Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Neuroscience

In utero Electroporation followed by Primary Neuronal Culture for Studying Gene Function in Subset of Cortical Neurons

Published: October 8, 2010 doi: 10.3791/2103

Summary

In utero electroporation is a valuable method for transfecting neuronal progenitor cells in vivo. Depending upon the placement of the electrodes and the developmental timepoint of electroporation, certain subsets of cortical cells can be targeted. Targeted cells can then be analyzed in vivo or in vitro for effects of genetic alteration.

Abstract

In vitro study of primary neuronal cultures allows for quantitative analyses of neurite outgrowth. In order to study how genetic alterations affect neuronal process outgrowth, shRNA or cDNA constructs can be introduced into primary neurons via chemical transfection or viral transduction. However, with primary cortical cells, a heterogeneous pool of cell types (glutamatergic neurons from different layers, inhibitory neurons, glial cells) are transfected using these methods. The use of in utero electroporation to introduce DNA constructs in the embryonic rodent cortex allows for certain subsets of cells to be targeted: while electroporation of early embryonic cortex targets deep layers of the cortex, electroporation at late embryonic timepoints targets more superficial layers. Further, differential placement of electrodes across the heads of individual embryos results in the targeting of dorsal-medial versus ventral-lateral regions of the cortex. Following electroporation, transfected cells can be dissected out, dissociated, and plated in vitro for quantitative analysis of neurite outgrowth. Here, we provide a step-by-step method to quantitatively measure neuronal process outgrowth in subsets of cortical cells.

The basic protocol for in utero electroporation has been described in detail in two other JoVE articles from the Kriegstein lab 1, 2. We will provide an overview of our protocol for in utero electroporation, focusing on the most important details, followed by a description of our protocol that applies in utero electroporation to the study of gene function in neuronal process outgrowth.

Protocol

The basic protocol for in utero electroporation has been described in detail in another JoVE article from the Kriegstein lab 1, 2. This technique was originally described in the Osumi lab 3 and our protocol is based upon one developed in the LoTurco lab 4. We will provide an overview of the our protocol for in utero electroporation of rat embryos, focusing on the most important details, followed by a description of our protocol that applies in utero electroporation to the study of gene function in neuronal process outgrowth.

