• No results found

Characterization of the tg(rgs4:mCherry) zebrafish line

N/A
N/A
Protected

Academic year: 2022

Share "Characterization of the tg(rgs4:mCherry) zebrafish line"

Copied!
26
0
0

Loading.... (view fulltext now)

Full text

(1)

Characterization of the tg(rgs4:mCherry) zebrafish line

Henrik Hallgren

Degree project inbiology, Master ofscience (2years), 2014 Examensarbete ibiologi 45 hp tillmasterexamen, 2014

Biology Education Centre and Department ofOrganismal Biology, Uppsala University Supervisors: Jonathan Sager and Lina Emilsson

External opponent: Bryn Farnsworth

(2)

i

Table of contents

List of abbreviations ... ii

Abstract ... iii

1 Introduction ... 1

1.1 G protein-coupled receptors ... 1

1.2 RGS proteins ... 2

1.2.1 RGS4 ... 2

1.3 Zebrafish as a model organism ... 3

1.4 rgs4 in zebrafish ... 4

1.5 Aims ... 5

2 Materials and methods ... 5

2.1 Zebrafish care and housing... 5

2.2 Imaging ... 5

2.3 In situ hybridization ... 6

2.4 Cryosectioning ... 8

2.5 Drug treatment ... 8

2.6 Morpholino injection ... 9

3 Results ... 9

3.1 Development of transgene expression ... 9

3.2 Rostrally located expression ... 10

3.3 Expression in mesencephalon, rhombencephalon and non-neuronal tissue... 12

3.4 In situ hybridization ... 14

3.5 Drug treatment and Morpholino injection ... 15

4 Discussion ... 17

5 Acknowledgements ... 20

6 References ... 20

(3)

ii

List of abbreviations

CISH Chromogenic in situ hybridization dFISH Double fluorescent in situ hybridization

DIG Digoxigenin

dpf Days post fertilization

FISH Fluorescent in situ hybridization GCL Ganglion cell layer

GPCR G protein-coupled receptor hpf Hours post fertilization HRP Horse radish peroxidase HYB+ Hybridization buffer INL Inner nuclear layer IPL Inner plexiform layer ISH In situ hybridization MAB Maleic acid buffer

MO Morpholino oligonucleotide ONL Outer nuclear layer

OPL Outer plexiform layer PBS Phosphate buffer saline PFA Paraformaldehyde

RGS Regulator of G protein-signaling

RT Room temperature

SFGS Stratum fibrosum et griseum superficiale sFISH Single fluorescent in situ hybridization

SO Stratum opticum

SPV Stratum periventriculare SSC Saline-sodium citrate

TSA Tyramide signal amplification

(4)

iii

Abstract

Cell-to-cell communication is one of the fundamental requisites of making multicellular organisms. G protein-coupled receptors (GPCRs) are one of the most abundant receptor-types within vertebrates. They canonically mediate their signal via hetrotrimeric G proteins, and G protein signaling is regulated by regulators of G protein-signaling (RGS). One of these RGS proteins, RGS4, is preferentially expressed in the central nervous system of humans and has been strongly connected to dopaminergic signaling, along with a number of severe neuronal diseases. rgs4 is not well studied in the model organism Danio rerio, the zebrafish, with only two publications. In this project, a newly constructed transgenic line, tg(rgs4:mCherry), with the fluorophore mCherry regulated by the promoter element of rgs4 was characterized in order to investigate fidelity to endogenous rgs4 expression and the utility of the transgenic line. The mCherry expression is apparent by 48 hours post fertilization, and expression is found mainly in neuronal tissue. Cell bodies are visible only in some labeled areas, while other areas show a more diffuse signal indicative of projections. There is only one

transgenically labeled area that also unambiguously expresses rgs4; the pronephric tubule.

This line is therefore not particularly well suited for rgs4-specifc studies, but this does not discredit the fidelity of the construct. A transgenic line made with a site-directed technique would most likely confer the fidelity of the promoter to the expression of the fluorophore. A way of increasing the labeling resolution includes exchanging the mCherry fluorophore for one with stronger signal and a lower tendency to aggregate, e.g. eGFP. Increasing the resolution of the characterization, e.g. to the level of sub-nuclei or neuronal types, would serve to enhance the utility of the line. As it is, the tg(rgs4:mCherry) zebrafish line has limited uses, and yet it is not without them.

(5)

1

1 Introduction

A fundamental function in a multicellular organism, and to some extent single cellular ones, is communication between cells. This is what enables the trillions of cells that make up the adult human body (Bianconi et al. 2013) to function together as a single being rather than a pile of unicellular loners. For a signal to elicit a response from the target it first needs to be registered by a receptor, and the receptor needs to be controllable to allow the response to cause an adaption for future responses. This is a basic argument for the importance of studying biological receptors and their regulators.

1.1 G protein-coupled receptors

Some of the most common types of receptors are the G protein-coupled receptors (GPCRs).

They comprise a family of receptors that have been studied for more than a century, though the term was coined later. There are four features common to the signaling pathways of all members of the GPCR family (Lodish et al. 2013). Firstly, all GPCRs have seven

transmembrane regions, which gave rise to their other name: 7-transmembrane receptors (7TMs). Secondly, GPCRs canonically interact with a trimeric G-protein to convey the signal, which is how they got their name. However this is not always true; they can participate in G protein-independent signaling, such as in some tyrosine kinase-dependent growth regulatory pathways (Luttrell et al. 1999). Thirdly, there is always a membrane-bound effector protein being activated which is responsible for carrying the signal on, either directly or via a secondary messenger. Lastly, the signaling pathway includes proteins that participate in its feedback regulation and desensitization. In short, GPCRs are activated when a ligand binds, causing a conformational change and enabling the binding of a heterotrimeric G protein to the receptor. This catalyzes an exchange of GDP to GTP in the G subunit, causing it to

dissociate from the G/ heterodimer and become active. It then activates or deactivates an effector protein, e.g. phospholipase C or adenylyl cyclase. The G contains intrinsic GTP hydrolysis properties, which will eventually deactivate the protein and enable it to again bind to the G/ heterodimer (Lodish et al. 2013).

