seizure-like events in ca1 of the rat hippocampus

75
The Pennsylvania State University The Graduate School Huck Institute of Life Sciences INTERACTIONS BETWEEN ORIENS, BASKET AND PYRAMIDAL CELLS DURING IN VITRO SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS A Dissertation in Neuroscience by Ruchi Parekh © 2010 Ruchi Parekh Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy December 2010

Upload: others

Post on 21-Nov-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

The Pennsylvania State University

The Graduate School

Huck Institute of Life Sciences

INTERACTIONS BETWEEN ORIENS, BASKET AND PYRAMIDAL CELLS DURING IN VITRO

SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

A Dissertation in

Neuroscience

by

Ruchi Parekh

© 2010 Ruchi Parekh

Submitted in Partial Fulfillment

of the Requirements

for the Degree of

Doctor of Philosophy

December 2010

Page 2: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

ii

The dissertation of Ruchi Parekh was reviewed and approved* by the following:

Steven J Schiff

Brush Chair Professor of Engineering

Professor of Engineering Science and Mechanics, Neurosurgery and Physics

Dissertation Advisor

Chair of Committee

Bernhard Luscher

Professor of Biology

Bruce Gluckman

Associate Professor of Engineering Science and Mechanics, Neurosurgery

Byron Jones

Professor of Biobehavioral Health/Pharmacology

John R. Cressman

Assistant Professor of Physics & Astronomy/Krasnow Inst. for Advanced Study

George Mason University, Fairfax, VA

Special Member

Jokubas Ziburkus

Assistant Professor of Biology and Biochemistry

University of Houston, Houston, TX

Special Member

Ping Li

Professor of Psychology, Linguistics, and Asian Studies

Co-Chair, Neuroscience Graduate Program

*Signatures are on file in the Graduate School.

Page 3: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

iii

ABSTRACT

Network activity in the brain is shaped by interactions between cells. There is limited

knowledge about such interactions in pathological conditions like epilepsy. Previous

results from our laboratory showed unique interplay between pyramidal cells and oriens

lacunosum-moleculare interneurons in rat hippocampal CA1 during in vitro seizure-like

events (SLEs). To further investigate the interactions between different sub-types of

interneurons and pyramidal cells, we performed simultaneous dual and triple whole cell

patch and extracellular recordings in pyramidal, basket and oriens cells in the CA1

region of the rat hippocampus. Triple immunofluorescence confirmed the identity of

patched cells. We measured spike frequency, spike and subthreshold correlation

between these cells before, during and after single seizure-like events (SLEs) and during

the inter-seizure intervals (ISIs). We confirmed our previous findings and found a

distinctly different firing pattern between the perisomatically-innervating basket cells

and the dendritically-innervating oriens cells during 4-aminopyridine induced SLEs in the

presence of low magnesium. At the pre-ictal stage of the SLE the subthreshold

correlation between pyramidal and basket cells is strong; oriens cells show high firing

frequency, whereas the basket cells show a gradual increase. At the start of the SLE, the

previously strong subthreshold correlations between pyramidal and basket cells

decrease; all 3 cells show increased firing rates with basket and oriens cells reaching

their maximum firing rates. Following this, subthreshold correlations for all 3 cells

decrease; oriens cells go into depolarization block that is characterized by total spike

failure, whereas pyramidal and basket cells continue firing. SLE termination is defined by

conditions returning to pre-ictal dynamics. During the ISI all three cell types increase

their firing activity at different times before onset of the subsequent seizure.

Our findings further emphasize the importance of cell-type specific differences in

interactions between excitatory and inhibitory cells during SLEs. Other similar cellular

dynamics may be representative of the hippocampal circuitry under conditions that

cause general hyperexcitability. We also show a significant gradual increase in spike

rates in both the inhibitory and excitatory cells, starting at least a minute before seizure

onset. These findings expand on the current knowledge of seizure formation patterns

and can be used in studies of seizure prediction models.

Page 4: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

iv

Contents

List of figures ………………………………………………………………………………………………………………..v

Introduction ………………………………………………………………………………………………………………….1

1. What is epilepsy?…………………………………………………………………………………...1

2. Electrophysiological seizures….……………………………………………………………….3

3. Region of interest ………………………………………………………………………………….5

i. The hippocampus in the normal brain ……………………………………….5

ii. The hippocampus and epilepsy ………………………………………………….6

4. Interneuron diversity …………………………………………………………………………….8

5. Seizure models …………………………………………………………………………………….11

i. High K+ ……………………………………………………………………………………..11

ii. Low Mg2+

………………………………………………………………………………….12

iii. Kainic acid ………………………………………………………………………………..13

iv. 4-Aminopyridine ………………………………………………………………………14

Research Design and Methods ……………………………………………………………………………………17

Animals ……………………………………………………..……………………………………………………17

Hippocampal slice preparation ……………………………………………………………………….17

Electrophysiology ……………………………………………………………………………………………17

Immunohistochemistry and confocal microscopy ……………………………………………21

Correlation Analysis ………………………………………………………………………………………..24

Statistical Analysis …………………………………………………………………………………………..28

Results ………………………………………………………………………………………………………………………..30

1. Spike Rate …………………………………………………………………….……………………………34

2. Correlation Analysis………………………………………………………………….…………………39

i. Pyramidal cell - Pyramidal cell …………………………………………………..……39

ii. Basket cell - Basket cell …………………………………………………………………..41

iii. Basket cell - Pyramidal cell ………………………………..……………………………43

iv. Oriens interneuron - Basket cell …………………..…………………………………45

v. Oriens interneuron - Pyramidal cell ………………………..………………………47

Summarized Results …………………..………………………………………………………………………………..50

Pyramidal cells ………………………………………………………………………………………………..50

Basket cells ……………………………………………………………………………………………………..54

Oriens interneurons ………………………………………………………………………………………..55

Discussion ..………………………………………………………………………………………………………………….57

Conclusion …………………………………………………………………………………………………………………..60

References …………………………………………………………………………………………………………………..65

Page 5: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

v

List of Figures

Figure 1: Diagram of location of electrodes ………………...................................................18

Figure 2: Electrical membrane properties of patched cells ………………………………………….20

Figure 3: Confocal images ………………………………………………………………………………………..…22

Figure 4: Single seizure and resample analysis ……………………………………………………………25

Figure 5: Interleaving pattern of interplay between OLM and pyramidal cells ……………31

Figure 6: Schematic diagram of cell-types investigated in our experiments ………………..32

Figure 7: Single seizure spike rate ……………………………………………………………………………….36

Figure 8: Inter-seizure interval spike rate ……………………………………………………………………37

Figure 9: Linear regression analysis of inter-seizure interval ……………………………………….38

Figure 10: Correlation analysis of pyramidal-pyramidal cell pairs ……………………………….40

Figure 11: Correlation analysis of basket-basket cell pairs ………………………………………….42

Figure 12: Correlation analysis of basket-pyramidal cell pairs …………………………………….44

Figure 13: Correlation analysis of oriens interneuron-basket cell pairs ……………………….46

Figure 14: Correlation analysis of oriens interneuron-pyramidal cell pairs ………………….49

Figure 15: Summarized resample analysis ………………………………………………………………….53

List of Tables

Table 1: Number of cell pairs patched and analyzed for SLEs ……………………………………23

Page 6: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

1

Introduction

1. What is Epilepsy?

Epilepsy is a chronic condition characterized by repeated, unprovoked seizures. It is a

disorder that results from many different underlying conditions. There are many causes

of seizures, like trauma, infection, drugs etc., however not all people that have seizures

are epileptic. More than 5% of all people suffer from at least one epileptic seizure

during their lives and at any one time the prevalence of epilepsy is 0.5-1.0% of the

general population (Somjen 2004, Sanders and Shorvon, 1996).

Depending on the area of the brain involved seizures can be classified as partial or

generalized. Partial seizures can be either simple or complex depending on whether the

patient retains consciousness, in the former, or partially or completely loses it, in the

latter. Temporal lobe epilepsy is so called because complex partial seizures originate

within the structures of the temporal lobe. Simple partial seizures that involve the

motor cortex are called “Jacksonian seizures” that manifest as a twitch in one part of an

extremity and slowly spread over the body. Partial seizures that start in a small area of

the brain and then spread to the opposite side of the brain are called secondary

generalized seizures. Generalized seizures can be either petit mal, where the patient

loses consciousness for a brief time or grand mal, where the patient has convulsive

seizures. Grand mal is also knows as generalized tonic-clonic convulsions. The condition

in which seizures that last for 30 minutes or longer is called status epilepticus. Epilepsy

can be a result of a known brain lesion or disease or it can be due to unknown causes

like genetic abnormalities.

Despite the different initial causes and differences in manifestations of the seizures,

certain underlying pathophysiological features are common. Excessive excitation,

reduced inhibition or an increase in both excitation and inhibition can result in seizures.

In 1929 Hans Berger first published an EEG recording from the intact scalp of a

schizophrenic patient. He described various waveforms seen in different states of

Page 7: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

2

wakefulness and sleep giving rise to the current conventions of brain wave frequencies.

In an EEG recording the ictal event of the seizure is seen as a sudden increase in

frequency and amplitude of the waves. The increase in frequency can vary for different

types of seizures. In humans a petit mal seizure is observed as a 3-5 Hz rhythm and a

generalized tonic seizure will have a frequency of 15-20 Hz. Interictal discharges

between seizure events are classified as EEG spikes, sharp waves, poly-spikes, isolated

large amplitude slow waves and isolated spike-wave complexes (Somjen 2004).

Matsumoto and Ajmone-Marsan (1964) first described paroxysmal depolarization shifts

(PDS) in the “epileptic neuron” of the cat cerebral cortex, as “the typical, most

characteristic epileptiform expression of the neuron”. Using topical application of

penicillin as an experimental reproduction of epileptogenic foci, they made extracellular

and intracellular recordings from the cerebral cortex of cats. They studied the

spontaneous and repetitive interictal and ictal activity. They described in detail the giant

depolarizing shift in the membrane potential of a neuron which would often increase

the firing level and then inactivate the spike mechanism. In other cases the spikes would

be preserved during the PDS. A steep repolarization of the membrane potential ended

the PDS. Traynelis and Dingledine (1988) reported novel spontaneous electrographic

seizures in the rat CA1 that contained components of discharges similar to in vivo

recordings during tonic-clonic motor seizures using the high extracellular K+

model.

Simultaneous recordings from the CA3 and CA1 regions showed that spontaneous

interictal bursts in from the CA3 region triggered the electrographic seizures in CA1.

Isolating the CA1 region stopped the seizures, and electrical stimulation of the Schaffer

collaterals re-established the CA1 seizures. They concluded that interictal activity in the

CA3 was necessary but not sufficient for CA1 seizures. Certain other conditions like

elevated extracellular K+([K

+] o) in the CA1 region, lower spike threshold, cell swelling

leading to a decrease in extracellular space, decreased IPSPs and depolarization leading

to NMDA receptor activation were also associated with CA1 seizures.

Page 8: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

3

Our knowledge of the complex interactions between different regions of the

hippocampus during seizures has since then steadily increased. Besides differences in

activity in different regions, we now recognize the differences in contribution of distinct

cell sub-types to seizure-like events. Studying these complex interactions at the local

network level is crucial to further our understanding of the formation of seizure

patterns.

2. Electrophysiological seizures

Seizures are the result of an imbalance in the excitation-inhibition ratio. It could be an

increase in excitation, a decrease in inhibition or an asymmetric change in both. Seizure

discharges are synchronized network activity of a population of neurons caused by the

pathology of these neurons. Experimental seizures are often identified by a group of

action potentials riding on a slow depolarizing wave called a paroxysmal depolarizing

shift (PDS). This wave starts with an initial fast depolarization, then continues with a

slow depolarization and end with a hyperpolarization (Somjen 2004).

Though the PDS is the experimental cellular correlate of clinical epileptiform discharges,

it is important to note that the underlying mechanism of PDS is different for different

seizure models. PDSs can be evoked synaptically or generated via intrinsic membrane

current or a combination of both. As Wong and Traub (1983) showed in a disinhibited

hippocampal slice, PDSs are generated via intrinsic membrane current in the CA2-CA3

region then synaptically transmitted to the CA1 region. Another explanation suggests

that the PDSs are giant EPSPs. However the source of the giant EPSPs remains unclear.

One possibility may be reduced inhibition resulting in an increase in excitation or an

overall increase in neurotransmitter release, like that seen in the 4-aminopyridine

seizure model. Then the obvious question to follow is which of these models correctly

represents the clinical epileptiform discharges observed in human patients? There may

not be a single model that correctly represents clinical epileptiform discharges, which

are not just one type. But what is agreed upon is that a PDS is generated by an increase

Page 9: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

4

in excitatory synaptic potentials followed by Ca2+

current-driven intrinsic membrane

currents (Somjen 2004).

Specifically in the hippocampus, pyramidal cells in the CA3 region have the intrinsic

ability to generate bursts (hence they are known as the pacemakers). CA1 cells are

unable to do so. The burst discharge is propagated from CA3 to CA1 via synaptic

connections (Wong and Traub 1983). Along with synaptic connections the seizure

discharges propagate via electrical interactions: ephaptic interactions and gap junctions.

