). Many TFs are involved in the maintenance of the MHB, but two crucial TFs are Orthodenticle homolog 2 (Otx2), found expressed in the midbrain, and Gastrulation brain homeobox 2 (Gbx2), found in the hindbrain. Mainly two TFs work cooperatively to induce the formation of DA neurons. These are called Fibroblast growth factor 8 (Fgf8) and Wingless-int1 (Wnt1) (Arenas et al. 2015).

Another TF in this region is Developing brain homeobox 1 (Dbx1), a TF that works in maintaining the FP. The FP is located anterior to the MHB and is defined by TFs such as Sonic hedgehog (Shh), Forkhead Box A2 (Foxa2) and LIM Homeobox Transcription Factor

1 Alpha (Lmx1a). More specifically, it has been shown that microdomains of the FP that are positive for Dbx1 has been proven positive for important TFs for DA formation, for example Engrailed 1 (En1) (Kouwenhoven et al. 2016; Nouri & Awatramani 2017). En1 itself has been shown to be involved in the maintenance of the MHB, but it is mostly known for being important for the survival of dopaminergic neurons (Kouwenhoven et al. 2016).

(4)

Dbx1 has also been proved to be involved in the formation of the forebrain. This was

confirmed after an ablation of Dbx1 in mice. Ablated mice showed a severe reduction in size of the forebrain and abnormal craniofacial structures already at E8.5. This indicates that Dbx1 positive progenitors would play an important role in the survival of early neurons of the forebrain (Causeret et al. 2011). In zebrafish, Dbx1 has two paralogues: Dbx1a and Dbx1b.

The formation of mDA neurons starts by the progenitor cells of mDA neurons, which is characterized by the gene Nuclear receptor related 1 (Nurr1). At E11.5, progenitor cells migrate to the mantle zone (MZ), a ventrally located area in the FP, where the progenitors differentiate and mature to mDA neurons expressing Tyrosine hydroxylase (Th). mDA neurons are then migrating towards dopaminergic structures such as the SNc (Arenas et al.

2015). Th is what characterize a DA neuron, because of its crucial involvement in DA synthesis. Th encodes for the rate-limiting enzyme TH. TH catalysed tyrosine to L-DOPA, which in turn is the precursor of DA (Lohr & Miller 2014). This differentiation of a mDA neuron, where the mDA neurons are Th positive, occurs at E10.5 in mice. In zebrafish dopaminergic neuronal populations could be detected at 24 hours post fertilization (hpf) (Du et al. 2016).

Dopamine synthesis and transport

Following the production of DA in the now mature dopaminergic presynaptic vesicle, Vesicular monoamine transporter 2 (Vmat2) is involved in the transportation of DA. Vmat2 allows DA to be packed in vesicles and transported from the presynapse to the postsynapse (Lohr & Miller 2014). The release of DA is maintained by Aldehyde dehydrogenase 1A1 (Aldh1a1), and the transportation of DA by Dopamine transporter (Dat) (Brichta &

Greengard 2014). Aldh1a1 is a part of a large family of Aldh genes. Aldh1a1 is almost exclusively expressed in the SNc (Cai et al. 2014), where it is involved in the production of retinoic acid, involved in neural development (Jacobs et al. 2007). It also converts toxic aldehydes metabolites, such as a variant of DA, into non-toxic variants by oxidation (Marchitti et al. 2007). Aldh1a1 has been reported to be involved in Parkinson’s disease (PD), as studies have shown that PD patients have reduced Aldh1a1 mRNA and protein levels in the SN. However, in Aldh1a1 null mice, there is an increase in extracellular DA and Th positive neurons in the SN (Anderson et al. 2011). PD is a disease related to a dysfunction

(5)

in the DA system and is caused by a degeneration of DA producing neurons in the midbrain, causing severe motor symptoms.

Parkinson’s disease

Parkinson’s disease (PD) affects 1.5% of the population above the age 65 (Pringsheim et al.

2014), being the second most common neurodegenerative disease after Alzheimer’s disease.

The very first symptoms of PD are sleep disturbances and olfactory dysfunction (Jin et al.

2017), while common symptoms in a progressed form of PD include tremors, rigidity, akinesia, and postural instability. Although, not only motor symptoms are seen in PD: also dementia (Hanagasi et al. 2017) and depression are common. It has been documented that up to 50% of PD patients are thought to be suffering from depression (Weintraub et al. 2008).

The symptoms of PD arise from the degeneration of dopaminergic neurons, mainly in the SNc which degenerates first in the disease. Pathological hallmarks of PD include, as mentioned before, degeneration of dopaminergic neurons, but also the generation of Lewy Bodies. Lewy Bodies are largely composed of the protein alpha-synuclein (α-syn). Mutations in the gene encoding for α-syn, Snca, has been documented involved for hereditary forms of PD. α-syn has also been linked to mitochondrial dysfunction, that might be contributing to PD (Rocha et al. 2018). Lately, not only the SNc has been shown to be affected in PD, but also the VTA. Considering that PD patients not only experience motor symptoms, but also cognitive and limbic symptoms such as depression, there is a need to investigate how degeneration in the VTA might cause non-motor symptoms in PD. Previous studies report that certain genes may have neuroprotective roles in different subpopulations of dopaminergic neurons of the SNc and VTA.