Since GPCRs are so wide spread, and involved in so many important functions, dysregulation or misfiring often leads to some kind of disorder or disease. Constitutively active GPCR have been connected to diseases such as familial male-limited precocious puberty and congenital night blindness (see Arvanitakis et al. 1998 for review), and mislocalization is associated with

(6)

2

diseases such as retinitis pigmentosa and diabetes (see Tan et al. 2004 for review). But even if the association between receptor and disease has been made, the actual mechanism behind the effect is often not completely known. To elucidate such mechanisms, in vitro studies might be useful, and while it today is possible to satisfactorily replicate the in vivo behavior of GPCRs it was not always so; the proteins regulating the receptors were not known. About 32 years ago, a gene was found that when mutated prevented recovery from pheromone-induced cell cycle arrest in Saccharomyces cerevisiae (Chan & Otte 1982). It’s product normally controls the desensitization of pheromone signaling and enable the cells to recover from the initial effect via interactions with the  subunits of G proteins (Dohlman et al. 1995). 14 years later, a family of homologous proteins in vertebrates was found, and named regulator of G protein signaling (RGS) (Druey et al. 1996).

1.2 RGS proteins

The RGS proteins function as the name implies: they regulate the signaling of G proteins.

They are negative regulators, meaning they act to stop the signal by catalyzing the intrinsic GTP hydrolysis of the G proteins  subunit and they are therefore classified as GTPase activating proteins (GAPs). They bind to the dissociated GTP-bound G and stabilize the transition state in the GTP hydrolysis reaction, increasing the reaction rate many-fold (Watson et al. 1996). Although it is their GAP function they are most known for, some RGS proteins can in fact inhibit the G protein dependent signaling in other ways. For example, RGS12 and RGS14 contain a domain that can stabilize the G in the GDP-bound state, and RGS16 can translocate the G to detergent-resistant membranes and thus prevent effector interactions (Riddle et al. 2005). There are more than 20 known RGS proteins today, and most of them have been shown to have region specific expression (Gold et al. 1997; Grafstein-Dunn et al.

2001), and so specific RGS proteins are responsible for specific signal transduction pathways (Taymans et al. 2003). Many of them have been associated with diseases, such as RGS1 with celiac disease and type 1 diabetes (Hunt et al. 2008; Smyth et al. 2008), and RGS9 with Parkinson’s disease (Tekumalla et al. 2001). RGS4, encoding a 201 amino acid large RGS protein, has been associated with severe neuronal disorders such as schizophrenia (Erdely et al. 2006) and Alzheimer’s disease (Emilsson et al. 2006).

1.2.1 RGS4

RGS4 is specific for Go/i, which inhibits adenylyl cyclase, Gq, which activates

phospholipase C, and transducin (Berman et al. 1996; Watson et al. 1996; Hepler et al. 1997).

(7)

3

All RGS proteins contain an RGS domain that is responsible for its GAP activity and the amino acid sequence of this domain, together with its small size and an absence of many other domains, puts RGS4 in the R4 subfamily (Riddle et al. 2005). In humans, it’s expression is highly concentrated to the central nervous system and its expression pattern in the human brain is similar to the pattern seen in rat (Larminie et al. 2004). RGS4 has been strongly connected to dopaminergic neurotransmission. It is involved in regulating the dopamine receptors D2R and D3R (Min et al. 2012), but is also itself affected by dopamine receptor signaling: D1R antagonists and D2R agonists cause fluctuations in RGS4 mRNA in the rat striatum (Taymans et al. 2003). It does not come as a surprise that a protein involved in such mechanisms is associated with a number of disorders, diseases and disabilities. As previously mentioned, it has been identified as an indicator of Alzheimer’s disease (Emilsson et al.

2006), and the RGS4 mRNA levels in the cingulate gyrus, the superficial frontal gyrus and the insular cortex of schizophrenia patients is significantly decreased (Erdely et al. 2006). It is also involved in modulating the response to drugs of abuse, which directly connects to its function in dopaminergic signaling (Hooks et al. 2008) and its negative regulation of opioid receptor signaling (Traynor 2010). It is a popular focus for a lot of research today, in the hopes of it leading to drug targets or better treatment for suffering patients. As seems to so often be the case; even though we know a lot about it, so much more is yet to be fully understood.

1.3 Zebrafish as a model organism

There will always be tradeoffs to consider when choosing which model organism to use for a given experiment. Low cost and small space requirements tend to enable larger sample sizes, as does high fecundity and short generation time. Better values for these traits is often associated with species from more evolutionary basal branches such as with Caenorhabditis elegans and Drosophila melananogaster, and extrapolating hypotheses from such

experiments to vertebrates and humans becomes difficult and risky. On the other hand, using species that are evolutionary closer to humans such as primates or mice involves greater strains on the facility and the techniques used, and setting up the experiment can prove more challenging. Danio rerio, the zebrafish, is a very good middle ground. It has relatively low housing requirements and low upkeep costs, a generation time of three to four months and a female can comfortably lay hundreds of eggs every 10 days. Zebrafish are ovuliparous, so the fertilization of the eggs and embryo development occurs externally, and the embryo is

completely transparent during its first days. This easy access to the eggs and embryos makes

(8)

4

manipulations much easier than on a viviparous animal, and many techniques of genome manipulations have been developed for, or adapted to, their use on zebrafish. The genome is fully sequenced, and a large library of already identified mutants is available.

Using transgenically modified zebrafish, with the expression of a fluorophore under the regulation of a promoter of your choice, makes visualizing your cells of interest in the living fish easy. This is a very powerful technique used e.g. to study the development of specific cell groups (Xi et al. 2011) or the morphological effect of a treatment (Cheng et al. 2013) as it happens. One way of making a transgenic zebrafish is with the Tol2kit (Kwan et al. 2007).

With this technique, a construct is created that contains the transgene, e.g. a promoter followed by a fluorophore and a polyadenylation signal, flanked by sites recognized by a transposase. This construct is injected together with the mRNA of the transposase into the embryo (preferably at the one-cell stage), and the founder (F0) of the transgenic line is done.

However, a very important note is that transposon integration is random. This means that the transgene can be inserted in a region that is silenced, or under a strong promoter that overrides the one in the construct, and thus exhibit an expression pattern not wholly controlled. There will always be a measure of randomness involved.

1.4 rgs4 in zebrafish

The studies done on rgs4 specifically in zebrafish are few. A query in the online database Web of Science, using the search phrase “rgs4 zebrafish”, comes up with only two hits. One is a study done by Serafimidis et al. (2011), where they investigated the role of Gi mediated GPCR signaling on the development of pancreatic endocrine cells in mice and zebrafish. They found that rgs4 is expressed in early epithelial endocrine progenitor cells, and that rgs4 loss of function negatively affected islet cell aggregation (Serafimidis et al. 2011). The second study was done by Cheng et al. (2013), in which they investigated rgs4’s expression pattern and role in the developing nervous system. They saw rgs4 expression in nuclei in the

telencephalon and diencephalon, in the pronephric duct and in the pancreas. They found that rgs4 expression knockdown and rgs4 protein inhibition both result in motility defects and stunted axon growth in spinal motor neurons (Cheng et al. 2013).

