SlideShare a Scribd company logo
1 of 20
The Use of Recombinant Adeno-Associated Viral Vectors in Gene Therapy to Treat

                                         Epilepsy

                                      Omega Cantrell

Abstract

   Epilepsy is defined as being recurrent, unprovoked seizures, and naturally occurs in a

wide range of species. It affects approximately 1% of the U.S. population, and is a good

target for gene therapy by recombinant adeno-associated virus (rAAV) vectors. rAAV is

capable of stably transferring foreign genetic material to neurons (transduction), in

addition to being able to infect a wide range of organisms and cell types, but, in vector

form, is not pathogenic. The neuropeptides galanin (GAL) and neuropeptide Y (NPY) are

found abundantly in the central nervous system, have strong anticonvulsant effects and

are promising transgene options for rAAV vectors. Galanin is thought to have more of an

impact than NPY. Studies have shown that GAL is able to reduce time spent in seizures

by 77% and number of EEG-detected seizures by 40%. Following seizures induced by

electrical stimulation in the perforant path of the hippocampus, the number of GAL-

positive neurons was 16 times that of the control group. Because GAL was able to more

significantly decrease the rate of seizure occurrence and total time spent in seizure

activity, it is a better option as a transgene for rAAV in gene therapy for epilepsy and

other seizure disorders.

Key words: epilepsy, gene therapy, recombinant adeno-associated virus (rAAV),

galanin (GAL), neuropeptide Y (NPY), transgene




                                                                                            1
Introduction

   Normal brain function involves neuron-to-neuron transmission, causing signals from

the brain to be sent to the rest of the body. For this to occur, sodium channels open,
  Figure 1. Normal brain wave patterns.
                                               allowing sodium (Na+) ions to enter. This

                                               operates on a negative feedback loop, such

                                               that potassium acts to cease signaling by

                                               closing the sodium channels. As Figure 1

                                               illustrates, on an electroencephalograph

                                               (EEG), normal brain patterns appear as small


  http:/www.epilepsy.org.au/images/ElectroEn
                                               waves. When abnormal brain waves are seen
  cephalogram.png
                                               on an EEG, a seizure disorder is usually to

blame (NIH, 2004; Hains, 2006).

   Epilepsy is a widely occurring seizure disorder in many species, including rats, dogs,

and cats, and is the most common acquired neurological disorder in humans (Chandler,

2006). It has been defined as recurrent               Figure 2. Brain wave patterns observed
                                                      during an epileptic seizure.
unprovoked seizures, and has a prevalence of

0.5-1.0% in humans, with a higher rate of

occurrence in the underdeveloped world

(Ransom and Blumenfeld, 2007).There are

many different kinds of epilepsy, and these

disorders are diagnosed based on many factors.

                                                  http://brain.fuw.edu.pl/~suffa/SW/SW_patt.gif



                                                                                                  2
Figure 2 illustrates EEG-detected abnormalities, and other factors include age of onset,

characteristics of the seizure, and seizure induction stimuli (Chandler, 2006). As can be

seen in figure 2, epileptic brain waves appear as high spikes in rapid succession on an

EEG. Seizure intensity is determined by the height of the spikes, coupled with the rate of

succession. Higher, more rapid spikes are indicative of a more severe seizure (Hains,

2006).

   Epilepsy is thought to be caused by an imbalance in neurotransmissions (Vezzani,

2004). As mentioned before, normal neurotransmissions are facilitated by a negative

feedback loop between sodium, which opens the channels, and potassium, which closes

the channels. In a normal brain, neurons fire approximately 30 times each second. During

a seizure, neuronal firing can occur as many as 500 times each second (NIH, 2004).

Normally, when excitatory neurotransmitters such as glutamate are released, neuronal

firing occurs. Once this reaches a certain level (which varies among individuals, and

according to the type of signaling that is occurring), another negative feedback loop

occurs. When this happens, an inhibitory neurotransmitter such as γ-aminobutyric acid

(GABA) serves to stop the action of the excitatory neurotransmitters and cease neuronal

firing (NIH, 2004). Because of this, another common theory is that either a high level of

excitatory neurotransmitters or low level of inhibitory neurotransmitters are responsible

for the abnormal neuronal firing that causes seizures to occur.

   Having just one seizure does not mean a person has epilepsy. In order to be

considered for a diagnosis of epilepsy, a person must have had two or more seizures.

There are two main categories of epilepsy – focal (or partial), which affects only one part

of the brain. This includes temporal lobe epilepsy, which, according to Ransom and



                                                                                            3
Blumenfeld (2007), is more common in adults, and is often resistant to medical therapy.

Generalized seizures affect both of the brain’s hemispheres simultaneously (NIH, 2004).

As detailed previously, a seizure occurs when an imbalance in neurotransmitters causes

sodium channels in the brain to remain open, resulting in abnormal neuronal firing. This

abnormal firing results in symptoms of the many different types seizures, including

muscle spasms, repeated movements (also called automatisms), loss of consciousness,

and/or temporary loss of muscle tone.

   These disorders are commonly controlled with medications that operate in a variety of

ways, and are prescribed according to the type or types of seizures the patient has. A

common mode of action for an antiepileptic medication is to either close or block the

sodium channels in the brain, preventing neuronal hyperactivity. In doing this, these

drugs can help to control seizures, but are not guaranteed to prevent seizure occurrence in

all who take them. According to Riban et al. (2009), about one-third of diagnosed

epileptics suffer from a form of the disease resistant to anticonvulsant drugs. In these

more severe cases, surgery may be necessary. This is often a last resort for doctors, and is
                                                                 Figure 3. The human brain,
usually done only if a patient continues to suffer from          arrow pointing to the corpus
                                                                 callosum.
seizures after years of treatment

with different types of

anticonvulsant drugs. The

National Institutes of Health

recognizes only three categories

of seizures that can be treated

successfully with surgery, one of

                                            http://static.guim.co.uk/sys-
                                            images/Guardian/Pix/pictures/2009/4/6/1239055
                                            717363/Cross-section-of-the-huma-001.jpg            4
these being focal seizure disorders, which includes temporal lobe epilepsy, and has a 64%

success rate when compared with treatment with prescription medication alone (NIH,

2004). Surgery should not be taken lightly, however, as it often involves either removal

of an area in the brain where seizures are observed to occur most frequently (seizure

focus area), or, in more severe cases, cutting the corpus callosum (illustrated in figure 3).

This disconnects the two hemispheres of the brain, and does not stop those seizures

focused in one area of the brain (focal seizures), but merely prevents them from

spreading across the whole brain (NIH, 2004). Because there are so few options for

epilepsy treatment, and such a high incidence of medication-resistant epilepsies, gene

therapy – especially when mediated by recombinant adeno-associated virus (rAAV) – is

considered to be a promising new option for those who suffer from a more severe seizure

disorder.

   To better understand how rAAVs are used for gene therapy, it is necessary to

understand how a vector is constructed. A virus operates by first infecting a host cell,

then replicating its own genome inside a host cell. This amplifies viral infection in the

host organism, and after the host cells are lysed, these viruses can further infect more host

cells, resulting in a widespread infection. Viral vectors are genetically engineered in the

sense that their life cycles are manipulated such that the beneficial stages responsible for

 Figure 4. Genome of an AAV vector, introverted terminal repeat sequences (ITRs) in red boxes.




 Gene Therapy Approaches in Neurology (2007).           genomic replication are retained, while

the deleterious stages responsible for viral replication and cell lysis are eliminated

(Burton et al., 2007). Figure 4 depicts the relatively small (~4.5kb) rAAV genome, which


                                                                                                  5
codes for just two genes: rep and cap, and has an introverted terminal repeat sequence

(ITRs) at each end of its single-stranded DNA (Carter, 2008). To construct a vector, rep

and cap are first removed and then replaced with a coding sequence of a similar length

(4.5kb), while the ITRs are retained as “stops” for the genetic code (Burton et al., 2007).

The removal of rep has two purposes: the first – and the most obvious reason – is to make

room for the coding sequence intended for therapy, and the second being to make rAAVs

incapable of attaching to human DNA. While not a problem with animals tested in the

laboratory, the rep gene allows rAAVs to adhere to a site on human DNA chromosome

19 (Carter, 2008). Because this is a potential risk, rep is removed.

   Recombinant adeno-associated virus (rAAV) is an example of a viral vector – a

genetically engineered virus (Burton et al., 2007) – and has garnered attention in recent

years as a promising tool in gene therapy to potentially treat disorders such as epilepsy.

  Figure 5. An adeno-associated virus and its structural components.   rAAV has several positive

                                                                         attributes for gene

                                                                         therapy, the most notable

                                                                         being the ability to stably

                                                                         introduce foreign genetic

                                                        material into neurons (called
  Gene Therapy Approaches in Neurology (2007).
transduction) (Carter, 2008) and the ability to remain in vivo as a latent form for long

periods of time (Samulski et al., 1999). Figure 5 is an illustration of an adeno-associated

virus. While it is not able to carry a large amount of genetic material, Dong et al. (1998)

have reported that recombinant AAV (rAAV) has a genome of approximately 4.5 kilo-

base pairs (kb) in length, and consequently, can hold a coding sequence of up to 5kb



                                                                                                   6
when the its genes are removed (Carter, 2008). This appears to be enough to package

most complementary DNA (cDNA) sequences (Burton et al., 2007). These vectors are

not capable of replication, which means that it will not be able to spread farther than the

site of injection (Vezzani, 2004). The rAAV vector also has the ability to infect a wide

variety of cell types (as well as host organisms), and this vector should be coupled with a

cell-specific promoter (such as a neuronal promoter) to restrict gene expression to the

desired area of infection (Vezzani, 2004).

    The AAV vector is incapable of replication, meaning that, when its genes have been

removed, it is not able to replicate the newly-inserted genome in a host cell without aid.