Due to the close apposition of the pyramidal cell somata electrical current can flow from

one cell to the next and can influence its timing of excitation. This mechanism is called

ephaptic interactions and it contributes to the initiation of electrographic seizures

(Traynelis and Dingledine 1988). The tightly packed pyramidal neurons in the

hippocampus are connected to each other via gap junctions in their axons. Traub et al

(2001) suggest that “very fast oscillations” that precede the onset of seizures occur due

to gap junctions. They demonstrated that “spontaneous very fast oscillations” were

readily expressed in the presence of a gap junction-opening compound

tetramethylamine (TMA) or when chemical synaptic transmission was suppressed. Using

experimental and simulation data they suggest that gap junction networks may become

pathologically active as a result of impaired synaptic transmission and may be

responsible for initiating seizures. Along the same lines gap junction blockers prevent

the transition from the interictal to the ictal stage in low Mg2+

-induced seizures in rat

hippocampal slices (Koehling et al 2001).

Seizures can also propagate chemically via volume transmission; i.e. the extra glutamate

and K+ in the extrasynaptic space can excite neighboring neurons. Astrocytic networks

may also contribute to the spread. K+ and Ca

2+ can spread through the gap junctions and

the latter can induce the release of glutamate from astrocytes causing further excitation

(Somjen 2004).

Page 10: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

5

3. Region of interest – The hippocampus

i. The hippocampus in the normal brain

Technological advances over the decades have expanded our knowledge about the

functional role of the hippocampus in the human brain. Though the human

hippocampus cannot be studied as in detail in vivo, animal in vitro studies have provided

a wealth of information about the neuroanatomy, neurophysiology and functional

characteristics of individual cells, pathways, local circuits and network activities. The role

of the hippocampus in memory is now unquestionable. Studies have established that

the hippocampus is involved in implementing some types of memory and in memory

processing. Its role in declarative memory, spatial memory, and LTP/LTD phenomena,

though not completely clear, is acknowledged.

The functional significance of the hippocampus in declarative memory is attributed to a

patient called H.M. He underwent a bilateral temporal lobe resection to control

intractable epilepsy. The resulting impairment in his memory was intensively studied by

Scoville and Milner (1957) and it revealed a pattern of anterograde and retrograde

amnesia in the patient. He was unable to retain some long-term memory for new facts

or events and he had lost some memory that was acquired before the operation. His

short-term memory remained intact and there was no loss of general intellect. The

information learned from H.M.’s case along with several other patients with similar

resections helped in the understanding of the functional role of the hippocampus.

The hippocampus is assumed to be involved in the initial encoding and storage of spatial

memory. The discovery of “place cells” in the hippocampus by O’Keefe and Dostrovsky

in 1971 led to the study of the hippocampus in spatial memory. On recording electrical

activity from individual cells in the hippocampus of freely moving rats, they found that

the firing patterns of certain cells increased when the rat moved through a particular

region in space. Different “place cells” reacted to different regions. They suggested the

firing of different groups of “place cells” might be the neural representation of spatial

maps of an environment. Many theories have attempted to delve into the implications

Page 11: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

6

of a spatial memory; however, how the space map may be represented or used by the

hippocampal circuitry is not yet fully understood (Morris 2007).

Long-term potentiation (LTP), a form of activity-dependent synaptic plasticity, is brought

about by a brief period of coordinated neuronal activity which leads to a long-lasting

increase in synaptic strength. Even though LTP has been studied extensively in the

hippocampus it also occurs in the cortex, thalamus, amygdala, ventral tegmental area,

nucleus accumbens, neostriatum and in the cerebellum. It was Hebb (1949) who

proposed the idea that simultaneous activity between two neurons would lead to

strengthening of their synapses, hence the phrase “neurons that fire together, wire

together”. LTP was first described in 1973 by Timothy Bliss and Terje Lomo and

colleagues at excitatory synapses between the perforant pathway fibers and the granule

cells of the dentate gyrus in the hippocampus and defined it as a persistent

enhancement of EPSP following brief tetanic stimulation of afferent pathway. LTP and

LTD result from experimental protocols that cause synaptic modifications. Thus the

question to what extent the physiological properties of synaptic plasticity are correlated

to learning at the behavioral level remains unanswered. Martin, Grimwood and Morris

(2000) suggest the synaptic plasticity and memory (SPM) hypothesis which states:

“Activity-dependent synaptic plasticity is induced at appropriate synapses during

memory formation and is both necessary and sufficient for the information storage

underlying the type of memory mediated by the brain area in which the plasticity is

observed”.

ii. The hippocampus and epilepsy

The hippocampus is secondarily implicated in diseases like Alzheimer’s and

schizophrenia, is vulnerable to damage as a consequence of trauma, hypoglycemia and

ischemia/hypoxia, but is directly involved in temporal lobe epilepsy (TLE). TLE represents

60% of patients affected with partial epilepsies (Walker, Chan, Thom 2007). Neuronal

cell loss and gliosis in the hippocampus, specifically in the CA1 region is a hallmark of

TLE; however if the sclerosis is the cause or the result of TLE remains unclear. The

Page 12: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

7

neuronal cell loss that does follow the onset of epilepsy is due to excitotoxicity. Besides

sclerosis other characteristic pathology of the epileptic hippocampus includes structural

and organizational changes, aberrant neuronal connections, changes in function and

changes in intrinsic membrane properties of the cells. These changes can further

contribute to the probability of epileptogenesis.

Epileptic patients demonstrate a high frequency of temporal lobe foci. Often when all

pharmacologic treatment methods fail, surgical resection of parts of the temporal lobes

controls the intensity and frequency of seizures in some patients. However this is hardly

the kind of treatment one should settle for.

This is why in vitro and in vivo experiments in the hippocampus have been crucial in

helping us understand the underlying dynamics of a seizure. By studying the cellular

correlates of epileptiform discharges, we now know about some of the mechanisms of

network activity during seizures. Experiments allow us to draw inferences about

network activity and its underlying mechanisms, which can be used to isolate

therapeutic targets and develop treatments to control seizures. Experimental data is

also used to create computational models that can simulate conditions that are not

possible to create in an experimental setup.

Besides being the structure most prone to seizures, the importance of the hippocampus

in experimental studies is also due its cytoarchitecture. The layout of the hippocampus

is a very ordered one, where the cell bodies are tightly packed together in one layer and

the dendrites and axons occupy other distinct layers with glial cells and interneurons

scattered throughout. This simple layout allows for easy access to the cell bodies for

intracellular recordings when compared to the neocortex for example, where the cell

bodies, axons and dendrites are found across all five layers (Somjen 2004).

Page 13: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

8

4. Interneuron diversity

Network activity in the brain is shaped by interactions between cells. Inhibition has an

important role in balancing excitation and in controlling spike timing (Mann et al 2007).

In the hippocampus, feedforward and feedback inhibition form the two fundamental

modes of inhibition (Elfant et al 2007). The type of inhibition is cell-type specific,

however the underlying principle stays the same, which is, inhibitory innervations on

pyramidal cells results in hyperpolarization independent of the location along the entire

somato-dendritic axis of the pyramidal cell (Glickfield et al 2009). There are at least 21

different types of inhibitory neurons in the rat hippocampal CA1 region (Klausberger et

al 2008) which have been identified based on their electrophysiological characteristics,

intrinsic cellular markers, location of the soma within the hippocampal layers, dendritic

morphology, axonal arborizations and axonal targets (Sik et al 1995; Buhl et al 1996;

Halasy et al 1996; Han 1996; Klausberger et al 2008). These distinct characteristics for

the cell sub-types define their cellular function and contribution to the physiology of the

CA1 network.

Different types of interneurons innervate different domains of the pyramidal cells and

coordinate the activity of pyramidal cell populations in a manner that is temporally

distinct and brain-state-dependent (Klausberger et al 2003). In the current study, we

focused on the oriens lacunosum-moleculare (OLM) and parvalbumin positive basket

cells. In the CA1 the soma of OLM cells are located in stratum oriens with horizontal

somatodendritic orientation. The dendrites span stratum oriens and the axons project

to stratum lacunosum moleculare innervating the distal pyramidal dendrites and are

involved in feedback inhibition. In contrast, basket cell soma are located in the stratum

pyramidale with vertically oriented dendrites that span stratum radiatum and stratum

oriens, and their axons innervate the proximal dendrites and somata of pyramidal cells.

Basket cells receive extrinsic excitatory inputs along with excitatory afferents from the

pyramidal cells (Buhl et al 1996); these extrinsic excitatory inputs and the axons

terminating on and near the soma of the pyramidal cells make it possible for basket cells

Page 14: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

9

to be involved in feedforward inhibition. Since the time window for neuronal integration

at the pyramidal cell is narrower for inputs closer to the soma and is broader for distal

inputs (Pouille et al 2001), inhibition is stronger at the soma than at the dendrites.

Dendritically innervating OLM cells control the efficacy and plasticity of glutamatergic

inputs, whereas perisomatically innervating basket cells control the synchrony of action

potential outputs of pyramidal cell populations (Miles et al 1996). Hence local

alterations in patterns of inhibition may result in pathological forms of population

synchrony.

Inhibitory neurons are involved in temporal synchronization of principal cells in the

hippocampus and play a crucial role in rhythm generation (Whittington et al 2003).

During local field gamma oscillations (Gillies et al 2002), in an in vitro model of gamma-

and theta-frequency (30-80 Hz and 5-12 Hz, respectively) oscillations in the

hippocampus, the soma of pyramidal cells receives gamma-frequency IPSPs while the

distal dendrites receive theta-frequency IPSPs. During local field theta-frequency

oscillations, the soma and distal dendrites of the pyramidal cells receive theta-frequency

IPSPs. In simultaneous recordings of pre-synaptic basket and axo-axonic cells and post-

synaptic pyramidal cells (Cobb et al 1995), stimulating the pre-synaptic interneuron at 1-

8 Hz phase-locked the post-synaptic pyramidal cells at theta frequencies; thus the

entrained pyramidal cell’s firing could be increased or decreased by the pre-synaptic

interneuron. Rhythmic activation of the pre-synaptic interneurons entrained both the

sub-threshold and supra-threshold activity of the pyramidal cells. Hence different

interneurons impose different rhythms on the pyramidal cells in a compartment specific

manner.

The differences between OLM and basket cells like location of soma, dendritic

morphology, axonal arborizations and targets, and intrinsic membrane properties also

influence the type of inhibition they impose. In kainate induced local field gamma

oscillations, Pike et al (2000) showed that these two types of interneurons receive

similar rhythmic EPSCs, however, because of the differences in their intrinsic membrane

Page 15: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

10

properties the output frequencies are completely different from each other. Given their

role in network synchronization (Gillies et al 2002; Pike et al 2000),in controlling the

global activity within the hippocampal formation, and their importance and vulnerability

in epilepsy (Freund et al 2007) understanding the role of both OLM and basket cells in

epilepsy is crucially important.

Impairment in inhibition function has been implicated in temporal lobe epilepsy. In the

pilocarpine model of chronic temporal lobe epilepsy, Stief et al (2006) showed that

interneurons located at the border between stratum radiatum and stratum lacunosum

moleculare, called SRL interneurons, undergo a reduction in synaptic inhibition and are

thus disinhibited, leading to an increase in the excitation-inhibition ratio in SRL

interneurons. This results in an increase in the feedforward inhibition onto pyramidal

cells. At the same time OLM cells that are involved in feedback inhibition die, thus

reducing the feedback inhibition onto pyramidal cells. This imbalance enhances

feedforward inhibition and contributes to the characteristic synchronized events of

seizure dynamics.

Feedforward inhibition is also a prime determinant controlling the speed of epileptiform

activity propagation. In a zero magnesium (0 Mg2+

) model of epilepsy, the number of

preictal feedforward inhibitory barrages inversely determines the propagation velocity

of epileptiform activity. A loss in pre-ictal feedforward inhibitory barrages is coincident

with an increase in the velocity of epileptiform activity in coronal slices of mouse

occipital cortex (Trevelyan et al 2007).

The current study explores how cell-type specific differences in interactions between

excitatory and inhibitory cells shape SLE dynamics.

Page 16: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

11

5. Seizure Models

In vitro and in vivo experiments to study seizure activity aim to create conditions that

are physiologically probable in order to study the mechanisms of seizure dynamics in

the hippocampal network. Hence models using high extracellular K+, low extracellular

Mg2+

, kainic acid injections or 4-aminopyridine perfusions replicate processes that are

present biologically.

i. High K+

The resting membrane potential of a neuron is between -60mV to -80mV and is a result

of net movement of various ions including K+, Na

+ and Cl

- across the membrane. At rest

typically the concentrations of K+, Na

+ and Cl

- ions inside the neuron is (in mM) 140, 15

and 8, and that outside the neuron is (in mM) 3, 150 and 130 (Shepherd 2004). Thus K+

is in higher concentration inside than outside and Na+ and Cl

- are higher outside. Under

normal conditions during the repolarization phase of the action potential the K+

channels open and K+ flows out to repolarize the membrane and return it toward the

negative resting potential.

Under abnormal conditions like paroxysmal discharges provoked by stimulation of the

angular bundle in the rat hippocampus an increase in extracellular K+ ([K

+]o) in the fascia

dentata was observed only in the cell body layer of the granule cells and not in the

dendritic layer (Somjen et al 1985). The sudden and very localized increase in K+ in the

cell body layer led the authors to conclude that the paroxysmal discharges were

associated with an outflow of K+ ions from the soma and not from the dendrites.