To aid in the research of rgs4 in zebrafish, a transgenic line with the fluorophore mCherry under the rgs4 promoter and, fused with a membrane-localization tag was created. This tg(rgs4:mCherry) was created with the Tol2kit and so the expression pattern of mCherry

(9)

5

needed to be mapped out, in order to both examine the fidelity to endogenous rgs4 expression and asses the utility of the transgenic line.

1.5 Aims

The objective of this project was therefore to characterize the tg(rgs4:mCherry) line, and establish the degree to which rgs4-positive cells were transgenically labeled. This was done primarily with confocal microscopy, combined with the aid of in situ hybridization (ISH) techniques and cryosectioning. Pharmacological treatment and Morpholino injections were applied in an attempt to affect rgs4-positive cells.

2 Materials and methods

All experiments were performed according to Statens jordbruksverks föreskrifter och allmänna råd om försöksdjur SJVFS 2012:26 and the European parliament and council directive 2010/63/EU, approved under the ethical permits Dnr:C262/11 and Dnr:C255/11.

The zebrafish strains used for this project were wildtype AB/AB, tg(rgs4:mCherry) and tg(olig2:dsRed). The tg(olig2:dsRed) has been described before (Shin et al. 2003). The tg(rgs4:mCherry) was made with the tol2-system as described in the introduction.

2.1 Zebrafish care and housing

When crossed, the adult fish were set up in breeding tanks, with a bottom surface such that any produced eggs would be inaccessible to them and thus prevent them from being eaten.

This was usually done in the afternoon, leaving the fish in the breeding tanks overnight. The embryos were collected at noon the day after and the adult fish returned to their system tanks.

Embryos were raised at 28 oC in the dark, in system water containing methylene blue to protect them from fungus, and treated with 0.003% phenylthiourea to prevent melanogenesis.

Embryos were euthanized by overexposure to tricane (0.015%-0.03%) overnight.

2.2 Imaging

A Leica TCS SP5 confocal microscope was used to image fluorescent samples. The fish were imaged live when possible. They were anesthetized with 0.013% tricane for 10 minutes before being mounted in low-melting agarose containing 0.1% tricane. Laser power was kept as low as possible while still providing a good signal. Temperature was not regulated. After imaging, the embryos were taken out of the agarose and either euthanized as previously described or returned to the incubator.

(10)

6

A Nikon SMZ1500 stereomicroscope equipped with a Nikon DS-Vi1 camera was used to image non-fluorescent samples. Images were edited with ImageJ.

2.3 In situ hybridization

The exon sequence for calcium-dependent secretion activator 2 (cadps2), distal-less homeobox 2a (dlx2a), T-box brain 1b (tbr1b), orthodenticle homolog 5 (otx5) and solute carrier family 17 member 7 (slc17a7) was retrieved from the Ensembl database

(www.ensembl.org), primers designed using the online tool Primer3Plus

(primer3plus.com/cgi-bin/dev/primer3plus.cgi), and the best candidate primers were analyzed for primer dimmers, hairpin structures and self-complementarily using PREMIER Biosoft’s online tool NetPrimer (www.premierbiosoft.com/netprimer). Primer sequences are presented in table 1.

Complementary DNA (cDNA) was synthesized in a T100 Thermal Cycler using

Multiscribe™ Reverse Transcriptase with random hexamers and RNA extract from a mix of embryos from 15 somites, 24 hours post fertilization (hpf), three days post fertilization (dpf) and 10 dpf stages. The cDNA and the primers (table 1) were used to amplify the probe template by polymerase chain reaction (PCR) with GoTaq® G2 Flexi DNA Polymerase (Promega). The amplicons were then subcloned into pGEM®-T vectors (Promega),

transformed into One Shot® TOP10 chemically competent Escherichia coli (Invitrogen), and plasmid was harvested from isolated, single colonies using a miniprep procedure. Probe sequence insertion was verified by sequencing performed by EurofinsGenomics using primers targeting the T7 and SP6 promoter sites flanking the multiple cloning site on either side. The digoxigenin (DIG) or fluorescein labeled RNA-probes were synthesized with Roche-reagents according to manufacturer’s protocol and stored either as aliquots in nuclease free water at - 80 oC or in 50% formamide and 5x saline-sodium citrate (SSC; 3 M NaCl and 0.3 M Na citrate in H2O) buffer at -20 oC.

(11)

7

Table 1. Primer sequences for the genes targeted during the in situ hybridization experiments.

Targeted gene Primer direction

Sequence, 5’-3’ Product size (base pairs)

rgs4 Forward ACCAGCAACATAACCCGCAT

Reverse TGGATGGCCAACAGAAAAACA 545

mCherry Forward GGGCGAGGAGGATAACATGG

Reverse CGTTGTGGGAGGTGATGTCC 620

cadps2 Forward CCTGAAAAGACTTCGGATGC

Reverse TTTTGATTCCCAGGATGTGC 619

dlx2a Forward CGAACCAGATTACCTCCAGC

Reverse TGTTCATTCTCTGGCTGTGC 621

tbr1b Forward TGCCATGACTGGTTCTTTCC

Reverse CCTTGGAGCAGTTTTTCTCG 731

otx5 Forward TTCTCCAAAACCCGATACCC

Reverse GGCGTTGAAGTTCAGTTTCC 648

slc17a7 Forward ACGACCACACGGTTTACTCC

Reverse TGCAGAAGTTGGCTACGATG 666 rgs4 (qPCR) Forward GACTTCTTTTCTCTTGCTTTGG

273/144 Reverse GTCGTTTTTGATCAGGTTGGTA

actb2 (qPCR) Forward GATGATGAAATTGCCGCACTG Reverse ACCAACCATGACACCCTGATGT 135

For the single chromogenic ISH (CISH), zebrafish embryos were fixed in 4%

paraformaldehyde (PFA) in phosphate buffer saline (PBS) overnight at 4 oC and dehydrated through washes of increasing concentrations of methanol in PBS (25%, 50%, 75% and 100%), then stored at -20 oC for at least 20 hours. Prior to incubation with labeled probe, embryos rehydrated though methanol washes with the concentrations in the reverse order (75%, 50%, 25% and 0%), washed in PBS containing 0.1% Tween-20 (PBST) and permeabilized with 5 g/ml proteinase K in PBST for a time appropriate for the

developmental stage of the embryos used. The permeabilization was stopped and the embryos were fixed for 20 minutes in 4% PFA in PBS, after which they were prehybridized in

hybridization buffer (HYB+; 50% formamide, 5xSSC, 0.1% Tween-20, 50 g/ml heparin sulphate and 5mg/ml torula RNA) at either 65 oC or 67 oC for one hour. 150 ng probe was then added, resulting in a final probe concentration of 150 ng/ml HYB+. The samples were incubated overnight at the same temperature used for prehybridization. Samples were gently brought back to the PBST buffer though washes with decreasing concentrations of formamide and SSC. Blocking was done with Blocking Reagent in maleic acid buffer (MAB; 1 M maleic acid and 1.5 M NaCl in H2O) for one hour at room temperature (RT), after which the alkaline phosphatase-conjugated antibody targeting the DIG or fluorescein was added at a 1/2500 or 1/500 concentration respectively. Samples were incubated at 4 oC overnight. Antibody