Therefore, a helper virus is required for AAV to replicate in vivo. Typically, this virus is

an adenovirus, which is how this vector came to be known as an “adeno-associated

virus”. However, a herpes simplex virus can also be used (Burton et al., 2007). With a

helper, AAV is now known as a recombinant adeno-associated virus (rAAV), and viral

replication can now occur, but does so only in the cell nucleus. It typically attaches to

non-mitotic (non-dividing) cells, neurons being a primary example (Carter, 2008). The

genetic material carried by the vector is then transferred to the host cell, and rAAVs are

noted for the ability to stably transfer foreign genetic material to a host cell with little

reaction by the host cell’s immune system (Vezzani, 2004). Because it is very small, an

rAAV vector is capable of infecting a wide range of cell type in a variety of organisms. A

cell-specific promoter should also be used, so as to restrict gene expression to a particular

area or specific cell type (Carter, 2008).

    The most common method of rAAV production is referred to as the triple plasmid

transfection method, and involves the transfer of three plasmids to the vector (Burton et



                                                                                               7
al., 2007). These plasmids consist of the following: (1) packaging signals and the

transferred coding sequence, (2) a code which expresses the functions of an rAAV’s rep

and cap gene functions, and (3) a code expressing the helper virus (adenovirus or herpes

simplex virus) helper functions (Burton et al., 2007). This vector is then best delivered

within the parts of the organ essential to its function, referred to as an intraparenchymal

injection. This must be done in small volumes (1-10 μl), and at a low flow rate (0.2-0.4

μl/minute). The particles then appear to diffuse to the target cells, but because of both the

cell-specific promoter in the vector and rAAVs’ limited replication, transduction is

typically limited to only a few millimeters from the injection site (Burton et al., 2007).

This results in a limited efficacy in areas requiring large volumes, but nonetheless

remains the best way to deliver the treatment to date.

   The most common choices for coding sequences to replace the rAAV vector’s

genome (called transgenes) are galanin (GAL) and neuropeptide Y (NPY). Galanin is a
    Figure 6. The human brain, with the
    hippocampus highlighted in blue.
                                                             neuropeptide composed of 29

                                                             amino acids. It is called a

                                                             neuropeptide because it is

                                                             concentrated in the central nervous

                                                             system (CNS), is expressed only

                                                             in neuronal cells, and is highly

                                                              expressed in neurons in the
  http://upload.wikimedia.org/wikipedia/commons/2/2e/Gray7
  39-emphasizing-hippocampus.png
                                                              region of the brain known as the

hippocampus (see figure 6)(Pieribone et al., 1998). In the CNS, galanin acts as an

inhibitor for excitatory neurotransmitters. Neuropeptide Y (NPY) is a 36 amino acid

                                                                                                 8
neuropeptide that is found abundantly in the CNS, and is also found in peripheral nervous

tissue. (Dumont and Quirion, 2006). Like galanin, NPY acts by inhibiting excitatory

neurotransmitters and is found abundantly in the hippocampus. Both galanin and NPY

have been shown to dramatically increase in concentration after a seizure (Scharfman and

Gray, 2006), and have been shown to elicit an anticonvulsant effect in the body.

   Two types of studies of transgenes paired with rAAV vectors will be discussed: those

with galanin (GAL), and those with neuropeptide Y (NPY).

Galanin (GAL)-focused studies

Lin et al. (2003)

   The work of Lin et al. (2003) utilized the hippocampus of adult male rats, with

experimental rats receiving an injection of           Figure 7. The dorsal portion of the
                                                      hippocampus (encircled in red).
rAAV-GAL and controls receiving

an injection of rAAV-empty

(rAAV, without the galanin

transgene). To restrict expression to

neuronal cells, neuron-specific


enolase (NSE) was used as a promoter in the
                                                    http://www.brainybehavior.com/blog/wp-
                                                    content/uploads/2008/11/gray747.png
transgene-carrying rAAV vector. Figure 7

illustrates the dorsal hippocampus, the section of the rats’ brains injected bilaterally with

either the rAAV-GAL or rAAV-empty vector. Later, these rats were subjected to seizures

to determine efficacy of the vector. Seizure analysis in these rats was determined via

electroencephalogram (EEG) detection. To accomplish this, the animals were implanted

with electrodes and a guide cannula 2.5 months after the initial injection of rAAV-NSE-

                                                                                                9
Figure 8. Effects of rAAV-empty and rAAV-GAL on
number of observed seizures in rats.
GAL or rAAV-empty (no transgene present). Following implantation, the rats were given

                                                  kainic acid to induce seizure activity, and

                                                  were then monitored on the EEG and

                                                  analyzed for number of seizures as well as

                                                  time spent in seizure activity, as detected

                                                  by EEG analysis by a blind party. Figure 8
   Lin et al. (2003)
depicts the 40% decrease in the amount of seizures observed, with an average of

25 seizures noted in the control group compared to an average of 15 seizures in rats

injected with rAAV-NSE-GAL. As shown in figure 9, there was a 55% decrease in total

time spent in kainic acid-induced seizure activity. To determine galanin expression,

another group of rats were injected unilaterally in the right dorsal hippocampus with

rAAV-NSE-GAL. These rats were not subjected to seizures, but were killed 2.5 months

  Figure 9. Effects of rAAV-empty and rAAV-GAL on total     after vector injection. Upon
  time in seizure activity.
                                          analysis of the hippocampal areas of these

                                          animals, it was found that there was a higher

                                          amount of GAL in the right dorsal side (injected



  Lin et al. (2003)    with rAAV-NSE-GAL) than in the non-injected (left dorsal) side of

the hippocampus.

Mazarati et al. (1998)

   Mazarati et al. (1998) also studied rAAV-NSE-GAL in the rat hippocampus. This

study was done on 8-10 week old rats, and was included as part of a later study of the

effects of four neuropeptides – including NPY – in the rat hippocampus during a

prolonged seizure, referred to as status epilepticus (SE). The animals were subjected to

                                                                                                10
Figure 10. The perforant path of the hippocampus (in blue).
either electrical

stimulation in the

perforant path to

induce status

epilepticus (SE). As

its name implies, perforant path stimulation (PPS)             http://www.nature.com/neuro/journal/
                                                               v10/n3/images/nn0307-271-F1.gif
occurs in the perforant path of the hippocampus

(see figure 10). This area is neuron-dense, and acts as the input

pathway in the hippocampus (MRC, 2003). As a result, excess stimulation here will

result in prolonged seizure activity. For quantitative analysis, the rats were implanted

  Figure 11. The dentate gyrus of the     with a bipolar stimulating electrode in the perforant
  hippocampus (encircled in red).
                                                path, as well as a bipolar recording electrode

                                                in the dentate gyrus of the hippocampus,

                                                encircled in red in figure 11. Subjects

                                                received an injection of varying concentration


                                            of galanin either 30 minutes before the beginning
  http://www.brainybehavior.com/blog/wp
  -content/uploads/2008/11/gray747.png      of PPS or 30 minutes after its conclusion.

Controls received an injection of 0.9% NaCl.
                                                            Figure 12. The effects of galanin on time
                                                            spent in seizure activity between controls
                                                            and GAL-treated rats.
Figure 12 illustrates the findings

that those rats treated with higher

concentrations of galanin 30

minutes prior to PPS were


                                                                                                        11

                                                                           Mazarati et al. (1998)
observed to have spent 95% less time in seizure activity than the controls. Animals were

killed at varying time intervals following the conclusion of PPS to determine the

concentration of GAL-infected neurons in and around the injection site. Figure 13

illustrates the concentration of GAL-positive neurons in control and GAL-treated rats. 24

hours after PPS, an average of 16 GAL-positive neurons were found in a slice of the

hippocampus of GAL-treated rats, while none were found in non-GAL treated rats at any
                                                                Figure 13. Difference in GAL-positive
time following the conclusion of                                neurons after SSSE.

PPS. However, 3 days after PPS,

this number has gradually

decreased to an average of 8 GAL-

positive neurons in GAL-treated

rats, and decreased to 6 GAL-


positive neurons 7 days after the conclusion of PPS. In
                                                                             Mazarati et al. (1998)

                                                          Mazarati and Wasterlain’s 2002 study of
   Figure 14. Difference in total time spent in seizure
   activity between control and GAL-treated rats.
                                                            rAAV-GAL, it was found that,

                                                            compared to 590 minutes spent in

                                                            seizure activity in the rAAV-empty

                                                            control, total time spent in seizures

                                                            for rAAV-GAL treated animals was


   Mazarati and Wasterlain (2002)
under 10 minutes, as is illustrated in figure 14.




                                                                                                        12
Haberman et al. (2003)

                                In another study of the effects of galanin, Haberman et al.
 Figure 15. The inferior
 collicular cortex (in
 yellow) of the             (2003) found that it is able to reduce seizure intensity. This
 brainstem.
                           experiment used a vector constructed with a fibronectin secretory

                           signal sequence (FIB) as its promoter. The rats used in this study

                           were implanted with a stimulating electrode, and the inferior

                           collicular cortex (as shown in figure 15, highlighted in yellow) of

                           these animals’ brainstems was infused with either rAAV-FIB-GAL

                           (experimental) or rAAV-GAL (control). Four days after treatment,

                           seizure threshold was determined, and the rats were re-examined

                                          once per week for 4 weeks thereafter. In this period,
 http://upload.wikimedia.org/wikipedia/
 commons/0/00/Gray685.png
                                          it was found that the threshold for seizure genesis

was 60% higher in those animals treated with rAAV-FIB-GAL. Following this, the

animals were given water with doxycycline,            Figure 16. Seizure threshold comparison of
                                                      control group and rAAV-FIB-GAL-treated rats.
which returned the threshold to

baseline levels within 1 week.

After doxycycline was removed,

rAAV-FIB-GAL-treated rats were

observed to have a gradually

increasing threshold for seizure

genesis, with a threshold
                                                                          Haberman et al. (2003)
approximately 30% higher in


                                                                                                13
galanin-treated rats than those in the control group. Figure 16 illustrates these results. To

determine if treatment was able to prevent seizures, animals were injected with either

rAAV-FIB-GAL, rAAV-GAL, or received no treatment at all. In vitro, rAAV-FIB-GAL

or rAAV-GAL was introduced into HEK 293 cells. Twenty-four hours later, the media

were analyzed via ELISA for the presence of galanin. The rAAV-GAL cells showed no

detectable amount of GAL, indicating that it was not expressed or secreted in the cells,

but those cells infected with rAAV-FIB-GAL showed a significant amount (32ng/mL).