1. In utero Electroporation

  1. Preparing DNA and loading needles
    The first step for in utero electroporations is to design your experiment to determine what DNA constructs you want to inject. This method is useful for both misexpressing or knocking down genes of interest. If you are planning on misexpressing or overexpressing a gene, be sure to use a promoter that is active in neuronal precursor cells. We recommend the CAGGS promoter, which consists of the chicken beta-actin promoter and the CMV enhancer 5. Since only a small subset of cells are transfected using in utero electroporation, it is critical to include a plasmid encoding a fluorescent protein such as GFP so that you can follow those cells that were successfully electroporated. For the plasmid encoding GFP, we recommend preparing the DNA at a concentration of 0.5 μg per microliter. For shRNA constructs, we have found that 0.5-1.0 μG per μL results in efficient knock down of your gene of interest. For overexpression or misexpression, we use between 1.0 and 3.0 μG per microliter, depending upon the size of the gene and the level of expression that the experiment calls for. DNAs are prepared using a Qiagen endotoxin-free prep kit, and diluted in 1 x PBS. We inject approximately 0.5-1.0 μL per embryonic brain, so, for a litter of animals we prepare 10 μL of DNA mixture for injection. We add 1 μL of Fast Green to the DNA so that we can follow the injected DNA.
    Pulling needles to the correct shape is a critical step. Walantus et al. uses a different system for delivering the DNA and so their needle prep is also slighly different then ours1,2. The settings that you use to pull your needles will depend upon the brand of needle puller that you have. We use Model 750 from David Kopf. The settings we use are : Heat 1: 9.0, Heat 2: 0, Soleniod: 0, Filament size 3.0 mm, Heater Proximity: 3 mm, Time: 10 sec. Once pulled, we cut our needles with a razor blade at a ~45 degree angle such that the distance from the largest part of the opening to the tip is 11 mm. We then load the DNA from the back end of the needle. We then fill the remaining space in the needle with corn oil. For DNA injection, we use a Picospritzer III. Depending upon the exact bevel that is cut for each needle, we set the Picospritzer from 4.0 to 6.0. We use a foot pedal to deliver the pressurized air that expels the DNA from the needle.
  2. Preparing animals for surgery
    We use pathogen-free Sprague Dawley rats exclusively for these surgeries. Several other labs use mice of varying genotypes as well. Here, we describe show our protocol for electroporation of E15 rat embryos, but in utero electroporation is routinely performed in rats between the ages of E13 and E18. While early stage electroporation targets deep layers of the cortex, later stage electroporations target more superficial layers.
    Animals are given a pre-operative dose of buprenorphine (0.05-0.1 mg/kg) before the surgery starts. There are multiple options for anesthetizing the animal. Walantus et al. utilizes isofuorane inhalation, while we routinely use intraperitoneal injection of ketamine (40-80 mg/kg) and xylazine (5-10 mg/kg)1,2. A toe pinch should always be performed to ensure that animals are fully anesthetized and unresponsive. Animals are kept on a heated pad throughout the surgical procedure.
    The animal's fur is shaved in the region of incision, and washed three times with ethanol followed by three times with iodine. An incision is made in the skin just lateral to the midline, followed by an incision in the muscle. The uterine horns are exposed very carefully. They are gently teased out of the body cavity using your fingertips. Keep embryos hydrated with sterile PBS while they are outside of the body cavity.
  3. Injecting DNA and electroporation
    When you first start performing these surgeries, the hardest part is becoming familiar with where you need to inject the DNA so that you fill the lateral ventricles, and getting used to how deep you inject your needle in order to hit the correct region. Embryos are gently manipulated with your fingertips so that you can identify where the head is, and if you look closely you will be able to see the midline suture. This serves as a general landmark that you can use to determine where the lateral ventricle is located. We inject the DNA through the uterine wall and into the lateral ventricle. We use a footpedal to control the injection of the DNA - multiple pulses of DNA are performed until the lateral ventricle is filled with the DNA/dye mixture. We then place paddle electrodes on either side of the head of the embryo and use another footpedal to deliver the pulse across the head of the embryo. The placement of the electrodes is critical in determining which region of the cortex is electroporated. Since DNA is negatively charged, the DNA will travel toward the positive electrode when a charge is dispelled across the paddles. Depending on the exact placement of the electrodes, different subsets of cells will be targeted. We routinely place the positive electrode near the dorsal-medial positions across the cerebrum. However, the LoTurco lab beautifully showed that if you place the electrodes in more ventral lateral regions of the cerebrum you can target the cells of the cortical-striatal boundary and hit cells of the lateral cortical stream 6. Each embryo can be electroporated, and different combinations of DNA constructs can be used in each embryo.
  4. Suturing and post-operative care
    Following electroporation of all embryos, the uterine horns are carefully returned to the body cavity, and both the muscle layer and the skin are sutured. The technique for this is outlined in Walantus et al1,2. Animals are monitored continuously until they recover from anesthesia, and the analgesic buprenorphine (0.05-0.1 mg/kg) is administered every 8-12 hours.

2. Culturing Electroporated Cortical Neurons

  1. Harvesting electroporated brains and dissecting electroporated region
    For in vivo analyses following in utero electroporation, animals can be harvested at any time point from 24 hours following electroporation to early after birth to adulthood. However, when culturing primary neurons we harvest 24 hours following electroporation at E16. At this time, the electroporated embryos are expressing detectable levels of GFP.
    Animals are euthanized using carbon dioxide inhalation and rapid decapitation. Embryos are dissected out of the uterus and placed in HBSS with divalent cations, keeping track of which embryos were electroporated with which DNA plasmids. It is critical to use filtered HBSS, sterile tubes and plates, and autoclaved tools for dissection. The cortices are dissected out and the meninges removed using a microscope in a hood. These cortices are then observed under a dissecting microscope with the capacity to visualize GFP. GFP positive regions of the cortex are identified, and we ue a pair of vanna scissors to cut out the GFP positive regions from the cortex. These pieces are placed in HBSS without divalent cations in a 15 ml conical tube.
  2. Dissociating and plating neurons
    Once all GFP positive regions are dissected, HBSS is replaced with 0.25% trypsin, and incubated at 37 degrees for 5 minutes. Trypsin is removed, replaced with plating media (DMEM + 5% FBS + Penn/strep + glutamine), and triturated 5-7 times to dissociate the cells. Volumes used depend upon the amount of tissue present. Dissociated cells are then plated on CC2 coated chamber slides. For two chamber slides, we plate 200,000-350,000 cells per chamber in a volume of 1.5 mL of plating media. After 4 hours, plating media is aspirated and replaced with 1.5 mL Neuronal culture media (Neurobasal media + B27 supplement + glutamax + gentamycin) per chamber.