Fertzinger and Ranck (1970) loaded radioactive K42

on brain surfaces of rats and cats and

used the efflux of the isotope from the brain as a measure of change in [K+]o during

seizures induced by tetanic stimuli or intravenously administered pentylenetetrazol

(Metrazol). They found a significant increase in interstitial K+ during seizures and

proposed a model of positive feedback loop to show that the increase in [K+]o reinforced

excitation of the cells and resulted in self-sustaining seizures. Termination of seizures

occurred when depolarization induced by K+ inactivated the Na

+ currents and the cells

Page 17: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

12

go into “cathodal block”. In the hippocampus, the positive feedback mechanism results

in an accumulation of [K+]o in the CA1 region. This prevents the efflux of K

+ from the cell

making the pyramidal cells excitable and vulnerable to the subsequent interictal burst

which results in a seizure (Traynelis and Dingledine 1988). These results established that

sustained seizure activity leads to the accumulation of [K+]o to 8-12mM and these levels

of [K+]o were sufficient to induce seizures in isolated brain slice preparations. In in vitro

experiments of hippocampal slices increasing bath concentrations of K+ from 3.5mM

(normal) to 7-8mM resulted in spontaneous bursting activity in pyramidal cells (Rutecki

1985, Korn et al 1987, Traynelis and Dingledne 1988). Raising the [K+]o from 7-10mM led

to increase in burst frequency and intensity, and at 12mM [K+]o the bursts become

irregular and less intense (Korn et al 1987). Increasing [K+]o also depolarizes presynaptic

terminals opening voltage-gated Ca2+

channels causing erroneous action potentials to

fire and the unnecessary release of neurotransmitters (Somjen 2004). This forces the

uptake of Cl- into the cell which shifts the reversal potential of inhibitory post-synaptic

potential (EIPSP) positively thus converting the inhibitory synaptic potentials to excitatory

(Chamberlin and Dingledine 1988, Korn et al 1987).

ii. Low Mg2+

Though the role of Mg2+

during seizures is not yet clear, lowering [Mg2+

]o has been

shown to induce interictal discharges and ictal events. Spontaneous interictal bursting in

the CA3 region of the rat hippocampus evolves after 6-21min in Mg2+

-free ACSF.

Following this initial phase ictal-like events formed which began 7-24min after Mg2+

-free

ACSF application (Anderson et al 1986). Epileptiform discharges were observed in

human epileptic neocortex 1.5-2hrs after perfusion of Mg2+

-free ACSF (Avoli et al 1991),

and lowering [Mg2+

]o resulted in epileptiform activity that initiated in the CA3 region and

spread to CA1 in the rat hippocampus (Mody et al 1987). The mechanism underlying the

epileptiform activity under these conditions include removal of Mg2+

block of the N-

methyl-D-aspartate (NMDA) receptors predisposing the cell to prolonged excitation

(Dingledine et al 1999) and also causing increased neurotransmitter release at pre-

Page 18: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

13

synaptic boutons (Somjen 2004). Low Mg2+

also eliminates surface charge screening

thus increasing the likelihood of voltage-gated channel activation and excitation on the

cell (Mody et al 1987, Somjen 2004).

iii. Kainic Acid

Kainic acid, isolated from a Japanese seaweed Digenea simplex in 1953, is now used as a

selective agonist at kainate receptors and is a potent neural excitant. Intracerebral

injections of kainic acid have generated epileptiform seizures in the CA3 region of the

rat hippocampus. Seizures propagated to other limbic structures and tissue

histopathology showed neuronal cell loss in the hippocampus as well as other limbic

structures. These are patterns consistent with those seen in human patients with

temporal lobe epilepsy (Ben-Ari 1985).

The highest level of binding sites of kainate is in the stratum lucidum in the CA3 region

of the hippocampus where mossy fibers from the granule cells of the dentate gyrus

synapse onto the somata of CA3 pyramidal cells. Here kainate binds to receptors on CA3

pyramidal cells that have the GluR6 subunit. Since CA3 pyramidal cells are connected to

each other via an extensive network of excitatory collateral synapses, this CA3 region

can serve as a pacemaker for generation of synchronized activity. Injecting kainic acid

intracerebrally (Ben-Ari 2000) at concentrations small enough to not diffuse to the

hippocampus and in structures that are distal from the hippocampus, the CA3 pyramidal

cells are damaged, and through their extensive collateral connections the pyramidal

cells generate synchronized excitatory currents that propagate to the CA1 region and to

other limbic structures.

In the CA1 region inhibitory interneurons expressing kainate receptors with the GluR5

subunit are specifically activated by kainate. Low concentrations of kainate produces

long lasting depolarization and sustained action potentials in the inhibitory interneurons

in the CA1 region. As a result of this increased interneuron activity spontaneous

inhibition is increased in the CA1 pyramidal cells thus reducing seizure propagation and

generation of synchronized activity.

Page 19: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

14

Hence, within the hippocampus kainate acts as a “double agent” by enhancing the

activity of both the inhibitory interneurons and the excitatory pyramidal cells via

different receptor subtypes (Ben-Ari and Cossart 2000).

iv. 4-Aminopyridine

4-Aminopyridine (4-AP) was found to increase the release of neurotransmitter in

cholinergic synapses at the neuromuscular junction and also at noradrenergic synapses

in various tissues including cat spleen, vas deferens of rabbit and the ventral horn of rat

spinal cord (Thesleff 1980). At these synapses they found the effect of 4-AP to be pre-

synaptic. 4-AP increased neurotransmitter release during evoked action potentials by

blocking K+ channels pre-synaptically hence increasing Ca

2+ influx. Blocking the K

+

channel prolonged the action potential duration and left the voltage sensitive Ca2+

channels open longer. In 1982 Galvann et al described the convulsant actions of 4-AP in

slice preparations of guinea pig olfactory cortex. Bath application of 4-AP led to an

increase in spontaneous neurotransmitter release, intracellular paroxysmal discharges

and loss of inhibitory post-synaptic potentials (IPSPs) just prior to the onset of these

paroxysmal discharges. The IPSPs then reappeared during the “interictal” event. Rutecki

et al (1987) studied the effect of 4-AP on CA3 pyramidal neurons in rats and showed

that 4-AP induces epileptiform activity by enhancing both excitation and inhibition. They

stated that “epileptiform activity can occur without impairment of synaptic inhibition”.

Perrault and Avoli first studied the effects of 4-AP on pyramidal cells in the CA1 region of

the rat hippocampus (1989). They found low levels of 4-AP resulted in an increase in

stratum radiatum-induced excitatory post-synaptic potentials (EPSPs) and both early

and late IPSPs evoked by orthodromic stimuli increased in amplitude and duration. 4-AP

blocks the transient, rapidly inactivating A-type K+ currents by intracellularly binding to

the voltage-gated K+ channel (Kv channel). In the CA1 hippocampal neurons the A-type

currents are important in dendritic signal processing, synaptic integration and in LTP.

Thus this type of current plays an important role in modulating neuronal excitability.

Page 20: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

15

4-AP specifically interacts only with channels that express the Kv1.4, Kv4.1, Kv4.2 and

Kv4.3 α-subunits that form the pore of the channel (Kim et al 2005). Kv4.2 subunits are

highly expressed in the somatodendritic membrane of the CA1 cells and correspond to

the A-type current recorded in these cells, whereas Kv4.3 are more prevalent in the

interneurons in the hippocampus. Kim et al (2005) expressed an EGFP-tagged Kv4.2 in

the CA1 pyramidal cells in an organotypic slice culture and on comparing it with the

dominant negative mutant of Kv4.2 they found that overexpression of Kv4.2 led to an

increase in the A-type current with narrower action potentials and less frequency-

dependent broadening of the action potentials, thus reducing the excitability of the cell.

The expression of the dominant negative mutant displayed broader action potentials

with an increase in frequency-dependent broadening of the action potential and

enhanced action potential back-propagation. Thus confirming previous molecular

localization studies, they showed that Kv4.2 channels specifically contribute to the A-

type current in the CA1 pyramidal neurons.

By using 4-AP to block the Kv channel in the hippocampus the efflux of K+ ions is

reduced resulting in the prolongation of action potential like that seen in the dominant

negative mutant of Kv4.2 mentioned above. 4-AP also leads to an increase in

neurotransmitter release by enhanced Ca2+

influx into the pre-synaptic terminal via

voltage-sensitive Ca2+

channels or due to the reduced K+

repolarizing conductance

(Perrault and Avoli 1989).

The above mentioned epilepsy models are all reliable in their results and give us

patterns of seizure activity that are observed in human patients. The overall significance

of these models is that they allow us to make inferences about seizure dynamics which

can then be applied to study intact brains. The 4-AP model of epilepsy is a suitable

model because it has reliably shown to generate epileptiform activity in in vitro and in

vivo experiments (Perrault and Avoli 1991) and also as a convulsant in clinical

experiments (Thesleff 1980). Some drugs induce epileptiform discharges by decreasing

inhibition, whereas in the 4-AP model both inhibition and excitation are enhanced.

Page 21: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

16

Our experiments specifically studied two inhibitory interneuron sub-types in the CA1

region during seizure-like events, and hence it was critical for our purposes to have an

intact inhibitory network to reflect physiological processes in the hippocampus.

Page 22: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

17

Research design and methods

Animals

Experiments were performed using male Sprague-Dawley rats (P21-P30).

Hippocampal slice preparation

Animals were anesthetized with diethyl-ether, decapitated and brains extracted into

cold dissection buffer aerated with 95% O2-5%CO2 (in mM: 2.6 KCl, 1.23 NaH2PO4,

26mM NaHCO3, 10 MgCl2, 0.5 CaCl2, 213 sucrose, 20 glucose). The hippocampi were

isolated and transverse slices of 450µm thickness were made using a LEICA vibratome.

Sections were incubated for one hour in artificial cerebrospinal fluid (ACSF) (pH=7.4,

30°C, in mM: 130 NaCl, 1.25 NaH2PO4, 26 NaHCO3, 3.5 KCl, 10 glucose, 1.2 MgSO4, 2

CaCl2) aerated with 95% O2-5%CO2. For recordings slices were transferred to a

submersion recording chamber and perfused with recording ACSF containing (in mM):

130 NaCl, 1.25 NaH2PO4, 26 NaHCO3, 3.5 KCl, 10 glucose, 0.6 MgSO4, 2 CaCl2 and aerated

with 95% O2-5%CO2.

Electrophysiology

Slices were viewed with a fixed-staged Olympus (BX51WI) microscope with a water

immersion objective using differential infrared contrast imaging. Cells to be patched

were identified based on morphological characteristics and location of the cell body

within the hippocampal layers. Putative OLM cells were identified based on the location

of the cell body in stratum oriens and their horizontal somatodendritic orientation.

Putative basket cells were identified based on their location of soma in stratum

pyramidale and the vertical bipolar somatodendritic orientation. Pyramidal cells were

identified based on the location of the cell body in stratum pyramidale, triangular-

shaped soma, and apical and basilar dendritic branching pattern.

Page 23: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

18

Whole cell recordings were made using borosilicate glass capillaries (World Precision

Instruments) pulled to obtain resistance of 4-8MΩ, containing (in mM): 116 K gluconate,

6 KCl, 20 HEPES, 0.5 EGTA, 2 NaCl, 10 sodium creatine, 4 MgATP, 0.3 NaGTP and 0.3%

Neurobiotin (pH 7.26, 290 mOsm). Extracellular recordings were made with the same

borosilicate glass capillaries but with 1-3MΩ resistance and containing 0.9% NaCl. Using

micromanipulators (Burleigh PCS-6000) simultaneous dual and triple whole cell and

extracellular recordings were made in the CA1 region of the rat hippocampus (Figure 1).

Figure 1. Diagram of location of electrodes. Extracelullar (black) and whole cell recordings (red) of oriens (left),

basket (middle) and pyramidal (right) cells made in the CA1 region of the rat hippocampus (figure adapted from

Andersen et al. 2007).

After a whole cell patch and holding the cell at -75 to -80mV, negative and positive

square wave current pulses were injected in increments of 50-100pA for 500msec to

determine the membrane properties of the cells and further confirm the identity of the

cell-type (Figure 2) (Ali et al 1998; Buhl et al 1996; Elfant et al 2008; Fujiwara-Tsukamoto

et al 2004). Connectivity between cells in dual and triple patch conditions was

determined by injecting a pulse train current (rate = 25 Hz, pulse width = 4 ms,

amplitude = 800 pA-1200pA) into one of two patched cells and the response was

recorded from the other. However, none of the cell pairs that exhibited SLEs were found

Page 24: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

19

to be connected. SLEs were induced using 4-AP (200µm) that has shown to induce

epileptiform discharges in the hippocampus (Perreault and Avoli 1991; Perreault and

Avoli 1992). Data were acquired using Multiclamp 700B amplifiers and pClamp10

acquisition software (Molecular Devices), high pass filtered at 4 kHz (whole cell) and 1

kHz (extracellular) and digitized at 10 kHz (Digidata 1440A, Molecular Devices).

Page 25: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

20

Figure 2. Electrical membrane properties of patched cells. Examples of membrane properties of pyramidal cells (a),

basket cells (b) and oriens interneuron cells (c) after 500ms negative and positive current injections.

a.

c.

b.