Neuropeptide Y (NPY)-focused studies

Richichi et al. (2004)

   Other studies focused on the effects of rAAV-delivered neuropeptide Y (NPY) in the

rat brain, more specifically, the hippocampus. Richichi et al. (2004) conducted an

experiment on adult male rats using rAAV-NSE-NPY. Neuron-specific enolase (NSE)

functioned as a cell-specific promoter in this vector. Two subtypes (called serotypes and

determined by protein markers on the cell surface) of rAAV were used in this experiment

– serotype 2, and a mixture of serotypes 1 and 2 (called serotype 1/2). The hippocampus

of these rats was injected bilaterally with the vector on each side. Eight weeks later, the

animals were implanted with electrodes and cannulas, and 4 days after implantation,

received a unilateral injection of kainic acid in either the dorsal hippocampus to induce
  Figure 17. Difference in observed seizure activity and onset time of seizures.




  Richichi et al. (2004)    seizure activity. Seizure activity was measured via

electroencephalogram (EEG) analysis by a blind party, and was done before injecting the

                                                                                              14
animals with kainic acid, and up to 3 hours following the injection. As shown in figure

17, time for seizure onset was delayed almost twofold in both serotypes of rAAV-NSE-

NPY, with an average of 11.5 minutes observed in serotype 1/2, compared to an average

of 6.2 minutes in the control group. In these subjects, those treated with serotype 1/2 had

no EEG-detected episodes of status epilepticus (SE), but in the control group, SE lasted at

least 60 minutes.

Mazarati and Wasterlain (2002)

   Mazarati and Wasterlain (2002) conducted a study of the effects of four

neuropeptides in the rat hippocampus, one of these neuropeptides being GAL, and

another being NPY. This study used 8-10 week old male rats, which were implanted with

a bipolar stimulating electrode in the perforant path of the hippocampus (see figure 10),

as well as a combination of a bipolar recording electrode and guide cannula in the dentate

gyrus of the hippocampus (see figure 11). These animals were subjected to 30 minutes of

perforant path stimulation (PPS) 7-10 days later, to induce a state of SSSE. Recordings of

brain activity were made via EEG by a blind party during PPS and 24 hours after its

conclusion. Spike distribution for this activity was measured in 30 minute periods, and

seizure activity was assessed according to total time spent in seizure activity, time

between the end of PPS and occurrence of the last seizure, and time spent in seizure

activity in a 1 hour period during a state of prolonged seizure activity (called self-

sustaining status epilepticus, or SSSE). Ten minutes after the conclusion of PPS, the

dentate gyrus of the rats were injected with NPY. Controls were injected with the same

dose of 0.9% NaCl. As illustrated by figure 18, there was no significant difference noted

in time spent in EEG-detected seizure activity between the control (average time in



                                                                                            15
seizure activity: 15 minutes) and NPY-treated group (average time in seizure activity: 13

minutes). NPY-treated animals were found to have spent 4 hours total in seizure activity,

and SSSE decreased to less than 20 minutes in these animals.

Discussion

    Mazarati et al. (1998) showed that, when injected        Figure 18. Time in seizures
                                                             following PPS treatment.
into the dentate gyrus, galanin has a seizure-protecting

effect. In this situation, galanin prevents the

initiation of self-sustaining status epilepticus

(SSSE). When injected after perforant path

stimulation was performed, galanin was able

to stop the maintenance phase of an

established SSSE, an accomplishment even

anticonvulsant drugs could not achieve.

Similarly, Lin et al. (2003) showed that when

rAAV-NSE-GAL was used, an over-
                                                            Mazarati and Wasterlain (2002)
expression of GAL by the vector was able to

drastically reduce seizure activity brought about by an intrahippocampal injection of

kainic acid, in addition to producing a strong anticonvulsant effect.

    Neuropeptide Y (NPY) in neuronal cells causes a decrease in both seizure

susceptibility and epileptogenesis, and therefore, the use of rAAV-NSE-NPY for over-

expression of NPY in rat hippocampal cells was used to inhibit seizures and

epileptogenesis (Richichi et al., 2004). In these rats, kainic acid-induced, EEG-detected

episodes of prolonged seizure activity (status epilepticus, or SE) were not observed in



                                                                                             16
NPY-treated rats, but episodes lasting at least 60 minutes were observed in the control

group. A delay in onset of seizures was observed in these animals, with NPY-treated rats

having an average delay time of approximately 11.5 minutes on average, while the

average delay for the control group was observed to be 6.2 minutes, almost half that of

the experimental group. Animals that received an injection of kainic acid showed a

decrease in EEG-detected seizure activity induced by the injection, due to rAAV vector-

mediated NPY over-expression. EEG-detected episodes of SE were eliminated in rats

expressing the NPY transgene in multiple areas of the hippocampus. Richichi et al.

(2004) concluded that the ability of NPY to inhibit the release of excitatory

neurotransmitters from brain cells may be the cause of the decrease in seizures noted in

rats injected with rAAV-NSE-NPY.

   In the hippocampus, NPY has inhibitory properties, acting as an anticonvulsant.

According to Mazarati and Wasterlain (2002), mice that lack NPY are more inclined to

develop seizures. In their study, Mazarati and Wasterlain (2002) were able to show that,

after injecting the dentate gyrus in rats with rAAV-delivered NPY, SSSE episodes were

significantly attenuated, though only a 2-minute difference in time spent in seizures was

observed between the experimental and control groups. This indicates that there was no

significant difference in time spent in seizures when comparing NPY-treated rats to the

saline-injected controls.

Conclusions

   In order to determine the most effective method of rAAV-mediated gene therapy,

many aspects must be considered. These include the level of expression of the transgene

included in the vector, length of time spent in seizure activity, length of time between



                                                                                           17
seizure activity, and number of seizures observed. By analyzing rAAV-mediated

experiments involving one or both of the neuropeptides of interest, NPY and GAL can be

compared and contrasted, and a conclusion regarding efficacy can be more easily

determined.

   In the studies conducted regarding rAAV vectors containing the GAL transgene, there

was a considerable reduction in seizure activity. There was an average decrease of 77%

in time spent in seizure activity, as well as a 40% decrease in number of seizures

observed. There was also found to be a large quantity of galanin expression and secretion

in cells in and around the injection site in the hippocampus, with 16 times more galanin-

positive cells found in galanin-treated animals than in the control group. The experiments

conducted with rAAV-NPY vectors also showed a decrease in EEG-detected seizure

activities, but it was only a 13% decrease – not nearly as vast as that observed in

experiments conducted with rAAV-GAL vectors.

   When compared, each neuropeptide was observed to have an anticonvulsant effect, as

well as the ability to decrease time spent in seizure activities, decrease time between

seizures, and to decrease the number of seizures observed in the tested animals compared

to the control. However, rAAV-GAL vectors were observed to have a greater effect than

the rAAV-NPY vectors and because of this, rAAV vectors utilizing galanin as a

transgene for expression and secretion in neuronal cells are the best choice for gene

therapy.

   While these vectors have worked well in rodent models of seizures, there are still

many obstacles that must be surpassed before this treatment can be applied to humans.

These include establishing an effective, minimally invasive method for vector delivery,



                                                                                          18
as well as receiving approval from the Food and Drug Administration (FDA) for

treatment. Once established, these tests may be able to help control seizures in humans

suffering from epileptic disorders. AAV-mediated gene therapy is a promising new

therapy, and will hopefully one day become a widespread, successful treatment for not

just epilepsy, but many other human seizure disorders.

References

Burton, E.A., J.C. Glorioso, and D.J. Fink. 2007. Gene Therapy Approaches in
Neurology. In Molecular Neurology. Ed. S.G. Waxman. Elsevier Academic Press.
Burlington, MA. 101-123.

Carter, B.J. 2008. Adeno-Associated Virus Vectors. In Concepts in Genetic Medicine.
Ed. B. Dropulic and B.J. Carter. Wiley-Liss. Hoboken, NJ. 61-68.

Chandler, K. 2006. Canine epilepsy: What can we learn from human seizure disorders?
The Veterinary Journal. 172: 207-217.

Dong, J.Y., P.D. Fan, and R.A. Frizell. 1996. Quantitative analysis of the packaging
capacity of recombinant adeno-associated virus. Human Gene Therapy. 7: 2102-2112.

Dumont, Y. and R. Quirion. 2006. An overview of neuropeptide Y: pharmacology to
molecular biology and receptor localization. In NPY Family of Peptides in Neurobiology,
Cardiovascular and Metabolic Disorders: from Genes to Therapeutics. Ed. Z. Zukowska
and G.Z. Feuerstein. Birkhaüser Verlag. Basel, Switzerland. 7-33.

Haberman, R.P., R.J. Samulski, and T. J. McCown. 2003. Attenuation of seizures and
neuronal death by adeno-associated virus vector galanin expression and secretion. Nature
Medicine. 9(8): 1076-1080.

Haberman , R.P., T.J. McCown, and R.J. Samulski. 1998. Inducible long-term gene
expression in brain with adeno-associated virus gene transfer. Gene Therapy. 5: 1604-
1611.

Hains, B. 2006. Brain Disorders. Chelsea House Publishers. Philadelphia. 37-38.

Lin, E.D., C. Richichi, D. Young, K. Baer, A. Vezzani, and M.J. During. 2003.
Recombinant AAV-mediated expression of galanin in rat hippocampus suppresses
seizure development. European Journal of Neuroscience. 18: 2087-2092.




                                                                                          19
Mazarati, A.M and C.G. Wasterlain. 2002. Anticonvulsant effects of four neuropeptides
in the rat hippocampus during self-sustaining status epilepticus. Neuroscience Letters.
331: 123-127.