(12)

8

solution was removed and the embryos washed in MAB for more than one hour, then briefly in PBST. BM purple was used as substrate to stain the samples, and staining was stopped with PBST when sufficient signal was seen. Samples were fixed for 20 minutes in 4% PFA in PBS and left in PBST at 4 oC overnight. They were mounted in glycerol, and then stored at -20 oC after being imaged.

Single fluorescent ISH (sFISH) was performed similarly to the single CISH, with a few exceptions. The washes after hybridization were gentler, with decreasing concentrations of HYB- buffer (HYB+ buffer without heparin sulphate and torula RNA) in 2xSSC followed by long washes with 0.2xSSC and then decreasing concentrations of 0.2xSSC in PBST. The antibodies used were conjugated to horseradish peroxidase (HRP) and the washing afterwards was done with washing buffer. Staining was done with tyramide signal amplification

(TSA™), the substrate being tyramide conjugated to an appropriate fluorophore (i.e either fluorescein or CY3). The staining was done for one hour, after which washing in PBST was done until background signal had been reduced as much as possible. Samples were mounted in low-melting agarose gel and imaged with confocal microscopy.

Double FISH (dFISH) included inactivation of the HRP by incubation in 2% H2O2 for 1 hour after a shorter wash following the first TSA incubation. After HRP inactivation the protocol was repeated from the blocking step, and antibodies targeting the second marker were used together with the second fluorophore. ¨

2.4 Cryosectioning

Zebrafish embryos were fixed for one to four hours in 4% PFA in PBS at RT then freeze- protected with washes of increasing concentrations of sucrose in PBS (10%, 20% and 30%).

They were then mounted in OCT Cryomount™ and frozen at -80 oC for at least one hour, after which they were moved to the Leica CM3050 S CryoStat and incubated in the cutting chamber for more than 30 minutes before being sectioned. The finished sections were incubated at 37 oC in the dark overnight and then stored at -80 oC.

2.5 Drug treatment

Embryos were treated with the drug CCG-4986 (ChemBridge), from here on referred to as CCG, at times ranging from eight hpf to four dpf. The drug was administered via immersion, and was added to the normal water the embryos were raised in to the final concentrations 10 and 50 M. Along with CCG, the final concentration of dimethyl sulphoxide was raised to

(13)

9

1% to increase the uptake of the drug. Before treatment, the chorion of the embryos were either left intact, punctured or removed completely.

2.6 Morpholino injection

The M2rgs4 splice blocking Morpholino oligonucleotide (MO), 5’-

TATTCTGGACCAGTCTTACCTGTTG-3’, was used as a means to knock down rgs4 expression. A solution containing 1/10 phenol red and 1.5ng/nl MO in water was injected immediately into the yolk of eggs in one to four cell stages with a Narishige IM-31 microinjector, in a few different volumes. The embryos were then raised as normal and imaged with confocal microscopy at two dpf.

MO efficiency was analyzed by two-step RT-PCR, requiring RNA to be extracted from all samples. This was done with a trizol-chloroform extraction, where acidic trizol was used to lyse the cells of the samples and chloroform used to separate the RNA into the aqueous phase.

Isopropanol was then used to precipitate the RNA. Complete cDNA was synthesized as mentioned previously and used as template for an RT-qPCR. The RT-qPCR was performed in a 7500 Real Time PCR System with Power SYBR® Green PCR Master MIX (Applied

Biosystems) and qPCR primers for rgs4 and actin 2b (table 1). No-template-controls and no- cDNA-controls were included. The data was analyzed with a 1-way ANOVA with a

Bonferroni’s Multiple Comparison Test using GraphPad Prism 5, where the threshold cycle (CT) was compared between the injected and non-injected sample groups as a relative measurement of the RNA concentration.

3 Results

As an initial part of the characterization, I investigated the number of transgene insertions by checking the frequency of transgenic offspring produced when the tg(rgs4:mCherry) were outcrossed to wildype AB/AB. Three independent outcrossings resulted in 3/6 (50%), 35/89 (39%) and 241/487 (49%) transgenic embryos for a mean percentage of 46%.

3.1 Development of transgene expression

No fluorescence can be seen at one dpf (data not shown), but most structures are present at three dpf (figure 1). There is weak signal in the mesencephalon and telencephalon at two dpf, with the epiphysis fully labeled and the hindbrain mostly so. The main differences after three dpf are enlargement and/or further development of already labeled areas. Images were also

(14)

10

taken at seven dpf, and the signal was consistent with that observed at five dpf (data not shown).

Figure 1. Maximum intensity projections of the head of two to five dpf tg(rgs4:mCherry) zebrafish embryos.

Labeling in the forebrain and midbrain can be seen weakly at two dpf, but no specific structures except the epiphysis (A). At three dpf, the structures labeled in the forebrain and midbrain are visible, and the development thereafter consists of an enlargement or further development of already apparent structures (B-D). Hindbrain structures are close to fully labeled from two dpf (A’-D’), with the addition of a bridge-structure developed at five dpf (D’). Dorsal view. Rostral is up. Scale bars are 100 m.

3.2 Rostrally located expression

The majority of signal originates from labeled neuronal tissue. The most strongly labeled structure is the epiphysis (figure 2D,G), with connective neurites extending ventrally and laterally outwards. An asymmetrical structure can be seen to the right of the epiphysis, and posterior to the pallium, roughly in the area of the right habenula (figure 2D). Other strongly labeled areas are the pallium and the ganglion cell layer (GCL) of the retina. The pallium signal clearly includes labeled cell bodies (figure 2E) and projects down into the subpallium where the signal is more diffuse (figure 2F), suggesting that the signal comes at least in part from neurites. There is also a concentrated mass of mCherry aggregates in the rostral and medial subpallium, possibly indicative of cell bodies although it could also be axonal termination sites. The subpallial signal connects between the hemispheres and continues caudally into the diencephalon.