Page 26: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

21

Immunohistochemistry and confocal microscopy

Post-hoc immunohistochemistry analysis further validated the identity of the patched

cells. On completion of recordings, slices were fixed and stored in 4% paraformaldehyde

in 0.1M phosphate buffer for 48 hours. Slices were then transferred to 0.1M phosphate

buffer solution (PBS, pH 7.4).

Slices were incubated in 10% normal goat serum in 0.1M PBS with 0.3% TritonX-100 on

a shaker for 90 minutes at room temperature then rinsed in PBS, following which they

were incubated in primary antibodies for 72 hours on shaker in 4°C. Primary antibodies

were used in the following concentrations: mouse monoclonal anti-parvalbumin

1:1000(Sigma P-3088), and rabbit anti-cholecystokinin 1:1000 (Sigma C-2581).

Slices were rinsed in PBS and incubated in secondary antibodies overnight on a shaker in

4°C. Biocytin filled cells were visualized using TRITC conjugated with avidin (Sigma A-

7169). Primary antibodies were visualized using Alexa Fluor 488 (1:500, Invitrogen A-

21121), and Alexa Fluor 647 (1:500, Invitrogen A-31573) (adapted from Losonczy et al

2002). Slices were rinsed in PBS, mounted on glass slides and coverslipped. Stained cells

were identified for dual or triple immunofluorescence using an Olympus FluoView

FV1000 laser scanning confocal microscope (Figure 3).

Page 27: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

22

a. b.

c.

Figure 3. Confocal images. Triple immunofluorescence validated the identity of the cells patched during recordings.

Cells were filled with neurobiotin (red) during the recordings and were stained for parvalbumin (PV, green) and

choleycystokinin (CCK, blue). All the cells that were impaled stain positive for neurobiotin. Pyramidal cells and oriens

cells stain negative for PV and CCK. Basket cells containing endogenous parvalbumin stain green, and blue if they

contain choleycystokinin.

Page 28: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

23

Triple immunofluorescence further validated the identity of the cells that were patched

based on morphological characteristics (Table 1). SLEs from only those cell pairs whose

identities were confirmed both morphologically and by using triple immunofluorescence

were included in the correlation analysis. Hence all cell pairs in Table 1 were first

identified based on their morphology (column 1) as seen under IR-DIC before patching

attempts were made. From this pool of cell pairs only some pairs exhibited SLEs (column

2). From the original group of cells (in column 1), we further validated the identity of the

patched cells using triple immunofluorescence (column 3). In doing so, some cell pairs

were found to be wrongly identified. Hence, the number of cell pairs identified as P-P,

which had been initially identified as B-P, increased (column 3). Column 4 represents the

cell pairs that were analyzed for SLEs; this includes cell pairs which exhibited SLEs and

whose identities were confirmed both morphologically and using triple

immunofluorescence. Columns 5 and 6 show the total number of SLEs included in the

single seizure and resample analysis from the cell pairs in column 4. 9 SLEs were

mistakenly omitted for the single seizure analysis but were included in the resample

analysis, hence there are 8 more ISIs in the resample analysis.

1. 2. 3. 4. 5. 6.

# of pairs

morphologically

identified

# of pairs

exhibiting

SLEs

# of pairs

after identity

confirmed

# of pairs

analyzed for

SLEs

# of SLEs

Single

event

analysis

# of SLEs

Resample

analysis

B-P 40 13 7 5 70 46

P-O 8 6 5 5 99 107

B-O 7 7 5 1 5 3

P-P 4 4 10 10 76 55

B-B 2 1 2 2 18 15

Total 61 31 29 23 268 226

Table 1. Number of cell pairs patched and analyzed for SLEs. B, Basket cell; P, Pyramidal cell; O, Oriens cell; SLE,

seizure-like events; ISI, inter-seizure interval.

Page 29: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

24

Correlation Analysis

Using subthreshold membrane potentials and spike times of pairs of neurons over

discrete time windows, we tracked the progression of synchrony between the cell types

during seizure-like events (SLEs). The presence of synchronization was determined using

crosscorrelation analysis (Netoff and Schiff 2002).

A seizure-like event was defined as a period of sudden and extreme increase in the firing

activity of a cell with a simultaneous fast positive extracellular shift (FPES; Ziburkus et al,

2006) seen in the extracellular recording, reflecting a network-wide event. We first

analyzed the SLE as single seizures and then resampled the data to study the interval

between two seizures (inter-seizure interval, ISI).

Single SLE analysis - The start of each SLE was determined using the FPES (Figure 4a, red

arrow). Correlation analysis was performed over 1s overlapping windows extending

from 30 sec before to 70 sec after the FPES. This created 100s long files that included

the SLEs.

Resample analysis - To analyze the interval between two consecutive SLEs (inter-seizure

interval) correlation analysis was measured on 1s overlapping windows beginning from

the start of the first SLE (determined using FPES, Figure 4b red arrows) to the start of

the consecutive SLE. The two start times were then fixed and the interval in-between

was resampled in MATLAB (MATLAB R2007a Student version). Inter-seizure intervals

shorter than 50 sec and longer than 300 sec were excluded.

Page 30: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

25

a.

b.

Figure 4. Single seizure and resample analysis. Single seizure analysis (a) of oriens interneuron (top trace) and

pyramidal cell (middle trace). Resample analysis (b) of two pyramidal cells (top and middle trace). Start times for both

analysis were determined using the fast positive extracellular shift (FPES) seen in the extracellular recording (bottom

traces in (a) and (b)).

20mV

20mV

0.4mV

10sec

20 mV

0.2 mV

20 sec

20 mV

Page 31: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

26

In addition to performing a standard crosscorrelation analysis, we also prepared the

voltage traces in two ways in an attempt to investigate the input and output signals

separately. Output correlations were investigated by measuring the correlations

between the spike rates of the two neurons. Input correlations were investigated by

measuring the correlation between the membrane potentials after removing all action

potentials.

To identify spikes, a threshold was used to determine spike times. The spike time data

was transformed into spike rate data by binning over 100 msec windows. This course

grained spike rate data was then used in our spike correlations. Subthreshold analysis

uses data were the entire action potential is removed from the voltage trace. To

determine spike initiation and spike termination the slope of the membrane potential

was measured over 0.1ms. The slope threshold of spike initiation was set between 20

and 25mV/ms to best detect spikes and the slope of spike termination was set to zero,

to confirm the end of this spike before the start of the subsequent spike.

Subthreshold and total crosscorrelation analysis was measured for each neuron i and j

over overlapping 1s windows using the equation:

/2

/2, 1/2 1/2

/2 /22 2

/2 /2

( ) ( )

( )

( ( )) ( ( ))

T

i j

t T

i jt T t T

i j

t T t T

x t x t

c

x t x t

ττ =−

= =

=− =−

+

=

∑ ∑

Here the voltage ( )x t for neuron ior neuron j has had its mean removed and τ is the

time lag. The Bartlett estimator of cross-correlation standard error ,i jσ (Bartlett, 1946):

Page 32: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

27

/2

, ,2 /2,

( ) ( )

( )( 1 )

T

i i j j

Ti j

c c

T

τ

τ τσ τ

τ=−=

+ −

where , ( )i ic τ and

, ( )j jc τ are the crosscorrelation and autocorrelation functions of each

signal and . indicates absolute value. The sum, S , of the cross-correlation values

greater than two times the standard error was calculated to determine the correlation

value for each window:

/ 2

, , ,

/ 2

( ) ( ( ) 2 ( ))T

i j i j i j

T

S c cτ

τ θ τ σ τ=−

= −∑

where θ ,the Heaviside function, takes on the following values: (1 if , ( ) 2 0i jc τ σ− > ,

and 0 otherwise).

To measure spike correlation for each pair of neurons iand j we binned time series of

spikes ( )s t by discretizing time into N intervals of 100ms. To account for spike

correlations we use the following point process correlation

/ 2

,

/ 2

1( ) ( ) ( )

T

i j i j

t T

nmp s t s t

nm Nτ τ

=−

= + −

(Brody, 1991). Here n and m are the average spike rates for neurons i and j across

the window. In order to avoid erroneous higher correlation values in the

crosscorrelation signal, we again used the Bartlett estimator to determine the

significance of , ( )i jp τ using:

Page 33: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

28

/ 2

, ,2 / 2,

( ) ( )

( )( 1 )

T

i i j j

Ti j

p p

T

τ

τ τσ τ

τ=−=

+ −

where , ( )i ip τ and

, ( )j jp τ are the correlation and autocorrelation functions of each

signal and . indicates absolute value.

The point process estimator was calculated in the same manner as the subthreshold

and total correlation, using the autocorrelation function

2/ 2

, 2/ 2

1( ) ( ) ( )

T

i i i i

t T

np s t s t

n Nτ τ

=−

= + −

where ,i ip is the autocorrelation function of neuron i.

For resample analysis the average length of the inter-seizure interval was determined.

Using the resample command in MATLAB (MATLAB R2007a Student version), all the

inter-seizure intervals were either stretched or contracted to fit the average.

Statistical Analysis

Averages of spike rates, spike, subthreshold and total cross-correlations were measured

and reported as the standard error of the mean. ANOVA was calculated to determine

significant differences between different times within a SLE and followed by Tukey

multicomparison tests (confidence limit 0.001p < ). Linear regression analysis was

performed (using GraphPad Prism version 5.04 (Trial) for Windows, GraphPad Software,

San Diego, California, USA, www.graphpad.com) for the spike rates during the ISI. For

linear regression analysis the start ISI was determined as the time when the spike rate of

the cell was at its minimum following seizure termination. The end of the ISI was

Page 34: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

29

determined to be the time just before the sudden increase in firing activity at seizure

onset.

Page 35: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

30

RESULTS

Previous findings (Ziburkus et al 2006) about the unique interleaving pattern of activity

between OLM and pyramidal cells are confirmed in the current experiments. OLM cell

firing reaches its peak just before the start of the SLE. At the start of the SLE the OLM

cells enter into depolarization block which is characterized by a significant drop in firing

activity. Severe depolarization inactivates the voltage-gated Na+ channels thus raising

the firing threshold and the OLM cells are rendered unexcitable. During this drop in

firing of the OLM cells, pyramidal cells display an increase in the firing activity at the

start of the SLE and the excitation comes to a stop when the OLM cells recover and

increase their firing again (Figure 5a).

Prior experiments also showed that spike correlation between pyramidal and OLM cells

showed a significant increase at the beginning and the end of SLEs (Figure 5b).

Subthreshold correlation between these cell types showed a similar pattern (Figure 5c).

Page 36: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

31

a.

b.

c.

Figure 5. Interleaving pattern of interplay between OLM and pyramidal cells. Inhibitory (blue) and excitatory

(orange) spike rates during the evolution of SLEs. Inset: average spike rates at expanded time scale (a). Cross

correlation averages for spike (b) and subthreshold (c) activity of pyramidal and OLM cells. Subthreshold correlation

between pyramidal and OLM cells increased at the start of the SLE (marked by inverted black arrow) and then again

towards the middle of the SLE (b). Spike correlation showed an increase at the start of the SLE (blue asterisk) and

after the DB in interneurons (green asterisk) (c). [Reproduced from Ziburkus et al. 2006]

Page 37: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

32

Current experiments extended the previous study of the OLM-pyramidal cell

interactions to include the parvalbumin positive basket cells (Figure 6).

Figure 6. Schematic diagram of cell-types in the CA1 region of the rat hippocampus investigated in our experiments.

The soma of these basket cells are located in the pyramidal layer, their dendrites span

the length of the stratum radiatum and also spread into the stratum oriens. Their axons

innervate the pyramidal cells perisomatically. Hence the microcircuit studied in the

current experiments involves two types of inhibitory networks that are complimentary

to each other in regard to their anatomical distribution and the axonal innervations onto

the pyramidal cells.

Schaffer collateral

inputs

Stratum lacunosum

moleculare

Stratum

radiatum

Stratum

pyramidale

Stratum

oriens

Long range

projections

Page 38: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

33

Given the differences between the two interneuron sub-types described earlier, we

hypothesized distinct patterns of interaction between pyramidal cells and each of these

interneurons.

We measured spike rates of all three cell types and also measured the total, spike and

subthreshold correlation, as described in detail in the Research Design and Methods

section. Spike rates were measured for individual cells and the cross-correlation was

measured between the different cell pairs to reveal the correlation in activity of the

pyramidal, basket and oriens cells in the different phases of the SLE.

Total correlation is a measure of the correlated activity between the cells. In order to

distinguish between input and output correlations, the subthreshold and spike

correlations are measured separately.

Subthreshold activity reflects the synaptic inputs into the cells and also the resulting

voltage-dependent subthreshold activation of the membrane potential. This analysis

measures the correlation in synaptic network inputs to the cells before, during and after

the SLE. The analysis allows us to observe any distinct patterns of correlated synaptic

input in the different stages of the SLE. Spike correlation analysis reflects the correlation

in neuronal output. We measured the spike cross correlation between the three cell-

types to observe any distinct patterns of correlated firing activity of the cells during the

different stages of the SLE.

As stated in the Research Design and Methods section, the start of each seizure was

determined using the fast positive extracellular shift (FPES). Correlation analysis for

single seizures was performed 30 seconds before to 70 seconds after the FPES.