Mazarati, A.M., H. Liu, U. Soomets, R. Sankar, D. Shin, H. Katsumori, Ü. Langel, and
C.G. Wasterlain. 1998. Galanin modulation of seizures and seizure modulation of
hippocampal galanin in animal models of status epilepticus. Journal of Neuroscience.
18(23): 10070-10077.

Medical Research Council. 2003. University of Bristol. Hippocampal Pathways.
http://www.bristol.ac.uk/synaptic/info/pathway/hippocampal.htm

National Institutes of Health. 2004. National Institute of Neurological Disorders and
Stroke. Seizures and Epilepsy: Hope Through Research. 13-27.

Pieribone, V.A., Z.D. Xu, X. Zhang, and T. Hökfelt. 1998. Electrophysiologic Effects of
Galanin on Neurons of the Central Nervous System. In Annals of the New York
Academy of Sciences. Ed. B.M. Boland, J. Cullinan, and A.C. Fink. New York Academy
of Sciences. New York. 264-273.

Ransom, C.B. and H. Blumenfeld. 2007. Acquired Epilepsy: Cellular and Molecular
Mechanisms. In Molecular Neurology. Ed. S.G. Waxman. Elsevier Academic Press.
Burlington, MA. 347-370.

Riban, V., H.L. Fitzsimons, and M.J. During. 2009. Gene therapy in epilepsy. Epilepsia.
50(1): 24-32.

Richichi, C, E.D. Lin, D. Stefanin, D. Colella, T. Ravizza, G. Grignaschi, P. Veglianese,
G. Sperk, M.J. During, and A. Vezzani. 2004. Anticonvulsant and antiepileptogenic
effects mediated by adeno-associated virus vector neuropeptide Y expression in the rat
hippocampus. Journal of Neuroscience. 24(12): 3051-3059.

Samulski, R.J., M. Sally, and N. Muzyczka. 1999. Adeno-Associated Viral Vectors. In
The Development of Human Gene Therapy. Ed. T. Friedmann. Cold Spring Harbor
Laboratory Press. Cold Spring Harbor, NY. 131-172.

Scharfman, H.E. and W.P. Gray. 2006. Plasticity of neuropeptide Y in the dentate gyrus
after seizures, and its relevance to seizure-induced neurogenesis. In NPY Family of
Peptides in Neurobiology, Cardiovascular and Metabolic Disorders: from Genes to
Therapeutics. Ed. Z. Zukowska and G.Z. Feuerstein. Birkhaüser Verlag. Basel,
Switzerland. 193-211.

Vezzani, A. 2004. Gene Therapy in Epilepsy. Epilepsy Currents. 4(3): 87-90.




                                                                                        20

More Related Content

What's hot

Disease modification in epilepsy therapy
Disease modification in epilepsy therapyDisease modification in epilepsy therapy
Disease modification in epilepsy therapywebzforu
 
Homocystinuria pdf
Homocystinuria pdfHomocystinuria pdf
Homocystinuria pdfTingYoongTee
 
Pathophysiology of migraine
Pathophysiology of migrainePathophysiology of migraine
Pathophysiology of migrainewebzforu
 
PET/SPECT in Epilepsy Surgery
PET/SPECT in Epilepsy Surgery PET/SPECT in Epilepsy Surgery
PET/SPECT in Epilepsy Surgery Lalit Bansal
 
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...MerqurioEditore_redazione
 
Transcranial direct current stimulation
Transcranial direct current stimulation Transcranial direct current stimulation
Transcranial direct current stimulation Andri Andri
 
Brain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainBrain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainRalph Sherman
 
Brief overview of brain stimulation techniques
Brief overview of  brain stimulation techniquesBrief overview of  brain stimulation techniques
Brief overview of brain stimulation techniquesSujit Kumar Kar
 
Epilepsy Surgery in Children
Epilepsy Surgery in ChildrenEpilepsy Surgery in Children
Epilepsy Surgery in ChildrenLalit Bansal
 
Deep Brain Stimulation surgery experience at Apollo Hospital, New Delhi
Deep Brain Stimulation surgery experience at Apollo Hospital, New DelhiDeep Brain Stimulation surgery experience at Apollo Hospital, New Delhi
Deep Brain Stimulation surgery experience at Apollo Hospital, New DelhiApollo Hospitals
 
Shawn dissertation NRSF
Shawn dissertation NRSFShawn dissertation NRSF
Shawn dissertation NRSFmidknightr
 
Spect in concussion, vertebro basilar insufficiency and other neurological di...
Spect in concussion, vertebro basilar insufficiency and other neurological di...Spect in concussion, vertebro basilar insufficiency and other neurological di...
Spect in concussion, vertebro basilar insufficiency and other neurological di...All India Institute of Medical Sciences
 
neuroinflammation
neuroinflammationneuroinflammation
neuroinflammationsarvesh022
 
Neurology 2nd investigation of neurological disease
Neurology 2nd investigation of neurological diseaseNeurology 2nd investigation of neurological disease
Neurology 2nd investigation of neurological diseaseRamiAboali
 

What's hot (20)

5. thomas lew how anaesthetic works (and why knowing matters)
5. thomas lew how anaesthetic works (and why knowing matters)5. thomas lew how anaesthetic works (and why knowing matters)
5. thomas lew how anaesthetic works (and why knowing matters)
 
Disease modification in epilepsy therapy
Disease modification in epilepsy therapyDisease modification in epilepsy therapy
Disease modification in epilepsy therapy
 
A Case of Chorea following ASV
A Case of Chorea following ASVA Case of Chorea following ASV
A Case of Chorea following ASV
 
Homocystinuria pdf
Homocystinuria pdfHomocystinuria pdf
Homocystinuria pdf
 
Pathophysiology of migraine
Pathophysiology of migrainePathophysiology of migraine
Pathophysiology of migraine
 
PET/SPECT in Epilepsy Surgery
PET/SPECT in Epilepsy Surgery PET/SPECT in Epilepsy Surgery
PET/SPECT in Epilepsy Surgery
 
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...
La conduzione del nervo surale dorsale in pazienti con carenza di vitamina B1...
 
Transcranial direct current stimulation
Transcranial direct current stimulation Transcranial direct current stimulation
Transcranial direct current stimulation
 
Epilepsy surgery
Epilepsy surgeryEpilepsy surgery
Epilepsy surgery
 
The brain and energy
The brain and energyThe brain and energy
The brain and energy
 
Brain stimulation therapies
Brain stimulation therapiesBrain stimulation therapies
Brain stimulation therapies
 
Brain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainBrain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spain
 
Brief overview of brain stimulation techniques
Brief overview of  brain stimulation techniquesBrief overview of  brain stimulation techniques
Brief overview of brain stimulation techniques
 
Epilepsy Surgery in Children
Epilepsy Surgery in ChildrenEpilepsy Surgery in Children
Epilepsy Surgery in Children
 
Deep Brain Stimulation surgery experience at Apollo Hospital, New Delhi
Deep Brain Stimulation surgery experience at Apollo Hospital, New DelhiDeep Brain Stimulation surgery experience at Apollo Hospital, New Delhi
Deep Brain Stimulation surgery experience at Apollo Hospital, New Delhi
 
NIBS
NIBSNIBS
NIBS
 
Shawn dissertation NRSF
Shawn dissertation NRSFShawn dissertation NRSF
Shawn dissertation NRSF
 
Spect in concussion, vertebro basilar insufficiency and other neurological di...
Spect in concussion, vertebro basilar insufficiency and other neurological di...Spect in concussion, vertebro basilar insufficiency and other neurological di...
Spect in concussion, vertebro basilar insufficiency and other neurological di...
 
neuroinflammation
neuroinflammationneuroinflammation
neuroinflammation
 
Neurology 2nd investigation of neurological disease
Neurology 2nd investigation of neurological diseaseNeurology 2nd investigation of neurological disease
Neurology 2nd investigation of neurological disease
 

Viewers also liked

Передача закупівель ліків міжнародним організаціям
Передача закупівель ліків міжнародним організаціям Передача закупівель ліків міжнародним організаціям
Передача закупівель ліків міжнародним організаціям UkrainianPhilanthropistsForum
 
ルイヴィトンとそのデザイナー·ジュリー·ド·リブラン
ルイヴィトンとそのデザイナー·ジュリー·ド·リブランルイヴィトンとそのデザイナー·ジュリー·ド·リブラン
ルイヴィトンとそのデザイナー·ジュリー·ド·リブランcanlie301
 
Công Nghệ Thuốc Giảm Cân Hiệu Quả Nhất
Công Nghệ Thuốc Giảm Cân Hiệu Quả NhấtCông Nghệ Thuốc Giảm Cân Hiệu Quả Nhất
Công Nghệ Thuốc Giảm Cân Hiệu Quả Nhấtjunior783
 
2014 December GrayReports - Student Demand Trends
2014 December GrayReports - Student Demand Trends2014 December GrayReports - Student Demand Trends
2014 December GrayReports - Student Demand TrendsGray Associates, Inc
 
Presentación1 nxt ggggggggggggggg 555555555555
Presentación1 nxt ggggggggggggggg 555555555555Presentación1 nxt ggggggggggggggg 555555555555
Presentación1 nxt ggggggggggggggg 555555555555Isaac Felipe Beltran Lopez
 
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)ΕΛΕΝΗ ΚΑΛΕΥΡΑ
 
Certificate- Advanced Purchasing and Procurement Training
Certificate- Advanced Purchasing and Procurement TrainingCertificate- Advanced Purchasing and Procurement Training
Certificate- Advanced Purchasing and Procurement TrainingMOHAMED KAMAL محمد كمال
 
Swissgolden englishofficial
Swissgolden englishofficialSwissgolden englishofficial
Swissgolden englishofficialsyacoub
 
Swissgolden company euro presentation
Swissgolden company euro presentationSwissgolden company euro presentation
Swissgolden company euro presentationsyacoub
 
Tramite de audiencia asociaciones de consumidores
Tramite de audiencia asociaciones de consumidoresTramite de audiencia asociaciones de consumidores
Tramite de audiencia asociaciones de consumidoresUnionConsumidoresMalaga
 