(15)

11

Figure 2. The labeled areas in the rostral part of the head of tg(rgs4:mCherry) zebrafish embryos. There are a few labeled cells in the olfactory epithelium (arrows in A), along with a few in the olfactory bulb (arrowheads in A). These connect to the subpallium which is weakly labeled (F). The subpallium is next to the pallium, which is more strongly labeled and cell bodies are readily apparent (E). Behind the pallium is an asymmetrical structure in the level of the right habenula (arrow in D). This is connected to the epiphysis (arrowhead in D,G), which is the strongest labeled structure in this fish line. In the ventral diencephalon only diffuse signal with no readily visible cell bodies can be seen (B,C). The outlined caudal zone of periventricular hypothalamus can be seen (arrow in B and C), as well as a more rostral and dorsally located area that connects the hemispheres via a commissure (arrowhead in B and C). There is strong labeling in the GCL and IPL of the retina as well as some labeling in the INL and weak signal in the OPL (H). In cryosections, the signal from photoreceptor cells become clearly visible (arrow in I), and the optic nerve, -chiasm and –tract are very bright (arrowheads in I and J). They end in retinal arborization fields, of which numbers 2, 5 and 7 can be seen here (arrows in J). Dorsal view in A-

(16)

12

G, lateral view in H, coronal view in I-J. Rostral is to the left for A-H, ventral is down for I-J. Scale bars are 50

m for A-D and H-J, 25 m for E-G.

A few cells are also visible in the olfactory epithelium and in the olfactory bulb, with connections to the subpallium (figure 2A). The diffuse labeling in the ventral diencephalon outlines the caudal zone of the periventricular hypothalamus and a more rostral and dorsal structure that extends projections connecting the hemispheres (figure 2B, C). The signal in the eye is strongest in the GCL and the inner plexiform layer (IPL), with weaker signal in the inner nuclear layer (INL) and outer plexiform layer (OPL). In cryosectioned samples the photoreceptor cells also appear strongly labeled (figure 2I), which is not seen in the live embryos. Retinofugal projections are also strongly labeled, making it easy to see the retinal arborization fields (AF) 7 and 10 (figure 3B), the optic tract and the optic chiasm (figure 2B- C, I-J). In sections, AF 2 and 5 can also be visualized (figure 2J).

3.3 Expression in mesencephalon, rhombencephalon and non-neuronal tissue There are labeled periventricular neurons in the stratum periventriculare of the optic tectum, with visible projections extending into the tectal neuropil (figure 3A). There are also a few other nuclei labeled in the mesencephalon, close to the border to the diencephalon; in the ventral thalamus and posterior tuberculum (figure 3D). In the rhombencephalon the labeling is rather widespread throughout the medulla oblongata, while still slightly more concentrated in the vagal lobe (figure 3C). Other notable structures in the hindbrain are the projection bridge connecting the two hemispheres (figure 3C), the projection arch (figure 3E, next to the scale), a rostro-lateral nucleus and the cerebellum (figure 3E). A weaker and more diffuse type of signal, indicative of projections, can be seen extending caudally along both sides of the notochord, with weakly labeled cell bodies scattered along the spinal cord (figure 3F).

(17)

13

Figure 3. The labeled areas in the midbrain, hindbrain and trunk of tg(rgs4:mCherry) zebrafish embryos.

Labeling in the optic tectum can be seen in both tectal neurons (arrows in A) and retinofugal axon arborization fields 7 and 10 (arrows in B). Beneath the optic tectum, a nucleus in the ventral thalamus, as well as in the posterior tuberculum, is labeled (arrows in D). There are labeled cells throughout the medulla oblongata, with an increased concentration in the vagal lobes (C), and labeled projections can be seen forming a bridge-like structure (arrow in C). Weak signal in the cerebellum (arrow in E) and a laterally positioned nucleus in the rostral part of the medulla (asterisk in E), and in distinct projections forming an arch (arrowhead in E). Some signal can also be seen running along the notochord, with both cell bodies and neurites labeled (F). The pronephric tubule is clearly labeled, with the signal gradually tapering off in the pronephric duct (G). The undifferentiated muscles in the pectoral fin are weakly labeled (H). Large peripherally located cell can be seen almost all over the body, here represented by some just behind the pectoral fin (I). Lateral view in A, B, D, F-H, dorsal view in C, E and I. Rostral is to the left. Scale bars are 50 m for A-F and I, 25 m for G, 100 m for H.

In the rest of the body, few structures are labeled. A weak signal can be seen in the

musculature of the pectoral fin (figure 3H), and in large cells in the skin scattered over the whole embryo (figure 3I). There is also some labeling in the pronephros, with the strongest

(18)

14

signal in the pronephric tubule and a gradual weakening of the signal in the pronephric duct (figure 3G). The glomerulus is not labeled.

3.4 In situ hybridization

To confirm some of the areas believed to be labeled in the tg(rgs4:mCherry), we used known marker genes for specific brain regions in ISH to see if overlapping expression occurred.

Based on CISH results, the mCherry signal (figure 4A, G) seems to overlap with tbr1b in the pallium (figure 4C, Mueller et al. 2008), otx5 in the epiphysis (Gamse 2003) and the eye (figure 4E), and with slc17a7 in the cerebellum (Bae et al. 2009), the eye, the epiphysis and the pallium (figure 4F). It might also overlap with rgs4 in the hindbrain and in the pronephric tubule (figure 4B), and with cadps2 in the habenula (figure 4H, Gamse 2003). There does not seem to be any overlap of mCherry and dlx2a in the subpallium (figure 4D, Mueller et al.

2008).

Figure 4. CISH on tg(rgs4:mCherry). mCherry expression is strong in the pineal gland, the pallium, the cerebellum, the hindbrain and the pronephric tubule (A, G). rgs4 in the forebrain is expressed in a nucleus more ventral than anything labeled with the mCherry probe, but they overlap in the pronephric tubule and possibly in hindbrain neurons (B). tbr1b expression seems to overlap with mCherry in the pallium (C). dlx2a is seen in the subpallium, but does not seem to overlap with any strongly labeled structures in the mCherry CHIS (D). otx5 expression overlaps with mCherry in the epiphysis and the eye (E). slc17a7 overlaps with mCherry in the cerebellum, the eye, the epiphysis and the pallium (F). cadps2 is expressed in the habenula (H), and could overlap with the mCherry expression even if the patterns look slightly different. Lateral view in A-F, dorsal view in G-H. Rostral is to the left. Scale bars are 150 m.