Calcium ion binding proteins such as Calbindin D28k (Calb1) and Calbindin 2 (Calb2) are both established VTA markers. Calb1 is found in dopaminergic neurons, and studies have found that Calb1 positive neurons survive in PD. This suggests that some Calb1 could have a protective effect on some dopaminergic neurons in PD (Mouatt-Prigent et al. 1994).

Another gene that might have a neuroprotective effect in PD is Gastrin Releasing Peptide (Grp). Grp mRNA appears to colocalize with Th mRNA, and Grp mRNA positive cells has been shown to be spared in non-melanized dopaminergic neurons in the SNc of human PD patients (Viereckel et al. 2016). Later studies confirm the possible neuroprotective effect of

(6)

Grp in PD, but also discusses the combinatorial expression of Grp and Neuronal Differentiation 6 (NeuroD6) in dopaminergic subpopulations of the VTA (Kramer et al.

2018). Lastly, G protein-coupled inwardly-rectifying potassium channel family 2 (Girk2) encodes for a protein controlling cellular electrical excitability via receptor stimulation, changing the neural circuit activity. Previous studies report that Girk2 was found to be expressed in the whole SN, and in 50-60% of the VTA (Reyes et al. 2012). Girk2 has also been discussed as one of the gene candidates in surviving dopaminergic neurons in PD (Brichta & Greengard 2014). These genes are established markers for studying the dopaminergic system of the midbrain.

Animal models for Parkinson’s disease

There are several animal models for PD, all mimicking different aspects of the disease.

Generally, animal models for PD can be divided into two groups: genetic and toxin-based models. In this study, one genetic model, called Line 61 (L61), and one toxin-based model, the 6-Hydroxy-Dopamine (6-OHDA) model, will be studied.

The L61 is a genetic animal model for PD, overexpressing the human wildtype form of α- syn under the Thy-1 promoter. Thy-1 is used as a promoter because it is ubiquitously expressed in the brain. This way, all structures of the brain are affected, and the typical α- syn pathology that is seen in PD patients post-mortem is generated. Using other promoters, such as Th, fails to do so, because it targets catecholaminergic neurons and is therefore not as widespread as with the Thy-1 promoter. The L61 model also causes symptoms that are typical in PD; both motor symptoms and non-motor symptoms are seen. For example, there is a progressive decrease in Th, and striatal DA release is decreased by 40% (Chesselet et al.

2012). For non-motor symptoms, the L61 model causes for example cognitive deficits (Magen et al. 2012), which is also seen in PD.

The 6-OHDA model is a toxin-based model of PD, where the neurotoxin 6-Hydroxy- Dopamine (6-OHDA) is injected in unilaterally in the brain. 6-OHDA is a DA agonist causing neuronal death of dopaminergic neuron by being absorbed by the DA transporter (Hernandez-Baltazar et al. 2017). 6-OHDA is commonly injected into the medial forebrain bundle (MFB), which ultimately leads to degeneration of 95% of dopaminergic neurons in the SNc and VTA. This leads to typical motor symptoms of PD. To confirm the success of

(7)

the unilateral injection, induced rotation is created by administering methamphetamine or amphetamine, that are DA acting drugs. The animals will then rotate ipsilaterally (Torres et al. 2011). Non-motor symptoms include anxiety and hypolocomotion (Vieira et al. 2019).

In this study, fluorescent in situ hybridization is used to study the expression pattern of before mentioned markers within the SNc and VTA in the L61 model and the 6-OHDA model.

Established DA markers are used to visualize the SNc and VTA.

Aims

1) Investigate the effect of the MHB in zebrafish after a CRISPR/Cas9 mediated knockout of Dbx1.

2) Investigate the expression pattern in the SNc and VTA in two common PD models, and to see whether Grp-positive neurons are spared in these models.

(8)

Method and materials

Dbx1 mutant zebrafish

Animals

All experiments were in accordance with Swedish regulations and European Union legislation. An ethics approval was obtained from the Uppsala Animal Ethical Committee.

The zebrafish were kept at 28.5 °C. The developmental stages were determined according to (Kimmel et al. 1995). TgDAT:eGFP zebrafish were generated according to the protocol from (Xi et al. 2011). CRISPR/Cas9 mutant zebrafish targeting Dbx1a and Dbx1b were generated using the protocol adapted from Varshney et al. (2016) (Shawn Burgess Lab/NIH-NHGRI) by the Zebrafish Facility, SciLifeLab, Uppsala University. Two targets for each paralogue were chosen based on their exon location within the target sequence (see table 1), number of off-targets and GC-content.