#GGDT9 "Sport, Tecnologia & Disabilità"
#GGDT9 "Sport, Tecnologia & Disabilità"#GGDT9 "Sport, Tecnologia & Disabilità"
#GGDT9 "Sport, Tecnologia & Disabilità"Deborah Sormani
 

Viewers also liked (15)

Habilidades infancia
Habilidades infanciaHabilidades infancia
Habilidades infancia
 
hii
hiihii
hii
 
Передача закупівель ліків міжнародним організаціям
Передача закупівель ліків міжнародним організаціям Передача закупівель ліків міжнародним організаціям
Передача закупівель ліків міжнародним організаціям
 
ルイヴィトンとそのデザイナー·ジュリー·ド·リブラン
ルイヴィトンとそのデザイナー·ジュリー·ド·リブランルイヴィトンとそのデザイナー·ジュリー·ド·リブラン
ルイヴィトンとそのデザイナー·ジュリー·ド·リブラン
 
Công Nghệ Thuốc Giảm Cân Hiệu Quả Nhất
Công Nghệ Thuốc Giảm Cân Hiệu Quả NhấtCông Nghệ Thuốc Giảm Cân Hiệu Quả Nhất
Công Nghệ Thuốc Giảm Cân Hiệu Quả Nhất
 
2014 December GrayReports - Student Demand Trends
2014 December GrayReports - Student Demand Trends2014 December GrayReports - Student Demand Trends
2014 December GrayReports - Student Demand Trends
 
Presentación1 nxt ggggggggggggggg 555555555555
Presentación1 nxt ggggggggggggggg 555555555555Presentación1 nxt ggggggggggggggg 555555555555
Presentación1 nxt ggggggggggggggg 555555555555
 
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)
Οι δώδεκα θεοί του Ολύμπου (Μαρία και Φαίη)
 
Certificate- Advanced Purchasing and Procurement Training
Certificate- Advanced Purchasing and Procurement TrainingCertificate- Advanced Purchasing and Procurement Training
Certificate- Advanced Purchasing and Procurement Training
 
Hadoop-Certificate
Hadoop-CertificateHadoop-Certificate
Hadoop-Certificate
 
Swissgolden englishofficial
Swissgolden englishofficialSwissgolden englishofficial
Swissgolden englishofficial
 
Swissgolden company euro presentation
Swissgolden company euro presentationSwissgolden company euro presentation
Swissgolden company euro presentation
 
Tramite de audiencia asociaciones de consumidores
Tramite de audiencia asociaciones de consumidoresTramite de audiencia asociaciones de consumidores
Tramite de audiencia asociaciones de consumidores
 
Urban and Rural Sources of Particulate Matter
Urban and Rural Sources of Particulate MatterUrban and Rural Sources of Particulate Matter
Urban and Rural Sources of Particulate Matter
 
#GGDT9 "Sport, Tecnologia & Disabilità"
#GGDT9 "Sport, Tecnologia & Disabilità"#GGDT9 "Sport, Tecnologia & Disabilità"
#GGDT9 "Sport, Tecnologia & Disabilità"
 

Similar to AVS 406 Review Paper

Pathophysiology and management of epilepsy
Pathophysiology and management of epilepsyPathophysiology and management of epilepsy
Pathophysiology and management of epilepsySoujanya Pharm.D
 
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...Dr. Rafael Higashi
 
Research paper on epilepsy
Research paper on epilepsyResearch paper on epilepsy
Research paper on epilepsylilysolomon
 
Septicemic encephalopathy
Septicemic encephalopathySepticemic encephalopathy
Septicemic encephalopathyNeurologyKota
 
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdf
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdfEPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdf
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdfAdamu Mohammad
 
Somatosensory evoked potentials
Somatosensory evoked potentialsSomatosensory evoked potentials
Somatosensory evoked potentialsManideep Malaka
 
Lecture 12. Antiepileptic drugs pharmacology.pptx
Lecture 12. Antiepileptic drugs pharmacology.pptxLecture 12. Antiepileptic drugs pharmacology.pptx
Lecture 12. Antiepileptic drugs pharmacology.pptxsathishvsathish1
 
Review Nature 399 A15
Review Nature 399 A15Review Nature 399 A15
Review Nature 399 A15MADHU.MATHEWS
 
Recent advances in the management of alzheimers disease
Recent advances in the management of alzheimers diseaseRecent advances in the management of alzheimers disease
Recent advances in the management of alzheimers diseaseParthajyoti Neog
 
Cns Intro Davidson Plus2.
Cns Intro Davidson Plus2.Cns Intro Davidson Plus2.
Cns Intro Davidson Plus2.Shaikhani.
 
Powerpoint
PowerpointPowerpoint
Powerpointerinhunt
 

Similar to AVS 406 Review Paper (20)

Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
 
E P I L E P S Y U P D A T E
E P I L E P S Y  U P D A T EE P I L E P S Y  U P D A T E
E P I L E P S Y U P D A T E
 
Pathophysiology and management of epilepsy
Pathophysiology and management of epilepsyPathophysiology and management of epilepsy
Pathophysiology and management of epilepsy
 
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
A guideline for discontinuing antiepileptic drugs in seizure-free patients – ...
 
Research paper on epilepsy
Research paper on epilepsyResearch paper on epilepsy
Research paper on epilepsy
 
Epilepsy
EpilepsyEpilepsy
Epilepsy
 
Septicemic encephalopathy
Septicemic encephalopathySepticemic encephalopathy
Septicemic encephalopathy
 
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdf
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdfEPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdf
EPILEPSY CLASSIFICATION, PATHOENESIS, AND MANAGEMENT.pdf
 
Somatosensory evoked potentials
Somatosensory evoked potentialsSomatosensory evoked potentials
Somatosensory evoked potentials
 
Epilepsy and Seizures, e-Medicine Article
Epilepsy and Seizures, e-Medicine ArticleEpilepsy and Seizures, e-Medicine Article
Epilepsy and Seizures, e-Medicine Article
 
7523850.ppt
7523850.ppt7523850.ppt
7523850.ppt
 
Lecture 12. Antiepileptic drugs pharmacology.pptx
Lecture 12. Antiepileptic drugs pharmacology.pptxLecture 12. Antiepileptic drugs pharmacology.pptx
Lecture 12. Antiepileptic drugs pharmacology.pptx
 
Review Nature 399 A15
Review Nature 399 A15Review Nature 399 A15
Review Nature 399 A15
 
Recent advances in the management of alzheimers disease
Recent advances in the management of alzheimers diseaseRecent advances in the management of alzheimers disease
Recent advances in the management of alzheimers disease
 
Cns Intro Davidson Plus2.
Cns Intro Davidson Plus2.Cns Intro Davidson Plus2.
Cns Intro Davidson Plus2.
 
Caser record... Rasmussen's encephalitis
Caser record... Rasmussen's encephalitisCaser record... Rasmussen's encephalitis
Caser record... Rasmussen's encephalitis
 
Epilepsy
EpilepsyEpilepsy
Epilepsy
 
Powerpoint
PowerpointPowerpoint
Powerpoint
 
Powerpoint
PowerpointPowerpoint
Powerpoint
 
Epilepsy
EpilepsyEpilepsy
Epilepsy
 

More from Omega Cantrell

Hyperlipemia in Equids
Hyperlipemia in EquidsHyperlipemia in Equids
Hyperlipemia in EquidsOmega Cantrell
 
Case Presentation: Pancreatic Clinical Chemistry Abnormalities
Case Presentation: Pancreatic Clinical Chemistry AbnormalitiesCase Presentation: Pancreatic Clinical Chemistry Abnormalities
Case Presentation: Pancreatic Clinical Chemistry AbnormalitiesOmega Cantrell
 
Client Handout - Fleas
Client Handout - FleasClient Handout - Fleas
Client Handout - FleasOmega Cantrell
 
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...Omega Cantrell
 

More from Omega Cantrell (13)

Hyperlipemia in Equids
Hyperlipemia in EquidsHyperlipemia in Equids
Hyperlipemia in Equids
 
Case Presentation: Pancreatic Clinical Chemistry Abnormalities
Case Presentation: Pancreatic Clinical Chemistry AbnormalitiesCase Presentation: Pancreatic Clinical Chemistry Abnormalities
Case Presentation: Pancreatic Clinical Chemistry Abnormalities
 
Client Handout - Fleas
Client Handout - FleasClient Handout - Fleas
Client Handout - Fleas
 
Liver Enzymology
Liver EnzymologyLiver Enzymology
Liver Enzymology
 
Liver Enzymology
Liver EnzymologyLiver Enzymology
Liver Enzymology
 
Unknown bacteria
Unknown bacteriaUnknown bacteria
Unknown bacteria
 
N059 06 17-2011
N059 06 17-2011N059 06 17-2011
N059 06 17-2011
 
N059 06-17-2011
N059 06-17-2011N059 06-17-2011
N059 06-17-2011
 
Osteochondrosis
OsteochondrosisOsteochondrosis
Osteochondrosis
 
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...
Ehrlichia canis in a dog with large granular lymphocytosis, thrombocytopenia,...
 