To further clarify possible overlapping signals we tried sFISH experiments with rgs4 and mCherry, as well as dFISH using the marker genes together with mCherry. The sFISH signal from the rgs4-pobe (figure 5C) differs from the signal of the dFISH using the same probe

(19)

15

(figure 5A’’). The sFISH shows labeling located more medial, ventral and caudal than the one in the dFISH, which would not co-localize with the labeling of the mCherry probe. The mCherry sFISH (figure 5B) shows a pattern similar to the dFISH (figure 5A-A’), suggesting that it is the rgs4 signal in the dFISH that is nonspecific. The dFISH of the other probes show a similar pattern as the dFISH of rgs4 and mCherry (data not shown), suggesting that only the mCherry labeled signal survived the dFISH protocol.

Figure 5. FISH on tg(rgs4:mCherry) using rgs4 and mCherry probes. The rgs4 signal of the dFISH (A’’) does not resemble the signal in the rgs4 sFISH (C). The mCherry signal of the dFISH (A’), however, does resemble the mCherry sFISH (B), indicating that the signal overlap seen in the dFISH (A) is likely a false positive. Dorsal view. Rostral is to the left. Scale bars are 100 m.

3.5 Drug treatment and Morpholino injection

As a means to affect rgs4-positive cell, I tried to inhibit the rgs4 protein and knock down the gene expression. I used tg(olig2:dsRed) zebrafish to visualize the effect on axonogenesis and motility.

Treating embryos with CCG had no effect on either motor axon development (figure 6B-B’’), or on locomotor behavior (data not shown). No effect could be seen with concentrations of CCG of up to 150 M, treatment initiation at 8 hpf or with chorion removed prior to treatment (data not shown). MO injections also failed to produce any effect on aforementioned motor axon development (figure 6A-A’’). The MO injection was also established not to have had an effect on the mRNA levels of the embryos (figure 7).

(20)

16

Figure 6. Motor axons of treated 2dpf tg(olig2:dsRed) zebrafish embryos. CCG-treatment had no effect on the development of motor axons. No change was seen when treated with 10 µM (A’) or 50 µm (A’’) compared to non-treated control (A). Nor did injections of a splice blocking MO affect the motor axon development. Neither of 0.75 ng MO (B’), 1.5 ng MO (B’’) or 3 ng MO (B’’’) resulted in a difference from the non-injected control (B). Lateral view. Rostral is to the left. Scale bars are 100 m.

Figure 7. A scatter plot of the CT values from a qPCR using cDNA from MO-injected embryos plotted against the sample groups. Slight variations can be seen, but is not statistically significant (P > 0.05).

(21)

17

4 Discussion

I have in this report provided a basic characterization of the tg(rgs4:mCherry) line. As summarized in table 2, several areas are visible, although many of them do not actually contain any mCherry-producing cells.

Table 2. A summary of the labeled areas, with notations of whether the signal comes from cell bodies, projections or both.

Area Cell bodies Projections

Olfactory epithelium X

Olfactory bulb X X

Pallium X

Subpallium X

Epiphysis X

Asymmetrical structure X

Eye X X

GCL X

IPL X

INL X

OPL X

ONL X

Optic nerve X

Optic chiasm X

Optic tract X

AF2 X

AF5 X

AF7 X

AF10 X

OT X X

SPV X X

SFGS+SO X

Ventral thalamus X X

Posterior tuberculum X X

Diencephalic nuclei X

Caudal zone of the periventricular hypothalamus X

Cerebellum X

Rostro-lateral nucleus of the rhombencephalon X

Medulla oblongata X X

Vagal lobe X

Projection arch X X

Projection bridge X

Spinal cord X X

Pronephric tubule X

Muscle cells of the pectoral fin X

Large peripheral cells X

(22)

18

The expression of the construct does not seem to overlap with endogenous rgs4 expression in areas other than the pronephric tubule, and potentially hindbrain neurons. The rgs4 positive area in the forebrain does not seem to overlap with mCherrry signal in the pallium, but

whether or not it does so in the subpallium has yet to be determined. The mCherry CISH does not show the subpallium, likely due to the signal in this region coming from projections. To investigate whether the insertion is in a region close to an external enhancer or repressor, you could sequence the area surrounding the insert, e.g. by inverse PCR (Hui et al. 1998). This also works as an alternative and more exact way of determining the number of transgene insertions. To circumvent the problem of random transgene insertion, the transgenesis could be repeated using a newer technique of producing transgenic zebrafish, which uses the bacterial phage phiC31 to introduce the transgene in a site-directed manner (Mosimann et al.

2013). This can be used to insert the transgene in a gene desert, where it will be free from external enhancers or repressors. Producing the transgenic line with this technique would most likely increase the fidelity of fluorophore expression to the endogenous rgs4.

The uses for a transgenic line such as this come from the visual information it provides about the labeled areas, e.g. in studies where you expect a morphological effect or alteration to occur, or as an aid in characterization of other expression patterns. In this line, single axons are difficult to see, the only regions where you can spot them easily being the optic tectum and the mesencephalic nuclei ventral of it. This diminishes the uses of this line, because it decreases the resolution of the information. Only in the optic tectum is it possible to perform experiments with the expected outcome of altered projections. In other areas, it would not be possible to visualize such results, and the level of detail one can get is on the level of cell bodies. This is true for the areas where such cell bodies are visible, which as seen in table 2 is far from all. In those areas where only diffuse labeling can be seen, it is the projections that could be studied by affecting the origin of the neurites rather than the diffusely labeled area itself. An example of possible studies using this zebrafish lineis retino tectal pathfinding. This entails a model system for axon guidance and requires the visualization of the optic nerve, the optic tract and its termination area (in this case the optic tectum) (Pittman et al. 2008). It might be possible to use the tg(rgs4:mCherry) to study general effects in this, if the study’s focus is on the tract or the AFs, rather than individual axons. Another, slightly broader, example is ablation studies. Such studies in the tectum have been used to investigate the functions it has in visuomotor behaviors (Roeser & Baier 2003), and the use of this line would remove the need to manually label the area with DiI injections.

(23)

19

The weak labeling of projections and the aggregation of the fluorophore, responsible for much of the reduced resolution, might be caused by the fluorophore itself rather than the regulation of it by the promoter. A way of solving this would be to alter the construct and make a new line with the promoter regulating a different fluorophore, e.g. eGFP. This could improve upon the quality of the signal produced by this promoter, but it could also change the expression pattern by inserting into a differently regulated area of the genome.