Table 1. Targets for Dbx1a and Dbx1b. Abbreviations: F: forward R: reverse. T: target.

dbx1a_T1/F GAAAGAGGCGTTGAATTTGG dbx1a_T2/R GAGAGTGCGCGGTTTGGCTC dbx1b_T1/F GAGGAGATGCACTTGGCGAC dbx1b_T2/R CGTCGCCATGTCTACTTCTT

Phenotype analysis

3 days post fertilization (dpf) old zebrafish from the group with a +1/-8 insertion and deletion (indel) mutation were phenotypically analysed and documented by confocal and fluorescence microscopes (Leica TCS SP5; Software Leica Microsystem LAS-AF; Leica M205FA; Leica DFC450C) to find potential mutants. 17 zebrafish were then chosen for genotyping based on their abnormal phenotype.

(9)

Genotyping

17 zebrafish were fin clipped using the protocol Standard Operating Procedure #C3 version 2 for SciLifeLab, Uppsala University. Biopsies were lysed in 30 µL NaOH (50 mM) and amplified with PCR. PCR products were sent to KI Gene CMM (Karolinska Institutet, Stockholm) for fragment length analysis (FLA) using CRISPR-somatic tissue activity test (STAT) (Carrington et al. 2015) to find somatic mutations. Zebrafish that were either heterozygotes for one or both paralogues were kept for further breeding.

Animal models for Parkinson’s disease

Animals

All experiments were in accordance with Swedish regulations and European Union legislation. An ethics approval was obtained from the Uppsala Animal Ethical Committee.

L61 project

In the L61 project, 5 months old transgenic C57BL6/DBA2 male mice overexpressing the human α-syn under the Thy-1 promoter (n=5) and littermate controls with the same sex and age (n=5) were used (Table 2).

6-OHDA project

In the 6-OHDA project, C57BL/6NTac male mice (n=4) (Table 2) were injected with 1 μl 6-OHDA (1.85mg/ml) in the medial forebrain bundle in the right hemisphere at the age of either 8 or 10 weeks. Control mice (n=4) were also injected in the same location but with saline solution and ascorbic acid. Both groups were given saline injections every day and sucrose solution and were sacrificed 2 weeks after the 6-OHDA or saline injection.

In both projects, the mice were sacrificed by cervical dislocation and the brains were dissected. The brains were then snap-freezed in isopentane (-35°C, >99% 2-methylbutane, Honeywell) and stored in a -80°C freezer until being sectioned coronally using a Leica Cryostat CM1950. 16 μm sections were placed on glass slides and stored in -80°C.

(10)

Table 2. Number of mice and sections for the L61 and 6-OHDA projects.

L61 6-OHDA

L61 Control 6-OHDA Control

Number of mice

5 5 4 4

Number of sections analysed per mouse

24 24 8 8

Table 3. PCR primer sequences for making PCR products of the DNA sequence of interest. PCR products are then used for mRNA probe synthesis.

Probe PCR Primer Sequence (S-T3) PCR Primer Sequence (AS-T7)

Dat AATTAACCCTCACTAAAGGGAGT

GGATCGATGCTGCCACCC

TAATACGACTCACTATAGGGAGC CAGTGACGCAGCGTGAA

Th AATTAACCCTCACTAAAGGGAGT

GCGTCGGGTGTCTGACGA

TAATACGACTCACTATAGGGTCC AAGGAGCGCTGGATGGTG

Vmat2 AATTAACCCTCACTAAAGGGATC

CGTGGCTGGGATGGGTATG

TAATACGACTCACTATAGGGCCTT TGCGATGGCACCACCA

rSNCA AATTAACCCTCACTAAAGGGAGA

GCCTTTCACCCCTCTTGC

TAATACGACTCACTATAGGGAGG TGCATAGTCTCATGCTCACA

Calb1 AATTAACCCTCACTAAAGGGAAG

CCCTCTCGCCCGAGGTTC

TAATACGACTCACTATAGGGCCC TCCATCCGACAAGGCCATTA Aldh1a1 AATTAACCCTCACTAAAGGGACT

GTGTCGCAGCATCCCGGA

TAATACGACTCACTATAGGGCCT GGGGAACAGAGCAGCTGAC

Girk2 AATTAACCCTCACTAAAGGGAGA

ACCGGCGAGTCGGAGCTG

TAATACGACTCACTATAGGGCGG GGCACTTGTCCGTGATG

Grp AATTAACCCTCACTAAAGGGACG

GCTCGGAGCTCTCGCTCT

TAATACGACTCACTATAGGGGAA TGGTAGCAAATTGGAGCCCTGA

(11)

Synthesis of riboprobes

To synthesise riboprobes, transcription reagents were added in the following order: 5X transcription buffer, 10X labelling mix with either DIG (red label) or Fluorescein (green label), 100 mM DTT, Rnasin (20U/μl), T7 RNA polymerase (20U/μl), the PCR product targeting the sequence of interest and water. The mix was then incubated in 37°C for 2 hours.

The riboprobe mix was then purified by centrifugation with an Illustra Probequant G-50 micro column and RNA level was subsequently measured using a Nanodrop. Hybridization buffer with formamide was then added to the riboprobes. The riboprobes (Table 3) were then stored in -80°C.