VPI-VBMA Case Study
VPI-VBMA Case StudyVPI-VBMA Case Study
VPI-VBMA Case Study
 
AVS 406 Presentation
AVS 406 PresentationAVS 406 Presentation
AVS 406 Presentation
 
AVS 406 Poster
AVS 406 PosterAVS 406 Poster
AVS 406 Poster
 

AVS 406 Review Paper

  • 1. The Use of Recombinant Adeno-Associated Viral Vectors in Gene Therapy to Treat Epilepsy Omega Cantrell Abstract Epilepsy is defined as being recurrent, unprovoked seizures, and naturally occurs in a wide range of species. It affects approximately 1% of the U.S. population, and is a good target for gene therapy by recombinant adeno-associated virus (rAAV) vectors. rAAV is capable of stably transferring foreign genetic material to neurons (transduction), in addition to being able to infect a wide range of organisms and cell types, but, in vector form, is not pathogenic. The neuropeptides galanin (GAL) and neuropeptide Y (NPY) are found abundantly in the central nervous system, have strong anticonvulsant effects and are promising transgene options for rAAV vectors. Galanin is thought to have more of an impact than NPY. Studies have shown that GAL is able to reduce time spent in seizures by 77% and number of EEG-detected seizures by 40%. Following seizures induced by electrical stimulation in the perforant path of the hippocampus, the number of GAL- positive neurons was 16 times that of the control group. Because GAL was able to more significantly decrease the rate of seizure occurrence and total time spent in seizure activity, it is a better option as a transgene for rAAV in gene therapy for epilepsy and other seizure disorders. Key words: epilepsy, gene therapy, recombinant adeno-associated virus (rAAV), galanin (GAL), neuropeptide Y (NPY), transgene 1
  • 2. Introduction Normal brain function involves neuron-to-neuron transmission, causing signals from the brain to be sent to the rest of the body. For this to occur, sodium channels open, Figure 1. Normal brain wave patterns. allowing sodium (Na+) ions to enter. This operates on a negative feedback loop, such that potassium acts to cease signaling by closing the sodium channels. As Figure 1 illustrates, on an electroencephalograph (EEG), normal brain patterns appear as small http:/www.epilepsy.org.au/images/ElectroEn waves. When abnormal brain waves are seen cephalogram.png on an EEG, a seizure disorder is usually to blame (NIH, 2004; Hains, 2006). Epilepsy is a widely occurring seizure disorder in many species, including rats, dogs, and cats, and is the most common acquired neurological disorder in humans (Chandler, 2006). It has been defined as recurrent Figure 2. Brain wave patterns observed during an epileptic seizure. unprovoked seizures, and has a prevalence of 0.5-1.0% in humans, with a higher rate of occurrence in the underdeveloped world (Ransom and Blumenfeld, 2007).There are many different kinds of epilepsy, and these disorders are diagnosed based on many factors. http://brain.fuw.edu.pl/~suffa/SW/SW_patt.gif 2
  • 3. Figure 2 illustrates EEG-detected abnormalities, and other factors include age of onset, characteristics of the seizure, and seizure induction stimuli (Chandler, 2006). As can be seen in figure 2, epileptic brain waves appear as high spikes in rapid succession on an EEG. Seizure intensity is determined by the height of the spikes, coupled with the rate of succession. Higher, more rapid spikes are indicative of a more severe seizure (Hains, 2006). Epilepsy is thought to be caused by an imbalance in neurotransmissions (Vezzani, 2004). As mentioned before, normal neurotransmissions are facilitated by a negative feedback loop between sodium, which opens the channels, and potassium, which closes the channels. In a normal brain, neurons fire approximately 30 times each second. During a seizure, neuronal firing can occur as many as 500 times each second (NIH, 2004). Normally, when excitatory neurotransmitters such as glutamate are released, neuronal firing occurs. Once this reaches a certain level (which varies among individuals, and according to the type of signaling that is occurring), another negative feedback loop occurs. When this happens, an inhibitory neurotransmitter such as γ-aminobutyric acid (GABA) serves to stop the action of the excitatory neurotransmitters and cease neuronal firing (NIH, 2004). Because of this, another common theory is that either a high level of excitatory neurotransmitters or low level of inhibitory neurotransmitters are responsible for the abnormal neuronal firing that causes seizures to occur. Having just one seizure does not mean a person has epilepsy. In order to be considered for a diagnosis of epilepsy, a person must have had two or more seizures. There are two main categories of epilepsy – focal (or partial), which affects only one part of the brain. This includes temporal lobe epilepsy, which, according to Ransom and 3
  • 4. Blumenfeld (2007), is more common in adults, and is often resistant to medical therapy. Generalized seizures affect both of the brain’s hemispheres simultaneously (NIH, 2004). As detailed previously, a seizure occurs when an imbalance in neurotransmitters causes sodium channels in the brain to remain open, resulting in abnormal neuronal firing. This abnormal firing results in symptoms of the many different types seizures, including muscle spasms, repeated movements (also called automatisms), loss of consciousness, and/or temporary loss of muscle tone. These disorders are commonly controlled with medications that operate in a variety of ways, and are prescribed according to the type or types of seizures the patient has. A common mode of action for an antiepileptic medication is to either close or block the sodium channels in the brain, preventing neuronal hyperactivity. In doing this, these drugs can help to control seizures, but are not guaranteed to prevent seizure occurrence in all who take them. According to Riban et al. (2009), about one-third of diagnosed epileptics suffer from a form of the disease resistant to anticonvulsant drugs. In these more severe cases, surgery may be necessary. This is often a last resort for doctors, and is Figure 3. The human brain, usually done only if a patient continues to suffer from arrow pointing to the corpus callosum. seizures after years of treatment with different types of anticonvulsant drugs. The National Institutes of Health recognizes only three categories of seizures that can be treated successfully with surgery, one of http://static.guim.co.uk/sys- images/Guardian/Pix/pictures/2009/4/6/1239055 717363/Cross-section-of-the-huma-001.jpg 4
  • 5. these being focal seizure disorders, which includes temporal lobe epilepsy, and has a 64% success rate when compared with treatment with prescription medication alone (NIH, 2004). Surgery should not be taken lightly, however, as it often involves either removal of an area in the brain where seizures are observed to occur most frequently (seizure focus area), or, in more severe cases, cutting the corpus callosum (illustrated in figure 3). This disconnects the two hemispheres of the brain, and does not stop those seizures focused in one area of the brain (focal seizures), but merely prevents them from spreading across the whole brain (NIH, 2004). Because there are so few options for epilepsy treatment, and such a high incidence of medication-resistant epilepsies, gene therapy – especially when mediated by recombinant adeno-associated virus (rAAV) – is considered to be a promising new option for those who suffer from a more severe seizure disorder. To better understand how rAAVs are used for gene therapy, it is necessary to understand how a vector is constructed. A virus operates by first infecting a host cell, then replicating its own genome inside a host cell. This amplifies viral infection in the host organism, and after the host cells are lysed, these viruses can further infect more host cells, resulting in a widespread infection. Viral vectors are genetically engineered in the sense that their life cycles are manipulated such that the beneficial stages responsible for Figure 4. Genome of an AAV vector, introverted terminal repeat sequences (ITRs) in red boxes. Gene Therapy Approaches in Neurology (2007). genomic replication are retained, while the deleterious stages responsible for viral replication and cell lysis are eliminated (Burton et al., 2007). Figure 4 depicts the relatively small (~4.5kb) rAAV genome, which 5
  • 6. codes for just two genes: rep and cap, and has an introverted terminal repeat sequence (ITRs) at each end of its single-stranded DNA (Carter, 2008). To construct a vector, rep and cap are first removed and then replaced with a coding sequence of a similar length (4.5kb), while the ITRs are retained as “stops” for the genetic code (Burton et al., 2007). The removal of rep has two purposes: the first – and the most obvious reason – is to make room for the coding sequence intended for therapy, and the second being to make rAAVs incapable of attaching to human DNA. While not a problem with animals tested in the laboratory, the rep gene allows rAAVs to adhere to a site on human DNA chromosome 19 (Carter, 2008). Because this is a potential risk, rep is removed. Recombinant adeno-associated virus (rAAV) is an example of a viral vector – a genetically engineered virus (Burton et al., 2007) – and has garnered attention in recent years as a promising tool in gene therapy to potentially treat disorders such as epilepsy. Figure 5. An adeno-associated virus and its structural components. rAAV has several positive attributes for gene therapy, the most notable being the ability to stably introduce foreign genetic material into neurons (called Gene Therapy Approaches in Neurology (2007). transduction) (Carter, 2008) and the ability to remain in vivo as a latent form for long periods of time (Samulski et al., 1999). Figure 5 is an illustration of an adeno-associated virus. While it is not able to carry a large amount of genetic material, Dong et al. (1998) have reported that recombinant AAV (rAAV) has a genome of approximately 4.5 kilo- base pairs (kb) in length, and consequently, can hold a coding sequence of up to 5kb 6
  • 7. when the its genes are removed (Carter, 2008). This appears to be enough to package most complementary DNA (cDNA) sequences (Burton et al., 2007). These vectors are not capable of replication, which means that it will not be able to spread farther than the site of injection (Vezzani, 2004). The rAAV vector also has the ability to infect a wide variety of cell types (as well as host organisms), and this vector should be coupled with a cell-specific promoter (such as a neuronal promoter) to restrict gene expression to the desired area of infection (Vezzani, 2004). The AAV vector is incapable of replication, meaning that, when its genes have been removed, it is not able to replicate the newly-inserted genome in a host cell without aid. Therefore, a helper virus is required for AAV to replicate in vivo. Typically, this virus is an adenovirus, which is how this vector came to be known as an “adeno-associated virus”. However, a herpes simplex virus can also be used (Burton et al., 2007). With a helper, AAV is now known as a recombinant adeno-associated virus (rAAV), and viral replication can now occur, but does so only in the cell nucleus. It typically attaches to non-mitotic (non-dividing) cells, neurons being a primary example (Carter, 2008). The genetic material carried by the vector is then transferred to the host cell, and rAAVs are noted for the ability to stably transfer foreign genetic material to a host cell with little reaction by the host cell’s immune system (Vezzani, 2004). Because it is very small, an rAAV vector is capable of infecting a wide range of cell type in a variety of organisms. A cell-specific promoter should also be used, so as to restrict gene expression to a particular area or specific cell type (Carter, 2008). The most common method of rAAV production is referred to as the triple plasmid transfection method, and involves the transfer of three plasmids to the vector (Burton et 7