Resampling analysis was performed over the inter-seizure interval beginning from the

start of one seizure to the start of the subsequent seizure. Also, inter-seizure intervals

shorter than 50 seconds and longer than 300 seconds were excluded. Hence in some

cases the number of SLEs is less in the resample analysis than the single seizure analysis.

Page 39: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

34

Since resample analysis was conducted on the interval between pairs of seizures, in

most cases for all analysis the second seizure in a given interval will be the first seizure

of the subsequent interval.

In the associated figures (Figures 7 - 14) we will denote statistical differences with

asterisks/circle/square, or when there are many points we will use a solid bar. The bar

indicates that there are a number of points that are statistically separated from the

point marked with an asterisk/circle/square. However it does not necessarily mean that

all points within the region denoted by the bar are statistically different from the point

marked with an asterisk/circle/square.

1. Spike Rate

Single seizure analysis of spike rates (Figure 7) for pyramidal (green trace, n=245),

basket (red trace, n=93) and oriens interneurons (black trace, n=104) shows a

statistically significant increase in firing rate at seizure onset (30s) for pyramidal (●) and

basket (○) cells and just before seizure iniZaZon for oriens interneurons (*) compared

to pre- and post-seizure (0-30s, 50-100s) intervals.

Resample analysis of the spike rates (Figure 8) for pyramidal (green trace, n=208),

basket (red trace, n=64) and oriens interneurons (black trace, n=110) shows the same

statistically significant increase (*-oriens, ● - pyramidal, ○- basket cells) in firing activity

that is seen in the single seizure analysis. (The start of the first seizure in the resampled

analysis is at 100 seconds which corresponds to the start of the seizure in the single

seizure analysis at 30 seconds, and the start of the second seizure is at 287 seconds).

Linear regression analysis of the spike rates during the ISI shows a statistically significant

increase in firing activity for all three cell types (Figure 9; basket cells (red trace, n= 64)

slope= 0.02175 ± 0.001280, r2= 0.9507, p < 0.0001; pyramidal cells (green trace, n= 208)

Page 40: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

35

slope= 0.004037 ± 0.0007630, r2= 0.6363, p < 0.0001; oriens interneurons (black trace,

n= 110) slope= 0.02379 ± 0.002449, r2= 0.8252, p < 0.0001).

Page 41: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

36

Figure 7. Single seizure spike rate. Single seizure analysis of the spike rates of pyramidal (green trace, n=245), basket

(red trace, n=93 ) and oriens interneurons (black trace, n=104 ). Oriens interneurons (black trace) show a statistically

significant increase (* in inset) in spike rates just before seizure onset (30s) compared to pre- and post-seizure

periods. Pyramidal (green trace) and basket cells (red trace) show a statistically significant increase (● for pyramidal

cells, ○ for basket cells in insets) in spike rates at seizure onset (30s) compared to pre- and post-seizure periods. Errors

bars=standard error of the mean. [ANOVA, Tukey multi-comparison test, p < 0.001).

0 10 20 30 40 50 60 70 80 90

5

10

15

20

25

30

35

40

45

50

Time (s)

Fre

qu

en

cy

(H

z)

0 10 20 30 40 50 6 0 70 80 90 10 0

10

20

30

40

50

T im e(s )

Fre

qu

en

cy(H

z)

* * *

0 10 20 30 40 50 60 70 80 90 100

2

4

6

8

10

12

14

Time(s)

Fre

qu

en

cy

(Hz)

● ● ●

0 10 20 30 40 50 60 70 80 90 100

5

10

15

20

25

Tim e(s )

Fre

qu

en

cy

(Hz)

○ ○ ○

Page 42: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

37

Figure 8. Inter-seizure interval spike rate. Resampled analysis of the spike rates of pyramidal (green trace, n=208),

basket (red trace, n= 64) and oriens interneurons (black trace, n=110). Oriens interneurons (black trace) show a

statistically significant increase (* in inset) in spike rates just before seizure onset (100s) compared to pre- and post-

seizure periods. Pyramidal (green trace) and basket cells (red trace) show a statistically significant increase (● for

pyramidal cells, ○ for basket cells in insets) in spike rates at seizure onset (100s) compared to pre- and post-seizure

periods. Errors bars=standard error of the mean. [ANOVA, Tukey multi-comparison test, p < 0.001).

0 50 100 150 200 250 300 350

5

10

15

20

25

30

35

40

Time (s)

Fre

qu

en

cy (

Hz)

0 50 100 150 200 250 300 350 400

5

10

15

20

25

30

35

40

Time(s)

Fre

qu

en

cy

(Hz)

* * *

0 50 100 150 200 250 300 350 400

2

4

6

8

10

12

Time(s)

Fre

qu

en

cy

(Hz)

○ ○

0 50 100 150 200 250 300 350 400

2

4

6

8

10

Time(s)

Fre

qu

en

cy

(Hz)

● ●

Page 43: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

38

Basket cells

Time (s)

Fre

qu

en

cy (

Hz)

0 50 100 150 2000

1

2

3

4

Oriens interneurons

Time (s)

Fre

qu

en

cy (

Hz)

0 50 100 150 2006

7

8

9

10

11

Pyramidal cells

Time (s)

Fre

qu

en

cy (

Hz)

0 50 100 150 2001.4

1.6

1.8

2.0

2.2

Figure 9. Linear regression analysis of inter-seizure interval. The spike rates during the ISI of basket (n=64), oriens

(n=110) and pyramidal (n=208) cells show a statistically significant increase during the ISI. The graphs in the left

column correspond to the traces in the right column. The ISI intervals used for linear regression analysis of each cell

were as follows (right column): basket cells= 70-150s, oriens interneueons= 45-150s, pyramidal cells= 65-150s. Basket

cells (top row, red trace) slope= 0.02175 ± 0.001280, r2= 0.9507, p < 0.0001; Oriens interneurons (middle row, black

trace) slope= 0.02379 ± 0.002449, r2= 0.8252, p < 0.0001; Pyramidal cells (bottom row, green trace) slope= 0.004037 ±

0.0007630, r2= 0.6363, p < 0.0001. (Start times of seizures in the graphs and traces above are at 0 and 187 seconds,

corresponding to seizure start times of 100 and 287 seconds, respectively, in Figure 8).

0 20 40 60 80 100 120 140 160 180 200

2

4

6

8

10

12

Time(s)

Fre

qu

en

cy

(Hz)

0 20 40 60 80 100 120 140 160 180 200

10

20

30

40

Time(s)

Fre

qu

en

cy

(Hz)

0 20 40 60 80 100 120 140 160 180 200

2

4

6

8

10

Time(s)

Fre

qu

en

cy

(Hz)

Page 44: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

39

2. Correlation Analysis

Pyramidal cell-Pyramidal cell (PP)

Total correlation

In both single seizure (n=76) (Figure 10a) and resample analysis (n=55) (Figure 10b) the

start of the seizure shows a significant decrease in total correlation (37s, asterisk)

compared to the pre-, post- (0-30s, 50-95s; bars with asterisks) and inter-seizure interval

(125-225s; bar with asterisk).

Subthreshold correlation

Similar to the total correlation, in both single seizure (n=76) (Figure 10c) and resampled

(n=55) (Figure 10d) analysis, the start of the seizure is characterized by a statistically

significant decrease in correlation (37s, asterisk) compared to the pre-, post- (0-30s, 50-

95s; bars with asterisks) and inter-seizure interval (125-225s; bar with asterisk).

Spike correlation

In the single seizure analysis (n=76) (Figure 10e) the start of the seizure shows a

statistically significant increase (30s asterisk) compared to the pre- (0s, asterisk) and

post- (60-100s) seizure levels. The resampled analysis (n=55) (Figure 10f) reflects this

same significant increase in spike correlation.

Page 45: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

40

a.

b.

c.

d.

e. f.

Figure 10. Correlation analysis of pyramidal-pyramidal cell pairs. Total (a, b), Subthreshold (c, d) and Spike

correlation (e, f) measured for single seizure events (left column, n=76) and resampled inter-seizure intervals (right

column, n=55). Error bars=standard error of the mean. [ANOVA, Tukey multi-comparison test, p < 0.001].

0 10 20 30 40 50 60 70 80 90 100

0.007

0.009

0.011

0.013

0.015

Time (s)

A.U

.

* * *

0 10 20 30 40 50 60 70 80 90 100

0.01

0.014

0.018

0.022

0.026

Time (s)

A.U

.

* * *

0 50 100 150 200 250 300 350 400

0.01

0.015

0.02

0.025

0.03

Time (s)

A.U

.

* *

0 10 20 30 40 50 60 70 80 90 100

0.002

0.003

0.004

0.005

Time (s)

A.U

. *

* *

0 50 100 150 200 250 300 350 400

0.006

0.008

0.01

0.012

0.014

0.016

0.018

0.02

Time (s)

A.U

.

* *

0 50 100 150 200 250 300 350 400

.001

.0015

.002

.0025

.003

.0035

Time (s)

A.U

.

* *

*

Page 46: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

41

Basket cell-Basket cell (BB)

Total correlation

Using our present conservative approach to significance testing both single seizure

analysis (n=18) (Figure 11a) and resample analysis (n=15) (Figure 11b) show no

statistically significant separations between the different phases of the SLE. However,

certain trends do emerge; the total correlation between basket-basket cell pairs

decreases at the start of the seizure event (37s) and remains low during the body of the

seizure. As the seizure terminates the correlation returns to pre-ictal level. The

correlation remains at this level during the subsequent inter-seizure interval (70-160s).

Subthreshold correlation

As in the total correlation, the subthreshold correlation is devoid of any statistically

significant separations between the phases of the SLE in both the single seizure analysis

(n=18) (Figure 11c) and resample analysis (n=15) (Figure 11d). The overall trend is

similar to that seen in the total correlation.

Spike correlation

Single seizure analysis (n=18) (Figure 11e) shows an increase in correlation that begins

at the start of the seizure, reaching a statistically significant value during the body of the

seizure (40s, asterisk) compared to pre- (0-30s; bar with asterisk) and post-seizure (60-

100s) levels. The resampled data (n=15) (Figure 11f) reflects this same statistically

significant increase in correlation during the body of the seizure (115s, asterisk)

compared to the inter-seizure interval (150-250s; bar with asterisk).

Page 47: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

42

a.

b.

c.

d.

e. f.

Figure 11. Correlation analysis of basket-basket cell pairs. Total (a, b), Subthreshold (c, d) and Spike correlation (e, f)

measured for single seizure events (left column, n=18) and resampled inter-seizure intervals (right column, n=15).

Error bars=standard error of the mean. [ANOVA, Tukey multi-comparison test, p < 0.001].

0 10 20 30 40 50 60 70 80 90 100

0.005

0.007

0.009

0.011

0.013

Time (s)

A.U

.

0 50 100 150 200 250 300 350 400

0.005

0.008

0.011

0.014

Time (s)

A.U

.

0 10 20 30 40 50 60 70 80 90 100

0.008

0.012

0.016

0.02

Time (s)

A.U

.

0 50 100 150 200 250 300 350 400

0.006

0.008

0.01

0.012

0.014

0.016

0.018

0.02

Time (s)

A.U

.

0 10 20 30 40 50 60 70 80 90 100

0.001

0.002

0.003

0.004

Time (s)

A.U

.

* * *

0 50 100 150 200 250 300 350 400

.001

.002

.003

.004

Time (s)

A.U

.

* *

Page 48: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

43

Basket cell-Pyramidal cell (BP)

Total correlation

In the single seizure analysis (n=70) (Figure 12a) the start of the seizure ( 37s, asterisk) is

characterized by a statistically significant decrease in correlation compared to both pre-

(0-30s; bar with asterisk) and post-seizure levels (50-60s, 80-90s; bars with asterisks)

The same statistically significant decrease in correlation (100s, asterisk) compared to

inter-seizure interval (150s-275s; bar with asterisk) is seen in the resampled data (n=46)

(Figure 12b).

Subthreshold correlation

Both single seizure (n=70) (Figure 12c) and resample (n=46) (Figure 12d) analysis of the

subthreshold correlation show a decrease in correlation at the start of the seizure (37s,

100s; asterisk) which is significantly different from the pre-, post- (Figure 12c, 0-30s, 80-

95s respectively; bar with asterisk) and inter-seizure (Figure 12d, 150-275s) intervals.

Spike correlation

Both single seizure (n=70) (Figure 12e) and resample analysis (n=46) (Figure 12f) show a

statistically significant increase in correlation at the start of the seizure (37s, asterisk)

followed by a statistically significant decrease during the body of the seizure (45s,

asterisk).

Page 49: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

44

a.

b.

c.

d.

e.

f. Figure 12. Correlation analysis of basket-pyramidal cell pairs. Total (a, b), Subthreshold (c, d) and Spike correlation

(e, f) measured for single seizure events (left column, n=70) and resampled inter-seizure intervals (right column,

n=46). Error bars=standard error of the mean. [ANOVA, Tukey multi-comparison test, p < 0.001].

0 10 20 30 40 50 60 70 80 90 100

0.008

0.012

0.016

0.02

Time (s)

A.U

.

* * * *

0 50 100 150 200 250 300 350 400

0.01

0.02

0.03

Time (s)

A.U

.