It was disappointing that neither CCG administration nor MO injections (figure 6, 7) managed to reproduce the effects seen in the paper by Cheng et al. (2013). As mentioned in the

introduction, they found that rgs4 inhibition led to stunted axon growth in motor neurons as well as motility defects. The tg(olig2:dsRed) transgenically labels the motor neurons in the spinal cord, which would have made any morphological alternation easy to spot. The effect Cheng et al. see in their experiment is significant, and they used a 15 M concentration of CCG and 1.5 ng MO. In previous batches of CCG, the LD50 was at approximately 50M (Jonathan Sager, personal communication), while I tried concentrations up to 150 M without any effect on motility or axonogenesis, let alone mortality. The next step to take with the drug treatment would be to use a new batch, in case the results are due to a poor synthesis reaction.

Regarding the MO, heating it up to 65 oC, or even autoclaving it, prior to injection can improve its efficacy, and would be the first step, before trying a new batch of MO as well.

As a continuation of this work, one could improve upon the characterization by confirming and increasing characterization resolution. One could define the neurotransmitter phenotypes in the labeled areas, establish whether or not the labeled cells are indeed neuronal, and confirm whether it is the whole nucleus/area or only a specific part of it that is labeled. For this, optimizing the dFISH protocol will be necessary, and the first thing to try would be decreasing the TSA incubation time and increasing the incubation time with the Blocking Reagent. TSA treatment could be lowered down to 20 minutes, while blocking could go on to 4-5 hours at RT or even overnight at 4 oC. This should help to decrease the background and enhance the signal to noise ratio, thus allowing for detection of weaker signals.

With increased characterization resolution and confirmation of the labeled areas, the

usefulness of this transgenic line would increase despite the relatively low label resolution. So even if this fish line did not turn out as planned, if used correctly it can still be a useful tool.

You just have to find a new angle. As could be true for life in general; if things don’t go as planned, use what you get to plan anew.

(24)

20

5 Acknowledgements

I would first like to thank my supervisor Jonathan Sager for great support and guidance throughout this thesis. You are a great teacher. I would also like to thank my senior supervisor Lina Emilsson, for making me continually consider my progress and so helping me keep on track. I thank my opponents Bryn Farnsworth and Argyris Spyrou for giving me constructive feedback and helping me improve. I would also like to thank Katarzyna Radomska for sharing her excellent RNA extract and access to the tg(olig2:dsRed) zebrafish, Helena Malmikumpu for the help with cryosectioning, and the staff at SciLife for the use of their locales and their excellent caretaking of the zebrafish.

Lastly I would like to thank the gang; JJ, Bryn, Giulia and Filipa, for great company and fun times.

6 References

Arvanitakis L, Geras-Raaka E & Gershengorn MC. 1998. Constitutively Signaling G-Protein-Coupled Receptors and Human Disease. Trends in Endocrinology & Metabolism 9: 27–31.

Bae Y-K, Kani S, Shimizu T, Tanabe K, Nojima H, Kimura Y, Higashijima S & Hibi M. 2009. Anatomy of zebrafish cerebellum and screen for mutations affecting its development. Developmental biology 330:

406–26.

Berman D, Wilkie T & Gilman A. 1996. GAIP and RGS4 Are GTPase-Activating Proteins for the Gi Subfamily of G Protein α Subunits. Cell 86: 445–452.

Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, Vitale L, Pelleri MC, Tassani S, Piva F, Perez-Amodio S, Strippoli P & Canaider S. 2013. An estimation of the number of cells in the human body.

Annals of human biology 40: 463–471.

Chan RK & Otte CA. 1982. Isolation and genetic analysis of Saccharomyces cerevisiae mutants supersensitive to G1 arrest by a factor and alpha factor pheromones. Mol. Cell. Biol. 2: 11–20.

Cheng Y-C, Scotting PJ, Hsu L-S, Lin S-J, Shih H-Y, Hsieh F-Y, Wu H-L, Tsao C-L & Shen C-J. 2013.

Zebrafish rgs4 is essential for motility and axonogenesis mediated by Akt signaling. Cellular and molecular life sciences : CMLS 70: 935–50.

Dohlman H, Apaniesk D, Chen Y, Song J & Nusskern D. 1995. Inhibition of G-protein signaling by dominant gain-of-function mutations in Sst2p, a pheromone desensitization factor in Saccharomyces cerevisiae. Mol.

Cell. Biol. 15: 3635–3643.

Druey KM, Blumer KJ, Kang VH & Kehrl JH. 1996. Inhibition of G-protein-mediated MAP kinase activation by a new mammalian gene family. Nature 379: 742–746.

Emilsson L, Saetre P & Jazin E. 2006. Low mRNA levels of RGS4 splice variants in Alzheimer’s disease:

association between a rare haplotype and decreased mRNA expression. Synapse (New York, N.Y.) 59:

173–6.

(25)

21

Erdely H a, Tamminga C a, Roberts RC & Vogel MW. 2006. Regional alterations in RGS4 protein in schizophrenia. Synapse (New York, N.Y.) 59: 472–9.

Gamse JT. 2003. The parapineal mediates left-right asymmetry in the zebrafish diencephalon. Development 130:

1059–1068.

Gold SJ, Ni YG, Dohlman HG & Nestler EJ. 1997. Regulators of G-protein signaling (RGS) proteins: region- specific expression of nine subtypes in rat brain. The Journal of neuroscience : the official journal of the Society for Neuroscience 17: 8024–37.

Grafstein-Dunn E, Young KH, Cockett MI & Khawaja XZ. 2001. Regional distribution of regulators of G- protein signaling (RGS) 1, 2, 13, 14, 16, and GAIP messenger ribonucleic acids by in situ hybridization in rat brain. Molecular Brain Research 88: 113–123.

Hepler JR, Berman DM, Gilman AG & Kozasa T. 1997. RGS4 and GAIP are GTPase-activating proteins for Gq and block activation of phospholipase C by -thio-GTP-Gq. Proceedings of the National Academy of Sciences 94: 428–432.

Hooks SB, Martemyanov K & Zachariou V. 2008. A role of RGS proteins in drug addiction. Biochemical pharmacology 75: 76–84.

Hui EK-W, Wang P-C & Lo SJ. 1998. Strategies for cloning unknown cellular flanking DNA sequences from foreign integrants. Cellular and Molecular Life Sciences (CMLS) 54: 1403–1411.