  • 8. al., 2007). These plasmids consist of the following: (1) packaging signals and the transferred coding sequence, (2) a code which expresses the functions of an rAAV’s rep and cap gene functions, and (3) a code expressing the helper virus (adenovirus or herpes simplex virus) helper functions (Burton et al., 2007). This vector is then best delivered within the parts of the organ essential to its function, referred to as an intraparenchymal injection. This must be done in small volumes (1-10 μl), and at a low flow rate (0.2-0.4 μl/minute). The particles then appear to diffuse to the target cells, but because of both the cell-specific promoter in the vector and rAAVs’ limited replication, transduction is typically limited to only a few millimeters from the injection site (Burton et al., 2007). This results in a limited efficacy in areas requiring large volumes, but nonetheless remains the best way to deliver the treatment to date. The most common choices for coding sequences to replace the rAAV vector’s genome (called transgenes) are galanin (GAL) and neuropeptide Y (NPY). Galanin is a Figure 6. The human brain, with the hippocampus highlighted in blue. neuropeptide composed of 29 amino acids. It is called a neuropeptide because it is concentrated in the central nervous system (CNS), is expressed only in neuronal cells, and is highly expressed in neurons in the http://upload.wikimedia.org/wikipedia/commons/2/2e/Gray7 39-emphasizing-hippocampus.png region of the brain known as the hippocampus (see figure 6)(Pieribone et al., 1998). In the CNS, galanin acts as an inhibitor for excitatory neurotransmitters. Neuropeptide Y (NPY) is a 36 amino acid 8
  • 9. neuropeptide that is found abundantly in the CNS, and is also found in peripheral nervous tissue. (Dumont and Quirion, 2006). Like galanin, NPY acts by inhibiting excitatory neurotransmitters and is found abundantly in the hippocampus. Both galanin and NPY have been shown to dramatically increase in concentration after a seizure (Scharfman and Gray, 2006), and have been shown to elicit an anticonvulsant effect in the body. Two types of studies of transgenes paired with rAAV vectors will be discussed: those with galanin (GAL), and those with neuropeptide Y (NPY). Galanin (GAL)-focused studies Lin et al. (2003) The work of Lin et al. (2003) utilized the hippocampus of adult male rats, with experimental rats receiving an injection of Figure 7. The dorsal portion of the hippocampus (encircled in red). rAAV-GAL and controls receiving an injection of rAAV-empty (rAAV, without the galanin transgene). To restrict expression to neuronal cells, neuron-specific enolase (NSE) was used as a promoter in the http://www.brainybehavior.com/blog/wp- content/uploads/2008/11/gray747.png transgene-carrying rAAV vector. Figure 7 illustrates the dorsal hippocampus, the section of the rats’ brains injected bilaterally with either the rAAV-GAL or rAAV-empty vector. Later, these rats were subjected to seizures to determine efficacy of the vector. Seizure analysis in these rats was determined via electroencephalogram (EEG) detection. To accomplish this, the animals were implanted with electrodes and a guide cannula 2.5 months after the initial injection of rAAV-NSE- 9
  • 10. Figure 8. Effects of rAAV-empty and rAAV-GAL on number of observed seizures in rats. GAL or rAAV-empty (no transgene present). Following implantation, the rats were given kainic acid to induce seizure activity, and were then monitored on the EEG and analyzed for number of seizures as well as time spent in seizure activity, as detected by EEG analysis by a blind party. Figure 8 Lin et al. (2003) depicts the 40% decrease in the amount of seizures observed, with an average of 25 seizures noted in the control group compared to an average of 15 seizures in rats injected with rAAV-NSE-GAL. As shown in figure 9, there was a 55% decrease in total time spent in kainic acid-induced seizure activity. To determine galanin expression, another group of rats were injected unilaterally in the right dorsal hippocampus with rAAV-NSE-GAL. These rats were not subjected to seizures, but were killed 2.5 months Figure 9. Effects of rAAV-empty and rAAV-GAL on total after vector injection. Upon time in seizure activity. analysis of the hippocampal areas of these animals, it was found that there was a higher amount of GAL in the right dorsal side (injected Lin et al. (2003) with rAAV-NSE-GAL) than in the non-injected (left dorsal) side of the hippocampus. Mazarati et al. (1998) Mazarati et al. (1998) also studied rAAV-NSE-GAL in the rat hippocampus. This study was done on 8-10 week old rats, and was included as part of a later study of the effects of four neuropeptides – including NPY – in the rat hippocampus during a prolonged seizure, referred to as status epilepticus (SE). The animals were subjected to 10
  • 11. Figure 10. The perforant path of the hippocampus (in blue). either electrical stimulation in the perforant path to induce status epilepticus (SE). As its name implies, perforant path stimulation (PPS) http://www.nature.com/neuro/journal/ v10/n3/images/nn0307-271-F1.gif occurs in the perforant path of the hippocampus (see figure 10). This area is neuron-dense, and acts as the input pathway in the hippocampus (MRC, 2003). As a result, excess stimulation here will result in prolonged seizure activity. For quantitative analysis, the rats were implanted Figure 11. The dentate gyrus of the with a bipolar stimulating electrode in the perforant hippocampus (encircled in red). path, as well as a bipolar recording electrode in the dentate gyrus of the hippocampus, encircled in red in figure 11. Subjects received an injection of varying concentration of galanin either 30 minutes before the beginning http://www.brainybehavior.com/blog/wp -content/uploads/2008/11/gray747.png of PPS or 30 minutes after its conclusion. Controls received an injection of 0.9% NaCl. Figure 12. The effects of galanin on time spent in seizure activity between controls and GAL-treated rats. Figure 12 illustrates the findings that those rats treated with higher concentrations of galanin 30 minutes prior to PPS were 11 Mazarati et al. (1998)
  • 12. observed to have spent 95% less time in seizure activity than the controls. Animals were killed at varying time intervals following the conclusion of PPS to determine the concentration of GAL-infected neurons in and around the injection site. Figure 13 illustrates the concentration of GAL-positive neurons in control and GAL-treated rats. 24 hours after PPS, an average of 16 GAL-positive neurons were found in a slice of the hippocampus of GAL-treated rats, while none were found in non-GAL treated rats at any Figure 13. Difference in GAL-positive time following the conclusion of neurons after SSSE. PPS. However, 3 days after PPS, this number has gradually decreased to an average of 8 GAL- positive neurons in GAL-treated rats, and decreased to 6 GAL- positive neurons 7 days after the conclusion of PPS. In Mazarati et al. (1998) Mazarati and Wasterlain’s 2002 study of Figure 14. Difference in total time spent in seizure activity between control and GAL-treated rats. rAAV-GAL, it was found that, compared to 590 minutes spent in seizure activity in the rAAV-empty control, total time spent in seizures for rAAV-GAL treated animals was Mazarati and Wasterlain (2002) under 10 minutes, as is illustrated in figure 14. 12
  • 13. Haberman et al. (2003) In another study of the effects of galanin, Haberman et al. Figure 15. The inferior collicular cortex (in yellow) of the (2003) found that it is able to reduce seizure intensity. This brainstem. experiment used a vector constructed with a fibronectin secretory signal sequence (FIB) as its promoter. The rats used in this study were implanted with a stimulating electrode, and the inferior collicular cortex (as shown in figure 15, highlighted in yellow) of these animals’ brainstems was infused with either rAAV-FIB-GAL (experimental) or rAAV-GAL (control). Four days after treatment, seizure threshold was determined, and the rats were re-examined once per week for 4 weeks thereafter. In this period, http://upload.wikimedia.org/wikipedia/ commons/0/00/Gray685.png it was found that the threshold for seizure genesis was 60% higher in those animals treated with rAAV-FIB-GAL. Following this, the animals were given water with doxycycline, Figure 16. Seizure threshold comparison of control group and rAAV-FIB-GAL-treated rats. which returned the threshold to baseline levels within 1 week. After doxycycline was removed, rAAV-FIB-GAL-treated rats were observed to have a gradually increasing threshold for seizure genesis, with a threshold Haberman et al. (2003) approximately 30% higher in 13
  • 14. galanin-treated rats than those in the control group. Figure 16 illustrates these results. To determine if treatment was able to prevent seizures, animals were injected with either rAAV-FIB-GAL, rAAV-GAL, or received no treatment at all. In vitro, rAAV-FIB-GAL or rAAV-GAL was introduced into HEK 293 cells. Twenty-four hours later, the media were analyzed via ELISA for the presence of galanin. The rAAV-GAL cells showed no detectable amount of GAL, indicating that it was not expressed or secreted in the cells, but those cells infected with rAAV-FIB-GAL showed a significant amount (32ng/mL). Neuropeptide Y (NPY)-focused studies Richichi et al. (2004) Other studies focused on the effects of rAAV-delivered neuropeptide Y (NPY) in the rat brain, more specifically, the hippocampus. Richichi et al. (2004) conducted an experiment on adult male rats using rAAV-NSE-NPY. Neuron-specific enolase (NSE) functioned as a cell-specific promoter in this vector. Two subtypes (called serotypes and determined by protein markers on the cell surface) of rAAV were used in this experiment – serotype 2, and a mixture of serotypes 1 and 2 (called serotype 1/2). The hippocampus of these rats was injected bilaterally with the vector on each side. Eight weeks later, the animals were implanted with electrodes and cannulas, and 4 days after implantation, received a unilateral injection of kainic acid in either the dorsal hippocampus to induce Figure 17. Difference in observed seizure activity and onset time of seizures. Richichi et al. (2004) seizure activity. Seizure activity was measured via electroencephalogram (EEG) analysis by a blind party, and was done before injecting the 14