3. Analyzing Neuronal Process Outgrowth

  1. Fixing and immunostaining cultures
    In order to measure short term effecs of genetic manipulation of these cells, we harvest the primary neurons after three days in vitro. If primary neurons are to be cultured longer for additional analyses, half of the media should be replaced every three days. For fixing cultures, we aspirate the media from the chambers, and fix the neurons in 4% paraformaldhyde for 15 minutes. Following fixation, cells are washed two times in PBS and then put in blocking solution (2% donkey serum with 0.1% Triton X-100 in PBS) for one hour. Cells are then incubated in primary antibody for 1 hour. For analyses of neuronal process outgrowth, we use anti-beta tubululin antibody to identify neurons - beta II tubulin immunostaining labels the neruonal cell body, dendrites and axons. Cells are then washed three times in PBS for five minutes, and then incubated in Cy3-anti mouse for 1 hour, followed by three more PBS washes, counterstaining nuclei with DAPI and mounting.
  2. Measuring neurite length
    Images of GFP positive, beta-III tubulin positive neurons are acquired on a Zeiss Axioskop with a MC100 camera system. Several variables can be examined in these GFP positive cells including length of all neurites, length of the longest neurite, branching of neurites, size of cell soma, etc. We have used this method to analyze neuronal process outgrowth upon knock down or overexpression of genes relating to neurodevelopment and neurodegeneration, with a focus on neuronal process length. In order to measure neuronal processes outgrowth, we use Axiovision LE 4.4 software (from Zeiss). Within this software, there is an option to select the "outline" tool. Using this tool, you can use your computer's mouse to trace the length of each neuronal process. It is critical to define the objective that you are using in order to get an accurate measure of your neurites. For these analyses, we usually use a 20x objective.

4. Representative Results

We have found that Sprague Dawley litter size ranges between 6 and 14 embryos. We usually electroporate all of the embryos. Each embryo can be electroporated with a different combination of DNAs. However, we usually electroporate at least four brains with the same condition and pool these brains before dissociating and plating.

We have found that with this technique approximately 75% of electroporated brains are targeted to the desired region of the cortex, whether that be dorsal medial or ventral lateral cortex (Figure 1). In addition, we have found early electroporations at E13-14 target deep layer neurons such as Tbr1 positive layer VI neurons, while later electroporations target CTIP2 positive, TBR1 negative layer V cells, and still later electroporations target Brn2 positive layer II/III cells. An excellent description of different markers and explanation of neuronal subtype specification in the cortex in found an article by Moleneaux et al 7. Figure 2 shows coronal sections of brains electroporated at either embryonic day 15.5 or 17.5 and harvested at postnatal day 5. Shown in red is immunostaining for Oct6. You can immunostain for markers in culture to confirm what cell layer populations you have targeted. We have found that you can expect to target the same cell layer population of cells in every embryo of the same litter (in other words, the targeting depends upon the embryonic timepoint rather then on other technical variations).

In culture, the percent of cells that are GFP positive can range widely depending on how conservative you are when dissecting out the GFP positive region (Figure 3). However, even when we are very conservative and dissect out only the GFP positive patch of cells, the highest percentage that we observe is 5-10% - although you are dissecting the region of the cortex that was electroporated, cells in only one layer will be targeted. This low transfection effciency is helpful in identifying which processes belong to the electoporated cell that you are analyzing. Plating cells at this higher density contributes to having heathier cultures, however, it is difficult to discern which process belongs to which cell body in the GFP negative cells (Figure 3).

If you have trouble seeing all of the fine processes of the electroporated cells, you can either increase the concentration of GFP DNA that you are electroporating to increase expression of GFP, or you can immunostain the dissociated cells using an anti-GFP antibody (from Invitrogen) along with a Cy2 secondary antibody.

Figure 1
Figure 1. E15.5 Sprague-Dawley rats were electroporated with GFP plasmid and harvested three days later. Based upon the placement of the electrodes, different regions of the cortex will be targeted. A-F show GFP fluorescence in whole brains.

Figure 2
Figure 2. E15.5 (A) or E17.5 (B) Sprague-Dawley rats were electroporated with GFP plasmid and harvest at postnatal day 5. Brains were fixed, sectioned coronally using a vibratome (100 micron sections), and immunostained for Oct6 (red). A and B show confocal images of immunostained sections.