Fluorescent in situ hybridization

Frozen (-80°C) sections on glass slides were thawed in room temperature (RT) before fixation treated with paraformaldehyde (4%). Sections were washed in 1X PBS and acetylated with triethylamine (TEA, pH=8.0) and acetic anhydride (0.25%) for 9 minutes and 45 seconds. Sections were washed in 1X PBS. Meanwhile, DIG- or fluorescein labelled riboprobes (kept on ice) were added to hybridization buffer with formamide (50/50 solution) and denaturated in 85°C for 10 minutes. Cold hybridization buffer with formamide was added to the riboprobes before being added to the sections. Sections were then hybridized in 65°C for 16-18 hours.

Sections were then washed in warm (65°C) 5X, 0.2X saline-sodium citrate (SSC) and subsequently 0.2X SSC in RT, before being washed in maleic acid buffer with Tween 20 (0.1%) (MABT). For the fluorescein-riboprobe revelation step, blocking reagent (BR) was added to the sections. The sections were incubated in horseradish peroxidase-conjugated anti-fluorescein antibody (anti-fluorescein-POD) in BR (1:1000) for 1 hour. Next, the fluorescent signal was amplified by adding a biotin conjugated tyramide signal amplification (TSA) with TSA diluent (1:75) to the sections. Sections were then treated with Neutravidin- Oregon-Green in PBST (1:500) for revelation of biotinylated targets. POD-inhibition was then performed to block the horseradish peroxidase by incubating sections in glycine (0.1M, pH=2.1) and H2O2 (3%). For the DIG-riboprobe revelation step, sections were incubated in anti-DIG-POD (1:1000) and Cy3 conjugated TSA in TSA diluent (1:150). Lastly, the sections were washed and then stained with DAPI (1:50,000) and mounted using Fluoromount mounting media.

(12)

Results

Dbx1 mutant zebrafish

The analysed zebrafish from the -1/+8 indel group showed, at 3 dpf, a decreased size of the forebrain, which is shown in figure 1. The MHB is hence located more anterior compared to the possible wildtype, and compared to reference literature (Kimmel et al. 1995). The straight line between the middle of pupils is a reference mark to visualize the difference in distance from the MHB and the anterior part of the forebrain (Fig. 1). As seen in figure 1, the MHB in the possible wildtype zebrafish is located posterior to the pupils of the eye, whereas in the possible mutant, the MHB is located more anterior. In the possible mutant, the MHB is instead found on the same location on the anterior-posterior axis.

Zebrafish were later genotyped using FLA to investigate somatic mutations and to see if they had mutations in either one or both of the Dbx1 paralogues, see table 2. Here, two zebrafish had a double mutation in both Dbx1 and Dbx1b, and several had a single mutation of either Dbx1a or Dbx1b (table 2).

Figure 1. 3 dpf tgDAT:eGFP Dbx1 mutant zebrafish and wildtype showing dopaminergic structures in the brain. Dotted line is placed between the pupils of the eyes as a reference point. As the genotype is unknown at this point, zebrafish are marked “possible wildtype” and “possible mutant” based on their phenotype only. Left: possible wildtype zebrafish where the MHB is located posterior to the eyes (posterior to dotted line). Right: possible hetero- or homozygous zebrafish where the MHB is

(13)

located between the eyes of the zebrafish (along dotted line). Abbreviations: A: anterior. F: forebrain.

M: midbrain. MHB: midbrain-hindbrain boundary. H: hindbrain. P: posterior.

Table 4. Fragment length analysis results. Every combination of number and letter equals one zebrafish. 17 zebrafish were genotyped to find somatic mutations within both Dbx1a and Dbx1b.

Cells marked either 8 or -1 shows insertion or deletion respectively, indicating a mutation. WT reference for Dbx1a is 311.41∓0.21 and for Dbx1b 303.99∓1.12, meaning no somatic mutation within the gene sequence was found. Bold marks zebrafish with a double mutation (cell C1 and B4).

Empty cells indicate no result.

Gene

1 2 3 4 5 6

Dbx1a

A 311.51 8.07 8.2 8.14 8.18 311.55 B 311.46 - 311.32 8.23 8.06 311.46 C 8.34 311.43 311.14 8.16 8.2 -

Dbx1b

A

- 304.59 - 303.38 304.54 303.61

B -1.99 305.59 303.38 -1.99 303.36 - C -1.04 - 304.5 303.54 303.38 -

(14)

Animal models for Parkinson’s disease

Dopaminergic mRNA levels are unaffected in L61 mice

By using fluorescent in situ hybridization, it could be shown that the DA markers Th, Dat and Vmat2 mRNA seem unaffected in L61 mice (Fig. 2). Both L61 mice and controls show expression of Dat, Th and Vmat2 mRNA in the SNc and VTA. Overall, a strong expression of mRNA can be seen in all these markers.

Figure 2. L61 mice showed no change in mRNA expression of the dopaminergic markers Dat, Th and Vmat2. A, C, E: Control mice at bregma -3.40. B, D, F: L61 mice at bregma -3.40 mm. A-B:

Expression of Dat mRNA. C-D: Expression of Th mRNA. E-F: Expression of Vmat2 mRNA.