  • 15. animals with kainic acid, and up to 3 hours following the injection. As shown in figure 17, time for seizure onset was delayed almost twofold in both serotypes of rAAV-NSE- NPY, with an average of 11.5 minutes observed in serotype 1/2, compared to an average of 6.2 minutes in the control group. In these subjects, those treated with serotype 1/2 had no EEG-detected episodes of status epilepticus (SE), but in the control group, SE lasted at least 60 minutes. Mazarati and Wasterlain (2002) Mazarati and Wasterlain (2002) conducted a study of the effects of four neuropeptides in the rat hippocampus, one of these neuropeptides being GAL, and another being NPY. This study used 8-10 week old male rats, which were implanted with a bipolar stimulating electrode in the perforant path of the hippocampus (see figure 10), as well as a combination of a bipolar recording electrode and guide cannula in the dentate gyrus of the hippocampus (see figure 11). These animals were subjected to 30 minutes of perforant path stimulation (PPS) 7-10 days later, to induce a state of SSSE. Recordings of brain activity were made via EEG by a blind party during PPS and 24 hours after its conclusion. Spike distribution for this activity was measured in 30 minute periods, and seizure activity was assessed according to total time spent in seizure activity, time between the end of PPS and occurrence of the last seizure, and time spent in seizure activity in a 1 hour period during a state of prolonged seizure activity (called self- sustaining status epilepticus, or SSSE). Ten minutes after the conclusion of PPS, the dentate gyrus of the rats were injected with NPY. Controls were injected with the same dose of 0.9% NaCl. As illustrated by figure 18, there was no significant difference noted in time spent in EEG-detected seizure activity between the control (average time in 15
  • 16. seizure activity: 15 minutes) and NPY-treated group (average time in seizure activity: 13 minutes). NPY-treated animals were found to have spent 4 hours total in seizure activity, and SSSE decreased to less than 20 minutes in these animals. Discussion Mazarati et al. (1998) showed that, when injected Figure 18. Time in seizures following PPS treatment. into the dentate gyrus, galanin has a seizure-protecting effect. In this situation, galanin prevents the initiation of self-sustaining status epilepticus (SSSE). When injected after perforant path stimulation was performed, galanin was able to stop the maintenance phase of an established SSSE, an accomplishment even anticonvulsant drugs could not achieve. Similarly, Lin et al. (2003) showed that when rAAV-NSE-GAL was used, an over- Mazarati and Wasterlain (2002) expression of GAL by the vector was able to drastically reduce seizure activity brought about by an intrahippocampal injection of kainic acid, in addition to producing a strong anticonvulsant effect. Neuropeptide Y (NPY) in neuronal cells causes a decrease in both seizure susceptibility and epileptogenesis, and therefore, the use of rAAV-NSE-NPY for over- expression of NPY in rat hippocampal cells was used to inhibit seizures and epileptogenesis (Richichi et al., 2004). In these rats, kainic acid-induced, EEG-detected episodes of prolonged seizure activity (status epilepticus, or SE) were not observed in 16
  • 17. NPY-treated rats, but episodes lasting at least 60 minutes were observed in the control group. A delay in onset of seizures was observed in these animals, with NPY-treated rats having an average delay time of approximately 11.5 minutes on average, while the average delay for the control group was observed to be 6.2 minutes, almost half that of the experimental group. Animals that received an injection of kainic acid showed a decrease in EEG-detected seizure activity induced by the injection, due to rAAV vector- mediated NPY over-expression. EEG-detected episodes of SE were eliminated in rats expressing the NPY transgene in multiple areas of the hippocampus. Richichi et al. (2004) concluded that the ability of NPY to inhibit the release of excitatory neurotransmitters from brain cells may be the cause of the decrease in seizures noted in rats injected with rAAV-NSE-NPY. In the hippocampus, NPY has inhibitory properties, acting as an anticonvulsant. According to Mazarati and Wasterlain (2002), mice that lack NPY are more inclined to develop seizures. In their study, Mazarati and Wasterlain (2002) were able to show that, after injecting the dentate gyrus in rats with rAAV-delivered NPY, SSSE episodes were significantly attenuated, though only a 2-minute difference in time spent in seizures was observed between the experimental and control groups. This indicates that there was no significant difference in time spent in seizures when comparing NPY-treated rats to the saline-injected controls. Conclusions In order to determine the most effective method of rAAV-mediated gene therapy, many aspects must be considered. These include the level of expression of the transgene included in the vector, length of time spent in seizure activity, length of time between 17
  • 18. seizure activity, and number of seizures observed. By analyzing rAAV-mediated experiments involving one or both of the neuropeptides of interest, NPY and GAL can be compared and contrasted, and a conclusion regarding efficacy can be more easily determined. In the studies conducted regarding rAAV vectors containing the GAL transgene, there was a considerable reduction in seizure activity. There was an average decrease of 77% in time spent in seizure activity, as well as a 40% decrease in number of seizures observed. There was also found to be a large quantity of galanin expression and secretion in cells in and around the injection site in the hippocampus, with 16 times more galanin- positive cells found in galanin-treated animals than in the control group. The experiments conducted with rAAV-NPY vectors also showed a decrease in EEG-detected seizure activities, but it was only a 13% decrease – not nearly as vast as that observed in experiments conducted with rAAV-GAL vectors. When compared, each neuropeptide was observed to have an anticonvulsant effect, as well as the ability to decrease time spent in seizure activities, decrease time between seizures, and to decrease the number of seizures observed in the tested animals compared to the control. However, rAAV-GAL vectors were observed to have a greater effect than the rAAV-NPY vectors and because of this, rAAV vectors utilizing galanin as a transgene for expression and secretion in neuronal cells are the best choice for gene therapy. While these vectors have worked well in rodent models of seizures, there are still many obstacles that must be surpassed before this treatment can be applied to humans. These include establishing an effective, minimally invasive method for vector delivery, 18
  • 19. as well as receiving approval from the Food and Drug Administration (FDA) for treatment. Once established, these tests may be able to help control seizures in humans suffering from epileptic disorders. AAV-mediated gene therapy is a promising new therapy, and will hopefully one day become a widespread, successful treatment for not just epilepsy, but many other human seizure disorders. References Burton, E.A., J.C. Glorioso, and D.J. Fink. 2007. Gene Therapy Approaches in Neurology. In Molecular Neurology. Ed. S.G. Waxman. Elsevier Academic Press. Burlington, MA. 101-123. Carter, B.J. 2008. Adeno-Associated Virus Vectors. In Concepts in Genetic Medicine. Ed. B. Dropulic and B.J. Carter. Wiley-Liss. Hoboken, NJ. 61-68. Chandler, K. 2006. Canine epilepsy: What can we learn from human seizure disorders? The Veterinary Journal. 172: 207-217. Dong, J.Y., P.D. Fan, and R.A. Frizell. 1996. Quantitative analysis of the packaging capacity of recombinant adeno-associated virus. Human Gene Therapy. 7: 2102-2112. Dumont, Y. and R. Quirion. 2006. An overview of neuropeptide Y: pharmacology to molecular biology and receptor localization. In NPY Family of Peptides in Neurobiology, Cardiovascular and Metabolic Disorders: from Genes to Therapeutics. Ed. Z. Zukowska and G.Z. Feuerstein. Birkhaüser Verlag. Basel, Switzerland. 7-33. Haberman, R.P., R.J. Samulski, and T. J. McCown. 2003. Attenuation of seizures and neuronal death by adeno-associated virus vector galanin expression and secretion. Nature Medicine. 9(8): 1076-1080. Haberman , R.P., T.J. McCown, and R.J. Samulski. 1998. Inducible long-term gene expression in brain with adeno-associated virus gene transfer. Gene Therapy. 5: 1604- 1611. Hains, B. 2006. Brain Disorders. Chelsea House Publishers. Philadelphia. 37-38. Lin, E.D., C. Richichi, D. Young, K. Baer, A. Vezzani, and M.J. During. 2003. Recombinant AAV-mediated expression of galanin in rat hippocampus suppresses seizure development. European Journal of Neuroscience. 18: 2087-2092. 19
  • 20. Mazarati, A.M and C.G. Wasterlain. 2002. Anticonvulsant effects of four neuropeptides in the rat hippocampus during self-sustaining status epilepticus. Neuroscience Letters. 331: 123-127. Mazarati, A.M., H. Liu, U. Soomets, R. Sankar, D. Shin, H. Katsumori, Ü. Langel, and C.G. Wasterlain. 1998. Galanin modulation of seizures and seizure modulation of hippocampal galanin in animal models of status epilepticus. Journal of Neuroscience. 18(23): 10070-10077. Medical Research Council. 2003. University of Bristol. Hippocampal Pathways. http://www.bristol.ac.uk/synaptic/info/pathway/hippocampal.htm National Institutes of Health. 2004. National Institute of Neurological Disorders and Stroke. Seizures and Epilepsy: Hope Through Research. 13-27. Pieribone, V.A., Z.D. Xu, X. Zhang, and T. Hökfelt. 1998. Electrophysiologic Effects of Galanin on Neurons of the Central Nervous System. In Annals of the New York Academy of Sciences. Ed. B.M. Boland, J. Cullinan, and A.C. Fink. New York Academy of Sciences. New York. 264-273. Ransom, C.B. and H. Blumenfeld. 2007. Acquired Epilepsy: Cellular and Molecular Mechanisms. In Molecular Neurology. Ed. S.G. Waxman. Elsevier Academic Press. Burlington, MA. 347-370. Riban, V., H.L. Fitzsimons, and M.J. During. 2009. Gene therapy in epilepsy. Epilepsia. 50(1): 24-32. Richichi, C, E.D. Lin, D. Stefanin, D. Colella, T. Ravizza, G. Grignaschi, P. Veglianese, G. Sperk, M.J. During, and A. Vezzani. 2004. Anticonvulsant and antiepileptogenic effects mediated by adeno-associated virus vector neuropeptide Y expression in the rat hippocampus. Journal of Neuroscience. 24(12): 3051-3059. Samulski, R.J., M. Sally, and N. Muzyczka. 1999. Adeno-Associated Viral Vectors. In The Development of Human Gene Therapy. Ed. T. Friedmann. Cold Spring Harbor Laboratory Press. Cold Spring Harbor, NY. 131-172. Scharfman, H.E. and W.P. Gray. 2006. Plasticity of neuropeptide Y in the dentate gyrus after seizures, and its relevance to seizure-induced neurogenesis. In NPY Family of Peptides in Neurobiology, Cardiovascular and Metabolic Disorders: from Genes to Therapeutics. Ed. Z. Zukowska and G.Z. Feuerstein. Birkhaüser Verlag. Basel, Switzerland. 193-211. Vezzani, A. 2004. Gene Therapy in Epilepsy. Epilepsy Currents. 4(3): 87-90. 20