* *

0 10 20 30 40 50 60 70 80 90 100

0.008

0.012

0.016

0.02

Time (s)

A.U

.

* * *

0 50 100 150 200 250 300 350 400

0.01

0.015

0.02

0.025

0.03

Time (s)

A.U

.

* *

0 10 20 30 40 50 60 70 80 90 100

0.002

0.003

0.004

Time (s)

A.U

.

*

*

0 50 100 150 200 250 300 350 400

.001

.0015

.002

.0025

.003

.0035

Time (s)

A.U

.

*

*

Page 50: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

45

Oriens interneuron-Basket cell (OB)

Total correlation

Single seizure analysis (n=5) (Figure 13a) shows no significant separations between the

different phases of the SLE. However, the resample analysis (n=3) (Figure 13b) shows a

statistically significant increase in correlation during the body of the seizure (Figure 13b,

110s, asterisk) compared to the inter-seizure interval (Figure 13b, 150-275s, bar with

asterisk).

Subthreshold correlation

Single seizure analysis (n=5) (Figure 13c) does not show significant separations between

the phases of the SLE, however resampled analysis (n=3) (Figure 13d) shows a

statistically significant increase in correlation during the body of the seizure (120s,

asterisk) compared to the inter-seizure interval (200-275s, bar with asterisk).

Spike correlation

Both single seizure (n=5) (Figure 13e) and resample (n=3) (Figure 13f) analysis show no

statistically significant separations between the phases of the SLE. However a trend

does emerge; correlation begins to decrease at the start of the seizure and is at its

lowest value during the body of the seizure when the oriens interneurons do not fire for

a while. The correlation recovers to pre-ictal values then decreases again as the seizure

terminates.

Page 51: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

46

a.

b.

c.

d.

e.

f.

Figure 13. Correlation analysis of oriens interneuron-basket cell pairs. Total (a, b), Subthreshold (c, d)

and Spike correlation (e, f) measured for single seizure events (left column, n=5) and resampled inter-

seizure intervals (right column, n=3). Error bars=standard error of the mean. [ANOVA, Tukey multi-comparison

test, p < 0.001].

0 10 20 30 40 50 60 70 80 90 1000

0.01

0.02

0.03

Time (s)

A.U

.

0 10 20 30 40 50 60 70 80 90 100

0.01

0.02

0.03

Time (s)

A.U

.

0 10 20 30 40 50 60 70 80 90 100

0.001

0.003

0.005

Time (s)

A.U

.

0 50 100 150 200 250 300 350 400

.001

.003

.005

.007

Time (s)

A.U

.

0 50 100 150 200 250 300 350 400

0.01

0.02

0.03

0.04

Time (s)

A.U

.

* *

0 50 100 150 200 250 300 350 400

0.01

0.02

0.03

0.04

Time (s)

A.U

.

* *

Page 52: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

47

Oriens interneuron-Pyramidal cell (OP)

Total correlation

For the single seizure analysis (n=99) (Figure 14a) the pre-ictal period shows a

statistically significant decrease in correlation (25s, asterisk) compared to the start (30s,

asterisk) and the body (50s, asterisk) of the seizure. The start of the seizure shows a

significant increase in correlation (30s, square) compared to the pre-ictal levels (25s,

square). The body of the seizure shows yet another increase (50s, circle) which is

significantly different from the pre- (0-30s, bar with circle) and post-seizure (65s, circle)

levels.

The resampled data (n=107) (Figure 14b) shows a significant increase in correlation at

seizure onset (100s, square) compared to the body (110s, square) and the inter-seizure

interval (175-250s, bar with square). This was not seen in the single seizure analysis due

to the coarseness of the binning. Also the correlation during the body of the seizure at

140s (circle) is significantly increased compared to the inter-seizure interval (175-250s,

bar with circle).

Subthreshold correlation

Single seizure analysis (n=99) (Figure 14c) shows statistically significant increase in

correlation just before seizure onset (30s, asterisk) compared to pre- (0-30s, asterisk)

and post-seizure (60s, asterisk) values. A second significant increase is seen during the

body of the seizure (50s, circle) compared to pre-seizure interval (0-30s, circle).

The resampled analysis (n=107) (Figure 14d) reveals a statistically significant increase in

correlation just before seizure onset (100s, asterisk) compared to the body of the

seizure and the inter-seizure interval (110-260s, bar with asterisk). As in the total

correlation, this peak was not observed in the single seizure analysis due to the

coarseness of the binning. A second increase during the body of the seizure at 145s

(circle) is significantly different from the inter-seizure interval (160-260s, bar with circle).

Page 53: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

48

Spike correlation

The start of the seizure in the single seizure analysis (n=99) (Figure 14e) shows a

significant decrease in spike correlation (37s, asterisk) compared to the pre- (0-30s, bar

with asterisk) and post-seizure (60-110s, bar with asterisk) intervals. The resampled

analysis (n=107) (Figure 14f) shows a statistically significant increase in correlation just

before seizure onset (90s, asterisk) compared to the start of the seizure (100s, asterisk)

and the inter-seizure interval (150-200s, bar with asterisk). A second increase in

correlation during the body of the seizure (125s, circle) is also statistically significant

compared to the decrease at the start of the seizure (100s, circle) and the inter-seizure

interval (150-200s, bar with circle).

Page 54: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

49

a.

b.

c.

d.

e.

f. Figure 14. Correlation analysis of oriens-pyramidal cell pairs. Total (a, b), Subthreshold (c, d) and Spike correlation (e,

f) measured for single seizure events (left column, n=99) and resampled inter-seizure intervals (right column, n=107).

Error bars=standard error of the mean. (p < 0.001 for Total and Subthreshold correlation, p = 0 for Spike correlation

since the oriens interneurons stop firing during the body of the seizure). (Each symbol represents significant

differences between points in the SLEs with the same symbol). [ANOVA, Tukey multi-comparison test].

0 10 20 30 40 50 60 70 80 90 100

0.003

0.004

0.005

0.006

0.007

Time (s)

A.U

.

*

*

*▪

▪ ○

0 50 100 150 200 250 300 350 400

0.004

0.006

0.008

0.01

Time (s)

A.U

.

▪○

0 10 20 30 40 50 60 70 80 90 100

0.004

0.006

0.008

0.01

Time (s)

A.U

.

*

* *

0 50 100 150 200 250 300 350 400

0.003

0.005

0.007

0.009

Time (s)

A.U

.

* *

0 10 20 30 40 50 60 70 80 90 100

0.001

0.002

0.003

Time (s)

A

.U.

*

* *

0 50 100 150 200 250 300 350 4000

.001

.002

.003

Time (s)

A.U

.

* *

*○

Page 55: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

50

SUMMARIZED RESULTS

The description of the results of the total, subthreshold and spike correlation analysis

between the different cell pairs presented in the above Results section is now discussed

in detail. Each of the three cells (pyramidal, basket and oriens cells) is discussed

separately here.

Pyramidal cells

To understand the role of each cell type in seizure dynamics we will focus on the inputs

and outputs of each one separately. Pyramidal cells receive inputs from Schaffer

collaterals, basket cells, and oriens interneurons and are also innervated by other CA1

pyramidal cells.

The spike correlation of pyramidal cells (Figure 10f and Figure 15c, purple trace) steadily

increases until seizure onset though the firing frequency shows a moderate increase

(Figures 8 and 9; green trace). Pyramidal cell firing rates increase sharply at seizure

onset, at which time their correlation begin a sharp decline. Therefore the strength of

the excitatory output will be increased by higher firing rates, but will be tempered by

the loss of correlated signaling between these cells. The increase in spike correlation

and firing activity seen here before and leading up to the SLEs is similar to the biphasic

network dynamics observed by Cymerblit-Saba and Schiller (2010). They showed an

initial decrease in neuronal synchrony followed by increase in firing activity and

synchronization between neurons leading up to the seizure.

Basket cells provide perisomatic inhibition. Our measurements show that spike

correlations between basket cells (Figure 11e,f and Figure 15c, red trace) as well as their

spike rates (Figures 7 and 8; red trace) reach a maximum value during the body of the

seizure and help control the excitatory output from the pyramidal cells.

Page 56: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

51

The oriens interneurons fire at a high frequency and their output correlation value

remains high for most of the SLE (Ziburkus et al 2006). They reach their maximum firing

frequency just before seizure onset before abruptly entering into depolarization block.

During depolarization block the oriens interneurons are unable to generate action

potentials, causing their output correlation to drop to zero. This drop in inhibition may

be responsible for the sudden increase in firing seen in pyramidal cells. As the SLE

terminates the oriens interneurons emerge from the depolarization block with a steady

increase in the firing frequency and the output correlation returns to pre-seizure levels.

Simultaneously the firing frequency of the pyramidal cells decreases. The return of

oriens interneuron firing activity after the depolarization block along with the basket

cells appear to be responsible for reigning in the runaway excitation of the pyramidal

cells.

Finally, the input or subthreshold correlations in the pyramidal cell network remain

relatively constant, and high, leading up to the body of the seizure falling dramatically at

the seizure onset and recovering through the body of the seizure (Figure 15b, purple

trace). This result demonstrates that the cells of the pyramidal network are receiving

common inputs, possibly from the Schaffer collaterals from CA3, leading up to the

seizures. The loss of correlation at the beginning of the seizure (Figure 15b, purple trace

at 100s) may reflect the input from CA3 being cut off by the sudden increase in oriens

cell activity. It could also be a result of intrinsic membrane properties as the pyramidal

cells are engaging in higher firing and are therefore more depolarized during the seizure.

Subthreshold correlation of the pyramidal cells returns to pre-seizure levels as the

seizure terminates possibly reflecting the simultaneous increase in the output

correlation in the basket cell network (Figure 15c, black trace) and oriens firing activity

returning to pre-ictal levels (Figures 7 and 8, black trace).

For the pyramidal cells, correlated excitatory inputs of the Schaffer collaterals are only

partially balanced by a smaller increase in the correlated inhibitory inputs from, first, the

Page 57: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

52

oriens cells (before seizure onset) and then later by the basket cell network (after

seizure onset). Inhibitory firing rates in the basket (Figures 7 and 8, red trace,

significance denoted by ○) and oriens (Figures 7 and 8; black trace, significance denoted

by *) networks increase leading up to the onset of the seizure, but oriens inhibition fails

as these cells enter depolarization block (Figures 7 and 8; black trace). Decreasing

excitatory spike correlation occurs at the same time as the basket cell correlation

increases perhaps leading to the end of the seizure.

Page 58: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

53

a.

b.

C.

Figure 15. Summarized resample analysis. Total (a), subthreshold (b) and spike (c) correlation of basket-basket (B-B),

basket-pyramidal (B-P), oriens-pyramidal (O-P) and pyramidal-pyramidal (P-P) cell pairs. Traces are the same as the

resampled graphs in figures 10-14, shown here without error bars.

0 50 100 150 200 250 300 350 400

0.005

0.01

0.015

0.02

0.025

Time (s)

A.U

.

B-B

B-P

O-P

P-P

0 50 100 150 200 250 300 350 400

0.005

0.01

0.015

0.02

0.025

Time (s)

A.U

.

B-B

B-P

O-P

P-P

0 50 100 150 200 250 300 350 400

.001

.002

.003

.004

.005

.006Spike Correlation

Time (s)

A.U

.

B-B

B-P

O-P

P-P

Page 59: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

54

Basket cells

Basket cells receive input from the perforant pathway, Schaffer collaterals, other

unidentified interneurons and form autapses (Somogyi 2005), although they receive

inputs mostly from the principal cells (Freund and Katona 2007) and these excitatory

inputs are almost twice as numerous as inhibitory inputs (Gulyas 1999). Also, basket cell

dendrites in the stratum radiatum are connected with an extensive network of

dendrodendritic gap junctions (Fukuda and Kosaka 2000). Basket cell axons innervate

the pyramidal cells perisomatically.

PV+ basket cells receive very little inhibitory input; however the largest proportion of

this inhibitory input is at distal dendrites located in the stratum lacunosum moleculare

(Gulyas 1999). If the basket cells do receive any inhibition from the oriens interneurons,

it would not be effective since the majority of the input to the basket cell dendrites is

from the perforant pathway. However basket cells do innervate each other thus possibly

governing their own output firing rate. This is reflected in the spike rate (Figures 7 and

8; red trace) and spike correlation (Figure 15c, black trace) of basket cells, which

gradually begins increasing at seizure onset. The spike correlation reaches its maximum

value shortly after seizure onset although the firing frequency has now returned to pre-

ictal values.

The output correlation of the pyramidal cells increases before seizure onset and this

increase along with increased firing rates in the pyramidal network may contribute to

the gradually increasing firing rates (Figures 7 and 8, red trace) and spike correlation

(Figure 11e, f) in the basket network.

The subthreshold correlation of the basket-basket cell pairs (Figure 15c; black trace and

Figure 11c, d) does not show any significant changes though a trend exists. During the

pre-ictal period the input correlation of these cells increases, then drops at the start of

the seizure, and the input correlation remains low during the body of the seizure. The

Page 60: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

55

correlation returns to pre-ictal levels as the seizure terminates and stays here during the

subsequent inter-seizure interval.