Abbreviations: SNc: substantia nigra pars compacta. VTA: ventral tegmental area.

(15)

α-syn is overexpressed in L61 mice

The rat Snca (rSNCA) sequence was used to design the probe for Snca mRNA to verify the L61 model as the Thy1 promoter is ubiquitously expressed in the brain. The results show that Snca was highly overexpressed in the L61 mice (Fig. 3). Structures that are most expressed are the cortex (Fig. 3B, 3D, 3F) and hippocampus (Fig. 3F), while it appears that Snca is relatively less expressed within the SNc and VTA (Fig. 4).

Figure 3. rSNCA mRNA overexpression at three different levels in control and L61 mice. A, C, D:

control where rSNCA is not overexpressed. B, D, F: L61 mice where rSNCA is overexpressed. A, B:

bregma 0.38mm. C, D: -1.06mm. E, F: 2.06mm. Abbreviations: SNc: substantia nigra pars compacta.

VTA: ventral tegmental area.

(16)

Figure 4. Snca is overexpressed in L61. A, C, E: control animals. B, D, F: L61 mice. A, B: overview of Snca mRNA expression at bregma -3.28 showing a clear overexpression of Snca in L61 mice. C, D: closer view of the SNc and VTA. E, F: colocalization of Th and Snca mRNA (yellow). In control (E) both Th and Snca mRNA can be seen in the SNc and VTA, while in L61 mice (F), no Snca is expressed within the SNc and VTA.

Markers Calb1, Aldh1a1 and Girk2 are unchanged in L61 mice

When compared to control, the markers Calb1, Aldh1a1 and Girk2 show the same mRNA expression in L61 mice (Fig. 5). Calb1 and Girk2 both have a relative weak expression compared to Aldh1a1, which is seen as highly expressed in the SNc. Calb1 is mainly found in the VTA and not SNc, and Aldh1a1 in the SNc and not VTA. Girk2 is ubiquitously expressed within both the SNc and VTA, but the expression was found low in this study.

(17)

Figure 5. Expression of Calb1, Aldh1a1 and Girk2 mRNA, that all are unchanged in L61 mice when compared to controls. A, C, E: control mice. B, D, F: L61 mice. A, B: Calb1 expression, bregma - 3.52mm. C, D: Aldh1a1 expression, bregma -3.08. E, F: Girk2 expression, bregma -3.08mm.

Abbreviations: SNc: substantia nigra pars compacta. VTA: ventral tegmental area.

(18)

Unilateral expression of dopaminergic markers Dat, Th and Vmat2 in 6-OHDA mice The 6-OHDA injection caused a unilateral expression of the mRNA of Dat, Th and Vmat2 (Fig. 6). All three markers are found in the SNc and VTA. For Vmat2, it appears that there is a neurodegenerative progression since the mice sacrificed 3 weeks after the 6-OHDA injection show less expression of Vmat2 mRNA compared to mice sacrificed 2 weeks after the injection (Fig. 7).

Figure 6. The injection with 6-OHDA results in a unilateral degeneration of dopaminergic neurons, which is shown by the dopaminergic markers Dat, Th and Vmat2, all expressed in the SNc and in the VTA. Mice sacrificed 2 weeks after injection. A, C, E: Control mice at bregma -3.40 mm. B, D, F:

6-OHDA injected mice at bregma -3.40. A-B: Expression of Dat mRNA. C-D: Expression of Th mRNA. E-F: Expression of Vmat2 mRNA. Abbreviations: SNc: substantia nigra pars compacta.

VTA: ventral tegmental area.

(19)

Figure 7. Comparison of Vmat2 mRNA expression in mice that were sacrificed either 2 weeks or 3 weeks after the 6-OHDA injection, and their controls, all bregma -3.40 mm. A, B: Control mice where no difference is seen between 2 weeks and 3 weeks after injection. C, D: 6-OHDA injected mice. A, C: Mice sacrificed 2 weeks after the injection. B, D: Mice sacrificed 3 weeks after the injection. D: Results suggest that the 6-OHDA injection caused more degeneration of Vmat2 positive cells in mice sacrificed after 3 weeks compared to 2 weeks (7C). Abbreviations: SNc: substantia nigra pars compacta. VTA: ventral tegmental area.

Grp and Aldh1a1 positive cells survive in 6-OHDA injected mice

Some Aldh1a1 and Grp mRNA positive cells can be seen in the injected hemisphere of the 6-OHDA mice (Fig. 8B, 8F, marked with arrowhead). Although, the number of surviving cells is few. This may suggest that Aldh1a1 and Grp positive cells survive in this model of PD. Girk2 mRNA shows no surviving neurons in the 6-OHDA injected hemisphere (Fig.

8D). For Grp, it appears that some, but not all, of the maintained cells are colocalized with Th compared to the colocalization in the controls (Fig. 9).