Figure 3
Figure 3. E15.5 Sprague-Dawley rats were electroporated with GFP plasmid. 24 hours following electroporation, brains were harvested and GFP-positive, electroporated regions were dissected and dissociated, as described in the video. After 3 days in vitro, cells were fixed and immunostained for bIII-tubulin (red) and staining nuclei with DAPI (blue) (A,C). The length of the longest neurite was measured using Axiovision software (B,D).

Subscription Required. Please recommend JoVE to your librarian.

Discussion

In vitro study of primary neuronal cultures allow for quantitative analyses of neurite outgrowth. In order to study how genetic alterations affect neuronal process outgrowth, shRNA or misexpression constructs can be introduced into primary neurons via chemical transfection or viral transduction. However, with primary cortical cells, a heterogeneous pool of cell types (glutamatergic neurons from different layers, inhibitory neurons, glial cells) are transfected using these methods. The use of in utero electroporation to introduce DNA constructs in the embryonic rodent cortex allows for certain subsets of cells to be targeted: while electroporation of early embryonic cortex targets deep layers of the cortex, electroporation at late embryonic timepoints targets more superficial layers. Further, differential placement of electrodes across the heads of individual embryos results in the targeting of dorsal-medial versus ventral-lateral regions of the cortex.

Targetting of specific layers:

When you inject DNA into the lateral ventricle and electroporate, only the cells immediately lining the lateral venrtricle are targeted: these cells are the radial glial progenitor cells of the neocortex, as well as cells that have just undergone their terminal mitosis. Interestingly, when examined in the days following electroporation, the cells targeted match the pattern of birthdated cells. In other words, cells that are born , that is to say those that undergo their terminal mitosis on the day of electropoartion, are the cells that are targeted. Since radial glial cells also are present at the ventricular surface, one might hypothesize that these cells would be targeted, and that all of the progeny from these cells would also be targeted. However, this is not the case, later generations of cells do not express GFP. It may be that multiple rounds of division in the radial glial cells dilutes out the plasmid.

Applications:

This method is an excellent way to examine the effects of gene knock down via electroporation of shRNA contructs, as well as by misexpression of cDNA constructs. We are applying this techniqe to the study of genes involved in neurodegeneration and psychiatric disease. Through this technique, we introduce both wild type and mutant forms of genes critical in these diseases, and examine the effects on neuronal morphology. In addition, we can examine the effects of mutation or alternative splice variant expression in the absence of the endogenous gene product by co-electroporating the cDNA and the shRNA contruct. Co-electorporation also can be utilized to look at genetic interactions between two gene products: by knocking down multiple genes and by attempting to rescue effects of knock down of one gene product with other gene products 8, 9.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

No conflicts of interest declared.

Acknowledgments

The authors would like to thank Joseph LoTurco and Dennis Selkoe for helpful discussions on this technique. The authors thank the donors of the American Health Assistance Foundation, for support of this research.

Materials

Name Company Catalog Number Comments
10X Hanks’ Balanced Salt Solution (HBSS) (Ca+2 /Mg +2 free) GIBCO, by Life Technologies 14185-052
10X Hanks’ Balanced Salt Solution (HBSS) (with Ca+2 /Mg +2 ) GIBCO, by Life Technologies 14065-056
1M HEPES pH 7.4 GIBCO, by Life Technologies 15630-080
100 x 15 mm Petri Dishes Fisher Scientific 08-757-12
60 x 15 mm Petri Dishes BD Biosciences 351007
15 mL conical vial Sarstedt Ltd 62-547-205
50 mL conical vial Sarstedt Ltd 62-554-205
Scissors Fine Science Tools 91402-12
Standard Forceps Fine Science Tools 11000-12
Curved Forceps Fine Science Tools 11273-20
Fine Forceps Fine Science Tools 11255-20
Vannas spring scissors Fine Science Tools 15000-00
.25% Trypsin-EDTA GIBCO, by Life Technologies 25200
Reichert Bright-Line Hemacytometer Hausser Scientific 1490
Hand-Held Tally Counter Sigma-Aldrich Z169021
Dulbecco’s Modified Eagle Medium (D-MEM) GIBCO, by Life Technologies 11960-051
Fetal Bovine Serum Sigma-Aldrich F4135
Penicillin-Streptomycin GIBCO, by Life Technologies 15140
L-glutamine GIBCO, by Life Technologies 25030
NEUROBASAL Medium GIBCO, by Life Technologies 21103-049
B-27 Serum-Free Supplement GIBCO, by Life Technologies 17504-044
GlutaMAX -I Supplement GIBCO, by Life Technologies 35050-061
Gentamicin Reagent Solution GIBCO, by Life Technologies 15750-060
Paraformaldehyde Sigma-Aldrich P6148
Phosphate Buffered Saline Sigma-Aldrich P4417
Triton X-100 Sigma-Aldrich T9284
Donkey Serum Jackson ImmunoResearch 017-000-121
beta-III tubulin antibody Chemicon International MAB1637
MAP2 antibody Chemicon International AB15452
Donkey Cy3 anti-mouse Jackson ImmunoResearch 715-166-151
Donkey Cy2 anti-chicken Jackson ImmunoResearch 703-226-155
DAPI GIBCO, by Life Technologies D3571
CC2 Coated Two-Chamber Slides Lab-Tek 154852
Fluorescent Mounting Media KPL Inc 71-00-16
24 x 60 mm Micro Cover Glasses VWR international 48393-106
Clear nail polish Electron Microscopy Sciences 72180
Ketamine Henry Schein 995-2949
Xylazine Henry Schein 4015809TV
buprenorphine Henry Schein 1118217
Picospritzer III Parker Hannifin Corporation
BTX square wave electroporator Fisher Scientific BTXECM830
Tweezertrodes, 7 mm, platinum Harvard Apparatus 450488