Oriens interneurons

The oriens interneurons receive inputs from the CA1 pyramidal cells as well as inputs

from the CA3 via the Schaffer collaterals. However these CA3 fibers are distinct from

those entering the radiatum, hence stratum oriens and stratum radiatum receive

distinct synaptic inputs from different CA3 cells. The dendritic trees of oriens

interneurons lie in the stratum oriens layer and are electrotonically coupled via gap

junctions (Zhang et al 2004). The axons of oriens interneurons in particular the OLM

interneurons, innervate the distal part of the pyramidal cell dendritic tree in stratum

lacunosum moleculare.

Leading up to seizure onset both the output correlation (Figure 10e, f) and the firing

frequency of the pyramidal cells (Figures 8 and 9; green trace) increases. It is possible

that the increasingly synchronized firing output of the pyramidal cells (Figure 15c, purple

trace) before the seizure excites the oriens interneurons and results in their massive

burst of firing activity at the start of the seizure (Figures 7 and 8; black trace).

The subthreshold correlation in the oriens interneuron network remains weak during

the pre-ictal stage compared to during the seizure (not shown here, ref. Ziburkus et al

2006). It increases suddenly and drastically for a few seconds at seizure onset and then

decreases, but to a level higher than the pre-ictal correlation value. The sudden increase

in subthreshold correlation of oriens interneurons may be a result of the pyramidal cell

spike correlation and the spike rate reaching its peak at the same time.

Before summarizing the above dynamics there are several remarkable patterns of

interaction that may further elucidate the network dynamics involved. One such

relationship to be noted is that between pyramidal and basket cells. The input

Page 61: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

56

correlation between the pyramidal-basket cell pairs follows that of the pyramidal-

pyramidal cell pairs very closely and is distinctly different from the input correlation

between basket-basket cell pairs. It is known that the apical and basal dendrites of

basket cells spanning stratum radiatum and stratum oriens, respectively, receive inputs

from different CA3 pyramidal cells (Somogyi 2005). We propose that, pyramidal cells

receive the same input and the basket cells receive a subset of that information,

however the different basket cells receive different subsets of that information.

Yet another relationship to note is that between excitation and inhibition during the

seizure. The increased spiking activity of the oriens interneurons before seizure onset

can lead to pyramidal cell depolarization due to GABA-A receptor-mediated influx and

subsequent accumulation of Cl- and an efflux of HCO3

- (Fujiwara-Tsukamoto et al 2003).

The positive shift in chloride reversal decreases the efficacy of inhibition or even making

it excitatory, thus contributing to the “runaway excitation” seen in pyramidal cells

during seizures.

Page 62: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

57

DISCUSSION

Dual and triple whole cell recordings of two sub-types of interneurons and pyramidal

cells were performed to explore the interactions between these three cell types before,

during and after an SLE. Basket and oriens interneurons are two functionally and

morphologically distinct sub-classes of interneurons in the CA1 region of the

hippocampus and inhibit pyramidal cells via disparate mechanisms. These three cell

types by no means make up the entire neuronal network but are important since these

two types of GABAergic cells have such a fine degree of specificity for their target

membrane compartments; basket cells innervate the pyramidal cells perisomatically

whereas the oriens interneurons innervate the distal dendrites located in the stratum

lacunosum-moleculare.

Current information tells us that each of the 21 interneuron sub-types identified in the

CA1 region of the hippocampus are morphologically and functionally diverse

(Klausberger and Somogyi 2008). Specific dynamic interactions between interneurons

and pyramidal cells cause distinct brain states. Hence, exploring the functional diversity

of these interneurons during seizures is imperative to further our knowledge about

seizure dynamics in the hippocampal network. Given the differences between oriens

interneurons and basket cells (Klausberger and Somogyi 2008), we hypothesized distinct

and unique roles for both cell types in seizure dynamics.

Since the perisomatic region of pyramidal cells integrates inputs that lead to action

potential generation, basket cells with axons innervating the perisomatic region are

strategically placed to synchronize pyramidal cells (Somogyi 2005). Both basket and

pyramidal cells receive inputs from Schaffer collaterals which would lead to generation

of repetitive action potentials in the pyramidal cells but the strong feedforward

inhibition acts to control this repetitive generation of action potentials. The dendrites of

the basket cells located in stratum lacunosum-moleculare receive extrinsic excitatory

Page 63: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

58

afferents and also receive excitatory inputs from the pyramidal cells that they innervate.

Buhl et al (1996) state that given this specifically-targeted excitatory input to the basket

cells and their connections to other interneurons, basket cells play “a key role in the

feedforward control of principal cells” and are also involved in shaping the inhibitory

network activity.

Given the above, it is possible that under normal physiological conditions, basket cells

are involved in both feedforward inhibition (due to the extrinsic excitatory afferents)

and local feedback inhibition (due to the local excitatory inputs from the pyramidal

cells).

In fact, a single perisomatic IPSP can suppress repetitive discharge (Cobb et al 1995). A

single perisomatically-innervating interneuron synapses onto 500-1650 pyramidal cells

whereas a single dendritically-innervating interneuron synapses onto 400-2000 neurons

(Miles et al 1996). Even though the number of synapses is comparable between these

two interneuron cell types, Miles showed that in the CA3, and under normal

physiological conditions, basket cells were far more effective at silencing the activity in

the principal cells of the CA3. This higher degree of efficacy is attributed to more

synchronized IPSPs generated by the basket as compared to the dendritically

innervating cells.

A previous study demonstrated the unique interleaving pattern of activity between OLM

and pyramidal cells (Ziburkus et al 2006). OLM cell activity was shown to book-end the

SLE with increased firing before and after the pyramidal cells show “runaway excitation”

during the body of the seizure. OLM cells receive at least 70% of their glutamatergic

inputs from the CA1 pyramidal cells (Somogyi 2005) and provide feedback inhibition to

distal dendrites of the pyramidal cells, located in the stratum lacunosum-moleculare.

Results from the current study show the spike correlation of basket cells peak during the

Page 64: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

59

body of the seizure (Figure 15c, black trace; Figure 11e) at which time the pyramidal

cells’ spike correlation starts to decrease (Figure 15c, purple trace; Figure 10e). From

this observation along with other conclusions stated above and in the Interneuron

Diversity chapter earlier, we can further conclude that, under normal physiological

conditions, perisomatic inhibition more closely regulates efferent signaling of pyramidal

cells than dendritic inhibition. Hence perisomatically-innervating basket cell activity

most likely plays an important role in seizure dynamics.

Page 65: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

60

CONCLUSION

In reviewing all of the above information we propose the following summary. During

the ISI the firing activity of the oriens interneuron network is characterized by an

increasing firing rate that is statistically significant (Figure 9, middle row). Concurrently

the firing activity of the pyramidal cells and basket cells also show a statistically

significant increasing firing activity during the ISI (Figure 9, top and bottom rows

respectively).

The output correlation of the pyramidal cell network and the basket cell network also

shows an increasing trend (Figure 15c, purple and red trace respectively). The increase

in the excitatory correlation and activity appears to drive the system into a pre-seizure

state. Seizures are ignited when the effectiveness of the excitatory network overcomes

inhibition as the oriens cells enter a hyper-excited depolarization block.

Inhibition however, is not entirely lost as the basket cell activity and spike correlation

increases past the beginning of the seizure; outlasting the excitatory ramp up. This

inhibition may be responsible for the termination of the seizure, which occurs at the

same time oriens inhibition returns and runaway excitation is reigned in.

In our study, we emphasize cell-specific interactions leading up to, during and following

an SLE. Conceivably each interneuron sub-type in the CA1 region of the hippocampus

plays a specific and unique role in seizure dynamics. Discovering the cellular

mechanisms underlying these seizure interactions is necessary in creating rational

pharmaceutical targets.

Currently, certain antiepileptic drugs like carbamazepine target the sodium channel by

inactivating them thus rendering the cells inexcitable. Others like valproate prevent

entry of calcium at axon terminals preventing neurotransmitter release and also activate

Page 66: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

61

potassium conductance thus decreasing excitatory activity. Although effective, these

drugs treat epilepsy by suppressing network-wide activity and can have serious side

effects on normal cognitive function. By delineating cell-specific activity we are creating

the opportunity to target and alter specific cell types without using intrusive treatments

that tamp down the entire network.

In order to minimize seizures one would like to reduce the synchronization and

excitation seen in pyramidal cells. This is a common target of epileptic drugs to reduce

neuronal activity (gabapentin, felbamate). However, reduction of excitatory activity will

inevitably have effects on normal cognitive activity.

In addition, the intense firing of oriens interneurons may lead to chloride-loading of

pyramidal cells. This causes a positive shift in the chloride reversal decreasing the

efficacy of inhibition from oriens interneurons and possibly even resulting in net

excitation (Rivera et al 2005). Diuretics like furosemide are known to be anti-convulsant

by inhibiting chloride transport however its exact mechanism of action remains unclear

(Staley 2002). It is possible that furosemide prevents chloride-loading of pyramidal cells

thus preventing the chloride reversal and contains the excessive excitatory activity of

pyramidal cells. Another mechanism of action of diuretics is to draw water out of the

cells thus increasing the surrounding extracellular matrix volume (Staley 2002). The

swelling of the cell soma results in enhanced ephaptic interactions that contribute to

the spread of paroxysmal discharges and enhanced synchronization in pyramidal cell

population, and diuretics are able to reduce the excitability of pyramidal cells by

shrinking them. This effect may contribute to the enhanced synchronization seen in our

results where the pyramidal cells display increase spike correlation before seizure onset.

Second we look at the oriens interneurons that go into depolarization block. These

interneurons may go into depolarization block due to the inactivation of action

potential-generating mechanisms brought on by a persistent-sodium current which

Page 67: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

62

provides the driving force for potassium efflux and maintains the open potassium

channels, resulting in [K+]o buildup (Bikson 2003) or due to the lack of oxygen locally.

The failure of oriens interneurons during the depolarization block releases inhibition on

pyramidal cells which display excessive excitation during seizures. One possible

therapeutic target would be to prevent depolarization block by reducing oriens

interneuron spiking activity. With reduced firing activity the action potential-generating

mechanisms will not be inactivated and hence prevent depolarization block.

Third, our results showed that basket cells are last to reach their maximum firing

frequency during the SLE (Figures 7 and 8, red trace). Basket cells are “strategically

placed” for perisomatic inhibition which is more effective than dendritic inhibition in

modulating the output of the pyramidal cells. Hence, one possible therapeutic option

would be to target the excitability of basket cells to increase their firing activity earlier,

thus cutting short the seizure or preventing it all together. This could be done by a cell-

targeted increase in efficacy of glutamate receptors on the basket cells. If basket cells

are more responsive to the pyramidal input they are more likely to fire earlier and thus

can terminate or prevent the seizure all together.

Fourth, any typical changes in activity of cells, if any, during the inter-seizure intervals

can be monitored to predict future seizures. Recent findings (Cymerblit-Sabba and

Shiller 2010) show a “typical network dynamics signature” that preceded seizures

induced in vivo. They made multi-electrode single-unit recordings in the rat

hippocampus and induced seizures using picrotoxin, kainate or pilocarpine. The typical

biphasic network dynamics they observed before seizure onset was a decrease in

interneuronal synchronization followed by an increase in the firing activity and

synchronization of the network leading up to the start of the seizure. Current data in the

literature about preictal states is confined to surface and intracranial EEG recordings but

Cymerblit-Sabba and Shiller (2010) extend that knowledge by using multielectrode

Page 68: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

63

single-unit recording to study these network dynamics at the single-neuron and multi-

neuron level.

We performed simultaneous extracellular and dual/triple whole-cell recordings from

three different cell types in the CA1 of the rat hippocampus and monitored the activity

before, during and after SLEs. Our findings confirm Cymerblit-Sabba and Shiller’s

excitatory cell rate and correlations findings, and furthermore we provide the first

contrasting patterns and pre-seizure interactions of two different inhibitory

interneurons. We demonstrate changes in firing activity and correlations of all three cell

types during the inter-seizure interval leading up to seizure onset. We observed a

significant increase in spike rates for all three cells approximately 60-105 seconds before

seizure onset (Figure 9). A similar increase in firing activity (Bower and Buckmaster

2008) was also observed in the granule cells of the dentate gyrus 4 minutes before the

onset of local seizures. Using tetrodes to record from single granule cells they measured

the firing rates of the granule cells before and during spontaneous seizures in the rat

pilocarpine model of temporal lobe epilepsy. They found significant changes in action

potential rates of granule cells before the onset of spontaneous seizures. The firing

rates of 19% of granule cells recorded did not change before or during seizures, many

showed a significant decrease in firing frequency before seizure onset, some showed an

increase followed by a decrease in firing frequency (“bimodal firing pattern”) before

seizure , however, 34% of granule cells showed an increase in firing frequency before

the seizure. This increase in firing began 4 minutes before seizure onset. Findings from

our experiments show a significant increase in firing activity of all three cell types that

begins 1 to almost 2 minutes before seizure onset in CA1.

In conclusion, the findings of the above two studies and the results from our

experiments underline the fact that transitions from interictal to ictal stages in an

epileptic brain can be monitored by the changes in firing activity of the cells before

seizure onset. However, a more sensitive approach to monitor changes in activity would

be the correlation measures, as we have described in our work. Using multi-contact strip

Page 69: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

64

electrodes to record multiple single cell activity, we can monitor and measure the

correlation of spike activity between multiple cells and thus predict seizures. Seizure

prediction algorithms can be used in novel treatments like closed-loop

neurostimulators, where the neurostimulators can detect typical network activity

preceding a seizure and can produce a specific stimulation protocol that prevents the

oncoming seizure (Cymerblit-Sabba and Shiller 2010).