(20)

Figure 8. Expression of Aldh1a1, Girk2 and Grp mRNA in 6-OHDA injected mice and control. Mice sacrificed 2 weeks after injection. A, C, E: Control mice at bregma -3.40 mm. B, D, F: 6-OHDA injected mice at bregma -3.40 to -3.50. A, B: Aldh1a1 mRNA is found in the SNc and VTA in control, while Aldh1a1 positive neurons are spared in the VTA in 6-OHDA injected mice (marked with arrowhead). C, D: Girk2 is expressed in the SNc and VTA in control and shows no spared neurons in the 6-OHDA injected mice. E, F: Grp positive neurons, found in the ventral VTA in control, are spared in the 6-OHDA injected mice. Spared neurons are found in the ventral VTA (F; marked with arrowhead). Abbreviations: SNc: substantia nigra pars compacta. VTA: ventral tegmental area.

(21)

Figure 9. Colocalization of Grp and Th in 6-OHDA injected mice and control mice. Mice sacrificed 2 weeks after injection. A: control where Grp is expressed in the medial VTA. B: 6-OHDA injected mice where Grp positive cells are expressed in the medial VTA (arrow). C: colocalization of Th and Grp mRNA in control mice (arrowhead). D: colocalization of Th and Grp mRNA (arrowhead).

Abbreviations: SNc: substantia nigra pars compacta. VTA: ventral tegmental area.

(22)

Discussion

This master thesis consists of two parts: 1) CRISPR/Cas9 mediated knockout of Dbx1 in zebrafish. 2) Expression pattern analysis of the midbrain dopaminergic system in two models for Parkinson’s disease in mice.

Dbx1 knockout in zebrafish

For the first part of this master thesis, the knockout of the two paralogues of Dbx1 resulted in an anterior shift of the MHB in the zebrafish brain. This was visualized using by fluorescent expression of Dat, marking dopaminergic structures. The MHB is located posterior to the eyes in wildtype but were in knockouts seen as located in the same location as the eyes on the anterior-posterior axis. This might be due to a reduction of the forebrain, causing an anterior shift of the MHB. Previous Dbx1 knockout studies, but in mice, have shown a reduction of size of the forebrain and craniofacial structures, suggesting that a knockout would have the same effect on zebrafish.

The limitations to this study are that the genotype of the zebrafish that were first phenotypically analysed (Fig. 1) is unknown. Figure 1 shows the phenotypic analysis and assumptions about whether the zebrafish are wildtype or mutants (hetero- or homozygous for Dbx1 paralogues) are therefore made, which is not reliable. However, the results based on the phenotypic analysis are promising, since the “possible mutant” shows severe changes in the location of the MHB. There were also limitations timewise, as the zebrafish had to mature before further breeding after the genotyping (Table 2) to generate more offspring with somatic mutations. In this study, this would be the next step. After a strain with double mutations has been generated, additional imaging and experiments can be performed. For example, in situ hybridization targeting a neurotransmitter found in large parts of the brain could be of interest to see whether the forebrain is smaller in size, and to see other potential effects of the knockout. Targeting the glutamatergic marker Vesicular glutamate transporter 2 (Vglut2) could be a good marker for that purpose. After generating a strain with double mutations, imaging could be done during several embryonic stages, for example from 3 dpf to 5 dpf and adult, to see whether the MHB remains to be located more anterior than the control in older embryos and adults.

(23)

However, the knockout of Dbx1 generated zebrafish with interesting phenotype that suggests that Dbx1 could play a role in maintaining the location of the MHB in an anterior-posterior axis. Future experiments will test this hypothesis.

Animal models for Parkinson’s disease

In the L61 study, it appears that the overexpression of α-syn did not have an effect on the SNc and VTA. The heterogeneity of DA neurons of the SNc and VTA appear to be unaffected in L61 mice, by looking at the expression pattern of dopaminergic markers. Since the L61 does not cause any degeneration of DA neurons in the SNc and VTA, this could be an explanation to the results. In addition, the mice used in this study were only 5 months old, while previous studies saw a decrease in older mice. Potential neuroprotective effects in genes are therefore difficult to study. However, in the 6-OHDA study, a few Aldh1a1 and Grp positive neurons appear to be maintained in the 6-OHDA mice when compared to controls. For Aldh1a1, this is in accordance with previous studies studying the vulnerability of Aldh1a1 positive cells. For Grp, some studies have also shown that Grp positive neurons survive in 6-OHDA mice. However, not all studies came to the same conclusion. One study found that neurons are both Grp mRNA positive and NeuroD6 mRNA positive are not any more resistant to degeneration than other neurons. This raises questions whether Grp itself is neuroprotective, or if other factors influence the resistance to neurodegeneration. Since NeuroD6 was not studied in this thesis, it is not possible to confirm that the surviving Grp positive neurons in our 6-OHDA mice are also colocalized with NeuroD6. Although, when colocalization of Th and Grp was studied, not all remaining Grp neurons in the 6-OHDA mice were Th positive. If this also is due to a combinatorial effect is a potential future investigation. The limitation to the 6-OHDA animal model study was that a quantification of the spared cells could not be made due to limited time.

As a conclusion, this PD animal model study confirms previous studies that show that some Aldh1a1 positive neurons are spared in a 6-OHDA model for PD. This study also gives new insight to the possibility that Grp positive cells are resistant to neurodegeneration.