Hunt KA, Zhernakova A, Turner G, Heap GAR, Franke L, Bruinenberg M, Romanos J, Dinesen LC, Ryan AW, Panesar D, Gwilliam R, Takeuchi F, McLaren WM, Holmes GKT, Howdle PD, Walters JRF, Sanders DS, Playford RJ, Trynka G, Mulder CJJ, Mearin ML, Verbeek WHM, Trimble V, Stevens FM, O’Morain C, Kennedy NP, Kelleher D, Pennington DJ, Strachan DP, McArdle WL, Mein CA, Wapenaar MC, Deloukas P, McGinnis R, McManus R, Wijmenga C & van Heel DA. 2008. Newly identified genetic risk variants for celiac disease related to the immune response. Nature genetics 40: 395–402.

Kwan KM, Fujimoto E, Grabher C, Mangum BD, Hardy ME, Campbell DS, Parant JM, Yost HJ, Kanki JP &

Chien C-B. 2007. The Tol2kit: a multisite gateway-based construction kit for Tol2 transposon transgenesis constructs. Developmental dynamics : an official publication of the American Association of Anatomists 236: 3088–99.

Larminie C, Murdock P, Walhin J-P, Duckworth M, Blumer KJ, Scheideler MA & Garnier M. 2004. Selective expression of regulators of G-protein signaling (RGS) in the human central nervous system. Brain research. Molecular brain research 122: 24–34.

Lodish H, Kaiser CA, Bretscher A, Amon A, Berk A, Krieger M, Ploegh H & Scott MP. 2013. Molecular Cell Biology. 7:th ed. W. H. Freeman and Company, New York.

Luttrell LM, Ferguson SSG, Daaka Y, Mille WE, Maudsley S, Della Rocca GJ, Lin F-T, Kawakatsu H, Owada K, Luttrell DK, Caron MG & Lefkowitz RJ. 1999. β-Arrestin-Dependent Formation of β2 Adrenergic Receptor-Src Protein Kinase Complexes. Science 283: 655–661.

Min C, Cheong S-Y, Cheong S-J, Kim M, Cho D-I & Kim K-M. 2012. RGS4 exerts inhibitory activities on the signaling of dopamine D2 receptor and D3 receptor through the N-terminal region. Pharmacological Research 65: 213–220.

(26)

22

Mosimann C, Puller A-C, Lawson KL, Tschopp P, Amsterdam A & Zon LI. 2013. Site-directed zebrafish transgenesis into single landing sites with the phiC31 integrase system. Developmental dynamics : an official publication of the American Association of Anatomists 242: 949–63.

Mueller T, Wullimann MF & Guo S. 2008. Early teleostean basal ganglia development visualized by zebrafish Dlx2a, Lhx6, Lhx7, Tbr2 (eomesa), and GAD67 gene expression. The Journal of comparative neurology 507: 1245–57.

Pittman AJ, Law M-Y & Chien C-B. 2008. Pathfinding in a large vertebrate axon tract: isotypic interactions guide retinotectal axons at multiple choice points. Development (Cambridge, England) 135: 2865–71.

Riddle EL, Schwartzman RA, Bond M & Insel PA. 2005. Multi-tasking RGS proteins in the heart: the next therapeutic target? Circulation research 96: 401–11.

Roeser T & Baier H. 2003. Visuomotor Behaviors in Larval Zebrafish after GFP-Guided Laser Ablation of the Optic Tectum. J. Neurosci. 23: 3726–3734.

Serafimidis I, Heximer S, Beis D & Gavalas A. 2011. G protein-coupled receptor signaling and sphingosine-1- phosphate play a phylogenetically conserved role in endocrine pancreas morphogenesis. Molecular and cellular biology 31: 4442–53.

Shin J, Park H-C, Topczewska J, Mawdsley D & Appel B. 2003. Neural cell fate analysis in zebrafish using olig2 BAC transgenics. Methods in Cell Science 25: 7 – 14.

Smyth DJ, Plagnol V, Walker NM, Cooper JD, Downes K, Yang JHM, Howson JMM, Stevens H, McManus R, Wijmenga C, Heap GA, Dubios PC, Clayton DG, Hunt KA, van Heel DA & Todd JA. 2008. Shared and Distinct Genetic Variants in Type 1 Diabetes and Celiac Disease. New England journal of medicine 359:

2767–2777.

Tan CM, Brady AE, Nickols HH, Wang Q & Limbird LE. 2004. Membrane trafficking of G protein-coupled receptors. Annual review of pharmacology and toxicology 44: 559–609.

Taymans J-M, Leysen JE & Langlois X. 2003. Striatal gene expression of RGS2 and RGS4 is specifically mediated by dopamine D1 and D2 receptors: clues for RGS2 and RGS4 functions. Journal of neurochemistry 84: 1118–27.

Tekumalla PK, Calon F, Rahman Z, Birdi S, Rajput AH, Hornykiewicz O, Di Paolo T, Bédard PJ & Nestler EJ.

2001. Elevated levels of ΔFosB and RGS9 in striatum in Parkinson’s disease. Biological Psychiatry 50:

813–816.

Traynor J. 2010. Regulator of G protein-signaling proteins and addictive drugs. Annals of the New York Academy of Sciences 1187: 341–352.

Watson N, Linder ME, Druey KM, Kehrl JH & Blumer KJ. 1996. RGS family members: GTPase-activating proteins for heterotrimeric G-protein alpha-subunits. Nature 383: 172–175.

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 : an official publication of the American Association of Anatomists 240: 2539–47.

References

Related documents

Ree, H.J., et al., Coexpression of Bcl-6 and CD10 in diffuse large Bcell lymphomas: significance of Bcl-6 expression patterns in identifying germinal center B-cell

Industrial Emissions Directive, supplemented by horizontal legislation (e.g., Framework Directives on Waste and Water, Emissions Trading System, etc) and guidance on operating

Re-examination of the actual 2 ♀♀ (ZML) revealed that they are Andrena labialis (det.. Andrena jacobi Perkins: Paxton & al. -Species synonymy- Schwarz & al. scotica while

This seems to indicate that CNN minimizes any negative agency of “our side” more than Fox does, although given that kill is also missing as a collocation for ISIS, the difference

Stöden omfattar statliga lån och kreditgarantier; anstånd med skatter och avgifter; tillfälligt sänkta arbetsgivaravgifter under pandemins första fas; ökat statligt ansvar

46 Konkreta exempel skulle kunna vara främjandeinsatser för affärsänglar/affärsängelnätverk, skapa arenor där aktörer från utbuds- och efterfrågesidan kan mötas eller

För att uppskatta den totala effekten av reformerna måste dock hänsyn tas till såväl samt- liga priseffekter som sammansättningseffekter, till följd av ökad försäljningsandel

Tillväxtanalys har haft i uppdrag av rege- ringen att under år 2013 göra en fortsatt och fördjupad analys av följande index: Ekono- miskt frihetsindex (EFW), som