In addition, though we are unable to fully explain the mechanisms underlying the

interactions we have observed in our experiments, distinct patterns of interactions

between the various cell sub-types do clearly emerge. Hence, seizures are a culmination

of a ramping up of excitatory activity and correlation which is initially kept in check by

interneuron activity. This balance however cannot be maintained as inhibition fails

leading to a release of the pyramidal cell activity which is only brought under control by

the increased activity in correlation in the basket cell network. Understanding the role

of feedback inhibition to suppress seizure onset and perisomatic inhibition to terminate

seizures may lead to novel and more effective treatments of epileptic disorders.

Page 70: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

65

References

Ali AB, Deuchars J, Pawelzik H, and Thomson AM. (1998) CA1 pyramidal to basket and

bistratified cell EPSPs: dual intracellular recordings in rat hippocampal slices. J

Physiology 507.1:201-217.

Andersen P, Morris R, Amaral D, Bliss T, O’Keefe J. (2007) The hippocampus book.

Oxford University Press.

Anderson WW, Lewis DV, Swartzwelder HS, Wilson WA. (1986) Magnesium-free

medium activates seizure-like events in the rat hippocampal slice. Brain Research

398: 215-219.

Avoli M, Drapeau C, Louvel J, Pumain R, Olivier A, Villemure J-G. (1991) Epileptiform

activity induced by low extracellular magnesium in the human cortex maintained in

vitro. Annals of Neurology 30: 589-596.

Bartlett MS. (1946) On the theoretical specification of sampling properties of

autocorrelated time series. J Royal Stat Soc B8, 27.

Ben-Ari Y. (1985) Limbic seizure and brain damage produced by kainic acid: mechanisms

and relevance to human temporal lobe epilepsy. Neuroscience 14(2): 375-403.

Ben-Ari Y and Cossart R. (2000) Kainate, a double agent that generates seizures: two

decades of progress. Trends in Neuroscience 23(11): 580-587.

Bikson M, Hahn PJ, Fox JE, Jefferys JGR. (2003) Depolarization block of neurons during

maintenance of electrographic seizures. Journal of Neurophysiology 90: 2402-2408.

Bower MR and Buckmaster PS. (2008) Changes in granule cell firing rates precede locally

recorded spontaneous seizures by minutes in an animal model of temporal lobe

epilepsy. Journal of Neurophysiology 99: 2431-2442

Brody CD. (1991) Correlations without synchrony. Neural Computation 11:1537-1551.

Buhl EH, Szilágyi T, Halasy K, and Somogyi P. (1996) Physiological properties of

anatomically identified basket and bistratified cells in the CA1 area of the rat

hippocampus in vitro. Hippocampus 6: 294-305.

Chamberlin NL and Dingledine R. (1988) GABAergic inhibition and the induction of

spontaneous epileptiform activity by low chloride and high potassium in the

hippocampal slice. Brain Research 445: 12-18.

Cobb SR, Buhl EH, Halasy K, Paulsen O, and Somogyi P. (1995) Synchronization of

neuronal activity in hippocampus by individual GABAergic interneurons. Nature 378:

75-78.

Page 71: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

66

Cymerblit-Sabba A and Shiller Y. (2010) Network dynamics during development of

pharmacologically induced epileptic seizures in rats in vivo. Journal of Neuroscience

30(5): 1619-1630

Dingledine R, Borges k, Bowie D, Traynelis SF. (1999) The glutamate receptor ion

channels. Pharmacological Reviews 51: 7-61.

Elfant D, Pál BZ, Emptage N, Capogna M. (2008) Specific inhibitory synapses shift the

balance from feedforward to feedback inhibition of hippocampal CA1 pyramidal

cells. European Journal of Neuroscience 27: 104-113.

Fertziger AP and Ranck Jr., JB. (1970) Potassium accumulation in interstitial space during

epileptiform seizures. Experimental Neurology 26: 571-585.

Freund TF and Katona I. (2007) Perisomatic Inhibition. Neuron 56: 33-42.

Fujiwara-Tsukamoto Y, Isomura Y, Nambu A, Takada M. (2003) Excitatory GABA input

directly drives seizure-like rhythmic synchronization in mature hippocampal CA1

pyramidal cells. Neuroscience 119: 265-275.

Fujiwara-Tsukamoto Y, Isomura Y, Kaneda K, and Takada M. (2004) Synaptic interactions

between pyramidal cells and interneurone subtypes during seizure-like activity in

the rat hippocampus. Journal of Physiology 557.3: 961-979.

Fukuda T and Kosaka T. (2000) Gap junctions linking the dendritic network of GABAergic

interneurons in the hippocampus. Journal of Neuroscience 20(4): 1519-1528.

Galvan M, Grafe P, Bruggencate GT. (1982) Convulsant actions of 4-Aminopyridine on

the guinea-pig olfactory cortex slice. Brain Research 241:75-86.

Gillies MJ, Traub RD, LeBeau FEN, Davies CH, Gloveli T, Buhl EH. (2002) A model of

atropine-resistant theta oscillations in rat hippocampal area CA1. Journal of

Physiology 543.3: 779-793.

Glickfield LL, Roberts JD, Somogyi P, and Scanziani M. (2009) Interneurons hyperpolarize

pyramidal cells along their entire somatodendritic axis. Nature Neuroscience 12: 21-

23.

Hajos N, Palhalmi J, Mann EO, Nemeth B, Paulsen O, and Freund TF. (2004) Spike timing

of distinct types of GABAergic interneuron during hippocampal gamma oscillations in

vitro. Journal of Neuroscience 24(41): 9127-9137.

Halasy K, Buhl EH, Lörinczi Z, Tamás G, and Somogyi P. (1996) Synaptic target selectivity

and input of GABAergic basket and bistratified interneurons in the CA1 area of the

rat hippocampus. Hippocampus 6: 306-329.

Han ZS. (1996) Morphological heterogeneity of non-pyramidal neurons in the CA1

region of the rat hippocampus. Neuroscience Research 25:51-65.

Page 72: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

67

Kim J, Wei DS, Hoffman DA. (2005) Kv4 potassium channel subunits control action

potential repolarization and frequency-dependent broadening in rat hippocampal

CA1 pyramidal neurons. Journal of Physiology 569.1: 41-57.

Klausberger T, Magill PJ, Márton LF, Roberts JDB, Cobden PM, Buzsáki G, and Somogyi P.

(2003) Brain-state- and cell-type-specific firing in hippocampal interneurons in vivo.

Nature 421: 844-848.

Klausberger T and Somogyi P. (2008) Neuronal diversity and temporal dynamics: the

unity of hippocampal circuit operations. Science 321: 53-57.

Koehling R, Gladwel SJ, Bracci E, Vreugdenhil M and Jefferys JGR. (2001) Prolonged

epileptiform bursting induced by 0 Mg2+

in rat hippocampal slices depends on gap

junctional coupling. Neuroscience 105: 579-587.

Korn SJ, Giacchino JL, Chamberlin NL, Dingledine R. (1987) Epileptiform burst activity

induced by potassium in the hippocampus and its regulation by GABA-mediated

inhibition. Journal of Neurophysiology 57(1): 325-340.

Losonczy A, Zhang L, Shigemoto R, Somogyi P and Nusser Z. (2002) Cell type dependence

and variability in the short-term plasticity of EPSCs in identified mouse hippocampal

interneurones. Journal of Physiology 542.1: 193-210.

Mann EO and Paulsen O. (2007) Role of GABAergic inhibition in hippocampal network

oscillations. Trends in Neuroscience 30(7): 343-349.

Miles R, Toth K, Gulyás AI, Hájos N, and Freund TF. (1996) Differences between somatic

and dendritic inhibition in the hippocampus. Neuron 16:815-823.

Mody I, Lambert JDC, Heinemann U. (1987) Low extracellular magnesium induces

epileptiform activity and spreading depression in rat hippocampal slices. Journal of

Neurophysiology 57(3): 869-880.

Morris R. 2007. Theories of Hippocampal function. In: Andersen P, Morris R, Amaral D,

Bliss T, O’Keefe J, editors. The Hippocampus Book. 1st

ed. Oxford University Press. p

617-622.

Netoff TI and Schiff SJ. (2002) Decreased neuronal synchronization during experimental

seizures. Neuroscience 22: 7297-7307.

Perreault P and Avoli M. (1989) Effects of low concentrations of 4-Aminopyridine on CA

1 pyramidal cells of the hippocampus. Journal of Neurophysiology 61(5):953-970.

Perreault P and Avoli M. (1991) Physiology and pharmacology of epileptiform activity

induced by 4-aminopyridine in rat hippocampal slices. Journal of Neurophysiology

65(4): 771-785.

Page 73: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

68

Perreault P and Avoli M. (1992) 4-Aminopyridine-induced epileptiform activity and a

GABA-mediated long-lasting depolarization in the rat hippocampus. Journal of

Neuroscience 12(1): 104-115.

Pike FG, Goddard RS, Suckling JM, Ganter P, Kasthuri N, and Paulsen O. (2000) Distinct

frequency preferences of different types of rat hippocampal neurons in response to

oscillatory input currents. Journal of Physiology 529.1: 205-213.

Pouille F and Scanziani M. (2001) Enforcement of temporal fidelity in pyramidal cells by

somatic feed-forward inhibition. Science 293: 1159-1163.

Rutecki PA, Lebeda FJ, Johnston D. (1987) 4-Aminopyridine produces epileptiform

activity in hippocampus and enhances synaptic excitation and inhibition. Journal of

Neurophysiology 57(6): 1911-1924.

Sanders JW, Shorvon SD. (1996) Epidemiology of the epilepsies. Journal of Neurology,

Neurosurgery & Psychiatry 61: 433-443.

Sik A, Penttonen M, Ylinen A, and Buzsáki G. (1995) Hippocampal CA1 Interneurons: an

in vivo intracellular labeling study. Journal of Neuroscience 15(10): 6651-6665.

Staley K. (2002) Diuretics as Antiepiletic drugs: Should we go with the flow? Epilepsy

Currents 2(2): 35-38.

Stief F, Zuschratter W, Hartmann K, Schmitz D, and Draughn A. (2007) Enhanced

synaptic excitation-inhibition ratio in hippocampal interneurons of rats with

temporal lobe epilepsy. European Journal of Neuroscience 25: 519-528.

Somjen GG and Giacchino JL. (1985) Potassium and calcium concentrations in interstitial

fluid of hippocampal formation during paroxysmal responses. Journal of

Neurophysiology 53(4): 1098-1108.

Somjen G. (2004) Ions in the brain-Normal function, seizures and stroke. Oxford

University Press.

Shepherd G. (2004) The synaptic organization of the brain. 5th

ed. Oxford University

Press.

Thesleff S. (1980) Aminopyridines and synaptic transmission. Neuroscience 5:1413-1419.

Traub RD, Whittington MA, Buhl EH, LeBeau FE, Bibbig A, Boyd S, Cross H and Baldeweg

T (2001) A possible role for gap junctions in generation of very fast EEG oscillations

preceding the onset of, and perhaps initiating, seizures. Epilepsia 42: 153-170.

Traynelis SF and Dingledine R. (1988) Potassium-induced spontaneous electrographic

seizures in the rat hippocampal slice. Journal of Neurophysiology 59(1): 259-276.

Page 74: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

69

Trevelyan AJ, Sussillo D, and Yuste R. (2007) Feedforward inhibition contributes to the

control of epileptiform propagation speed. Journal of Neuroscience 27(13): 3383-

3387.

Voskuyl RA and Albus H. (1985) Spontaneous epileptiform discharges in hippocampal

slices induced by 4-Aminopyridine. Brain Research 342: 54-66.

Walker, Chan, Thom. (2007) Hippocampus and human disease. In: Andersen P, Morris R,

Amaral D, Bliss T, O’Keefe J, editors. The Hippocampus Book. 1st

ed. Oxford

University Press.

Wilcox WR. (2006). Website: http://people.clarkson.edu/~wwilcox/ES100/datapoly

Whittington MA, and Traub RD. (2003) Interneuron diversity series: Inhibitory

interneurons and network oscillations in vitro. Trends in Neuroscience 26(12): 676-

682.

Wong RKS and Traub RD. (1983) Synchronized burst discharge in disinhibited

hippocmapal slice.1.Initiation in CA2-CA3 region. Journal of Neurophysiology 49(2):

442-458.

Ziburkus J, Cresmman JR, Barreto E, and Schiff SJ. (2006) Interneuron and pyramidal cell

interplay during in vitro seizure-like events. Journal of Neurophysiology 95: 3948-

3954.

Page 75: SEIZURE-LIKE EVENTS IN CA1 OF THE RAT HIPPOCAMPUS

70

VITA

Ruchi Parekh

The author was born and raised in Bombay, India. She attended Ohio Wesleyan

University in Delaware, Ohio where she earned her B.A. in Psychology in 2002. At

Pennsylvania State University she conducted her doctoral research in epilepsy in the

laboratory of Dr. Steven J. Schiff. While finishing this work, she accepted a postdoctoral

position with Dr. Giorgio Ascoli at George Mason University.