(24)

Acknowledgements

I would first like to thank Prof. Åsa Mackenzie for welcoming me in her research group, and for letting me work with interesting projects. I am thankful for the experience I have gained.

I am also very thankful for the advice, feedback and supervision from Åsa during this master thesis project, it has been extremely valuable to me. Additional thanks to my co-supervisor Dr. Maria Papathanou, who gave me valuable advice during this thesis and during previous projects. I also thank Bianca Vlcek for teaching me fluorescent in situ hybridization and for always being supportive in the lab, and Adriane Guillaumin, for doing the 6-OHDA injections and helping with the experiments. I would also like to thank Laura Waldmann for her help and guidance regarding the Dbx1 project. Further, I thank the SciLifeLab zebrafish facility, and the rest of the Mackenzie lab.

(25)

References

Anderson DW, Schray RC, Duester G, Schneider JS. 2011. Functional significance of aldehyde dehydrogenase ALDH1A1 to the nigrostriatal dopamine system. Brain research 1408: 81–87.

Arenas E, Denham M, Villaescusa JC. 2015. How to make a midbrain dopaminergic neuron. Development 142: 1918–1936.

Brichta L, Greengard P. 2014. Molecular determinants of selective dopaminergic vulnerability in Parkinson’s disease: an update. Frontiers in Neuroanatomy, doi 10.3389/fnana.2014.00152.

Cai H, Liu G, Sun L, Ding J. 2014. Aldehyde Dehydrogenase 1 making molecular inroads into the differential vulnerability of nigrostriatal dopaminergic neuron subtypes in Parkinson’s disease. Translational Neurodegeneration, doi 10.1186/2047-9158-3- 27.

Carrington B, Varshney GK, Burgess SM, Sood R. 2015. CRISPR-STAT: an easy and reliable PCR-based method to evaluate target-specific sgRNA activity. Nucleic Acids Research 43: e157.

Causeret F, Ensini M, Teissier A, Kessaris N, Richardson WD, Lucas de Couville T, Pierani A. 2011. Dbx1-expressing cells are necessary for the survival of the mammalian anterior neural and craniofacial structures. PloS One 6: e19367.

Chesselet M-F, Richter F, Zhu C, Magen I, Watson MB, Subramaniam SR. 2012. A progressive mouse model of Parkinson’s disease: the Thy1-aSyn (”Line 61”) mice.

Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics 9: 297–314.

Du Y, Guo Q, Shan M, Wu Y, Huang S, Zhao H, Hong H, Yang M, Yang X, Ren L, Peng J, Sun J, Zhou H, Li S, Su B. 2016. Spatial and Temporal Distribution of

Dopaminergic Neurons during Development in Zebrafish. Frontiers in Neuroanatomy, doi 10.3389/fnana.2016.00115.

(26)

Gibbs HC, Chang-Gonzalez A, Hwang W, Yeh AT, Lekven AC. 2017. Midbrain-

Hindbrain Boundary Morphogenesis: At the Intersection of Wnt and Fgf Signaling.

Frontiers in Neuroanatomy, doi 10.3389/fnana.2017.00064.

Hanagasi HA, Tufekcioglu Z, Emre M. 2017. Dementia in Parkinson’s disease. Journal of the Neurological Sciences 374: 26–31.

Hernandez-Baltazar D, Zavala-Flores LM, Villanueva-Olivo A. 2017. The 6-

hydroxydopamine model and parkinsonian pathophysiology: Novel findings in an older model. Neurología (English Edition) 32: 533–539.

Jacobs FMJ, Smits SM, Noorlander CW, von Oerthel L, van der Linden AJA, Burbach JPH, Smidt MP. 2007. Retinoic acid counteracts developmental defects in the substantia nigra caused by Pitx3 deficiency. Development (Cambridge, England) 134: 2673–2684.

Jin H, Zhang J-R, Shen Y, Liu C-F. 2017. Clinical Significance of REM Sleep Behavior Disorders and Other Non-motor Symptoms of Parkinsonism. Neuroscience Bulletin 33: 576–584.

Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. 1995. Stages of embryonic development of the zebrafish. Developmental Dynamics: An Official Publication of the American Association of Anatomists 203: 253–310.

Kouwenhoven WM, Veenvliet JV, van Hooft JA, van der Heide LP, Smidt MP. 2016.

Engrailed 1 shapes the dopaminergic and serotonergic landscape through proper isthmic organizer maintenance and function. Biology Open 5: 279–288.

Kramer DJ, Risso D, Kosillo P, Ngai J, Bateup HS. 2018. Combinatorial Expression of Grp and Neurod6 Defines Dopamine Neuron Populations with Distinct Projection Patterns and Disease Vulnerability. eNeuro 5: ENEURO.0152-18.2018.

Lohr KM, Miller GW. 2014. VMAT2 and Parkinson’s disease: harnessing the dopamine vesicle. Expert Review of Neurotherapeutics 14: 1115–1117.

Magen I, Fleming SM, Zhu C, Garcia EC, Cardiff KM, Dinh D, Rosa KDL, Sanchez M, Torres ER, Masliah E, Jentsch JD, Chesselet M-F. 2012. Cognitive deficits in a

(27)

mouse model of pre-manifest Parkinson’s disease. European Journal of Neuroscience 35: 870–882.

Marchitti SA, Deitrich RA, Vasiliou V. 2007. Neurotoxicity and metabolism of the catecholamine-derived 3,4-dihydroxyphenylacetaldehyde and 3,4-

dihydroxyphenylglycolaldehyde: the role of aldehyde dehydrogenase.

Pharmacological Reviews 59: 125–150.

Mouatt-Prigent A, Agid Y, Hirsch EC. 1994. Does the calcium binding protein calretinin protect dopaminergic neurons against degeneration in Parkinson’s disease? Brain Research 668: 62–70.

Nouri N, Awatramani R. 2017. A novel floor plate boundary defined by adjacent En1 and Dbx1 microdomains distinguishes midbrain dopamine and hypothalamic neurons.

Development (Cambridge, England) 144: 916–927.

Pringsheim T, Jette N, Frolkis A, Steeves TDL. 2014. The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Movement Disorders: Official Journal of the Movement Disorder Society 29: 1583–1590.

Reyes S, Fu Y, Double K, Thompson L, Kirik D, Paxinos G, Halliday GM. 2012. GIRK2 expression in dopamine neurons of the substantia nigra and ventral tegmental area.

The Journal of Comparative Neurology 520: 2591–2607.

Rocha EM, De Miranda B, Sanders LH. 2018. Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson’s disease. Neurobiology of Disease 109: 249–257.

Theiler K. 1989. The House Mouse: Atlas of Embryonic Development. Springer-Verlag, Berlin Heidelberg.

Torres EM, Lane EL, Heuer A, Smith GA, Murphy E, Dunnett SB. 2011. Increased efficacy of the 6-hydroxydopamine lesion of the median forebrain bundle in small rats, by modification of the stereotaxic coordinates. Journal of Neuroscience Methods 200: 29–35.

(28)

Varshney GK, Carrington B, Pei W, Bishop K, Chen Z, Fan C, Xu L, Jones M, LaFave MC, Ledin J, Sood R, Burgess SM. 2016. A high-throughput functional genomics workflow based on CRISPR/Cas9-mediated targeted mutagenesis in zebrafish.

Nature Protocols 11: 2357–2375.

Vieira JCF, Bassani TB, Santiago RM, de O Guaita G, Zanoveli JM, da Cunha C, Vital MABF. 2019. Anxiety-like behavior induced by 6-OHDA animal model of Parkinson’s disease may be related to a dysregulation of neurotransmitter systems in brain areas related to anxiety. Behavioural Brain Research 371: 111981.

Viereckel T, Dumas S, Smith-Anttila CJA, Vlcek B, Bimpisidis Z, Lagerström MC, Konradsson-Geuken Å, Wallén-Mackenzie Å. 2016. Midbrain Gene Screening Identifies a New Mesoaccumbal Glutamatergic Pathway and a Marker for Dopamine Cells Neuroprotected in Parkinson’s Disease. Scientific Reports 6:

35203.

Weintraub D, Comella CL, Horn S. 2008. Parkinson’s disease--Part 1: Pathophysiology, symptoms, burden, diagnosis, and assessment. The American Journal of Managed Care 14: S40-48.

Xi Y, Yu M, Godoy R, Hatch G, Poitras L, Ekker M. 2011. Transgenic zebrafish expressing green fluorescent protein in dopaminergic neurons of the ventral diencephalon. Developmental Dynamics 240: 2539–2547.

References

Related documents

Syftet eller förväntan med denna rapport är inte heller att kunna ”mäta” effekter kvantita- tivt, utan att med huvudsakligt fokus på output och resultat i eller från

Generella styrmedel kan ha varit mindre verksamma än man har trott De generella styrmedlen, till skillnad från de specifika styrmedlen, har kommit att användas i större

I regleringsbrevet för 2014 uppdrog Regeringen åt Tillväxtanalys att ”föreslå mätmetoder och indikatorer som kan användas vid utvärdering av de samhällsekonomiska effekterna av

a) Inom den regionala utvecklingen betonas allt oftare betydelsen av de kvalitativa faktorerna och kunnandet. En kvalitativ faktor är samarbetet mellan de olika

• Utbildningsnivåerna i Sveriges FA-regioner varierar kraftigt. I Stockholm har 46 procent av de sysselsatta eftergymnasial utbildning, medan samma andel i Dorotea endast

Den förbättrade tillgängligheten berör framför allt boende i områden med en mycket hög eller hög tillgänglighet till tätorter, men även antalet personer med längre än

På många små orter i gles- och landsbygder, där varken några nya apotek eller försälj- ningsställen för receptfria läkemedel har tillkommit, är nätet av

Figur 11 återger komponenternas medelvärden för de fem senaste åren, och vi ser att Sveriges bidrag från TFP är lägre än både Tysklands och Schweiz men högre än i de