DOWNLOAD MATERIALS LIST

References

  1. Walantus, W., Castaneda, D., Elias, L., Kriegstein, A. In utero intraventricular injection and electroporation of E15 mouse embryos. J Vis Exp. , (2007).
  2. Walantus, W., Elias, L., Kriegstein, A. In utero intraventricular injection and electroporation of E16 rat embryos. J Vis Exp. , (2007).
  3. Takahashi, M., Sato, K., Nomura, T., Osumi, N. Manipulating gene expressions by electroporation in the developing brain of mammalian embryos. Differentiation. 70 (4-5), 155-1562 (2002).
  4. Bai, J., Ramos, R. L., Ackman, J. B., Thomas, A. M., Lee, R. V., LoTurco, J. J. RNAi reveals doublecortin is required for radial migration in rat neocortex. Nat Neurosci. , 1277-1283 (2003).
  5. Okada, A., Lansford, R., Weimann, J. M., Fraser, S. E., McConnell, S. K. Imaging cells in the developing nervous system with retrovirus expressing modified green fluorescent protein. Exp Neurol. 156 (2), 394-406 (1999).
  6. Bai, J., Ramos, R. L., Paramasivam, M., Siddiqi, F., Ackman, J. B., LoTurco, J. J. The role of DCX and LIS1 in migration through the lateral cortical stream of developing forebrain. Dev Neurosci. , 144-156 (2008).
  7. Molyneaux, B. J., Arlotta, P., Menezes, J. R., Macklis, J. D. Neuronal subtype specification in the cerebral cortex. Nat Rev Neurosci. , 427-437 (2007).
  8. Young-Pearse, T. L., Bai, J., Chang, R., Zheng, J. B., Loturco, J. J., Selkoe, D. J. A Critical Function for -Amyloid Precursor Protein in Neuronal Migration Revealed by In Utero RNA Interference. J Neurosci. 27, 14459-14469 (2007).
  9. Young-Pearse, T. L., Chen, A. C., Chang, R., Marquez, C., Selkoe, D. J. Secreted APP regulates the function of full-length APP in neurite outgrowth through interaction with integrin beta1. Neural Develop. 3, 15-15 (2008).

Tags

In Utero Electroporation Primary Neuronal Culture Gene Function Subset Of Cortical Neurons Neurite Outgrowth Genetic Alterations ShRNA CDNA Constructs Chemical Transfection Viral Transduction Heterogeneous Pool Of Cell Types Glutamatergic Neurons Inhibitory Neurons Glial Cells Embryonic Rodent Cortex Deep Layers Of The Cortex Superficial Layers Of The Cortex Dorsal-medial Regions Of The Cortex Ventral-lateral Regions Of The Cortex Quantitative Analysis Step-by-step Method
<em>In utero</em> Electroporation followed by Primary Neuronal Culture for Studying Gene Function in Subset of Cortical Neurons
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Rice, H., Suth, S., Cavanaugh, W.,More

Rice, H., Suth, S., Cavanaugh, W., Bai, J., Young-Pearse, T. L. In utero Electroporation followed by Primary Neuronal Culture for Studying Gene Function in Subset of Cortical Neurons. J. Vis. Exp. (44), e2103, doi:10.3791/2103 (2010).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter