HIPPO

THE

M.

CAMPAL

Sc.,

MB.

REGULA

A

Suez

THESIS

BCh.,

THE

NEURONS

THE

THE

Canal

TION

KHALED

We Cairo

Khaled

REQUIREMENTS

SUBMITTED

UNIVERSITY

FACULTY

accept

OF

Medical DOCTOR (Department

Medical

to

INTRA Mohammed

MOHAMMED

AND

the

December

this

required

School,

OF

THE

School, CELL

OF

IN

thesis

OF

BY

GRADUA

in

PARTIAL

of

PHILOSOPHY

FOR

INFLUENCE

BRITISH ULAR

Abdel-Hamid,

Physiology)

Suez

standard

1994

ABDEL-HAMID

as

Cairo

THE

conforming

CALCIUM

TE

Canal

FULFILLMENT

DEGREE

COLUMBIA University

STUDIES

OF

University

CALCIUM

1994.

IN

OF

CUL (1982)

OF

TURED

(1989)

BUFFERS ______

In presenting this thesis in partial fulfilment of the requirements for an advanced

degree at the University of British Columbia, I agree that the Library shall make it

freely available for reference and study. I further agree that permission for extensive copying of this thesis for scholarly purposes may be granted by the head of my department or by his or her representatives. It is understood that copying or publication of this thesis for financial gain shall not be allowed without my written permission.

(Signature)

(r.’ Department of 11 4 I

The University of British Columbia Vancouver, Canada

Date 9

DE-6 (2/88)

competition the

Such

secondary

buffering to

in

hypothesis,

expressing

or

enhancing

loading

Manipulation

responses,

in

expression

(CaBP).

load

difference

subject

central

Neuronal

of

pathological

has

the

cultured

glutamate-induced

by

calcium

prolongation

been

handling

a

loading

enhancement

Excessive

prolongation

role

neurons

to

This

capacity

populations

blamed

to

neuronal this

has,

CaBP

hippocampal

and

(Ca 2 i,

of

were

between

in

situations

the

of

neurons

work

capacity

the

the

form

controversially,

on

neuronal

with

of

or

impairment

activation

more

as

high-capacity

are

triggering

the

the

aims

and

loaded

Ca 2 +

of

may

of

a

the

BAPTA-like

neuronal

differ

likely

with

causative

vulnerability

neuronal

of

vulnerable excessive

via

recovery

the

neurons

at

recovery

be

Ca 2

neurons.

buffering

a

with

BAPTA-like

studying

of

in

to

consequent

a

of

process

of

their

result

the

include

death

a

buffering

been

Ca 2 -mediated

calcium-binding

loss an

phase

mechanism

on series

Ca 2

activation

to

major

mechanisms

ABSTRACT

artificial

vulnerability

of

capacity,

The

agonist-

of

the

under

excitotoxicity.

in

attributed

known

these

an

at

the

buffers,

vitro.

buffers.

of

loss

latter

neurotransmitter

potential

capacity

increase

the

events

increased conditions

Ca 2 +

have

neurons

as

of in

and

end either

We

may

a

to

on

excitotoxicity.

Ca 2 +

will

inhibition

to

proteins

We

large

depolarization-induced

been

ending

influence

of

buffer,

the

found

could

in

one

excitotoxicity

possibly

render

by

The

tested

the

the

to

Ca 2 +

homeostasis

of

variability

number

suggested

side,

the

excitotoxic

also

that

excitotoxic

increases

mechanism(s)

net

such

in

contrary

of

glutamate

them

expression

influx

of

neuronal

the

reflect

Ca 2

and

influx

be

neurons

CaBP

as

of

Excessive

hypothesis

achieved

in

more

the

and

calbindin-D28K

neuro

and

to

to

influx,

Ca 2 + the

in

of

the

stimuli.

stimulus. receptors

expression

play

large

death.

our

neurons

this

Ca 2 ,

resistant

of

calcium-

level

involved

Ca 2 +

CaBP

and

a

influx

by

that of III number of high affinity 2Ca binding sites of the buffer on the other. In neurons that express CaBP, an added vulnerability factor may be the consistently higher peak 2+Ca responses to glutamate receptor agonists and to depolarization observed in these neurons when compared to responses in neurons lacking CaBP. The biological effects of enhanced 2Ca buffering capacity, particularly using fast and mobile buffers, would be subject to the cell-specific nature of the spatial and temporal pattern of the 2Ca signal. Thus, the influence of 2Ca buffering on neuronal vulnerability may be difficult to predict on a theoretical basis particularly in view of our results indicating the large impact of many factors in the culture environment on the vital properties of neurons in vitro. iv

TABLE OF CONTENTS Pages

ABSTRACT ii

TABLE OF CONTENTS iv

LIST OF TABLES ix

LIST OF FIGURES x

ACKNOWLEDGMENTS xiii

DEDICATION xiv

1. INTRODUCTION 1

GENERAL INTRODUCTION 2

1A. INTRANEURONAL CALCIUM AND ITS REGULATION 4 I. 2+Ca influx through neuronal membranes 5 A. Voltage-gated 2Ca channels 5 B. Ligand-gated 2Ca channels 9 II. 2Ca efflux mechanisms 12 A. Ca-ATPase 12 B. 2Na/Ca2 exchanger 13 Ill. Intracellular 2Ca sequestration 15 A. Mitochondria 15

B. Endoplasmic reticulum 18

IV. Calcium-binding proteins 20 V. Intracellular role of 2Ca 20 VI. Recapitulation 21 lB. EXCITOTOXICITY 23

I. History and definition 23

II. Evidence for glutamate involvement 23 V

A. In vivo 23

B. In vitro 26

Ill. Phases of glutamate-induced neurotoxicity: time course and ionic dependence 27

IV. A three-stage model of glutamate excitotoxicity 29 V. Limitations of the excitotoxic hypothesis 30 VI. 2Ca as the common final pathway and alternative hypotheses 31 VII. Clinical implications 33

VIII. Therapeutic approaches to excitotoxic injury 34 IX. Recapitulation 35 ic. CALBINDIN-D28K 36

I. Introduction 36

II. CaBP History and distribution 36

Ill. Chemistry and molecular biology of CaBP 38

IV. CaBP expression in the central nervous system 41 V. Developmental expression of CaBP and related CBPs 42 VI. Modulation of CaBP expression 43

VII. Biological role of CaBP 45

1D. CALCIUM BUFFERS AND BIOLOGICAL SYSTEMS 47 Theoretical considerations 47

Buffers in non-biological systems 47

Buffers in biological systems 48 Microdomains and immobile buffers 48

1E. HYPOTHESIS AND OBJECTIVES OF THE STUDY 52 2. MATERIALS AND METHODS 54

I. Tissue culture 55 a. Fetal cultures 55 b. Postnatal cultures 59

3.

CHARACTERIZATION

II.

I. Sources

VII.

Preparation VI.

V.

IV.

Ill.

II.

Survival

Ca 2

Induction

Intracellular

Staining

FURA-2

Loading

Statistical

a.

d.

c.

b.

a.

a.

b.

a.

b.

f.

e.

d.

c.

b.

a.

c.

for

binding

Calbindin-D28k

Effects

Sensitivity

Age

Identification

Relocation

Viability

Exposure

Background

Kinetic

Fura-2

Test

Image

Fura-2

Principle

bFGF-treated

of

of

Materials

data

neurons

and

of

hippocampal

dependence

media

methods

Calcium

excitotoxicity

Acquisition

of

proteins

calibration

photography AM

analysis

analysis

assessment

OF

of

protocols

basic

to exposure

experiments

and

with

Loading

Fura-2

correction

HIPPOCAMPAL

of

glutamate-induced

cultures

Measurement

(CaBP)

fibroblast

BSS

neurons

in

BAPTA

of

neurons

cultured

calcium

neuronal

protocols

and

in or

growth

in

cultures

hippocampal

its

calibration

measurement

culture

NEURONAL

density

and

analogues

excitotoxicity

factor

perfusion

in

culture

(bFGF)

neurons

CULTURES

93

93

93

92

91

90

90

89

78

78

77

75

72

71

71

70 69

70

69

68

67

66

64

64

60

60

60 vi vii

b. Parvalbumin 94 c. Effects of bFGF on 2Ca binding proteins 94 Ill. 2+Ca responses in cultured hippocampal neurons 95 a. 21+][Ca in non-stimulated neurons 95 b. Effects of excitatory amino acids on 2[Ca 96 c. Depolarization-induced 2Ca fluxes j 98 d. Effects of the absence of extracellular 2Ca 100 e. Effects of other 2Ca release agonists 101 Discussion 103 4. THE EFFECTS OF ARTIFICIAL BUFFERS ON NEURONAL Ca2+ RESPONSES 134 I. BAPTA effects on short 2Ca transients 136 II. Baseline 2Ca changes during the BAPTA loading procedure 138 III. Buffer effects on extended 2+Ca responses 140 IV. BAPTA effects on 2Ca levels during excitotoxic glutamate exposure 142 Discussion 144 5. CALBINDIN-D28K EXPRESSION AND NEURONAL 2Ca RESPONSES 163

I. The Association beteen CaBP expression and resting intraneuronal [Ca ] 164 Il. The Association between CaBP expression and transient increases in 21[Ca in cultured fetal hippocampal neurons 164 a. jNMDA-induced 2Ca transients 164 b. Depolarization-induced 2Ca transients 166 Ill. The Association between CaBP expression and transient exposure of cultured postnatal hippocampal neurons to NMDA 167 IV. The Association between CaBP expression and prolonged 2+Ca responses 169 Discussion 170

ABBREVIATIONS

BIOGRAPHICAL

BIBLIOGRAPHY

7.

6.

GENERAL

EXCITOTOXICITY

VI.

Discussion

Ill.

II.

I.

Excitotoxicity

Excitotoxicity

The

Effects

a.

b.

neurotoxicity

c.

b.

a.

analogues

d.

c.

b.

a.

DISCUSSION

association

INFORMATION Fetal

Postnatal

Correlation

Fetal

Postnatal

Effects

[Ca 2 1

Fetal

Postnatal

of

neurons

bFGF

cultures

cultures

cultures

IN

in

of

in

changes

HIPPOCAMPAL

cultures

cultures

‘naive’

cultures

neurons

between

treatment antagonists

AND

with

CONCLUSIONS

Ca 2

under

neuronal

loaded

CaBP

on

changes

excitotoxic

glutamate

NEURONAL

immunoreactivity

with

cultures

BAPTA

in

BAPTA-loaded

toxicity

conditions

CULTURES

and

and

its

glutamate

259

241

238

220

201

199

198

198

198

198

197

197

196

195

195

194

193

193

192 VIII ix

LIST OF TABLES Pages 2.1 Problems associated with fura-2 fluorimetry, and their remedies 87 2.2 Outline of the cresyl violet staining method. 88 4.1 The percentage chaes in the kinetic parameters of transient NMDA-induced Ca responses in neurons loaded with BAPTA or with DMB. 162 5.1 Comparison of the kinetics of the Ca responses in fetal CaBP CaBPN 2 and neurons exposed to NMDA. 188 5.2 Comparison of the kinetics of the 2Ca responses in fetal CaBP and CaBPN neurons depolarized by exposure to 50 mM 0[K] 189 5.3 Frequencies of the rlative magnitudes of kinetic parameters. in CaBP’ and CaBP’ fetal neurons. 190 5.4 Comparison of t[’ie kinetics of he 2Ca responses in cultured 4-day postnatal CaBPH and CaBP’ neurons exposed NMDA. 191 x

LIST OF FIGURES Pages

2.1 The excitation spectrum of fura-2. 81 2.2 Changes in fura-2 fluorescence in a typical 2+Ca response. 82 2.3 Illustrated diagram of the imaging hardware. 83 2.4 Schematic diagram of the perfusion chamber. 84

2.5 Schematic diagram of a perfusion-head assembly. 85 2.6 Kinetic parameters used in the description of 2+Ca responses. 86 3.1 Postnatal neuronal cultures stained with (a) NSE and (b) GFAP. 109 3.2 Neuronal survival in vitro varies with the different culture conditions used. 110

3.3 BFGF enhances neuronal survival in vitro. 11 2 3.4 Survival and number of CaBP cultured neurons varies in different culture types. 113 3.5 BFGF increases neuronal CaBP expression in vitro. 115

3.6 In fetal face-down cultures, bFGF effect on CaBP expression is enhanced in the presence of media conditioned by postnatal neurons. 116

3.7 NMDA induces a oncentration-dependent increase in intraneuronal [Ca 1. 117 3.8 APV and 2Mg abolish completely NMDA-induced 2Ca responses. 11 9 3.9 NMDA-induced 2Ca responses are modulated by changes in the extracellular pH. 120 3.10 Quisqualate is highly potent in inducing intraneuronal 2Ca responses. 121 3.11 Quisqualate-induced 2Ca responses show that QUIS does not have a homogeneous potency on hippocam pal neurons. 122 3.12 NMDA-induced 2+Ca responses in neurons are more homogeneous than Quis-induced responses. 123 3.13 Nifedipine inhibits depolarization-induced 2Ca responses. 124 3.14 Multiple types of VGCC contribute to HK-induced 2Ca responses in cultured hippocampal neurons. 125 xi

3.15 HK- and NMDA-induced 2Ca responses are modulated by changes in extracellular pH. 126 3.16 HK-induced and EAA-induced 2Ca responses are abolished in 2Ca-free media. 127 3.17 The effects of Ca-free medium on NMDA-induced 2Ca responses. 2 128 3.18 The effect of extracellular 2+Mg on the calcium responses to NMDA and ACPD isomers. 129

3.19 MK8O1 inhibited ACPD induced rises in neuronal free calcium. 130 3.20 An example of a single cell where MK8O1 did not abolish the calcium response to 5 mM 1S,3R-ACPD. 131

3.21 Caffeine induces only a small increase in intraneuronal 132 4.1 The effects of BAPTA loading on short-duration 2Ca transients. 150 4.2 Changes in ]2[Ca during exposure to BAPTA-AM. 151 4.3 Heterogeneity of the responses to BAPTA-AM exposure. 152 4.4 reverses BAPTA-AM induced increases in 21.[Ca 154 4.5 Ca buffering and buffer-AM induced increases in 2[Ca are independent phenomena. 155 4.6 Thei effect of Ca buffering on short- and extended- 2+ duration 2Ca responses. 157 4.7 The effect of 2+Ca buffers on ‘excitotoxic’ exposure to glutamate. 160 5.1 NMDA-induced 2+Ca responses are larger and longer-lasting in CaBP fetal hippocampal neurons. 179 5.2 Larger and longer-lasting +Ca responses are observes in CaBP 2 + neurons even in the absence of initial high resting Ca levels. 180 5.3 Comparison of the mean 2+Ca response in CaBPN and CaBP neurons following exposure to 2OpM of NMDA for 25 seconds. 181 5.4 Depolarization-inducçd increases in +][Ca are larger and recovery is slower in compared to CaBPN2 CaBP’ fetal neurons. 183 5.5 Mean [Ca in cultured 4-day postnatal hippocampal neurons (8-DIV)2 exposed to 20 pM NMDA for 25 seconds is larger in CaBP(H] CaBPN neurons than in neurons. 185 xl’

6.1 Glutamate toxicity in face-down fetal neurons: protective effect of extracellular 2Mg 210 6.2 Glutamate toxicity. in postnatal culture. 211 6.3 Changes in 2[Ca during glutamate toxicity. 212 6.4 Time course i of glutamate-induced neuronal death is widely variable in different cells. 213

6.5 Neurons hat collapse early in response to glutamate have higher peak Ca + responses. 214

6.6 BAPTA and DMB promote glutamate toxicity in fetal neurons. 215 6.7 BAPTA shifts the concentration-response curve of glutamate toxicity to the left. 216

6.8 BAPTA and DMB promote glutamate toxicity in postnatal neurons. 217 6.9 CaBP(H postnatal neurons are more sensitive to glutamate toxicity. 218

6.10 Effect of bFGF treatment on glutamate toxicity in postnatal neurons. 219 7.1 Simulated 2Ca changes in fetal neurons control, BAPTA-loaded and CaBP, in response to a short-duration exposure to NMDA. 237 XIII

ACKNOWLEDGEMENT

I would like to express my gratitude to Dr. Ken Baimbridge for his

continuous help and advice throughout all the stages of the research work I have undertaken for the last 5 years. During the numerous discussions we had, his criticism and insightful comments were invaluable in formulating the ideas presented in this work. His support, however, extends beyond the science arena and his guidance has been extremely valuable for me as a foreign student settling in and trying to understand a new society. His friendliness and considerate attitude have made my stay here a very enjoyable experience.

To Miss Stella Atmadja I owe a lot of thanks. The excellence of her technical expertise in tissue culture and histological techniques cannot be overstated. Her supportive attitude and enthusiasm for new ideas, including those that impose a lot more demand on her time, were always encouraging.

I also want to express my deep appreciation to Dr. John Church and Dr. Steve KehI for the time and effort they spent proof-reading the manuscript of the thesis and their extensive and invaluable comments and suggestions. Many thanks are also due to the members of my advisory committee for their helpful comments and appraisal of my work, from a research proposal to a completed thesis.

I am very grateful to the technical and secretarial staff of the Department of Physiology at the University of British Columbia for their continuous help and support.

No doubt that my greatest debt and ultimate gratitude should go to my wife, Dr Lamice El-Kholy, for her endless dedication and sacrifice, and her unlimited support I have been receiving over the years. Her effort in proof reading the manuscript and her critical comments are also highly appreciated. xiv

DEDICATION

This work is dedicated to my wife Lamice, may God bless her with happiness and success, and to the joy of my life, my sons Omar and Mostafa. 1

CHAPTER 1

INTRODUCTION 2

GENERALINTRODUCTION

The experimental work presented in this thesis will be concerned primarily with investigations of the role of artificial 2+Ca buffers and neuronal calcium- binding proteins that have been proposed to act as neuronal 2+Ca buffers, on transient or prolonged changes in 2+][Ca induced by membrane depolarization or specific ligand-receptor interactions.

Inside the cell, 2+Ca can function as a charge carrier and as an intracellular second messenger. In the latter role, changes in 21[Ca are sensed by one or more of a number of high affinity 2Ca +bindingi sites, such as those found on the ubiquitous protein calmodulin, and then translated into a cellular response. To some extent the binding of 2+Ca to these sites can also be considered as 2Ca buffering since a rise in 21[Ca will be limited by the removal of 2Ca from the ionic pool within the cytoplasm.j With proteins such as calmodulin this buffering action, while not necessarily unimportant, is essentially secondary to their enzyme modulatory actions. However, there are members of the Ca-binding protein family for which a high capacity for binding (and buffering)2 2Ca has been demonstrated and which appear not to modulate enzyme activity. It is therefore conceivable that the latter group of proteins have evolved as natural 2Ca buffers. This possibility, and the ensuing biological consequences will be discussed after first reviewing our current understanding of the mechanisms involved in the regulation of intraneuronal 2Ca The potential role of 2Ca buffering, both natural and artificial, in neurons. during periods of prolonged neuronal stimulation which, if uncontrolled, can lead to cell death will be examined. This form of neuronal 3 death, which has been termed excitotoxicity, will therefore be reviewed with an emphasis on the role played by intraneuronal 2Ca Finally, and in view of this laboratory’s interest in 2+Ca buffering, a theoretical. framework describing the concept of 2Ca buffering and its implications for biological systems will be presented. components regulating membrane binding inside individual simultaneously, Ca 2 + integrated terminals morphologically particular several of gradient, activation, and important recognized Ca 2 even In While A regulatory the order sites change intracellular binding represent [Ca 2 j 1 components cell importance cell, and system cellular must modulation vital for 1A. contributing or to death shaping the many sequestered in maintain and and in eventually INTRANEURONAL mechanisms. molecules is intracellular events is vast all specialized sequestration tightly (Choi, each functionally. not years living in that an of majority neurons. fully to long such membrane compartment intracellular regulated 1988). be as regulate with cells, intracellular inside understood term ionized an extruded as compartments Coordination of diverse coordinated important cell and Dendrites, Neurons membrane-bound the In CALCIUM balance, [Ca 2 ] via ionic the division calcium Ca 2 release ‘extra is into Ca 2 + physico-chemical the although face endowed conductances, are signal than the interplay Ca 2 + signal of somata, Ca 2 ’ AND regulation that mechanisms, and concentration homeostasis, of very such extracellular a about for differentiation, entering perform more will huge ITS with heterogeneous is an organelles. the of axons associated now REGULATION their extremely of multiple is inward an cell control properties. known diverse [Ca 2 ] 1 through and optimized be and and ([Ca 2 +1) integration. space. response discussed. a electrochemical those presynaptic influx The of enzyme with large about polarized functions is cells, many the While of has subset Ca 2 to routes, number plasma the both injury, been and of 4 5

I. Ca INFLUX THROUGH NEURONAL MEMBRANES

2+Ca influx through neuronal membranes can take place via two major classes of 2+Ca channels. The first are the voltage-gated 2+Ca channels (VGCC) that are activated by membrane depolarization, and the second are 2Ca channels which are dependent on binding of extracellular, biologically active molecules, such as neurotransmitters, for their activation; the ligand gated 2Ca channels (LGCC). A. Voltage-gated 2Ca channels Different types of voltage-gated 2Ca channels have been identified and well characterized. These include the four well established classes of VGCC: L-,

T-, N-, and P-channels as well as the recently reported Q- and R- channels. Identification of the type of 2Ca channels is based on biophysical characteristics (gating kinetics and ionic conductance) and pharmacological criteria. Q and R types have been expressed on Xenopus oocytes using variants of 2+Ca channels a1 subunits, namely alA and doe-i, and were recently also reported in cerebellar granule cells (Zhang eta!, 1993). For the purpose of this review, only a brief description of the basic pharmacological and biophysical characteristics of the more established L-, N-, P- and T channels will be presented.

I) L-type channels: In 1980, Llinas and Sugimori, identified a high-voltage activated (HVA) 2+Ca conductance in mammalian Purkinje cells. Since then, the presence of this HVA 2+Ca current, also known as L-type current, has been confirmed in many other preparations. L-type current is characterized by slow activation with a threshold of -50 mV, half-activation at -20 mV, and a large unitary conductance (25-27 pS in 100 mM 2j.Ba With 2Ba as the 6 charge carrier, current through L-type 2+Ca channels inactivates very slowly (r > 500 ms). The channel is more permeable to 2+Ba than to 2+Ca and is blocked by 2Cd and ,2Ni being more sensitive to the former. An extracellular dihydropyridine (DHP)-binding site allows for pharmacological modulation of L-channels. While Bay K8644, a DHP agonist, evokes sustained activation of the L-channels, these channels are blocked by other DHP derivatives such as nifedipine and nimodipine in the low micromolar range, and by other organic blockers such as Verapamil and flunarizine. Some neuronal L channels, e.g. in chick dorsal root ganglion (DRG) neurons, but not muscle fiber L-channels, can also be blocked by the omega-conotoxin GVIA; an antagonist generally considered to be a selective N-channel blocker (Tsien et a!, 1988).

ii) N-type 2Ca channels are also HVA channels that have been identified only in neurons. The single channel conductance is lower than that of L channels (13-15 pS in 100mM i2Ba while their activation and inactivation kinetics are faster although both conductance and kinetics of N-channels vary widely in different preparations.. For example, the inactivation time constant is

50-80 ms in chick DRG neurons and approximately 500 ms in rat sympathetic neurons (Tsien eta!, 1988). It has been suggested that N-channels are preferentially distributed in the presynaptic terminals and are believed to be, at least in part, responsible for 2+Ca flux that triggers neurotransmitter release

(Miller, 1987; but see Luebke et a!, 1993, and Takahashi and Momiyama, 1993). N-channels are resistant to DHP antagonists but are relatively selectively blocked by the marine snail polypeptide toxin, omega-conotoxin GVIA. Like L-channels, N-channels are more permeable to 2+Ba than to 2+Ca and are blocked by 2Cd and 2Ni (Tsien eta!, 1988). 7

iii) P-channels: In addition to L- and N-types of HVA voltage-gated 2Ca channels, a third type has been initially identified in Purkinje neurons using a polyamine toxin from the funnel-web spider (FWS), Agelenopsis aperta (Llinas etal, 1992). These channels, named P-channels, show little or no voltage- dependent inactivation and are insensitive to DHP antagonists and omega conotoxin GVIA but are selectively blocked by omega-Aga-IVA fraction of FWS venom with a Kd of 2-10 nM (Mintz eta!, 1992). Single channel studies in

Purkinje cells showed the existence of 3 subconductance states: 10, 14 and 19 pS in the presence of 80 mM 2Ba + (Llinas et at, 1992). P-channels are located on neuronal somata and dendrites in many other CNS regions including the dentate gyrus, CAl and CA3 region of the hippocampus as well as in spinal and dorsal root ganglion neurons (Mintz et a!, 1992; Takahashi and Momiyama, 1993). Recent reports have also suggested that P-channels may play an important role in mediating 2+Ca fluxes responsible for neurotransmitter release in spinal, cerebellar and hippocampal neurons (Luebke et a!, 1993; Takahashi and Momiyama, 1993)

iv) T-channels are low-voltage activated (LVA) 2Ca channels with an activation threshold of about -60 mV. They exist in both neuronal and non neuronal cells and have a small single channel conductance (7-9 PS). Their most prominent feature is the transient nature of their activity, hence the name

‘T’. T-channels display a strong, and purely-voltage dependent inactivation at holding potentials positive to -50 mV. In dissociated rat hippocampal CAl neurons held at -100 mV, activation of T-channels starts at -60 mV and reaches a peak at -30 mV. T-currents inactivate with r of 25-30 ms (Takahashi etal, 1989). T-channels are equally permeant to 2+Ba and 2+Ca and are very sensitive to blockade by 2Ni though less sensitive to .2Cd Like L-channels 8

T-channels are blocked by verapamil but are more sensitive to the blocking effect of phenytoin. They are also sensitive to amiloride (Tang et a!, 1988) and medium-chain alcohols (Llinas and Yarom, 1986).

Modulation of VGCC 2+Ca channels are targets for modulation by a variety of neurotransmitters and metabolites. For example, noradrenaline and isoprenaline, acting via r-adrenergic receptors, enhance the activity of L- and N-channels. The path for activation involves cAMP-sensitive protein kinase (Gray and Johnston, 1987). Adenosine has been reported to inhibit N-channel activity (Madison et a!, 1987) while acetyicholine in rat sympathetic neurons, was found to inhibit N- , but not L- or T-type of 2Ca channels. In peripheral sensory neurons, many neurotransmitters (e.g., GABA, dopamine, serotonin), acting on specific subsets of receptors, have been found to inhibit the activity of both L- and T-type 2Ca channels. Cyclic-AMP, diacyl-glycerol (DAG), protein kinase-C as well as various G-protein subunits have been implicated in such modulation.

Aside from their dependence on the membrane voltage, the gating of VGCC can be profoundly modulated by the increase in 21[Ca The -2Ca dependent inactivation of 2+Ca channels can be either .]irreversible, resulting +activated from a Ca proteolysis of the channel molecules, or reversible as a result of2 channel dephosphorylation by a Ca-dependent phosphatase (Chad, 1989). In addition, a recent study presented2 evidence that a direct, non enzymatic, mechanism of 2Ca +.dependent inactivation of VGCC also exists (lmredy and Yue, 1994). 9

Spatial heterogeneity and voltage responses mediated by VGCC Several lines of evidence from electrophysiological studies performed at various developmental ages in cultured neurons and hippocampal slices suggest spatial heterogeneity of the distribution of VGCC. For example, neuronal somata may have a larger proportion of T-channels than L- and N- channels, while in neuronal processes the reverse is true. This spatial heterogeneity may also reflect the functional specialization of the different types of VGCC. Sustained HVA 2Ca currents may underlie the 2Ca dependent slow spikes while the T-currents are predominant in sub-threshold and pacemaker potentials (Brown eta!, 1990).

B. Ligand-gated 2Ca channels (LGCC)

In contrast to VGCC, LGCC have little or no sensitivity to membrane potential and are less selective for 2+Ca over monovalent cations. The two major types of LGCC are those coupled to the NMDA-subtype of glutamate receptor or to nicotinic acetylcholine receptors (nAchR). The former is more relevant to the present work and will be discussed in detail.

Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system. Two major classes of GluRs exist: lonotropic GluRs (iGluR) with glutamate-activated cation channels, and metabotropic receptors (mGluR) that are not linked to an ion channel but activate phospholipase-C (PL C), and possibly other G-protein mediated processes. lonotropic GluRs are further classified into NMDA-, AMPA- and Kainate-receptor subtypes according to their sensitivity to the corresponding agonist. A different class of GIuR is the AP4 receptor subtype which is a presynaptic GIuR involved in the autoregulation of glutamate release (Monaghan eta!, 1989). 10

I) NMDA receptor-associated 2Ca channels: In 1986, MacDermott et a! reported that NMDA receptor-operated channels were particularly permeable to 2+Ca compared to non-NMDA receptor-linked ionophores. Later analysis using the Goldman-Hodgkin-Katz constant field equation suggested a Ca’Na of 0.15 for AMPA/Kainate receptors compared to 10.5 for NMDA receptors (Mayer and

Westbrook, 1987). However, the fractional contribution of 2Ca + to the cation current through AMPA/KA and NMDA receptor-linked ion channels has recently been estimated to be 1.5% and 7 % of the total current, respectively

(Schneggenburger eta!, 1993). The NMDA channel conductance is relatively high (45-50 pS with a 38 pS sub-conductance state) and has a unique pharmacological profile. At physiological concentrations, extracellular 2Mg is a potent blocker of NMDA receptor-linked open channel. This open channel blockade is, however, a voltage dependent one, and at depolarized membrane potentials the channel blockade by 2Mg is relieved giving the I-V relation for NMDA receptor channels in the presence of 2+Mg a region of negative slope conductance (RNSC) prominent at membrane potentials less than -30 mV. This RNSC is absent if I-V data are recorded in the absence of extracellular 2+Mg but in both cases the reversal potential for ‘NMDA is sensitive to extracellular 2Ca reflecting the high NMDA-receptor permeability to 2Ca Another class of open channel blockers bind to the PCP-site within the .pore. These include (MK-801), and .

An interesting modulatory site on the NMDA receptor is the binding site. The strychnine-insensitive binding of glycine is an absolute requirement for NMDA receptor activation (Johnson and Ascher, 1987). At physiological concentrations of glycine, the NMDA-glycine site is saturated which raises questions about the relevance of glycine to the modulation of 11

NMDA receptor activity under physiological conditions (but see Kemp and

Leeson, 1993). The NMDA receptor is also subject to modulation by a number of other factors including 2+,Zn polyamines, p1—I,the redox state, nitric oxide, arachidonic acid and protein kinase-C with evidence for allosteric interactions between different sites, e.g., glutamate and glycine binding sites (Wroblewski and Danysz, 1989). The extensive characterization of the role of NMDA receptors in various physiological and pathological situations has been aided by the early availability of selective pharmacological antagonists, including the competitive antagonists at the glutamate/NMDA binding site such as the amino phosphonovalerate and amino-phosphonoheptanoate (APV and APH; Davies et a!, 1981). At the present time, NMDA receptor activity has been implicated in a large number of major neuronal processes from growth cone development and neuronal differentiation to activity-dependent adaptation and neuronal death. In all these situations, the role of NMDA receptors is believed to be dependent on its 2+Ca conductance. ii) Non-NMDA receptor-associated 2+Ca channels: These channels have traditionally been considered impermeant to any significant 2Ca fluxes and the increases in 2[Ca observed with non-NMDA receptor activation were considered toj be secondary to depolarization of the target cells and the consequent recruitment of VGCC. However, Murphy and Miller (1989) have demonstrated that VGCC activation may not account for all the 2[Ca in ] increases induced by activation of non-NMDA receptors various preparations.

1mb eta! (1990), using cultured hippocampal neurons also found that a subset of kainate receptors was characterized by high 2+Ca permeability and an inwardly rectifying I-V relationship. The authors labelled these as type-Il kainate receptors to distinguish them from the ‘usual’ type-I 12

channels which are not permeable to 2+Ca and have a linear I-V relationship. Recent studies on the molecular biology of mammalian glutamate receptors are beginning to identify the molecular basis of this receptor heterogeneity (Sommer and Seeburg, 1992).

iii) Metabotropic glutamate receptors: In addition to iGluR-mediated increases in 2+],[Ca metabotropic GluRs have been reported to increase 2[Ca via activation of a G-protein/PL-C pathway with subsequent release of 2Caj from 3lP sensitive stores (Sugiyama eta!, 1987). Although quisqualate is the ‘typical’ metabotropic receptor activator, NMDA has also been linked to the mobilization of 2Ca from 31P sensitive intracellular pool (Harada eta!, 1992).

II. Ca EFFLUX MECHANISMS

After a period of influx, the restoration of cellular 2+Ca homeostasis requires all the 2Ca that entered the cell be extruded. This is achieved, against a huge electrochemical gradient, at the expense of energy stores, by both primary and secondary active transport. A. 2Ca-ATPase Direct coupling between ATP and 2Ca extrusion is mediated by a uniporter pump 2(Ca +ATPase) that is a member of the plasma membrane ATPases such as Na/K ATPase and K/H ATPase which are inhibited by vanadate (Kd=2-3pM) and 3La on the cytoplasmic side. A minimum concentration of 0.2 mM 21+Mg is an absolute requirement for the function of the plasma membrane Ca-ATPase. The 2Ca pump is distinguished from other membrane ATPases2 by a higher molecular weight (140 kDa compared to 100 kDa for other pumps), and by its sensitivity to calmodulin. Calmodulin 13 binding causes a 30-fold increase in the affinity of the pump to 2[Ca and a 10-fold increase in ] (Kd=0.2pM) the Vmax. Sensitivity to calmodulin and a 1:1 2Ca:ATP stoichiometry distinguish the plasma membrane Ca-ATPase from organellar membrane 2+Ca pumps which are insensitive to calmodulin2 and have a 2:1 2Ca:ATP ratio (Carafoli, 1987). Besides calmodulin, a multitude of factors contribute to the regulation of plasma membrane 2Ca pump activity. Phosphorylation of the pump by cAMP-dependent protein kinase results in an increased affinity for 2Ca while phosphorylation by protein kinase-C increases its Vmax (James et a!, 1989; Smallwood eta!, 1987). Poly-unsaturated long chain fatty acids as well as membrane phospholipids of the inositol phosphate pathway: phosphatidyl inositol and its mono- and di-phosphate derivatives (P1, PIP and )2PIP promote Ca-ATPase activity unlike the 2PIP hydrolysis products, 31P and DAG, suggesting2 a functional linkage between Ca-ATPase activity and inositol phosphate pathway activity (Penniston, 1982).2

The sensitivity of membrane 2Ca +.ATPase to calmodulin links an increase in ]2[Ca to 2Ca pump activation. However, this process is probably too slow to contribute significantly to the recovery of fast 2+Ca transients and, in effect, pump activity may reflect the time-averaged 2Ca level in the cytoplasm rather than the instantaneous 2Ca level. Also the small capacity of the pump and its high affinity for 2Ca makes it more suitable for the fine tuning of resting intracellular 2+][Ca rather than the fast recovery required after large 2Ca loads which are probably handled better by the 2Na/Ca exchanger. B. 2+Na+/Ca exchanger The 2NaICa exchanger is a low-affinity high-capacity bi-directional

types

analogues

exchanger

displaced

from

and

Allen the

and

hundred

phosphorylation

analogues

Ca 2 ,

that

influx),

and

process

potential

the

[Ca 2 the

antiporter

electrochemical

cannot

chelation

activity

electrochemical

DiPolo,

Little

this

the

In

+1,

While

and

of

reducing

reverse

[ATP]

antiporters

nanomolars

is

transported

[Na+]

is

in

stems

Baker,

of

selective

although

and

system

3:1

not

be

symmetric

the

and

1984).

the

of

was

displaced

other

(Na+:Ca 2 j,

the

and

mode

Na 0 :Na

the

from

of

Ca 2 1

the

latter

1985).

that

the

case.

found

pharmacological

(Taglialatela

gradient

membrane

La 3 +

Kd

operating

gradient

of

Ca 2

nucleotide

the

(i.e.,

show

operation

exchanger

with

from

couples

‘regulatory’

by

This

fact

and

to

In

Ca 2 +

binding

operating

high

EGTA

enhance forward

is

selectivity

30

i.e.

of

regulatory

that

amiloride

mode

highly

potential.

ionic

pM

phosphates

Ca 2 +

etal,

the

was

Na

influx

may

the

abolishes

site.

[Ca 2 j

to

control

exchanger

(forward/reverse)

operating

movements

initially

favorable

significantly

mode.

(100

1990).

and

0.75 stoichiometry

be

for

and

can

Ca 2

Regulatory

The

required

Na+.

Na+/Ca 2 +

mM)

pM.

Na+

is

cannot

block

is

the

assumed,

dependence

required

available

site

mode

activity

(DiPolo

while

reverse

efflux),

Three

in

Non-hydrolysable

its

the

is (DiPolo

replace

a

Ca 2

of

activity

intracellular

(i.e.,

direction

exchanger

exchanger

transported

of

for

and

later

is

the

over

major

mode

a

the

electrogenic

is

exchanger

Ca 2 +

minimum

on

and

ATP

exchanger

Beauge,

evidence

not

Na+ICa 2 +

and

exchanger:

factors

membrane

activity

determined

Beauge,

indicating

transported

efflux

affinity

over

some

and

Ca 2

ATP

of

1993;

function,

determine

distinct

indicated

other transport

(Baker

even

several

and

1993).

for

can

that

by

Na+

if

be 14 15 In spite of its high capacity, the low affinity of the exchanger for 2Ca (0.75 pM) means it cannot contribute to the regulation of resting intracellular 2+Ca levels but its role in recovery after large intracellular 2+Ca loads is widely accepted. Also, there is no evidence for its operation in the reverse mode in vivo although an interesting model has been presented by Blaustein (1988a) suggesting that during an action potential, changes in membrane potential and [Na+] could result in a biphasic operation of the exchanger promoting 2Ca influx at the beginning and 2Ca extrusion during later stages of the electrical response. The net result would be to enhance the 2+Ca signal with a faster recovery of 2+].[Ca However, this interesting model has not yet received experimental support (DiPolo and Beauge, 1993).

Mill (1993), using Heilsoma neurons in vitro, reported a large variability between different neuronal types in their capacity to recover from ‘equal’ 2+Ca loads induced by exposure to the 2Ca ionophore, A23187. The author attributed this variability to the differences in the 2Na/Ca exchanger activity in those neurons and demonstrated that interneuronal variability was markedly reduced, though not completely abolished, by removing Na from the extracellular environment. The author suggested that different neuronal 2+Ca set points, also observed by Guthrie eta? (1988) during 2Ca responses, may be attributed to different exchanger activity in different neurons.

III. INTRACELLULAR 2Ca SEQUESTRATION

A. Mitochondria

Mitochondrial respiratory chain enzyme activity is associated with pumping of H from inside the mitochondrial matrix into the cytosol thus generating a high trans-mitochondrial potential gradient of 150-1 80 mV with

effect

released of

calcium/phosphate slightly

McCormack, capacity

with

[Ca 2 jm certain

cytosolic

their

between

intramitochondrial

exchanger

generation

potential

proportion

compartment

cause

in

the

1966)

1969).

Ca 2 +

the

inside

the

response

of

Once

transport

reverse

this

level

higher.

into

According

at

is

Ca 2

the

(i.e.,

the

energy

[Ca 2 j

of

saturated

that

to

to

[Ca 2

high

inside

of

mitochondria.

the

1985

properties

the

the

the

using

net

mode

to

influx

[Ca 2 +] 1 ,

has

Once

+1

electrical

of

cytosol

across

to

mitochondrion

[Ca 2 +] 1

third

accumulation

trans-mitochondrial

ICa 2 ]

the

and

complex

Ca 2 +

above

to

a

be

an

to

and

resulting

sigmoidal

cytosolic

the

mitochondria,

accompanied

power

uniporter

1990;

generate

of

the

known

as

Ca 2 +

from

and

and

the

chemiosmotic

the

(lCa 2 jm)

the

Excess

mitochondrial

whose

set

[Ca 2 jm

of

McCormack

chemical

mitochondrial

in

the

Ca 2

as

other

accumulates

response of

being

[Ca 2 ].

the

Ca 2

ATP.

point

the

Ca 2 +.

cytosol

nature

mitochondrial

dynamic

by

+.ATPase

Ca 2 +

and

Ca 2 +

exceeds

flux

set-point,

negative

respectively,

The

can

gradient

larger

model

activity

a

membrane

Under

is

equals

to

and

can

saturable

be

same

regulatory

uniporter

not

inside

the

equilibrium

changes

the

considered

Denton,

of

whose

be

(Mitchell

exchanger

drives

yet

extreme

intramitochondrial

curve

zero)

calcium

driving

ATP

set

extruded

the

causes

clear

can

capacity.

point,

and

mechanisms

activity

generation

mitochondria

although

a

in

in

1990).

and

membrane

force,

between

be

conditions,

[Ca 2 ]m.

can

(Denton

relation

as

the

activity

changes via

the

diverted

Moyle,

a

be

exchanger

increases

Ca 2 +

however,

a

The

The

exchanger

the

slowly

Na+/Ca 2 +

(Mitchell,

to

and

[Ca 2 ]

restore

can

H-ATPase

differences

net

Up

[Ca 2 ]m

as

1967

in

from

buffering all

a

cope

to

influx

the

in

in

can

a

and

ATP

and

is 16 17 2[Ca to below the set point, thus causing prolongation of the 2Ca signal andi a delay in the recovery process.

Although the set point has not been firmly established in vivo, evidence obtained from cultured neurons suggest that it is in the range of 0.75 iiM of cytosolic 2Ca corresponding to approximately 1 pM 2]m.[Ca Thayer and Miller (1990), demonstrated that the mitochondrial uncoupler CCCP caused a marked increase in 2[Ca during the plateau phase of 21[Ca observed during a 2-mm K-induced j depolarization indicating that the plateauj level was set by mitochondria and probably corresponded to the mitochondrial set-point. In

Na+free medium, the set point was reduced in accordance with the prediction that the relative activities of 2Na/Ca exchanger and Ca-ATPase determined the mitochondrial set point. The increase in22[Ca+]m may serve a metabolic purpose since at least three major mitochondrial enzymes are regulated by 2Ca with a peak activation at 1 pM 2im.[Ca These enzymes are pyruvate dehydrogenase, oxoglutarate dehydrogenase and NAD-isocitrate dehydrogenase. Thus, the increase in 2[Ca+]m, within limits, is associated with increased production of ATP. With the slow mitochondrial transport systems, compared to the 2+Ca signal, the activation of mitochondrial metabolism follows a time-averaged 2+Ca signal and is independent of changes in ADP/ATP and NAD/NADH which are the major stimuli for mitochondrial metabolism.

Because of the low-affinity of the mitochondrial 2Ca buffering system, the physiological significance of this buffering has been debated. Levels of 2[Ca that are necessary for mitochondria to function as a 2+Ca buffer, i.e., 2i+][Ca above 1 pM may not be reached in neuronal somata under normal conditions. However, in presynaptic terminals, smaller dendrites and dendritic

to

and share

as thapsigargin-sensitive

the

mitochondria

as

found

have

mitochondrial

detected.

and

1980,

B.

neurotransmitter

that

work

sufficient

into

and

spines,

play

ryanodine

methyixanthine-sensitive

in

two

Endoplasmic

dantrolene

ruthenium

the

The

a

mitochondrial

a

dendrites.

In

been

many

of

in

McGraw

prolonged

role

many

systems

evidence

response

submembraneous,

Nishimura

ER

numbers

These

associated

in

characteristics

associated

and

suggesting

tissues,

the

stores

red-resistant

(Palade,

et

In

Reticulum

their

(McGraw

maximal

release

non-mitochondrial

for

modification

al, (i.e.,

Ca 2

many

(1986)

and

discussed

such

1980).

but

anatomically

cytoplasmic

Ca 2

transient

Ca 2 +

in

1987).

the

and

cases buffering

resistant

response

close

high

and

eta!,

with

perinuclear

release

Ca 2 +

release

presence

possibly

They

stores

of

Biscoe

above

they

Ca 2 +

The

sarcoplasmic

proximity

1980).

very

the

sequestration

high-affinity

may

to

with

have

and

provided

stores

can

ER

exist

are

temporal

levels

mitochondrial

intense

(Brinley,

be

and

of

and

stores contribute

the

the

be

A23187

a

some

involved

to

in

Duchen

diffuse

have

triggered

synaptic

inhibition

suggests

endoplasmic

close

the

that

reticulum

supra-maximal

can

relationship

1980;

functional

(0.1

a

mechanisms

source

releasable

mitochondria

subcellular

much

in

association

to

also

(1990)

pM

uncouplers.

terminal synaptic

by

the

of Verma

that

be

(SR)

or

of

release

high

lower

regulation

reticulum

relationship

mitochondria

led

emptied

less), Ca 2 +

between

and

Ca 2 +

et

stimulus

[Ca 2 ] 1 ,

regions,

distribution

have

plasticity.

to

with

capacity

a!,

are

by

vanadate-

ATP-dependent

the

entry).

ER

stores

1990)

been

procaine

present

(Henkart

by

of

the

stores

suggestion

translated

between

as

i.e.

exposure

than

could

stimulus

The

well

being

and

such

and

also

in

the 18 19 Ca-induced 2Ca release (McBurney and Neering, 1987). Mapping the distribution2 of ER stores using ryanodine binding proved that many peripheral and central neurons show some degree of ryanodine binding but it is exceptionally high in Purkinje cells (Ellisman eta!, 1990).

As high-affinity storage sites with specific release mechanisms, these stores can play the double role of a 2+Ca buffer and a 2+Ca signal generator depending on their state of filling. For example, their role as possible 2Ca buffers has been illustrated by the ability of caffeine treatment to increase the 2+Ca buffering capacity for subsequent 2+Ca loads in sympathetic neurons (Friel and Tsien 1990). C. 31P-sensitive intracellular 2+Ca release In addition to the caffeine-releasable intracellular 2Ca lP-sensitive intracellular 2+Ca stores have been identified in many ,cell types.3 These stores have occasionally been named calciosomes (Satoh et a!, 1990; Hashimoto et a!, 1988) and they share many characteristics with other ER associated 2Ca stores such as the presence. of calsequestrin and calreticulin as well as a vanadate-sensitive 2Ca +...ATPaSe and they, too, exist in high abundance in cerebellar Purkinje cells.

Although 31P receptors are structurally similar to ryanodine receptors (Tsien and Tsien, 1990), 1P-releasable 2+Ca stores seem to be anatomically and functionally distinct from3 the caffeine releasable stores since lP-induced release could 3 2+Ca occur after caffeine stores were depleted and they have distinct subcellular distributions (Thayer eta!, 1988). Release of 2Ca from 1P-sensitive stores normally follows generation of 31P subsequent to activation of3 phospholipase-C, (PL-C), a G-protein mediated process that has been linked allocation to mechanisms (affinity, consistent V. 1990). The Ca 2 studies, latter modulator, IV. Ca 2 + central mechanisms, 1990). activity. alpha-adrenergic to mediate many INTRACELLULAR most CALCIUM-BINDING Although three The buffers. signal A theme there large kinetics, neurotransmitter in relevant large with inside and proteins the will exemplified their is number in They lP 3 - the calbindin-D28k, various number little depend many the receptor functional of cooperation and large but are cell. ROLE these evidence of models caffeine-sensitive subcellular they PROTEINS of discussed not at Ca 2 number activation receptor Functional mechanisms OF an to capacity, only +.binding have our extreme attempting to Ca 2 (in parvalbumin of on support discussion a been in domains activation diverse positive their and detail integration etc.) by proteins involved considered metabotropic pools an inherent to intracellular but later. vital (for and feed-back explain enzyme-modulating are such also are have example functions in calmodulin, of calretinin. biophysical as the by regulated such critically spatial been muscarinic Ca 2 + many glutamate manner) regulation regulatory see [Ca 2 ] identified heterogeneity In waves on to a Lederer by properties spite known be their has different role acetyicholine, receptor of is ‘passive’ (Berridge, of been [Ca 2 +] 1 in believed spatial for et enzyme intensive neurons. a!, the a of is the 20 21 In spite of the diversity in the known functions of 21Ca the major roles of 2+Ca can be summarized as follows: ,

1. Charge carrier with direct effects on membrane potential.

2. Ion channel modulator where 2Ca can directly modify the gating of other channels (e.g., 2+Ca dependent K+conductance, 2Ca +dependent Cr-conductance or 2Ca + release activated 2+Ca conductance).

3. Enzyme, protein and gene activity modulator: e.g., phospholipases, protein kinases, genes (c-fos), proteases and endonucleases.

VI. RECAPITULATION

Increases in neuronal [Ca can be a consequence of activation of a 2+] (1) variety of calcium influx mechanisms such as VGCC, LGCC, and 2Ca +.release activated calcium channels or (2) release of intracellular calcium stores by depolarization, 3PL-C/1P pathway activation and calcium-induced calcium release. Restoration of calcium homeostasis requires the extrusion of ‘excess’ calcium across the cell membrane and the replenishment of intracellular stores. This energy-consuming restoration process must of course be appropriately adjusted by the cell so that the purpose of the calcium signal is realized, with adequate specificity/selectivity, otherwise futile calcium cycling would eventually drain the cellular energy resources. In spatially heterogeneous neurons, and possibly in most other cells, high spatial (nanometer) and temporal (sub-millisecond) resolution of the calcium signal require functional clustering of the signal source (influx and/or release), effector machinery (enzymes, genetic material, presynaptic vesicles, etc) and the recovery mechanisms (efflux/sequestration/buffering).

visualizing

regulatory are

the

as

least

mitochondrial

physiologically

of

ATPase

excess

following

then

of

dissipation

binding

Blaustein

small mitochondria

approximately

an

low-affinity

ensue

in

General

oversimplification.

Considering

calcium

restricted

proteins

can

dendrites)

0.1

(1988)

mechanisms

the

of

during

fine-tune

to

Ca 2

the

models

potential

relevant

mitochondrial

restoring

in

1

(e.g.,

has

1

diffusional

calcium

the

s,

the

where

only

ms.

uptake

the

suggested

the

of

following

restoration

parvalbumin)

under

significance

stimulation

the

Sequestration

the

Na/Ca 2

the

last

localized

However,

signal

capacity

affinities

spaces

‘natural

[Ca 2 +1 1

certain

uptake

stages

tens

that

away

of

[Ca 2 +]j

is

and

with

such

might

exchanger

has

Ca 2 +

history’

system.

and

diffusional

physiological

to

of

of

controversial.

from

into

near

the

milliseconds.

relative

made

large

the

models

be

homeostasis

can

ER-associated

recovery

the

the

of

binding

the

surface

such

may

be

a

calcium

fluxes

contribution

resting

can

fastest

calcium

or

high

a

extrude

However,

reaction

process.

pathological

be

role

Finally,

and

to

enough

following

signal

level

conceptually

mechanisms

volume

calcium

signal,

a

buffering

viable

the

of

where

constants,

over

source

the

The

to

various

bulk

of

ratios

conditions.

stores

make

large

possibility

the

involvement

necessity

Ca 2 +

by

of

with

of

helpful

Ca 2

(such

calcium

the

use

might a

+

of

r

at

in 22 23

1B. EXCITOTOXICITY

I. HISTORY AND DEFINITION

The earliest report that the acidic amino acid glutamate (GLU) was toxic to neurons was presented by Lucas and Newhouse in 1957. In their search for compounds that may be useful in the amelioration of retinal dystrophy they discovered that systemic GLU administration was associated with neuronal degeneration in their experimental preparation, the immature mouse retina. A series of pioneer studies by Olney and his colleagues (Olney, 1969; Olney and Sharpe, 1969; Olney eta!, 1971; Olney eta?, 1974; Olney, 1978) confirmed and established a phenomenon that was named excitotoxicity.

Excitotoxicity in principle links the excitatory properties of a compound to its neurotoxic sequels. The relationship between agonist-induced over- stimulation and cell injury is not limited to neurons or to glutamatergic synapses. For example, excessive adrenergic stimulation can cause injury of cardiac muscle fibers and the role of cholinergic agonists in the induction of muscle dystrophy has been well established (Bloom and Davies, 1972; Leonard and Salpeter, 1979).

II. EVIDENCE FOR GLUTAMATE INVOLVEMENT

A. In vivo

Glutamate is the major excitatory neurotransmitter in the central nervous system (Curtis and Johnston, 1974). In addition, it has a metabolic role and serves as an intracellular anion with an average tissue concentration of approximately 1 mM while in glutamatergic nerve endings tissue GLU levels

the after

number

Olney

substantiated

treatment.

neuroprotection

using

hypoglycemic

ligation

detected

phosphonoheptanoate

specific The

indicates

not

However,

powerful

degeneration,

variety

other

evidence,

may

ubiquitous

NMDA

removed

selectivity

the

Injection

Direct

be

different

et

endogenous

of

(Simon

of

types

as

at,

ischemic

in

that

uptake

direct CNS

specific

both

receptor

amino

high

The

CAl

injection

1989).

by

by

injury of neurodegeneration

of

even

hypoxic/ischemic

of

eta!,

diseases.

in

neuroprotective

was

as

such

injection

pyramidal the

mechanisms

a

GLU

the

acid

insult

vivo

competitive

large

excitatory

10

subtype

at

The

of

specific

evident

of

1984).

neuronal

uptake

receptors

high

mM.

(APH)

such

the

and

GLU

was

majority number

of

rat

neurons

in

concentration,

as

in

GLU

Although

into

even

NMDA

delivered

in

mechanisms

In

amino

vitro,

both

caudate

the

and

populations

glutamate

the

1985,

(GluRs).

properties

of

animal

the

of

is

analogues,

when

pathogenesis

in

experiments

non-competitive

hippocampus

a

in supports

these

acids,

antagonist,

nervous

response

neurons

rats

it

Wieloch

(McDonald

could

models.

may

the

can

studies

causes

most

exposed

are

of

damaged

antagonists

e.g.

also

the seem

system

GluR

be

and

involved

to

demonstrated

in

likely

2-amino-7-

of

In hypothesis

neurotoxic,

reduced

be widespread

concentrated the

et

kainate

different

glia

antagonists

counter-intuitive

neuronal

some

to

antagonists

a!,

ameliorated

by

fails

excessive

due

transient

(Schousboe,

in

such

1

were

987;

cases,

the

or

cell

to

to

animal

that

injury

NMDA,

induce

the

accumulating

procedures

that

neuronal

pathogenesis

damage

administered

Gill

have

on

carotid

stimulation

have

presence

by

GLU

et

since

the

species

that

1981).

neuronal

APH

been

that

at,

been

and/or

role

normally

damage.

artery

1

a

a

also

are

988;

large

of

and

of

of

of

24 a

suffered

patterns

pM

1983;

induced

hypoglycemic excitatory

high

concentration

ischemia concentrations

the

response

prerequisite

useful

following

that

AMPA

nitro-7-sulfamoyl-benzo(F)quinoxaline

pharmacological

(Honore

available

(Benveniste

use

These

enough

non-NMDA

Deafferentiation

Petito

in

receptor

of

neuronal

from

of

eta!,

it

demonstrating

to

for

ischemic

amino

in

neuronal

has

and

for

ischemic

etal,

to

vivo

this

cerebral

neuronal

increases

1988),

of

been

kill

other

etal, the

blocker

receptors

acids

tools

death.

receptor

amino

microdialysis

1987).

most

insults

induction

loss

shown

experiments

1984; insults

experiments

particularly

ischemia

(EAAs) for

damage

from

that

that

hippocampal

acids

observed

Experimental

the

(Sheardown subtype

may

that

Hagberg

intact

(Johansen

crosses

less

of

manipulation

in

in

for

also

and

and

neuronal

the

after

the

than the

have

of

in

direct

excitatory

since

status

in

mediate

pathogenesis

hippocampus

the

post-mortem

etal,

glutamatergic

neurons

recent

10

(NBQX),

the

results

et

5

et

established

pM

the

blood

measurement

damage

mm

a!,

a!,

generation

epilepticus

1985).

of

some

development

early

to

1990).

1986;

synaptic

also

of

in

non-NMDA

brain

which

30

ischemia

vitro.

in

of

bear

brains 1980’s.

during of

pM

Such

pathways

the

Onodera

the

barrier,

ischemic/hypoxic

the

of

is

(Corsellis

transmission

and

of

many

role

epilepsy-

a

hippocampus

concentrations

of

neuronal

induction

the

receptor

extracellular

of

potent

after

The

of

patients

has

2,3-dihydroxy-6-

et

similarities

GLU

have

endogenous

and

a!,

use

30

established

and

and

degeneration

subtypes

1986).

also

of

mm

is

Bruton,

of

who

selective

trauma

in

a

cerebral

new

and

been

to

are

to

500

By

the 25 26

Several characteristic features of excitotoxic lesions are evident from the accumulated experimental findings. First, excitotoxicity was found to be a primarily dendrosomatic lesion that spared axons traversing the site of the lesion. Second, the selective vulnerability of certain neuronal populations determined the final outcome of an excitotoxic lesion. This vulnerability was, amongst other factors, based on the receptor profile and the density of the synaptic inputs. Third, the induction of excitotoxic damage in vivo did not end with the termination of the primary insult (e.g., transient ischemia) possibly because excitotoxicity is, to some extent, a self-propagating process. This aspect may be of crucial value in the design of therapeutic approaches since post-insult therapeutic measures may still be capable of limiting the extent of the lesion. In other words, the therapeutic window may extend beyond the duration of the initial insult (Albers eta!, 1989).

B. In vitro Primary neuronal cultures from different brain regions (e.g., neocortex, hippocampus, striatum, spinal cord) have been used extensively to study the physiology, pharmacology and pathology of EAA in vitro. These studies have also suggested that EAA can induce neuronal injury in a concentration dependent manner, and that EAA-induced neurotoxicity can be specifically attenuated by GIuR antagonists. In addition, many elaborate in vitro models have been devised to ‘simulate’ specific in vivo situations like ischemia (Goldberg and Chol, 1993), hypoxia (Dubinsky and Rothman, 1991) and even mechanical trauma (Tecoma et a!, 1989). Though elaborate in vitro experiments cannot reproduce the complexity of the clinical phenomena, the fact remains that in the majority of such experiments, performed under diverse experimental conditions, EAAs were found to contribute to the neuronal injury and EAA antagonists were capable of alleviating, at least in part, the damage.

swelling,

distension

neurons

neuronal

phenomena.

the

during

delayed

neuronal

associated

suggesting

passive

this

could

(Rothman,

within

prolonged

is

experimental

isolation.

IONIC

III.

associated

Ca 2

swelling

PHASES

Neuronal

Removal

A

be

the

minutes

DEPENDENCE

major

death

influx

with

swelling

yet

degeneration

prevented

ionophore,

can

exposure

A

depolarization

with

1985;

that

delayed

brief

was

For

NMDA

OF

environment

with

advantage

of

of

swelling

is

itself

of

the

the

entirely

GLU

cr

GLUTAMATE-INDUCED

example,

without

Choi,

dependent

exposure

the

the

by

swelling

reversal

be

to

and

in

A23187

neuronal

or

exposure.

and

early

the

glutamate

removal

and

lethal,

1987b).

other

dependent H 2 0

of

of

the

so

a

absence

subsequent

delayed

in

of

neurons

development

of

was

brief

on

(Rothman,

that

delayed

the

(Rothman,

vitro

death

excitants

cultured

the

of

the

This

Ionic

and induced

relevance

exposure

these

relevant

swelling,

experiments

of cell on

presence

by

still

can

swelling

neuronal

Na+ 0

substitution

the

death

neurons

exposure

death

from

ions

1985).

occurred.

1985;

NEUROTOXICITY:

of

by

be

presence

parameters

to

of

neuronal

but

and

the

from

mimicked

over

the

of

appear

kainate

this

can

loss,

Choi,

to

is

extracellular

is

Cr 0

influx

bathing to

a

the

the

then

high

experiments

phase

also

Although

veratridine

period

of

while

prevented

to

swelling

1987b).

was

culture

ability

can

of

extracellular

by

concentrations

followed

be

be

medium

of

Na

exposing

treatment

associated

produced

of

be

independent

TIME

excitotoxicity

to

Na+

severe

medium

hours.

evaluated

that

followed

or

control

indicated

neuronal

by

COURSE

is

high

and

appears

Ca 2

neurons

a

usually

neuronal

of

by

This

with

delayed

cr

[K+1 0

of the

+

by

in

that

GLU

and

to

AND

the

to 27 operating dependent was However, to Tymianski death mechanism(s) development comparable brief Ca 2 development experiment the Ca 2 intracellular intense presence magnitude permeability and extracellular the the accumulation EAA-mediated prolonged glia Unlike exposure A in influx channel port unique exposure cultured in in of their eta!, on of vivo (Choi, demonstrated NMDA to the accumulation is the to space of of Ca 2 the by important. blockade an to feature (Koh excitotoxic Ca 2 delayed reverse delayed may 1993). radioactive cortical which AMPA, of of Ca 2 NMDA-induced 1992). agonists, in and entry eta!, neurons 45 Ca not vivo (MacDermott of the mode Ca 2 influx was The neurodegeneration excitotoxic allow neurons kainate the into not of and 1990). Such neuronal potency extensive tracer non-NMDA 45 Ca. latter in not NMDA only through (Choi, induces the the such vitro a effective or increase conclusion if This neuron. delayed 45 Ca group that increased eta!, injury the Specific high neuronal swelling receptor-associated to 1991). structural cell VGCC slowly GLU Ca 2 + duration in agonists [Ki 0 of 1986). is (Kurth the in death neuronal (Michaels authors doubtful, NMDA or [Ca 2 +] to is damage, or markedly influx triggered extracellular could NMDA also take support via of may eta!, are When the antagonists Na/Ca 2 death is also supported place without incapable raise and be since essential but resulted 1989; exposure when excitotoxicity performed by speculated ionophore spatially Rothman, while also or [Ca 2 1 1 medium, adjacent the the severely the Choi, by that in exchanger of for limited abolished voltage-gated is subsequent exposure the tightly the inducing to in the brief. is 1991). the that brief 1990; neurons the levels its may fact limit route linked high the and both that be time its This of 28

generating

dehydrogenase

increases

feature

perpetuation

cytoskeletal

proteases,

dependent

initial

early

responses

kinase-C,

GLU

are

and

‘positive

receptors,

of

changes

exposure

concentrations

expression.

neurotoxicity

IV.

the

triggered

diacylglycerol.

A

The

and

gene

Choi

injury

major

THREE-STAGE

of

feedback’

take

other

expression

in

calmodulin

this

determines

to

free

lipases

(1990)

expression. and

cytotoxic

is

[Ca 2 +]j

elements,

of

GluR

The

to

ambient

in

place.

not

stage

neurotransmitters,

include

oxygen

into

the

of

promote

which

induction

and

severe.

presented

subtypes: GLU

damage.

amplification

xanthine

The

which

is

phase

mechanisms

sensitive

These

the

GLU.

MODEL

endonucleases

plasma

the

accumulation

there

radicals

or

The

result

further

speed

other

involvement

result

proceeds

stage

amplification

changes

The

a

NMDA,

oxidase

were

These

membrane

OF

model

kinases,

of

(FOR).

endogenous

with

increases

effector

phase

in

such

is

and

GLUTAMATE

and

three

a

a

oxidative

AMPA/Kainate

of

with

which

reflect

for

(Olanow,

calpain-mediated

direct

are

the

even

an

High phospholipases,

of where

intracellular

stages:

the

integrity limbs

phase

involved

subsequent

generalized

accumulation

oxidative

in

a

enhance

the result

[Ca 2 ] 1

evolution

EAAs.

[Ca 2

mechanisms

set

several

1993) of

may

effects

EXCITOTOXICITY

induction,

of

+1

these

and

of

in

Ca 2 +,

mechanisms

and

primary

The

also

be

exposure

the

this

enhance

that

activation

Ca 2 +

irreversible

of

genomic

conversion

of

calpains is

more

changes

metabotropic

intensity

postsynaptic

GLU-mediated

activates

phase.

the

excessive

is

are

Na+,

amplification

sensitive

intracellular

capable

important

initiation

activated

to

the

DNA.

of

Cr,

and

in

include

A

high

damage

of

of

(CHOI,

release

all

the

major

activation

water,

this

of

xanthine

immediate

pathways

Ca 2 +

Many

of

if

and

by

protein

the

a 1990)

to

of

1P 3 29 the antagonists failure, infarcts, the that (Wieloch of 1985). receptor white cytotoxic example, major V. 1988). structural damage the dismutase, of activate membrane peroxidation phospholipase white hydroxyl insult release LIMITATIONS capacity can The matter limitations and Furthermore, White NMDA by matter be profile

nitric eta!, the in ‘pure’ mechanisms protein of lipids perpetuated peroxidases, to propagating initiated the radicals. of of with excitotoxic low-molecular-weight be matter, oxide 1988; A 2 that antagonists can cellular core the excitotoxic which (Chan effective and OF substantial which brain be is during (NO) of in THE Buchan Excessive DNA being a such explained anti-oxidant the

require eta!, models during the prerequisite ascorbic hypothesis liberates in synthase EXCITOTOXIC or ischemia, ischemic damage hypothesis have an lipid as formed and it 1985) FOR the ischemic/hypoxic the an of may production peroxidation, by not acid, Pulsinelli, the global expansion production. reperfusion oxidative which, defences iron (Olanow, of and the cannot be for lesion secondary been cytotoxic oligodendroglia of Vitamin that a oxidative complexes initiates ischemia HYPOTHESIS GLU-induced GLU-mediated unequivocally in may of explain 1990). vascular stress of excess, (glutathione-SH, 1993). the enzyme phase Increased E, the to arachidonic be lesion a stress and etc) highly chain loss too discussed the hypothesis associated However, factors (Halliwell promotes focal and promotes inactivation severe neuronal (Auer of damage and injury. reaction reactive neuroprotective [Ca 2 j Ca 2 + axons, ischemia acid and lipid and above. it for superoxide with to and the observed is This regulation damage neuronal from acidosis of FOR peroxidation can Siesjo, include the lacks possible and Gutteridge, production energy lipid damage with NMDA For also beyond major the has other in are or that the 30

general

development

unique

central

VI.

determining

HYPOTHESES

under

of

Excitotoxicity

again

reperfusion.

as

have

models,

why

thus

to

energy

focal

the

Chol

oxygen,

well

acidosis

Ca 2

main

It

Last,

hippocampal

Another rendering

a

(1991)

ischemic

contribute

such

can

for

role

high

mediator

as

depletion)

while

determinants

AS

neurons

but

the

be

even

conditions.

in

(or

the

density

of has

per

THE

the

limitation

concluded

functional

dentate

not

even

injury,

NMDA

neuronal

relative

to

if

of

suggested

se

formulation

may

CAl

least,

they

COMMON

since

the

cell

of

to

is

down

antagonists

granule

NMDA

reduce

not

dephosphorylation

of

pattern

neurons

injury

of

have

vulnerability

Rather,

all

an

capacity

injury

from

neuronal

the

enough

mammalian

that

increase

modulation

no

FINAL

in

of

receptors

excitotoxicity

the

the

cells

of

and

excitotoxicity

are

other

GluRs

the

in

cell

of

discussion

contribution

less

to

global

loss

death

are

selectively

PATHWAY

excitotoxicity

of

cell-specific

in

explain

damaged

cell

or

protective.

neurons

neurons

[Ca 2 i

spared

in

of

(Cotman

if

ischemia,

in

types. of

such

NMDA

GluRs

general.

hypothesis

NMDA

above

the

may

damaged

in of

is

in

extreme

AND

to

will

recovery

eta!,

spite

mechanisms

NMDA

The

widely

receptors

are the

hypothesis

various

be

or

that

eventually

receptors

ALTERNATIVE

This

a

effectively

post-ischemic

hypothesis

in

of

1987).

contributing

is

situations.

in

Ca 2 +

the

believed receptors

the

its

mechanisms,

situation

insults.

global

activity

infarct

inability

fact

and

secondary

of

occupies

die

Vascular

blocked.

cell

that

ischemia

to

that

in

if

to

is

zone

in

factor Indeed,

phase

be

deprived

fact

death

to

injury,

not

an

response

both

a

could

explain

a

factors

to

of

in

of

in

the 31 32 increase in 2[Ca +1 is ‘the final common pathway’ to cell death was presented by Schanne et al in 1979 and has generally been accepted by many researchers.

In a recent review, Dubinsky (1993b) presented a compilation of the experimental findings that could not be reconciled with the generalized 2Ca hypothesis. For example, Michaels and Rothman (1990) reported that a GLU induced rise in 2[Ca could be prevented using a combination of CNQX and APV while neuronalj loss was only attenuated. In addition, a chemical hypoxia model, using iodoacetate and rotenone to inhibit the glycolytic pathway and the mitochondrial respiratory chain reaction, was lethal to neurons and yet removal of extracellular 2Ca while preventing the rise in 2],[Ca did not alter the fate of energy depleted, neurons (Nedergaard, 1991).

Marked increases in 2+][Ca have also been observed in situations that did not end in cell loss. For example, prolonged depolarization of neurons using high extracellular K is not neurotoxic (Dubinsky and Rothman, 1991; + Tymianski et al, 1993a) even though the 2Ca responses are comparable to those induced by lethal concentrations ot GLU or NMDA. Also, in peripheral neurons deprivation of neurons of nerve growth factor (NGF) results in apoptotic cell death that appeares to be completely dissociated from any measurable increase in j21[Ca (Eichler etal, 1992). While the involvement of 2[Ca in the initiation of many cytotoxic biochemical pathways cannot bej denied, the exact relationship between the increases in 21[Ca and cell injury cannot be established with certainty. Cellular derangementj of any kind is likely to be associated with the loss of 2Ca homeostasis since the latter is tightly regulated against a large trans such Garthwaite, EAA Alzheimer’s’ induced neurodegenerative cerebral lesions VII. early and pathways still measurements death. injury cell oxygen This other exact an membrane increase limited. spatial injury metabolism. CLINICAL A as Technical is events mechanisms add sequence potential has not The sulfite neurotoxicity radicals ischemia/hypoxia, need as to been resolution unique electro-chemical 1991). most in disease leading Ideally, the much oxidase [Ca 2 +] IMPLICATIONS but, limitations to and role of discussed problem advanced Sufficient be disorders to of events as at Some to multiparameter and for severe Ca 2 + used is which neuronal the they deficiency and neuronal a excitotoxic amyotrophic of factor on present hereditary leading hypoglycemic, for above. an are intracellular of since evidence gradient. such do establishing the increase extended these injury symptoms not death. in measurement are as the to time, the Many interfere neuronal measuring techniques Huntington’s cell neurological such also same also lateral This pathogenesis in their acidosis periods the death studies [Ca 2 i epileptic believed of exists as two-way argument with sclerosis sequence this injury spatial excessive techniques of extremely of are which to injury. also disorders intracellular causing critical disease, and time to in establish fluorimetric and of relationship, the be could suggest (Meldrum of traumatic chronic could production in temporal mediated parameters events pathogenesis difficult injury, with order Parkinson’s of be a be signaling metabolic that causal made high Ca 2 + and to causative leading makes neuronal injury to resolution by discern GLU of temporal establish. of about disorderly free disease, of cell causing the origin to in the cell are 33

enhancement well

processes Schwartzkroin,

enhancing

use

function.

while

for

are,

function.

transmission

and

approach

selective

in

control

1988;

excitotoxic

VIII.

poisoning

disorders

relationship

some

the

of

as

theoretically,

consequently,

Another

The

minimally

THERAPEUTIC

VGCC

Olney

of

abnormally

this

animal

and

phospholipases

large

is

such

The

such

the

(Meidrum

mechanisms

pathogenic

that

between

may

etal,

blockers

of

approach

potent

Ca 2 +

use-dependent

1989;

number

as

interfering

models

as

FOR

EAAs

advantageous

have

the

stimulated

Guam

massive

1989).

buffering

and

excitatory

APPROACHES

removal,

Tymianski

many

Ca 2

(Deshpande

are

is

profound

of

of

process.

has

Garthwaite,

disease,

directed

C

excitotoxic

neurological

+

with

the

A

and

and

environmental

mediated

encouraged

major

EAA

open-channel

capacity

are

major

other amino

powerful

A 2 ,

in

eta!,

and

The

neurolathyrism

also

at

synapses

and

this

problem

TO

or

excitatory

reducing

synapses

unacceptable

1991).

insults

most

acid

activation

1993b).

being

diseases

Wieloch,

the

in respect

EXCITOTOXIC

the

antagonism

neurons

excitotoxins

inhibition

antagonists

direct

antagonists

with

mediating

considered

(McDonald

search

the

since

that

neurotransmitters

Also,

that

1986)

of

and

this

approach

increases

(Scharfman

side

protein

are

for

of

may

they

of

inhibition

domoate-induced

receptor

the INJURY

and that

lipid

and

required

therapeutic

effects

of

et

EAA

(Braughler

in

may

the

a!,

disease

kinases

possibly

certain

is

have

in

part

peroxidation

synaptic

the

1

and

[Ca 2 ]

PCP-site

blockade

show

of

987;

on

for

be

proven

in

down-stream use

normal

process

C

eta!

neurological

measures

mediated

normal

the

by

Gill

selectivity

and

of

by

shell

ligands

CNS

1987).

et

useful

and

A

the

a!,

as

fish

by

to 34 35

Recently, the use of neurotrophic factors for the control of excitotoxic neuronal damage has been proposed under the assumption that it is the imbalance between restorative and EAA-mediated catabolic functions that causes cell death. Thus, reduction of neuronal loss may be achieved by restoring the balance through supplementation of injured neurons with neurotrophic factors (Mattson eta!, 1993a).

IX. RECAPITULATION

Excitotoxicity is a mechanism of neuronal injury that is probably involved, to variable degrees, in the pathogenesis of a large number of acute and chronic neurological disorders. Evidence from in vitro as well as in vivo studies indicate that endogenous excitatory amino acids (e.g., glutamate and/or aspartate) may be the prime mediators of this phenomenon and that in many cases the antagonism of EAA-mediated primary, and/or more downstream effects, may be helpful in the amelioration or reversal of the disease process. Derangement of the intracellular calcium homeostasis is a central mechanism in the EAA mediated neurotoxicity and restoration of this homeostasis is the focus of many lines of therapeutic approaches including the use of intracellular calcium chelators. Other cytopathic mechanisms such as overproduction of free oxygen radicals and lipid peroxidation may also be involved in neuronal injury.

The issue of primary versus secondary effects in neurotoxicity is complicated by the extensive network of positive feedback regulatory mechanisms between the various causes of cell injury which are responsible for the perpetuation and amplification of the primary insult. The resolution of the cause-effect relationship issue must await the availability of less intrusive high spatial/temporal resolution, multi-parameter intracellular monitoring techniques.

the

metabolite,

associated

locations

distal

Wasserman

II.

parvalbumin fairly

intracellular

calcineurin

activation

proteins

in

proteins that

binding

nervous

number

I.

the

CaBP INTRODUCTION

induction

Morrissey

are

CaBP

Calbindin-D28K

large

convoluted

nervous

site

of

characterized

(Dalgarno

system

(CBPs)

HISTORY

CaBP

while

with

was

intracellular

vertebrate

and

1

and

Ca 2 +

known

(PV),

,25-dihydroxy-cholecalciferol.

of

system

and

synthesis

first

recoverin)

tissues

chicken

exist,

Taylor

the

and

tubules

buffers.

cairetinin

eta!,

AND

Wasserman

as

buffer

discovered

renal

(CaBP)

tissues

the

by

the

and

concentrations

in

performing

intestinal

DISTRIBUTION

1984).

is

and

the

1966.

EF-hand

are

cortex.

EF-hand.

proteins

they

To

totally

(CR)

is

with ic.

presence

in

known

this

a

and

(1971)

the

can

The

Ca 2

and

It

CaBP

CALBINDIN-D28K

dependent

particularly

proteins

transcellular

It

buffer

was

have

chicken

isolated

generally

Although

is

trigger

CaBP.

to

and

binding

reported

of

a

expression

subsequently

mediate

no

member

protein

a

can

are

specialized

proven

oviduct.

from

proteins

on

be

high

other

protein

potentially

the

Ca 2 +

a

the

Ca 2 +sensitive

classified

group

chick

of

close

most

and concentrations

function

families

active

a

(e.g.

found

Its

transport,

large

that

high

the

intestinal

belong

association

studied

expression

function

as

vitamin

calmodulin,

exists

enhanced

family

affinity

in

of

but

trigger

the

proteins

Ca 2 -binding

enzyme

and

they

group

epithelium

in

avian

in

of

as

D

between

Ca 2 -

a

has

in

the

or

proteins

exist

Ca 2

large

these

of

such

buffer

been

renal

CBPs

in

by

as 36

system

populations Christakos

neurons

standpoint

its

the

and

(Buchan

mammalian

and

detected

mathematically

facilitation The

Feher for

promotes

while

protein

expression

studies

(Spencer

transport

role

expression

CaBP:

central

genetically

mammalian

latter

for

CaBP

et having

is

synthesis

demonstrated

CaBP

is

and

a!,

in

et

not

a

induced

the

study

not

and

that

is

of

and

by

most

enterocytes,

(Jande

diffusional

1992)

a!,

Baimbridge,

not

no

dependent

Ca 2

in

reduction

in

universal

vitamin

Norman,

unrelated,

peripheral

it

by

1

intestinal

Ca 2 +

also

effect

Ca 2 +

978)

vertebrate

restricted

was

inhibitor,

by

a

eta!,

transport.

number

proposed

vitamin

and

originally

that

transport D

transport

Ca 2 +

on

since

of

1980,

it

on

lagged

1981;

1988).

epithelium

nervous

calbindin-D9k.

enhanced

that

has

intra-mitochondrial

in

to

cycloheximide,

vitamin

tissues

of

flux

vitamin

D.

anti-CaBP

avian

been

Varghese

an

the

Despite

authors

Roth

a

behind

tissues,

across

This

has

It

neuronal

alternative

system,

two

has

with

Ca 2 +

D

replaced Ca 2 +

(Wasserman

eta!,

D-deficient

been

led

(Baimbridge

that

the

processes

been the

(Bronner

antibodies

exceptions

expression

eta!,

to

In

transport

transport

which

1981;

protein including

presented

enhancement

intestinal

controversy

all

the

argued

and

by

Ca 2 +

1988).

vertebrates,

establishment

the

et

animals

Cello,

prevented

and

controversial

could

and

(Parmentier,

label

a!,

tissues

of

such

accumulation

the smaller (Morrissey

from

epithelium.

and

Fullmer,

1986;

CaBP

Also,

Parkes,

1990).

the

specific be

enteric

induction

of

as

modelled

an

uncoupled

and

CaBP

CaBP

Ca 2 +

idea

molecular

striated

in

its

evolutionary

Feher,

1982).

of

functional

1981;

the

eta!,

1989).

it

nervous

distribution

neuronal

that

an

However,

is

has

expression

and

transport

of

nervous

found

1983;

muscle essential

1978).

CaBP

CaBP

been

by

the

weight,

In

Unlike

system

the

role

sub-

in

in

later 37 38

III. CHEMISTRY AND MOLECULAR BIOLOGY OF CaBP

CaBP is an acidic 28 kDa protein composed of a single polypeptide with an isoelectric point of 4.2. In the purified form it is free from lipids, carbohydrates and phosphates (Bredderman and Wasserman, 1974). It is fairly heat stable up to 80°C and has an overall apparent affinity of 0.5 pM for 2Ca This high affinity for 2Ca is also associated with higher 2Ca selectivity. over other divalent cations such as (pK 2.44 versus 6.30 for 2Mg 2j,Ca 2Mn and .2Zn Human CaBP has been sequenced and cloned (Wilson et a!, 1985, Takagi eta!, 1986). Its gene has been located on chromosome 8 and is 79% identical to chick CaBP and 98% identical to bovine and mouse CaBP. CaBP is composed of 271 amino acids with six EF-hand domains, four of which are functional (numbers 1, 3, 4 and 5) resulting in a high 2Ca binding capacity with one CaBP molecule capable of binding 4 2Ca ions. Although CaBP is not as highly conserved as calmodulin, evolutionary rate analysis indicates that

CaBP is highly conserved compared to many other CBPs such as PV or calbindin-D9k. Based on the fairly high conservation of CaBP, Parmentier (1989) has suggested that, in addition to a potential 2Ca buffering function, which does not represent a strong pressure for conservation (see later), CaBP may be involved in other vital protein-protein interactions that are specific for the sites of expression of CaBP.

The molecular basis of 2Ca affinity: CaBP and many other 2Ca + binding proteins owe their 2+Ca binding properties to a conserved amino acid structural motif known as the EF-hand. This structure was first identified in 39 carp parvalbumin and it was named after the E and F a-helices in this protein (Kretsinger and Nockolds, 1973).

An EF-hand is composed of two a-helices (E and F with 9 and 8 amino acids respectively) linked to each other by a loop of 12 amino acids rich in acidic oxygen-donor groups that form the six coordination sites for 2+.Ca Five of the coordination sites come from carboxyl or hydroxyl side-chain oxygen of serine, threonine or acidic amino acids while the sixth coordination site comes from a carbonyl group on the main chain. The oxygen-donor amino acids are precisely spaced at positions 10, 12, 14 16, 18 and 21 in the EF-hand domain forming the vortices of an octahedron site for 2+Ca binding. EF-hands are almost always found in pairs as a result of the hydrophobic bonding between several hydrophobic amino acids across the two hands. Such pairing results in a positive cooperativity between the binding sites with an increase in the affinity and selectivity of the 2-hand assembly for 2Ca as well as an increased sensitivity to small changes in its concentration (Williams, 1986).

While six copies of the EF-hand exist in each CaBP molecule, other members of the EF-hand protein family have different numbers of EF-motifs per molecule of protein, ranging from 2 domains (calbindin-D9k) to 8 domains (e.g., 1LpS in Lytechinus pictus). The primary sequence of EF-hands varies in different proteins. For example, PV has a pair of ‘normal’ EF-hands and a third mutated non-functional hand that is packed against the globular structure formed by the two functional hands. Another variety exists in intestinal vitamin

D-sensitive calbindin-D9k which has two EF-hands, the first domain of which is considered atypical because the loop between the two a-helices contains 14 amino acids instead of the usual 12 amino acids. This atypical 14-amino acid

site

region

hand

According

Ca 2

and

gene

eta!,

others of

domain

members)

likely

proteins.

EF-proteins

substitution

the

required

primary

proteins.

EF-hand

proteins

exerts

others

tertiary

Beyond

proteins

mutations,

Genetics

It

similar

1990).

binding

as

are

is

proteins.

amino

for

is

well

important

All

that

to

only

a

have

that,

characteristic

structure

has

re!ative!y

Ca 2

of

to

the

Heizmann

are

Some

site,

have

as

of

underwent

acid

the

that

expressed

been

some

retained

over

genetic

in

presumed

the

Gene

binding.

without

amino

undergone

non-EF-hand

sequence

to

proteins

of

EF-hand

of

used

time,

weak

of

stress

calmodulin,

and

duplication

the

sequence

the

the

of

acids

in

multiple

for

retaining

acquired

to

Hunziker

conservation

a

EF-hand

a

are

tertiary

EF-hands

that

proteins:

of

subfamily

have

the

tremendous

species-

which

universally

sequences.

the

in

construction

coding

reiterations

since

and

evolved

the

over

quite

EF-motif

that

conformation

(1991)

allows

have

Genetic

and/or

mutation

actual of

it

pressure

is

200

for

diverse

is

diversification

the

expressed conserved

from

lost

‘maintenance

the

Undoubtedly,

retaining

the

is

proteins

tissue-specific

to

EF-proteins

primary

of

analysis

not

their

most

then

EF-hand, produce

a

expression

a

of

on

single

conserved

‘family

the

Ca 2

the

produced

conserved

(e.g.

the

via

of

amino

of

EF-hand,

both

primary the

prototypic

the

appropriate

coordination

of

known

genes

a

this

tree’

binding

calmodulin)

large

manner.

patterns

a

EF-family,

acid

multiple

in

but

calcium-binding

diversity

the

for

the

of

of

structure’.

as

number

identity.

and

rather

capacity

multitude

the these

the

gene

promoter

S100

(Moncrief

EF

Through

the

sites

the

family

EF

while

it

(most

of

is 40

conserved

scattered

gyrus

superficial

neurons

which

collateral

1992). pyramidale

the

bundle

differences,

purposes

hippocampus

view,

including

antibodies

any

specific-expression

IV.

reflects

granule

particular

CaBP

CaBP

Expression

CaBP

and

makes

do

of

The

are

functional

fibers)

interneurons

not

of

axons

mossy

throughout

pyramidal

in

EXPRESSION

in

to

of

is

cell,

CaBP

CaBP

this

albeit

other

CaBP

the

contain

a

CaBP

will

the

membranes

highly

terminate

and

is

of

work,

fibers

rat

CAl

be

staining

less

deeply

animals.

specialization

an

CBPs

show

(Baimbridge

and

fine

hippocampus:

CAl

presented

CaBP.

the

water

ideal

(Baimbridge

region

diverse

only

that

their

IN

dendrites

in

hippocampus

uniform

neurons

stained

on

or

pattern

THE

adult marker

synapse

CaBP

For

soluble

Axons

cytoskeletal

the

physiological

(Baimbridge

than

(for

CENTRAL

etal,

example,

of

brains

moderately

with

expression staining

found

are

which,

and

for

The

the

a

that

of

protein

with

comprehensive

1991).

CaBP

the

CA3

CaBP.

Miller,

individual

proper,

principal

is

in

in

pyramidal

elements.

none

and

subject

from

specific

of

NERVOUS

the

function.

the

that

pyramidal

CaBP

the

and

Their

In

1982;

Miller,

developing

while

rat

CaBP

of the

has

the

cell

whole

proteins

its

to

the

hippocampus

populations.

neuroanatomist’s

CaBP

neurons

hilar

not

distribution

species-specific

type

Neurons

Miettinen

the

review

SYSTEM

is

1982).

neurons

CAl

neurons

also

cytoplasmic

been

deep

zone

of

for

brain.

neurons

present

see

the

of

axons

Only

their

that

associated

pyramidal

in

and

of

(Schaffer

the

is fascia

in

Celio,

the

For

the

react

not the

tissue

the

Freund,

CA3

form

contains

volume

in

stratum

the

point

dentate

exactly

rat

dentata,

1990).

with

region

a

with

of 41 42

CaBP in the guinea pig (Rami et a!, 1987) while in the domestic pig all pyramidal and dentate granule cells lack CaBP (Hoim eta!, 1990).

V. DEVELOPMENTAL EXPRESSION OF CaBP AND RELATED CBPs

An extensive literature has accumulated concerning the expression of the three major neuronal CBPs with no known specific functions (CaBP, PV and CR) in the developing nervous system. In their review, Celio and colleagues (Andressen et a!, 1993) state that the ‘complex variety of [developmental] patterns does not allow us to associate any of the three CBPs with a specific phenomenon in the developing brain’. They presented, however, a few general principles related to CBP expression. CBPs can be transiently expressed at various stages of development and CR is the most precocious member of the group while CaBP expression precedes PV expression in the central nervous system and follows it in the peripheral nervous system. The pattern of expression of a particular CBP in a certain population of neurons during development bears little relation to the final expression profile of CBPs in the adult neuronal population, which in some cases (e.g., in primates) is not reached before 1-2 years postnatal. Transient expression of CaBP and PV has been observed, for example, in neocortical pyramidal neurons of layer V in both the monkey and neonatal kitten (Hendrickson eta!, 1991; Hogan and Berman, 1991). Developing non-neuronal elements, e.g. spiral limbus and crista supporting cells of the fetal mouse inner ear, can also express CBP5 transiently while they do not, under physiological conditions, do so in the adult animal (Dechesne and Thomasset, 1988).

CaBP in the developing hippocampus: CaBP immunoreactivity can be detected in the 18-day fetal rat hippocampus (Enderlin etal, 1987). During the

found Very

under

CaBP-D28k

enough

VI.

in

slowly

adult

pyramidal

CA3

CAl

observations).

(Baimbridge important

pronounced

pyramidal

closest

neurons

(Baimbridge,

granule

later which

first

pattern

vitamin

MODULATION

Expression

little,

pyramidal

few

pyramidal

developing

pattern

physiological

to

(Baimbridge

develop

to

to

seen

cells

alter

can

weeks

D

to

layer

induce

neurons

stratum

if

is

depleted

and

temporo-septal

any,

note

be

of

but

1992).

in

neuronal

not

neurons.

neurons

post-natally),

For

in

staining

the

of

seen

post-natum,

Miller,

granule

its

CaBP

is

sensitive

that,

the

oriens).

renal

develops

OF

KG,

conditions

the

adult

known

synthesis

animals.

At

in

first

CaBP

CaBP

in

purpose

expression

1982;

unpublished

the

(together

of

and

three

cells

Further,

rat.

vivo,

week

superficial

to

about

The

deepest

gradient

slowly

EXPRESSION

intestinal

expression.

CaBP the

vitamin which

Miettinen

and

Within

days

is

A

CaBP

of

adult

post-natum

delayed

expression

single

the

with

the

very

the

within

is

after

post-natum,

observations).

layers

form

(Baimbridge

is the

produced

regulation

pattern

neurons

D

CAl

calbindins

work

a

few

expressed

dose

depletion

and

few

about

until

dentate

hours

a

of

pyramidal

layer

experimental

presented

of

Freund,

do

scattered

of

of

the

the

that

CaBP

in

14

so

vitamin

while

of

staining

numerous

(D9k

KG, adjacent

CAl granule

or

large

third

only

days

only

neuronal

do

repletion

1992),

gradually

neurons

unpublished

in

express

and

pyramidal

in

interneurons)

or

in

amounts

transiently,

D

post-natum,

the

of

this

tools

cells

a

fourth

in

to

D28k)

intensely

the

small

CaBP

and

nervous

these

the

thesis

(see

develops

CaBP

(the

assumes

have

superficial

never

week

in

hilar

neurons

number

is

expression

animals

above).

the

majority

while

depressed

it

stained

been

in

and

system

zone

is

in

in

initial

the

quite

the

with

the

the

the

of

(i.e.

CAl

is

CAl

of

43 a cells factors expression expression was target of the contrast, cerebral expression concluded prolonged frequency eta!, regulation content or development seizures convulsive certain adrenalectomy septum hippocampus. found (neurotrophin-3, In Chronic Kindling 1985). of have cultured neural of cortex in CaBP adrenalectomy changes of electrical that to is indicating (Barakat the of response threshold also associated mRNA corticosterone of be CaBP However, is pathways depletion. dentate or CaBP chick a a required been was the convulsive notable As in and and stimulation of to a of expression Lindholm dorsal cerebellum [Ca 2 j 1 . in small reported CaBP role granule a the Droz, its Lowenstein with resulted kindling, using for previously-subconvulsive However, example mRNA for pathway treatment and root the in a response, 1989). subconvulsive target eta!, cells dramatic the of to only can induction in ganglion in the levels the induce where dentate the rats eta! (Miller Baimbridge be 1993), and transient. elements dentate perforant Recently, has reduction kindling modulated (lacopino and in (1991) eventually CaBP repeated neurons, but been the and granule cultured specific stimuli granule not hippocampus in Baimbridge, expression of path (1992) reported several reported the of and the the stimulus. by electrical cells a CaBP results gradually hippocampal depletion resulted regulation muscle cells stimuli maintainance Christakos, perforant reported neuronal 24 to that and were in in enhance 1983; hours but stimulations extract that Besides cultured in fully in reduce mRNA of of an path, the that not the growth the induce later. neurons 1990a). Baimbridge CaBP developed up- of main in rat, the factor the CaBP levels the the amygdala Purkinje CaBP the They high effect of and In in 44 45 in hippocampal organotypic cultures (neurotrophin-3, basic fibroblast growth factor, brain-derived neurotrophic factor, Collazo et a!, 1992)

VII. BIOLOGICAL ROLE OF CaBP

The functional role of CaBP in neurons is not known. Because of its high concentration in certain cell types (e.g., Purkinje cells and dentate granule cells) and its high affinity and 4:1 stoichiometry for Ca-binding, it has repeatedly been proposed that its primary function is to buffer2 2Ca While 2Ca buffering must be a property for a protein with the aforementioned. biophysical properties, it probably is not the sole function since the structure of CaBP is extremely conserved, a feature more consistent with a possible vital protein- protein interaction function than with a passive 2+Ca buffering role. The functional contribution of a 2+Ca buffer in neurons is not, however, a trivial one, and many researchers have reported experimental findings suggesting that CaBP, via its high 2Ca buffering capacity, is associated with regulation of 2Ca fluxes through cell membrane (Kohr eta!, 1991; Lledo eta!, 1992) as well as with neuroprotection of neurons against excitotoxic injuries (Baimbridge and Kao, 1988; Mattson eta!, 1991), cerebral ischemia (Goodman eta!, 1993) and in neurodegenerative diseases (Yamada eta!, 1990). However, excitotoxicity, ischemia and neurodegenerative diseases are very complex processes that are not well understood and their pathophysiotogy cannot be explained on the basis of a single neuronal characteristic such as CaBP immunoreactivity and the consequent higher neuronal 2+Ca buffering capacity.

The complexity of the clinical phenomena no doubt underlies the fact that many studies have failed to detect a global neuroprotective role for CaBP in

disorders

discussed

causal

condition

CaBP

systemic

midbrain

SN

neurons

comes

in

(lacopino

dorsalis,

Alzheimer’s

loss

immunoreactivity

lacking.

disorders,

are

very

resistant

neurological

mRNA

were

strongly

of

Many

sensitive

and

relationships.

from

CaBP

in

have

but

spared

While

injections

is

and

to

may

and

an

in

evidence

the

of

presented

the

disease,

human

Andressen

CaBP

not

insults.

improved

immunoreactivity

these

protein

Christakos,

reflect

also

to

SN

lchimiya

same

(Yamada

in

ischemic

in

pars

been

of

the

brains

observations

for

several

other

A

associative

levels

For

(Freund

insult

MPTP

in

or

cerebral

compacta

detailed

eta!

a

et

Heizmann

reported

example,

generalized

reduced

eta!,

1990b)

with

injury

authors

a!

of

in

neocortical

(Lavoie

(1993).

(1988)

eta!,

CaBP

spite

in

Parkinson’s

1990).

cortex.

discussion

whereas

or

and

the

in

and

and

neuronal

and

hippocampal

(lacopino

coincidental

1990).

of

monkeys

were

and

reported

striatum,

neuroprotective

associations

the

yet

the

Braun

areas

Sparing

Another

Parent,

ventral

the

detected

pigmented

fact

of

survival

disease,

Also

and

in

CBP5

rendered

(1992)

dentate

a

hippocampus

that

marked

of

brains

CAl

phenomena

1991).

in

example

Christakos,

tegmental

between

dopaminergic

in

neurodegenerative

in

both

under

where

CaBP

and

pyramidal

role

the

granule

neurodegenerative

from

parkinsonian

loss

neuronal

some

SN

of

for

a

marked

the

area

patients

neuropathological

of

rather

conflicting

and

and

1990b)

CaBP

cells

neurons

CaBP

aspects

presence

neurons

CaBP

of

nucleus

hippocampus

populations

than

are

reductions

is

the

after

with

reported

still

in

highly

results

are

any

are

the

of

raphe

46 a

the

affinity

constants,

total

time

buffer

and

where of

buffer technical

which

enhancing

Ca 2 )

binding

passive

membrane-bound

passive mechanisms,

of

active’ THEORETICAL

a

Ca 2 +

steady

reverse

passive

course

Ca 2 +

Buffers

Calcium

concentrations,

and

are

the

regardless

constant.

Ca 2 +.

is

buffers

buffers

1D.

in

reasons.

determined

difficult

total

will

state

its Ca 2 +

content

the

of

reaction

buffer

in

affinity

where

buffering

CALCIUM

the

be

non-biological

This

system

CONSIDERATIONS

Ca 2 +

satisfy

which

equilibrium

buffering

of

more

organelles)

to

buffering

can

is

general

the

the

rate

isolate

for

fast

content

on

faster

bind

the

results

be

effective

is

energy

‘buffered’

Ca 2

the

constants

a

compared

adequately

capacity BUFFERS

physico-chemical

Ca 2

will

mechanism

definition

action,

and

buffers,

basis

systems:

or

of

in

are

state

be

extruded

test

the

during

a

reversibly

of

smaller

determined known.

calcium

are

is,

particularly

to

system

experimentally,

i.e.

of

concentration

described encompasses

AND

however,

these

Under

also

by

the

the

those

(by

change

which

is

initial

Under

without

cell.

required

BIOLOGICAL

is

definition

rate

sequestered

Ca 2

equilibrium

solely

changing,

when

with

if

complicated

The

a

+..ATPase

period

constants.

both

non-equilibrium

in

change

of

active

sequestration.

in

at

larger the

on

the

biological

the

of

order the

least

the

of

free

values

conditions

a

rate

(in

available

buffering

forward buffer

change,

concentration in

SYSTEMS

or

basis

in

to

by

intracellular

‘biologically

the

For

of

exchangers),

part

describe outcome

of

many

change

total

equal

as

of

conditions,

the

ligand

rate

Only

although

for

the

the

their

factors

content

forward

total

effects

in

the

of

of

Ca 2

(i.e.,

the

the

and 47

While

concerned

capacity

Baker

shield,

Experimental

Ca 2

functional

be

localization

processes. the

receptors

function).

and/or

allowing

usually

function

with

and

Cells

applicable

end-point

effectively

+mediated

Ca 2 +

transient.

A

Microdomains

many

Buffers

are

no

and

isolating

large sequestered

achieved

of

an

attempt

outside

in

resulting

signal,

Umbach,

microdomains

steady

with

This

to

the

specialized

of

Specificity

a

intense

number

evidence

in

a

state

buffered

all

immobile

At

biophysical

a

cell

biological

microdomains

intense

the

the

Ca 2 +

by

will

particular

state

a

in

of

by and

death.

flux

1987;

activation

sub-microscopic

of

cell)

relevant

be

activation

continuous

for

sub-cellular

active

to

of

and

may

mathematical

and

immobile

of

made

Ca 2

near

avoid

the but

systems:

both

Llinas

Ca 2

reaction

aspect

Immobile

spatial

brief

function

Ca 2

buffering

integrated

machinery

the

of

to

of

buffers

the

unwanted

of

etal,

intense

Ca 2

for

present

an

buffers:

Ca 2 +

change,

any

of

homogeneity

compartments

microdomain

In

signal

mixture

influx

a

Ca 2 +

as

level,

models

the

a

short

exists

1992;

of

systems.

flux

source. biological

a

in

for

activity

Ca 2 +

an

a

messenger

An

can

effects

and

the

or

cells

buffers

large

is

a

can

period

exhaustive

have

in

Zhou

release

increase

particular

dissipated

only

overall

equilibrium

signal

the

are are

never

concept

from

In

number

During

that

such

system

been

can

and

theory,

of

be

literature

extremely

probably

channel

by

cell

time

and

the

are

maintained

in

be

as

be

Neher,

review process

published

binding

the

by

cytosolic

and

of

certain

met.

performance.

rest

the

autonomous

can

the

used

stray

(a

diffusional

Ca 2

brief

(for

the

the

(a

probability

activation

effect

of

only

heterogeneous

1993).

of

Of as

+

point

within

to

Ca 2 +

assumptions

the

example:

least

significant

life

these

by

each

mediated

Ca 2 +

an

these,

specific

be

of

cytosol.

co

span

source)

effective

dynamic

cogent.

should fluxes

(i.e.,

buffers.

of

is

an

of

can 48

case

channels)

(e.g., extrusion/sequestration

negative spreading

bound

above the

concentration

concentration

with

receptor

entry/release

Ca 2 +

high

is

is extremely

conclusions

(1990),

evaluation

models,

short-lived

never

extent

of

The

high concentration

Near

reduction

signal

by

to

Ca 2 +

Feher

feedback

reached

sites

the

the

addition

facilitating

and

temporal

the

of

high

the

of

and

are

presentation

mobile

the

source

and

transport

signal),

the

by

for

high gradient,

point

eta!

values

of

based

maintains

functional

preventing

in

Ca 2

of quickly

experimental

VEm).

on

enough

of

precision

the

(1992),

sources

buffer,

diffusion

a

and

influx

with

a

mobile

(hundreds

on

across

fluxes,

bulk

stray

fixed

within

the

handled

of

While

the

several

to

a

will

isolation

by

and

the

cytosol

some

of

and

rest steep

Ca 2 +

buffer

Ca 2

of

evoke

work

intestinal

the

and

high

results

Ca 2 +

escape

reduction

Nowycky

the

Ca 2

of

specificity.

of

by

interesting

microdomain general

concentration

probably

from

of

micromolars).

local

buffer

helps

the

increased under

an

of

extrusion

entry

presented

Roberts

from

from the

‘unintended’

and cell.

nearby

[Ca 2 ]

restrict

of and

conclusions

to

normal

microdomain

or

the

enhancing

renal

the

the

the

Because

consequences.

In

release,

Ca 2

Pinter

(1994),

mechanisms.

microdomain

can

this

sources

microdomain

driving (e.g.,

in

cell

gradient

the

tubular

conditions

This

this

be

scheme,

flux

response.

(1993).

results

spatial

of

[Ca 2 +]

inactivation will

Sala

triggered

influx

concentration

thesis.

force

structure

cannot

the

is

epithelium

between

be

desirable

and

in

spread

steep

as

The

(and

low

by

First,

thus

for useful

can

a

These

the

reach

Hernandez-Crus

removal

reduction effectively

the

affinity

existence

discussed

thus

of reach

reducing

the

more

Ca 2 +

of

(Feher

for

in

Ca 2

Ca 2

of

a

the

the

the

Ca 2 +

of

Ca 2 +,

flux

in

et

flux

the

of

the

49 a

Relative

depend

been

isolation

effect

constants

and

extremely

activation

physiological

summation

loss

the

for

presynaptic the

in

reducing

concentration

attempt

al,

the

Ca 2 +

1992),

recovery

source

nanometer

of

The

It

loaded

Third,

Second,

and

loss

is

precision

on

affinities,

and

to

outcome

important

the

may

and

fast,

of

the

the

in

of

(i.e.,

of

restore

the

with

terminals,

concentration

other

process,

functions

peak

other

a

the

a

buffer

buffer

buffer

gradient

be

dimensions).

short

dissipation

physiological

fast

a

and

the

unnecessarily

rate

Ca 2 +

of

stray

signal

processes

cellular

cell

to

mobile

affinity

concentration specificity

the

range

buffer,

properties

stress

may

constants

while

may

types

between

signal).

signal.

whole

in

of

Ca 2 +

gradient,

lead

and

the

Ca 2

be

particularly

to

other

that

function,

Under

may

the

it

interrupted

Ca 2

may

microdomain

array

non-equilibrium

as

of

to

activated

This

the

This

and

homeostasis.

concentration

all

buffer

occur.

the

Ca 2 -mediated

an

much

are

these

the

source

result

and

may

buffering

of

plays

change

enhancement

Ca 2

such

the

the

derangements

processes

results

as

increased

(e.g.,

circumstances, not

in

and

major

a

of

it

cell’s

as

signal.

while

excessive

minor

in

depends

be

Ca 2 +

capacities

unintended,

neurotransmitter

reactions

gradient

‘remote’

in

concentration

desirable

determinants

endogenous

the

processes

discussed

increasing

signal

role,

of

Consequently,

entry

dissipation

spatial

on

(loss

waste

and

in

proteases

of

(with

summation)

binding

the

since

and

any

possibly

endogenous

of

the

with

above

and

[Ca 2

cell

of

profile

buffers.

of

microdomain

bulk

microsecond

at

release

it

prolongation

energy

the of

rate

temporal

that

all.

higher

means

+1

or

cytosol

are

the

harmful

may

away

buffer

will

kinases).

has

in

in

affinity

the

result

an

from

thus

of 50

buffer

mechanisms

Indeed,

important

properties subcellular

biologically

versus

buffers

loading

soma

will

for

in

of

domain

all

relevant

vary

determining

and

or

artificial

a

practical

priori

dendrites).

from

binding

to

events.

buffers

is

another

one

purposes,

impossible.

the

sites

cell

Similarly,

response

Additionally,

will

within

that

type

ultimately

predicting

mediate

to

the

in

to

another

experimental

same

the

the

Ca 2

determine

the

addition

properties

and

cell

+.sensitive

net

(e.g.

probably

‘biological’

situations,

of

their

artificial

of

presynaptic

processes

Ca 2

effects

from

effect

the

buffers.

one

regulatory

on

precise

terminal

will

of

the

be

as 51

and

I.

designed

neuronal

CaBP

injury,

intraneuronal

experiments

This

It

e)

and ci b) a)

d)

induced

To

Ca 2 +

as

less

like

depolarization-evoked

the

‘Increasing

especially

has

the

the

the

the

the

a

Ca 2 +

characterize

to

1E.

hypothesis

vulnerability expression

molecules,

vulnerable

Ca 2

been

stores).

test

effects

dependence

types

effect survival

calcium

excitotoxicity.

HYPOTHESIS

buffering

the

will

established

buffer,

during

neuronal

and

of

of

hypothesis

of

be

will

of

to

culture

will

primary

homeostasis

bFGF

sources

of

neurons

designed

CaBP

excito excitotoxic

and

of

capacity,

be

enhance

neurons

Ca 2

CaBP

tested

treatment

Ca 2

the

with

type

or

hippocampal

toxic AND

+

of

that: under

+

availability

by

buffering

to

expression

reasonable

Ca 2

to

on

loads

their

the

is

in

injury’.

insults.

artificially

satisfy

glutamate-induced

OBJECTIVES

an

the

primary

different

work

on

ability

signals

and

important

vulnerability

neuronal

capacity,

neurons

the

of

presented

In

consequently

on

certainty

cultures

loading

tools

to

view

following

culture

(VGCC,

culture

handle

step

survival,

in

to

of

either

OF

of

them

of

vitro

that

enhance

in

the

conditions.

in

type

excitotoxicity.

LGCC

agonist-

neurons

THE

hippocampal

objectives:

this

the

render

as

potential

the

with

with

CaBP

and

thesis

a

development

STUDY

and

loss

result

artificially

respect BA

or

age.

to

them

expression

intracellular

PTA

of

glutamate

has

role

of neurons

-

been

of

to:

of 52 53

II. To determine the effect(s) of artificially increasing the neuronal 2Ca buffering capacity using 1,2-bis-(O-aminophenoxy)ethane-N,N,N’,N’-

tetraacetic acid (BAPTA) and 5,5’-dimethyl BAPTA (DMB) on short and prolonged 2Ca + responses evoked by either depolarization or exposures to EAAs (NMDA and glutamate).

Ill. To determine the potential neuroprotective role of increasing the neuronal 2Ca buffering capacity using BAPTA and DMB on glutamate-mediated neurotoxicity.

VI. To determine the potential functional association(s) between the expression of CaBP in cultured hippocampal neurons and the kinetics of depolarization- and EAA-induced 2Ca + responses.

V. To determine the potential neuroprotective role of CaBP expression on the vulnerability of cultured hippocampal neurons to glutamate-induced excitotoxicity. 54

CHAPTER 2

MA TERIALSAND METHODS 55

I. TISSUE CULTURE

All experiments were performed on primary cultures of rat fetal and/or 4- day postnatal hippocampal neurons. a. Fetal cultures

Fetal hippocampal neuronal cultures were prepared according to Banker and Cowan (1977) with modifications (SoIc etal, 1993) as detailed below.

I) Dissection

Eighteen-day pregnant female Wistar rats were anesthetized using (intraperitoneal, 50 mg/kg body weight) and the abdominal wall was shaved and disinfected with 70% ethanol. A lower transverse abdominal incision through the skin, abdominal muscles and peritoneum was performed and the pregnant uterus, usually containing 13 to 17 fetuses, was excised. The mother was then euthanized with an intracardiac injection of pentobarbital.

All subsequent steps were performed under sterile conditions in a laminar flow hood.

In turn, each uterine sac was incised and the fetus was decapitated. The separated head was gently stabilized with a medium size forceps grasping the snout. Using fine scissors, a mid-line slit of the scalp and thin skull bones was extended in a caudo-rostral direction to the bregma then antero-laterally on both sides. The brain was exposed by peeling apart the skull flaps and a spatula was used to free the brain from the underlying structures. The brain was picked up by the brain stem and rapidly immersed in ice cold Ca/Mg free buffered salt solution (CMF BSS, 2 ml in a 35 mm-diameter plastic-2 tissue culture plate). 0.20 longer sterile of terminated at 0.5 (DMEM). 12 culture then DNAase/trypsin dish ii) dissection. hippocampi scissors hippocampi then dissected structure pia/arachnoid side 35°C Dissociation 10% strokes mg/mI and transferred of mm) dissected fire-polished visible Hippocampi Cerebral tube, each fetal for and chopped surrounding After for away respectively) followed by were 20 to bovine Dumont allowed hemisphere, whichever a replacing meninges a out minutes similar gentle hemispheres mixture using to 1 pooled 5 into were Pasteur a using ml by serum fresh to #5 two the mixing, number smaller tissue trituration as with were settle in transferred came in forceps. and a the bisected CMF much amount the pairs combination (FBS) pipette were periodic kept added culture hippocampus first. the and pieces vicinity of BSS of as in strokes through hippocampal separated in A excess (internal of Dumont corpus Dulbecco’s with possible to (final ice Pasteur tube using agitation. ice of the of cold ice the cold or a CMF containing enzyme sharp callosum. hippocampal a tip #5 was narrower until cold of by pipette hippocampus CMF pair CMF diameter forceps. modified the tissue BSS blunt Enzymatic visible and pieces CMF of BSS concentrations enzyme BSS was iris was blunt ice The diameter dissection. was BSS as for scissors. 0.45-0.50 of pieces in Eagle’s Each cold decanted. used vascular a a triturated tissue the dissection were digestion into mixture whitish 1 CMF 5 hippocampus duration to pipette and a ml medium gently were transfer 35 On 25 Pieces mm) tissue BSS. incubated with C-shaped Six through was mm pg/mI the with (0.15- no of for ml 3 were All medial Petri the iris ml of and 10- was a 56 the attachment Coverslips 0.2-0.3 drained few grade was borate and iii) and (dH 2 O). neurons/mi. and a method were the repeated cell in diluent a Plating coated cell cleaned hours 22 suspension a followed refrigerated performed water, Calculated Sixty standard The buffer mm-diameter off ml pellet (50 The for gentle before were coverslips of cell immediately and to p1 by cell arranged coverslips (0.15 laminin by was eighty of suspension laboratory boiling were with incubated pipetting on centrifuge suspension extensive volumes cell cell re-suspended M, a were 1 solution then to glass suspension plating, sample in at pH ml before were reach 100 60°C through of of hemocytometer. 8.4) for transferred mounted was rinsing coverslips at to gently FBS of then coverslips mm-diameter (16.7 4°C. 2 the neuronal containing attain for purified the hours in in and a desired (more autoclaved 30 1 mixed 450 pg/mI) suspension 5 The in ml the centrifuging (Baxter ml to in minutes a using plating. were p1 of custom than supernatant 6 5% pipette. final final poly-D-lysine neuronal of or prepared DMEM Petri DMEM CO 2 0.4% #1, 1 an rinsed 20 and cell 2-well in plating using dishes designed FBS times) concentrated 0.17 at Viability density for then containing with suspension wlv in twice 35°C was gradient tissue a 8-10 density L-15 mm and trypan kept (PDL, in trypan 10% carefully stainless of distilled and in to minutes thickness) each (Gibco), culture for tissue 1 allow 10% FBS 15 (see by .25x10 6 blue cell were nitric blue 24 pg/mI). covered underlying was water FBS counting below). removed steel cell exclusion hours culture- at plates in which acid. plated of 150x saline) used viable using rack 18 A in with This was on G and the as 57

processes

configuration,

vitro

originally

low above

surface

neurons/cm 2 distinct

surface

cell

iv)

neurons/cm 2

Large

culture

medium

hours

addition

specified.

35°C

serum

changed

(Falcon)

Variations

density

density

with

22

Smaller,

Two

and after

and

Unless

(HS)

age,

facing

amongst

facing

with

of

for

developed

to

mm-diameter

clear

that

the

5%

different

5-fluoro-deoxyuridine

was plating.

neuronal

in

N2

a

plating

and

and

fresh

otherwise

neurons

further

down,

plastic

up.

order

tended

C0 2 195%

18

background,

supplemented

high

the

maintained

maintained

mm-diameter

In

N2

day

in

to

Nutrients

plating

closely

cultures

and

i.e.

surface

this

3

to

our

remained

supplemented

coverslips

improve

hours

was

indicated,

form

Air

neuronal

cells

configuration

laboratory

protocols

distinct

packed

counted until

in

in

(see below.

with

were

DMEM

neurite

were

tissue

tissue

coverslips

neuronal

in

(5-FDU,

were

used

also

glial

excellent morphology

2

replenished

neurons.

neuronal

sandwiched

mI/well

in

DMEM

and

This as

were

bundles,

culture

culture

Brewer

multiplication

7

plated

order

neurons

day

attachment.

to

the

20

were

‘face-down’

used

10

condition

twice

zero

pg/mI)

of

to

morphology plates

plates

neuronal

at

and

particularly

and days

DMEM

by

plated

enhance

survived

a

with

between

in

density

weekly.

replacing

Cotman,

neurites

to

with

with

vitro

except

was

fetal

for

Finally,

the

at

cultures

containing

culture

the

neuronal

the

for

a

(0

up

inhibited

and

the

of

in

culture

neuronal

low

In

where

Dlv).

were

1989).

half

to

cell-coated 3-4

older

cell-coated

2-3x10 5

the

extensive

glass

specifying

method

incubated

density

6

the

weeks,

weeks

5%

not medium

medium

survival

cultures.

otherwise

by

coverslip

cultures.

In

culture

visually

a

horse

this was

of

single

in

final

at

1O 5

was

48

in 58

A

FBS/HS)

cell

centrifugation

After

replaced supplemented

(25

initiated

centrifuge

chopped

Petri

individual

out

atmosphere.

technical

however,

Baughman,

neuronal

These

b.

procedures

calculated

pg/mI)

pellet

Postnatal

and

dish

incubation

At

Four-day-old

The

‘face-down’

by collected

was

with

into

the

cultures.

was

differences

filled

brains

required.

tube

medium.

stages

replacing

were

1

volume

end

added

986)

slices

cultures

The

serum-free

on

with

re-suspended

with

filled

for

on

a

of

performed.

in

of

neonatal

were

FBS

papain

A

Wistar

less cultures

filter

to

30

the

ice ice

of

Mechanical

preparation

with

the

between

short

adjust

EMEM

minutes

gradient

cold

cold

dissection

then

essentially

ice

paper

Eagle’s

ice

(1

rat

rats

description

were

in

cold

L-15. L-15

mg/mI),

the

1

cold

pups

supplemented

our

EMEM

soaked

mm

were

at

were

trituration

of

minimal

cell

L-15

turned

medium.

35°C L-15

handling

procedure

With the

were

postnatal

in

density

decapitated

BSA

and

performed thickness

medium

in

of

same

medium.

the

fine

right

anesthetized

essential

ice

this

viable

(0.2

and

of

Hippocampi

papain-BSA-DNAase

with

cold

scissors,

to as

enzymatic

neuronal

side

procedure

fetal

with

cell

mg/mI)

4x10 5

for

and

cell

as

and

5%

L-15

medium

up

purification

and

the

previously

5

collected

counting

the

FBS

before

ml

by

hippocampi

cultures

neuron/mI.

and

and

preparation

postnatal

dissociation

were

highlighting

pro-warmed

exposure

brains

and

(EMEM)-DNAase

DNAase

transferred

any

dissected

was

in

described.

5%

using

(Huettner

were

1

mixture experimental

cultures

5

were

performed.

Cell

HS

to

of

ml

(25

was

the

L-15

high

fetal

dissected

(EMEM

to

from

pg/mI).

and

was

a

is,

The

CO 2 59 a. were II. days with of was schemes. determined concentrations was (CaBP) the c. glial then (EMEM-N2). plates, suspensions previously culture Principle INTRACELLULAR bFGF-treated medium supplemented used cell 1 later, the treated The For Basic For ml in face proliferation. medium the of medium the to fluorescent the On 2Opl described, for of fibroblast with up, EMEM-N2 increase with were Arabinoside-C postnatal first the first Fura-2 the (1-10 in of cultures fresh was 20 basis in 24 then an with use 24 bFGF CALCIUM each UI ng/mI the calcium hours incubator and Ca 2 + growth hours, replaced Replenishing of containing EMEM-N2 cultures of of plated bFGF stock percentage bFGF well. the 0.5% these in after (Ara-C, sensitive cells factor (2.5 0.5% measurement coverslips on MEASUREMENT solution by in at (Collazo In trials, BSA plating twice 18 our 10pM were an 35°C ng/ml). all nutrients 10pM) BSA) (human mm-diameter of equal dye (vehicle experiments cultures. the weekly. (125 maintained neurons eta!, in the were Ara-C was fura-2 following The an volume was culture recombinant ng/ml) was solution). atmosphere then 1992). tested medium and expressing was added achieved using coverslips in of incubated 2.5 medium were treatment the as 1 N2-supplemented A 48 ml was ng/ml well bFGF, range probe bFGF, added of hours of by calbindin-D28k (EMEM-FBS/HS) totally as 5% prepared EMEM-FBS/HS in of replacing protocol of control used several R&D 6 to later bFGF. CO 2 or each replaced for 12 Systems) to in as cultures was dosage EMEM Two half well all 1 inhibit air. ml 60 61

experiments involving intraneuronal 2+Ca measurements. Fura-2 is a 2+Ca chelator with a BAPTA-like 2Ca binding site coupled to a fluorophore in a way that makes its fluorescence sensitive to 2+Ca binding. In the absence of free 2Ca fluorescent emission at 510 nm wavelength 510(EM will have a peak intensity, when fura-2 molecules are excited at 362 )nm while +bound 2Ca fura 2 molecules will have a peak emission intensity when excited at 335 nm wavelength. This 2Ca dependent shift in peak excitation wavelength is associated with a negligible shift in the peak emission wavelength from 510 nm in the 2Ca-free fura-2 to 505 nm in Ca-bound fura-2. These properties make fura-2 a very useful probe for free2 2+Ca measurement using a dual- excitation/single-emission wavelength protocol (Fig 2. 1).

As the free Ca concentration 2+ 2+])([Ca around fura-2 increases, the EM510 at 380 nm excitation wavelength )(Fl380 decreases while fluorescence intensity at 335 nm excitation wavelength )(Fl334 increases. The ratio between Fl334 and Fl380 /F1=Fl3343803341380 which also increases with increased [Ca has a wider dynamic range than the individual 2+], ),(R fluorescence intensity values (Fig 2.2). Assuming that Ca:fura-2 complex has a stoichiometry of 1:1, 3341380 is related to 2 according to the following equation R 2[Ca (Grynkievicz eta!, 1985): j

* * ]21[Ca = ( Kd (R3341380 - Rmin) / (Rmax - )R3341380 In this equation the proportionality coefficient 1, is the ratio of Fl380 in zero 2[Ca to Fl380 in saturating 2],[Ca Rmin and Rmax are R3341380 ] +1 fluorescence ratios in zero and saturating 2[Ca respectively, and Kd is the 62 affinity constant of fura-2 for 2+Ca under specific experimental conditions of pH, temperature, viscosity and ionic strength.

Rmin, Rmax and f are influenced by the optical properties of the experimental equipment (e.g., transmittance and reflectance of filters, mirrors, prisms and lenses) as well as by the spectral sensitivity of the photo-sensitive device and the electronic gain level in the acquisition and digitization equipment.

Intraneuronal free 2+Ca level can be probed using fura-2 if the dye is introduced into the cytoplasmic compartment. However, fura-2 is a highly charged polyanionic tetra-carboxyl molecule and is thus plasma membrane impermeant. A variety of loading techniques have been devised to accommodate particular experimental paradigms and cell types. These methods include direct intracellular injection of fura-2 penta-potassium salt, cell permeabilization and the use of the acetoxymethyl ester of fura-2, fura-2 AM (Tsien, 1981). The latter is the preferred method for loading a large number of cells, especially small cells, simultaneously and is the method of choice for neuronal cultures. The membrane permeant fura-2 AM diffuses easily into the cytoplasmic compartment where it is quickly de-esterified by cytosolic esterases to yield the anionic membrane impermeant fura-2, that becomes trapped inside the cell, and formaldehyde that quickly diffuses out. The use of R3341380 as the experimental measure of free 2+Ca (ratiometry) has significant advantages over the use of raw fluorescent intensity values. In addition to the wider dynamic range mentioned above, ratiometry corrects for a variety of confounding factors encountered with the use of all ion-sensitive fluorescent probes. Cell-to-cell variability in fura-2 fura-2 fura-2 measure active of wide remedies, characterized free probe the bleaching. index single-excitation/single-emission be is dependent correction are uniformities intensity result path loading a fura-2 performed. excitation also minor Ca 2 + variety through for of Although extrusion. AM If in in (e.g., fura-2 corrected [Ca 2 +] 1 choice cultured artifactual requires that the are problem efficiently concentration as processes in of detection in long welt a are different path. the is spectral single other for the changes excited The hippocampal only automatically established as without optical for many differences literature This with they such Ca 2 + affinity cell standard ratiometry of cell characteristics (Fig at is reasons. over intra-experiment minimal system (edge-to-center affect the as esterase the 362 sensitive 2.1) so of photo-bleachin:g, (Table a neurons need and corrected isosbestic UV fura-2 that nm, ion wide Fl 334 while (e.g. and subcellutar Our apparent optical activity sensitive for EM 510 2.1). potential fluorescent range is and and for and it laboratory mathematically non-homogeneous a is by increase point Ca 2 + function dye elements, Fl 380 affinity Neurons a the and becomes of ratiometry. gradients sequestration major probes problems, loss neuronal leakage dye and cell makes equally. probes has in coefficients, of due confounding has it are size) where and intracellular independent the can long in intensive or it to known been activity. are as Moderate raw routines and sensitive amount extrusion be leakage illumination Uncontrollable experience and well ratiometry available a extensively fluorescence variable very to fura-2 practically as factor shading light Also, de-esterify for of of or enough non- their of useful fura-2 ambient with photo- its optical was path) cannot patterns) the with for the use time a dye no our to use in can 63 distance microscope fluorescence c. de-esterification warmed before compartmentalization temperature, in hydrophobic mixing test pipette (30 b. more necessary IND0-1 imaging are BSS Image Fura-2 pg implemented tube expensive The Fura-2 Five any (1.5 on in tip offers BSS 40x system. and Acquisition 25 a imaging AM to acquisition Ca 2 + ml equipped ml/coverslip) vortex p1 fura-2 imaging objective fluorescence and neurons produce stirred of all Loading anhydrous of than BSS measurements in allowed the The hardware fura-2 stirrer AM. almost vigorously system with advantages that supplemented of were a hardware single-excitation/dual-emission (Zeiss final fura-2. at Neuronal AM. were to for a was DMSO) every well 33°C 100 cool was solution using LD fura-2 captured required using for loaded were After UV-Achroplan watt commercially down of built for was cultures Attofluor dual-emission with ratiometry dual-excitation of 60-70 a attempted mercury loading, around added with vortex to to 6 on 0.04% pM room achieve were a minutes. no imaging ZeissAttofluorTM slowly, fura-2 a stirrer. cultures arc available 0.6, but visible Zeiss temperature in BSA then microscopy lamp implementation dispersion order systems. Ph2). AM. software. Ca 2 while were Axiovert-lO incubated At The subcellular were and digital to this + BSA Excitation fura-2 placed sensitive stirring, ensure a for transferred concentration of is long fluorescence and digital significantly 60-90 with the AM fluorescent in of complete working vigorous using probe a light the the solution 15 minutes to fura-2 a was ml pre fine and 64 65 filtered through 10 nm band pass 334 nm or 380 nm excitation filters which were positioned into the light path using a computer-controlled solenoid filter changer. A 395 nm dichroic mirror and a 510 nm long pass filter completed the optical requirements for fura-2 excitation and emission. The photosensitive detector was a high sensitivity, Attofluor intensified charge-coupled device

(CCD) camera and images were digitized on a Matrox-AT image processor. The imaging software controlled the excitation filter changer, camera gain, image processor in addition to data processing, display and storage. The software was operating on a dual-monitor IBM-compatible 80486-33MHz. A schematic illustration of the imaging hardware is shown in Fig 2.3.

Using the imaging software, suitable gain and exclusion values were selected. Up to 99 regions of interest (ROls) could be positioned over the cell images in the selected experimental field. ROls were generally placed within the bounds of cell somata and average fluorescence intensities of all the pixels within the ROl were obtained at 334 nm and 380 nm wavelength. The frequency of ratio image acquisition was varied dynamically between 1 ratio every 1.5 seconds to a ratio every 900 seconds depending on the experimental protocol. Less frequent acquisitions were used whenever possible to reduce the risk of UV-induced photo-bleaching and photo-toxicity. For the same reasons, neutral density filters in the excitation path were used whenever fluorescence intensity was sufficiently high. On-line background correction was never necessary since cellular autofluorescence was below detection.

Background (BG) readings for both excitation wavelengths were, however, recorded from several cell-free regions in the field and used later for BG correction during off-line data analysis. The major source of these BG values was electronic noise in the acquisition hardware, although other factors such 66 as autofluorescence in the perfusion media and glass coverslips and light scatter in the optical path made some contribution. Although frame averaging could be performed using Attofluor software it was not used since illumination intensity and signal-to-noise ratio were very stable. d. Test drug exposure protocols and perfusion

Several perfusion chambers were used in experiments involving intracellular 2+Ca measurements. The most elaborate chamber was used for experiments involving transient (less than 30 seconds) exposure to test . The chamber was manufactured in the workshop of the Department of Physiology (UBC) according to our design specification. The chamber design permitted excellent laminar flow, relatively rapid medium exchanges, regulated temperature and control over CO2 around the perfusate (Fig 2.4). An alternative chamber with a fixed glass bottom (0.17 mm thick) was also used which was more secure against leakage, especially during 24-hour experiments, and allowed for a much faster and more efficient exchange of medium as well as a simpler coverslip replacement. However, it lacked temperature and atmospheric control.

A peristaltic pump was used to superfuse cells with BSS at a rate of 1.8- 2 mI/mm at 21-22°C. Unless otherwise indicated, BSS for perfusion was 2Mg free and contained 1 pM tetrodotoxin (TTX) to block voltage-gated sodium channels. Drugs that required transient application were prepared in

BSS and applied into the chamber through a 1 ml syringe connected to a 1 Y2” 30G blunt needle pointing to the inlet of the chamber. The drug application assembly was fixed to the microscope stage using a small C-clamp. The peristaltic pump was stopped during drug application and the exposure to test 67

drugs was terminated by resuming perfusion together with a flush of 1 ml of BSS through the drug application syringe.

In some experiments where long exposures to test drugs were needed, an alternative perfusion method was employed. The glass coverslip was glued onto the bottom of a well in a 12-well tissue culture plate using a trace of silicon rubber (Dow Corning). A small perfusion-head assembly was attached to the wall of the well with one perfusion inflow line and two suction lines. One of the suction lines was positioned near the bottom of the well and could be used to empty the well completely while the other suction line was well above the bottom and allowed about 1 ml of BSS to remain in the well at all times during perfusion. With the use of a 1 ml pipette and properly timed control of which line was open, a total exchange of the medium in the well could be achieved in less than one second (Fig 2.5). This configuration was particularly useful in experiments where relocation of the cells under examination was required following subsequent immunohistochemical staining (see below). e. Fura-2 calibration

An in situ calibration method was used to convert fluorescence ratios to absolute 21[Ca Fura-2 loaded cells were treated with 5-10 pM of the non fluorescent] ionophore, 4-Bromo-A23187 in BSS while recording fura-2 . 2Ca fluorescence ratio. In the presence of standard BSS 2[Ca +1 of 1.8 mM, the acquired ratios represented Rmax. The medium was then replaced by 2Ca +4ree BSS, supplemented with 200 pM EGTA, and Rmin was obtained. The proportionality coefficient, f, was calculated as the ratio of Fl380 Zero Calciumto 68

Fl380,MaximumCalcium We used the published fura-2 Kd values of 135 nm at 20°C and 224 nm at 37°C (Grynkievicz eta!, 1985).

Calibration was not performed routinely at the end of every experiment but generally when there was any change in the optical properties of the system (e.g., re-alignment or installation of a new lamp). Although our 2+Ca imaging results are presented as absolute 2+Ca concentration in nM, it is important to stress that, in most experiments, control results were obtained from sister cultures under identical experimental conditions and thus trends of change have more relevance than the absolute values of change in 2+.Ca A variety of calibration techniques are employed by different laboratories (e.g., in situ, in vitro and spectrofluorimetric). Each of these methods relies on a different aspect of the experimental setup, i.e. cells, dye and hardware properties, and they tend to produce different calibration parameters even when performed in the same laboratory. Indeed, problems with different calibration methods have led some authors, e.g. Dubinsky (1991a), to report data as fluorescence ratios rather than absolute 21+][Ca together with a sample of the calibration values (Rmin, Rmax and ,f values) to facilitate ‘coarse’ comparisons of results across different laboratories. In our imaging system

Rmin, Rmax and I averaged 0.2, 10 and 8 respectively. f. Relocation Experiments

In order to relocate a particular field on a coverslip following 2+Ca imaging and subsequent immunohistochemistry (IHC), the coverslip was attached with a minimal amount of silicon rubber to another coverslip with hand-made diamond pen grid scratches. Care had to be taken to avoid having the silicon rubber in the way of the test fields as it impaired microscopic 69 visualization. Also while the rubber was setting, two exchanges of the medium were performed to remove the traces of acetic acid. With the two coverslips attached back-to-back, the grid was visible and the assembly was sufficiently sturdy for handling during imaging and IHC.

An alternative marked field setup was used for experiments performed using the multi-well culture plates. In this case the grid was drawn with a fine tip permanent marker pen on the undersurface of the tissue culture plate and the test coverslip was attached, with the same precautions as above, to the plate using traces of silicon rubber. The use of the culture plate as an attachment platform had the advantage of making relocation extremely easy as orientation in the horizontal plane was maintained by the mechanical stage of the microscope, i.e., no field rotation errors existed.

Ill. FURA-2 DATA ANALYSIS a. Background correction and calibration

Experimental data of 2Ca imaging experiments were initially stored in binary Attofluor data files (ADF files) that could be converted into standard comma delimited ASCII file formats. The magnitude of the data generated made it practically impossible to use commercial spreadsheets and statistical packages to perform data processing and analysis. To automate and speed up this process several programs were written in Microsoft QuickBASICTM v4.5 to perform off-line BG correction, selected ROls separation, ratio calibration, descriptive statistical calculation, and preliminary graphical display of results. Final graphs and tables were generated using Borland Quattro ProTM v5.O or Poly Software International PsiPlotTM v2.O. temperature, AM prepared acetoxymethyl capacity buffers control used IV. were Ca 2 + measured the respectively, Ca 2 + (change/mid-span) rate responses [Ca 2 +] Ca 2 BAPTA b. Kinetic stock increase LOADING of to performed Two BAPTA, response response buffering For molecule, change were study loading) over of as solution the experiments in analysis artificial cultured 20 (slope-up) prepared unless the the of the time NEURONS dimethyl-BAPTA esters of mM to was the properties using several response neurons N-(o-methoxyphenyl)-iminodiacetic consequences [Ca 2 in its was course Ca 2 solutions peak otherwise neurons. described BSS. duration. and a by studying and used parameters Quick WITH change vigorously studied stock buffers, between during (e.g. duration, Neurons recovery as in specified. BASIC using and BAPTA The anhydrous solutions a Ca 2 different of in which (Fig descriptor BAPTA 25% artificially Ca 2 . half-BAPTA were were calculations slope mixing mid-span phases program 2.6). transients OR [Ca 2 +] 1 and After of used CaBP incubated and of DMSO. ITS Finally, a These these of (slope-down). [Ca 2 +] 75% calculated increasing and 5,5’-dimethyl-BAPTA, written to ANALOGUES loading, the immuno-reactivity were remained and in describe buffer-AM of low-span the included relation Working populations in acid statistical changes the all for the aspect cultures amount the obtained peak (half-BAPTA), that above mixture and of The basal Ca 2 solutions parameters compounds the measured ratio purpose. change compare comparisons with duration were of 50% magnitude as and levels buffering at the different and room rinsed or of peak during buffer were that Ca 2 + as or these 25%, of one were the the of 70 71 twice in BSS and left for 20-30 minutes to ensure complete hydrolysis of the AM ester before testing. When loading was conducted at 33°C, at least 60 minutes were allowed for complete de-esterification of the buffer ester.

V. INDUCTION OF EXCITOTOXICITY a. Exposure protocols

Two paradigms were used for exposing hippocampal cultures to excitotoxic amounts of glutamate or NMDA. ii 30-minute exposure protocol

Test coverslips were transferred from culture medium into 2 ml of a pre warmed BSS and allowed to cool down to room temperature. BSS was then replaced with an equal volume of BSS containing the excitotoxic drug. After

30 minutes a triple exchange of the BSS was performed to remove all traces of the excitotoxic drug and cultures were incubated for 20-22 hours at room temperature prior to neuronal viability assessment.

II) Extended exposure protocol

This alternate exposure protocol was devised to avoid some problems encountered with survival of control cultures using the above method. A lOx concentrated solution of the excitotoxic drugs in BSS was filter-sterilized (Costar Syrifil-MF 0.22 pm) and appropriate volumes were added directly to the test cultures without replacing the culture medium. The cultures were incubated for the test period (20-24 hours) in 5% 2CO at 35°C prior to viability assessment. satisfactory plating well) extremely culture Wroblewski LDH when using technique negligible. glial ii and neurons. b. injury where exposure temperature), neurons no protocols. Lactate Viability alteration sensitive contribution release used 0.1 compared site. Several This increases released to and % the dehydrogenase method LDH high assumes in In protocol It Triton-X. results and assessment method some glia of index other. our required techniques and release neuronal endogenous environmental LaDue to the to may hands, also no of the using that of LDH in has LDH These the loss Total have neuronal minimal in simulated our total (1955), LDH plating many release the LDH an previously releasable were (LDH) of hands accumulated differences releasable releasable LDH excitotoxins culture is any release advantages manipulation conditions tested was released density injury and in more release may potential both also fraction cited medium Trypan in in for be LDH, closely LDH in (up control control culture only method. extremely in are understandable use (pH, the publications over growth the to pool blue of assayed and has from not usefulness in in 106 bicarbonate cultures: medium and cultures total (Koh viability measured vivo exclusion probably removed been injured High factors neurons/35 variable treatment using medium-exchange and excitotoxic that reported density during no was assessment and in Choi, neurons which diminishes quickly the after content reported medium from view unacceptably efficiency groups mm incubation. method 1987). as neuronal culture the situations one of from and a diameter and exchanges, the reliable cultured of culture sister is that of the This of cultured lysis the This high the to 72 73 culture LDH content variability that may have been caused by the variable glial number in our cultures.

While the rationale behind the use of the LDH release method is the ability of healthy cells to retain macromolecules, viability assessment using the trypan blue exclusion method relies on the ability of healthy neurons to exclude extracellular trypan blue (0.04% in BSS) from their cytoplasm unless their membrane integrity is breached. We used trypan blue exclusion in some preliminary experiments but live/dead cell contrast was too low for photography and trypan blue itself was toxic, making it impossible to use for more than a single time point assessment of viability in the same sample. ii) Fluorescent viablilty probes

Fluorescent live/dead cell markers offer a significantly improved performance over traditional viability assessment methods. They have very high contrast and sensitivity while maintaining extremely low cell toxicity levels. Combining live and dead cell markers in the same procedures gives simultaneous indications of viability index, i.e. percentage survival, that can be easily followed as a function in time. The probes we selected for use were calcein AM and ethidium homodimer (EthD).

Calcein is a fluorescein analogue with high fluorescence yield, slow fading and long retention half-life inside the cytoplasm. As a lipophilic acetoxymethyl ester, calcein AM has access to the cytoplasmic compartment where it is hydrolyzed by cell esterases to produce the polyanionic membrane impermeant calcein molecule. In healthy cells with an intact cell membrane, calcein is retained and fills the entire cytoplasmic compartment including the finest neuritic processes. When excited at 460-490 nm, calcein emits a bright

Criteria

fixation.

Nikon

iii)

conditions,

‘viability’

dead

yellow/green

recorded

Contax

viability

of

EthD

excited

access When

have when

EthD

calcein

morphology

yellow/green

Morphological

1

.0

cells

were

intact

undergoes

TMS

Morphological

excited

pM

In

present

EthD

for

to

camera.

at

even

was

order

Cell

on

assessment

was

nuclear

and 490

viable

added

inverted

progress

membranes.

is

color

of

assessed

while

if

morphology

fluorescence

using

a

to in

0.5

facilitated

nm.

they

the

water

a

When

the

neurons

viability

assess

to

slide

DNA,

several

pM

dead

living

tissue

a

of the

retained

criteria

extracellular

of

rhodamine

on

soluble,

respectively.

film

neuronal

excited

and

fixed

bathing

cells

the

cells.

by

assessment In

an

was

included

orders

culture

which

using

dead

were

the

viability

projection

Zeiss

enough

appeared

cultures,

examined

high

at

Dead

injury

culture

DNA-EthD

filter

of

cells,

Fujichrome

environment, can

particularly

490

microscope

Axiovert

a

magnitude

affinity

After

phase

of

esterase

or

be

set,

during

nm

of

i.e.

loss

as

a

medium

dying

using

used

neuronal

the

living

bright

30

with

complex

bright

DNA

condition

35M

of

1600D

useful

excitotoxicity

slides

minutes

and

activity

to

cells

membrane

the

enhancement

EthD

a

cells

or

stain.

red

microscope

identify

peak

halo

either

culture,

phase

BSS

are

onto

for

fluoresces

and

spots.

is

fluoresced

immediately

to

at

around

emission

excluded

incapable

assessment

On

at

cleave

lOx

counting

room

a

contrast

the

integrity

a

screen.

calcein

binding

experiments,

Results

final

in

fitted

number

or

neuronal

temperature,

bright

fluorescence

calcein

20x

bright

at

from

concentration

of

optics

before

of

allows

AM

with

620

to

of

were

retaining

objectives.

live

and

red

DNA,

cells

culture

and

AM.

soma,

nm.

a

of

and

and

when

EthD

that

a 74 75 fairly homogeneous cytoplasm with minimal vacuolation, one or more neuritic extensions at least twice as long as the neuronal soma, and a general outline of the soma not indicative of excessive cell swelling. Dead or dying neurons showed distended spherical somata, dark granular cytoplasm and loss of neurite integrity. The results of morphological assessment correlated very closely with those of fluorescent viability assessment methods.

VI. STAINING AND PHOTOGRAPHY a. Immunohistochemistry

Cultured neurons intended for histological processing were fixed by replacing the bathing medium or BSS with 2% calcium acetate in 4% paraformaldehyde at room temperature for 1-2 hours. The cultures were then rinsed twice with phosphate buffered saline-azide (PBS-Azide, 0.04% sodium azide in 0.1 M PBS, pH 7.4) and stored at 4°C until processed.

Three-layer IHC was performed at room temperature except where specified (Sloviter and Nilaver, 1987). The staining procedure was initiated by replacing PBS-Azide with Tris buffer (0.1 M, pH 7.6) for 15 minutes. The endogenous peroxidase activity was removed by incubating cultures with freshly prepared 20H (1 % in Tris) for 30 minutes. Residual 20H was removed and cultures were incubated with Tris-A followed by Tris-B for 15 minutes each on a 60 RPM rotating shaker. Primary antibody in Tris-B was then added and allowed to react with its target protein for 48 to 72 hours at

4°C. After the cultures were rinsed again in Tris-A and Tris-B, the second layer of the reactants (biotinylated Protein-A, 1:400 in Tris-B) was added for 1 hour. This was followed by washing in Tris-A and Tris-B (15 minutes each) and then incubation for 1 hour in avidin/biotin-peroxidase (1:1000 Vectastain 76

Elite ABC Kit, Vector Laboratories, CA) in Tris-B. A double rinse in Tris (15

minutes each) was then followed by colour development using a DAB/glucose

oxidase mixture for 30-90 minutes. The colour reaction was terminated by the

removal of DAB reaction mixture and rinsing twice in 0.1 M Tris buffer.

Cultures, on coverslips, were then dehydrated in , cleared in and mounted on standard microscope glass slides.

No attempt was made to quantify the intensity of neuronal staining and the neurons were classifies only into immunopositive or immunonegative sub- populations for the antigen under study (e.g., CaBP and CaBPN).

Primary antibody sources and dilutions

Primary antibodies for CaBP and parvalbumin (PV) were raised and characterized in our laboratory by Dr. KG Baimbridge. They were used at dilutions of 1:1500 and 1:800 respectively. Neuron-specific enolase (NSE) was obtained from Dako, Copenhagen and was used at 1:2000 dilution. Monoclonal antibodies against glial fibrillary acidic protein (GFAP, Sigma) were used at a dilution of 1:100. b. Cresyl violet (CV) staining

CV staining was performed according to standard procedures (Culling,

1974) alone or as a counter stain after other immunohistochemical methods. A schematic of the CV staining procedures is shown in Table 2.2. c. Photography

For black and white photographs, Kodak TekPan ASA 50 was exposed at the specified sensitivity rating and developed using HC-1 10 developer, dilution 77

D. Final prints were made on glossy Ilford Multigrade Ill paper (RC Deluxe). Colour slides were taken using Fujichrome-1600d exposed at 800 ASA

(fluorescent microscopy) or Kodak Tungsten 160 exposed at 160 ASA (light microscopy). Both were processed commercially.

VI. STATISTICAL METHODS

Statistical comparisons between different treatment groups (e.g., CaBP versus CaBPN neurons) were performed using unpaired t-tests. Multiple comparisons (e.g., in some excitotoxicity experiments) were performed using Newman-Keuls multiple comparisons test. In order to test the effects of buffer-loading on the kinetic parameters of 2+Ca responses, comparisons were made between the parameter values in the same group of neurons before and after the buffer-loading procedure. Estimates of 50EC values were obtained by fitting the experimental data using non-linear regression to Hill’s equation. Two statistical programs were used for data analysis: KwikStat v3.3 (for t-tests and multiple comparisons) and NonLin v2.1 (for non-linear regression fitting). 78 Sources for Materials

Gibco: CMF BSS, DNAase, DMEM, EMEM, Fetal bovine serum, HBSS, Horse serum, Laminin, L-15 medium, Trypsin. Molecular Probes: BAPTA-AM, DMB-AM, BAPTA-AM, Calcein-AM, Ethidium Homodimer, Fura-2 AM. MTC Pharmaceuticals: Pentobarbital. R&D Systems: bFGF.

Sigma: 5-FDU, Ara-C, DNAase, N2 supplement, PDL , TRIS base, and other unlisted laboratory chemicals.

Preparation of media and BSS

Media sterilization: All liquid media were filter sterilized (Costar, 0.22pm filters) and stored at 4°C for up to 4 weeks.

Dulbecco’s Modified Eagle’s Medium (DMEM) Diluted as per instructions and supplemented with: HEPES 10mM NaHCO 22 mM Serum3 (HS 5% and FBS 5%) or N2 supplement pH adjusted to 7.35 and stored at 4°C.

Minimal Essential Medium with Earl’s salts (MEM) Diluted as per instructions and supplemented with: Glutamine 0.4 mM D-Glucose 10 mM HEPES 10mM NaHCO 22 mM Serum3 (HS 5% and FBS 5%) or N2 supplement pH adjusted to 7.35 and stored at 4°C.

N2 Supplement (concentrations in working media)

Insulin 5 pg/mI Transferrin 100 pg/mI Progesterone 20 nM 100 pM Sodium Selenite 30 nM BSS for perfusion in 2+Ca measurement and excitotoxicity experiments NaCI 139mM KCI 3.5mM 79 NaHPO 3mM 24NaHCO 2 mM HEPES3 acid 6.7 mM HEPES-Na 3.3 mM D-Glucose 11 mM 2CaCI 1.8 mM 2MgCI 0.8 mM Glycine 2pM TTX 1pM pH is adjusted to 7.35 with 0.1 N NaOH Notes: - For Ca-free BSS, 2CaCI was omitted and 200 g1M2 EGTA was added. - Mg’ was omitted from BSS to obtain Mg free BSS. -2 - High extracellular potassium medium (HK) was prepared by equimolar replacement of 46.5 mM NaCI with KCI to reach a final concentration of 50mM K. Other solutions Calcein-AM and Ethidium homodimer solutions Stock solutions: Calcein-AM 1 mg/mI DMSO Ethedium homodimer 1 mg/mI 2OdH store at -80°C. Working stock solution: Calcein AM Stock solution 100 p1 Ethedium homodimer stock 50 p1 0.9% Saline 850 p1 store at 4°C.

Note: - Final cell labelling solution is prepared as lOOx dilution of the working stock in culture medium or BSS. Borate buffer

Sodium tetraborate 150 mM pH is adjusted to 8.4 using HCI

DAB Glucose oxidase mixture (per 100 ml of Tris-B)

DAB 5 mg/dl Glucose oxidase 0.3 mg/dl Ammonium chloride 40 mg/dI I-D(+) Glucose 200 mg/dl 80 Phosphate buffered saline (PBS, pH 7.4) NaCI 150mM NaHPO 8mM 24POKH 2mM Tris Buffers

Tris (Tris Base) 0.1 M adjusted to pH 7.6 Tris-A Tris with 0.1 % Triton-X Tris-B Tris-A with 0.005% BSA 81

Figure 2.1: The excitation spectrum of fura-2.

The emission intensity at 510 nm is plotted (ir arbitrary units) as a function of the excitation wavelength for fura-2 under Ca + free and saturating’ high 2Ca + conditions (reconstructed from Molecular Probes Catalog 1992-94 by Richard P Haugland, figure 20.1, page 114). The major feature of this excitation spectrum is the shift in peak intnsity from 362 nm to a shorter wavelength of 335 nm, as the ambient Ca’ changes from 0 to a saturating concentration of several millimolars. Another feature is the presence of an isosbestic wavelength of 362 nm where fluorescence is independent of 2+Ca (dotted line and arrow head). Dashed line A and B (at 334, and 380 nm respectively) represent the wavelengths used for fura-2 excitation in our imaging system. These wavelengths are chosen because they offer a wider dynamic range of the fluorescence ratio than the range offered by the peak emission lines (362 and 335 nm)

A B

> (I) C U) C C 0 1-’ (‘3 C.)x w

250

Excitation Wavelength (nm) 82

Figure 2.2: Changes in fura-2 fluorescence in a typical 2+Ca response. An idealized 2C response in a fura-2 loaded cell is illustrated showing that an increase in [Ca j1 is detected by an increase in the ratio (thick line) of the fluorescence intensity at 334 nm to the fluorescence intenity at 380 nm (F1IF1334380 thin lines). Note that with the increase in [Ca’], the Fl34 is increased while Fl380 is decreased. The change in the fluorescence ratio has a wider, dynamic range than the change in the individual fluorescence intensities.

240 2.4

200 2.0

Cl) C D Cl) 160 1tI.’J 0) C.) C C D a) C.) 0 U) 0) 120 (‘3 0 1.2 U-

80 0.8

40 0.4 0 50 100 150 200

Time (s) filter) epifluorescence filter and controls The operation. processing Figure I right 3 changer and c. b. a. functional 2.3: the components Optical Computer Excitation an This unit Illustrated filter intensified mechanisms. (CPU, microscope unit blocks unit changer, unit light also that diagram 80486-33MHz), of CCD are: including source block includes includes shutter optics camera. of A). that the the a activation (dichroic the data includes imaging image Input/output neutral monitor mirror, processor and hardware. the density and the Hg UV controller Arc intensified an filters (ImPrc), objective image Bulb, (NDF), (I/O) the monitor central CCD and CCD which shutter emission camera Camera (top and left 83 84

Figure 2.4: Schematic diagram of the perfusion chamber.

Side View Gas Mixture Heating

Glass CoverSlip

OutFlow InFlow Rubber 0-ring

Top View

well

and This

perfusion

respectively.

plate.

Figure

complete

and

C-clamp

2.5:

Arrow

a

and

1

Schematic

ml

perfusion

orientation

Thin

exchange

suction

pipette

dashed

Objective

at

diagram

(top

and

lines:

a

(in

rate

arrows

suction

left)

or

suction

out

of

of

allowing

a

1-2

indicate

of

head

perfusion-head

the

mI/mm.

line

is

well)

for

1’

reaching

the

fitted

fast

lines

Thick

Tissue

4

indicates

to

exchange

assembly.

close

used

a

solid

side

culture

inflow

to

for

arrows

C-clamp

well

the

of

continuous

well

all

or

of

bottom

the

indicate

suction

a

with

12-well

medium.

of

cells

gentle

fast

the 85 86

Figure 2.6: Kinetic parameters used in the description of 2+Ca responses. Note that the percentage values specified represent the level of the percentage of change from basal to peak values (and not from zero to peak) in the relevant units used, i.e., ratio or nM calcium. The solid horizontal bar represents the duration of exposure to the agonist (e.g., NMDA).

C E :3 C)

C-)

(Es :3 0G) (Es C

ci) a) U-

100 150 200 250 300 350 400 450 Time (s) 87

Table 2.1: Problems associated with fura-2 fluorimetry, and their remedies Compiled from Roe etal(1990).

Problems Solution(s) Interference with he 2Ca signal Minimize loading, use high (i.e., buffering Ca ) sensitivity detectors (e.g., a intensified CCD).

Sequestration of fura-2 in Reduce loading concentration of subcellular organelles. Fura2-AM and loading temperature.

Phototoxicity of the cells and Reduced UV exposure using neutral photobleaching of intracellular fura-2 density filter, use shorter (UV light effects) experimental protocols, reduce frequency of acquisition and use high sensitivity photo-detectors. Active extrusion of the fura-2 by Use anion exchange blockers (e.g., anion exchangers (uncommon in SITS and DIDS) neurons) Incomplete hydrolysis of the fura-2 Use other loading methods (e.g., AM ester. microinjection and ATP permeabilization). Quenching of fura-2 fluorescence by Use the heavy metal chelator, TPEN. heavy metals. 88

Table 2.2: Outline of the cresyl violet staining method.

1. The tissue is dehydrated by passing through a series of graded alcohol solutions (70%, 90%, 100% and 100%).

2. Tissue is de-fatted in xylene (twice).

3. Tissue is rehydrated through graded alcohol solutions (100%, 100%, 90%, 70% and 35%). 4. After rinsing in 2O,dH stain in 0.1% (w/v) CV in 2OdH (30 seconds to 2 minutes. 5. Rinse in 2OdH to remove excess CV. 6. De-stain in CV differentiator with frequent inspection until the required de staining level is reached.

7. Dip a few times in 95% alcohol, 100% alcohol and then in xylene. 8. Mount on glass slide using Permount. 89

CHAPTER 3

CHARACTERIZATION OF HIPPO CAMPAL NEURONAL CULTURES 90

The objectives of these experiments are to characterize relevant aspects of the cultured hippocampal neurons prepared as described previously. This characterization will cover the survival of neuronal cultures, neuronal 2+Ca binding proteins as well as 2+Ca responses induced by exposure to excitatory amino acids (EAA) and high extracellular K (HK).

I. SURVIVAL OF HIPPOCAMPAL NEURONS IN CULTURE

Different types of primary hippocampal neuronal cultures were used in the course of the present experiments. Culture conditions were varied to suit the particular need of different experimental paradigms. Lower density cultures

(10 neurons/cm were plated on 18 mm diameter number 1 glass coverslips and maintained)2 in a face-down position (sandwich method) in 6-well tissue culture (TC) plates with 1.8-2 ml of TC medium until used. Higher density fetal 5(2x10 neurons/cm and postnatal 5(4x10 neurons/cm cultures were routinely)2 maintained in a face-up configuration)2 on 18 or 22 mm diameter glass coverslips in a volume of TC medium adjusted to the total number of cells plated (0.8-2 ml). a. Identification of neurons in cultures

Using phase contrast microscopy together with light microscopy of cresyl violet stained cultures, neurons and glia could be distinguished on a morphological basis. The use of neuron specific enolase (NSE) and glial fibrillary acidic protein (GFAP) immunohistochemistry confirmed that the distinction between neurons and glia on a morphological basis was accurate and it was thus used routinely for experiments requiring neuronal counting. At

7 Dlv, neurons constituted approximately 90%, 80% and 80% of the cells in face-down fetal cultures, face-up fetal cultures and 4-d postnatal cultures, 91 respectively. Different neuronal types were also identified on the basis of their morphology. Pyramidal and bipolar neurons represented the vast majority of neurons and were near equally prevalent (50% and 45%, respectively) while multipolar neurons represented only a small fraction (< 5%) of the identified neurons. These proportions were closely maintained in different culture types and over different culture ages (Fig 3.1). b. Age dependence of neuronal density in culture

As the cultures grew older more neuritic arborization was evident but the total viable neuronal density, as estimated morphologically, was greatly reduced. Fetal neurons cultured using the ‘sandwich’ method showed the best survival rates. After an initial rapid loss of neurons with only 75% and 52% of the initially plated neurons remaining morphologically normal after 1 and 3 DIV respectively, the rate of cell loss slowed down with 48% and 37% of the neurons remaining alive after 7 DIV and 14 Dlv, respectively.

Fetal neurons cultured in the face-up configuration suffered a much higher initial loss and only 28% and 11 % were alive after 1 and 3 Dlv, respectively. One week after plating 8.7% of the neurons were alive but these were almost completely lost during their second week in vitro with only a 1.4% survival rate after 14 DIV. Although face-up cultures initially had twice as many cells as the face-down cultures, the latter had 12 times more surviving neurons after 2 weeks in culture.

In four-day postnatal cultures only 20% of the originally plated neurons survived after 1 DIV and only 5.4% were viable on day 3 in vitro. After 7 and 14 DIV 3% and 2.5% of neurons were still alive. 92

Fig 3.2 shows the progress of survival of neurons in the three culture types tested. c. Effects of basic fibroblast growth factor (bFGF)

Addition of bFGF to the culture medium resulted in a major change in the survival pattern and the glia/neuron ratio of postnatal cultures. Several concentrations of bFGF were tested by adding small volumes of a concentrated stock solution of bFGF containing 5% BSA to the culture medium in the wells on days 0, 1 and 3 after plating. Final bFGF concentrations were 0.1, 1, 2.5 and 5 ng/ml of the culture medium.

Aside from the lowest concentration of 0.1 ng/ml, which had no noticeable effect on the cultured cells, bFGF produced a concentration dependent increase in neuronal density and glial/neuronal ratio compared to time-matched controls. Glial overgrowth was so excessive that antimitotic treatment with Ara-C had to be instituted after 24 hours rather than the usual

48 hours. Glial morphology was also different from that in untreated cultures.

Glia were larger, more phase bright and many had a spindle-shaped appearance. At a concentration of 2.5 ng/mI, neuronal density after 3 DIV was 125% of the BSA treated control cultures (p < 0.05).

With continued examination after the cessation of bFGF treatment these cultures displayed a faster loss in neuronal viability compared to their BSA treated counterparts so that on day 6 in vitro, (i.e., 3 days after the last bFGF treatment) both cultures had an approximately equal density of viable neurons.

This trend of accelerated neuronal loss in bFGF-treated cultures continued and after 12 Dlv, neuronal survival of the bFGF-treated cultures was only 25.5% of the BSA-treated control cultures with p < 0.001 (Fig 3.3). 93 d. Sensitivity to glutamate-induced excitotoxicity

All types of cultured hippocampal neurons were sensitive to glutamate- induced neurotoxicity when tested after 7 DIV. Toxicity could be demonstrated either by brief exposures to high concentrations of glutamate or by continuous exposure for 24 hours to lower concentrations of glutamate. Details of this process will be discussed in a later section.

II. Ca1 BINDING PROTEINS INCULTURED HIPPOCAMPAL NEURONS

In vivo, immunoreactivity to a variety of 2+Ca binding proteins has been demonstrated in different hippocampal neuronal populations with varying developmental patterns. The most relevant of these 2+Ca binding proteins to the present work are calbindin-D28k and parvalbumin. a. Calbindin-D28k (CaBP)

CaBP was expressed in at least a small percentage of neurons in all the culture types tested. The frequency of its expression was a function of culture type and culture age (Fig 3.4).

In face-up fetal cultures, the number of CaBP neurons declined progressively from 59 ± 11 neurons/mm (4.9 ± 0.7% of total) after 1 DIV to 8 ± 4.1 neurons/mm (2.1 ±2 1.1% of total) after 7 DIV. After 14 DIV no CaBP neurons2 were detected in the cultures. Thus, the reduction in the number of CaBP neurons in the face-up fetal cultures continued throughout all culture ages tested (0-14 DIV) and was always larger than the reduction in the number of CaBPN neurons, i.e., the percentage of neurons expressing CaBP decreased with time in vitro. 94

In fetal cultures maintained face-down, the change in the number of

CaBP neurons had a very different pattern. From an initial plating density of 75 ± 18.8 2cells/mm (3.9 ± 1.3%), the number of CaBP neurons fell to 16 ± 12.5 2cells/mm (1.8 ± 0.8%) at 1 Dlv, and to 8 ± 7.3 neurons/mm (0.8 ± 0.7%) at 3 DIV. By the end of the first week in vitro, this2 trend of decline has given way to increased CaBP expression and the number of CaBP neurons markedly increased to 60 ± 14.3 neurons/mm (6.4 ± 1.8%) and 50 ± 16.9 neurons/mm (6.7 ± 2.0%) at 7 and2 14 Dlv, respectively (Fig 3.4A,B). 2

In postnatal neurons (face-up), the temporal pattern of CaBP expression was more like the fetal neurons in the face-down cultures than like the fetal neurons in the face-up cultures. After an initial loss of CaBP expression that peaked after 3 DIV (6 ± 5.6 neurons/mm on day 3, down from 75 ± 14.4 2cells/mm at plating time), there2 was a progressive increase in the number of neurons expressing CaBP reaching a peak after 12 DIV when 235 ± 30.2 neurons/mm were CaBP, representing 100% of all the surviving neurons. At2 19 DIV the postnatal neuronal density fell to 38.5 ± 10.0 neurons/mm but all of these neurons were still CaBP (Fig 3.4A,B). 2 b. Parvalbumin

Unlike CaBP, parvalbumin immunoreactivity, which is a generally accepted marker for subpopulations of GABAergic neurons (Celio, 1990), could not be demonstrated in any of our cultures at any culture age. c. Effects of bFGF on 2Ca binding proteins At discussed above, bFGF enhanced neuronal survival in postnatal cultures but its withdrawal caused a more rapid neuronal loss. Following 95

treatment on days 0, 1 and 3 in vitro with 2.5-5 ng/ml bFGF, the number of CaBP neurons increased dramatically such that after 6 and 12 Dlv, 69.6 ± 8.8% and 100% of the neurons were CaBP, respectively. These numbers were significantly higher (p < 0.001 at both ages) than those obtained from the BSA-treated cultures at the same ages in vitro (7.3 ± 4.9% and 59.8 ± 10.8%, respectively) (Fig 3.5).

The effects of bFGF on fetal cultures were much less dramatic and more variable from one culture to the other. However, we tested the effect of culture media conditioned with postnatal cultures treated with either bFGF or BSA, by collecting half the culture medium from these cultures twice weekly (as part of the routine of culture feeding) and freezing the harvested medium at -80°C. The conditioned media were then used to supplement the feeding medium for fetal cultures (face-down) treated with additional bFGF or BSA. While bFGF and conditioned media from BSA- and bFGF-treated postnatal cultures were capable of enhancing CaBP expression independently, combining bFGF with either of the postnatal culture-conditioned media was far more effective in increasing the number of fetal neurons expressing CaBP (Fig 3.6).

The effect of bFGF on glia, neuronal survival and CaBP expression in

postnatal cultures contrasted sharply with its total lack of effect on PV expression in the same cultures. Even in the presence of 5 ng/ml bFGF, no PV immunoreactivity could be detected.

III. Ca RESPONSES IN CULTURED HIPPOCAMPAL NEURONS a. 2+][Ca in non-stimulated neurons In all types of hippocampal cultures tested, the average resting ]2[Ca varied within a narrow range (50-150 nM). Occasionally, some cells showed 96

significantly higher fluorescence ratios which were associated with a swollen appearance and cytoplasmic vacuolation of these cells. Such cells were

excluded from subsequent data analysis. Fig 3.7B illustrates the degree of 1 variability in 2[Ca ] at rest and during an NMDA-induced 2Ca response within a set of neurons being recorded simultaneously from a single coverslip.

The tight regulation of 2+][Ca observed in our neurons appears to be dynamically controlled in the face of on-going tonic 2Ca fluxes. Thus in many cultures, changing 2[Ca +1 of the perfusion medium was accompanied by a corresponding change in the recorded 21[Ca Also the addition of EAA antagonists (e.g., MK-801 or APV) or VGCC.] blockers (e.g., nifedipine) was sometimes associated with some reduction in ]2[Ca (Fig 3.8). b. Effects of excitatory amino acids on 21[Ca In the presence of extracellular 2+,Ca agonists of all the major classes of glutamate receptor subtypes were capable of increasing neuronal 2+]j[Ca in a concentration dependent fashion. Unless otherwise indicated, all 2+Ca responses were measured in the absence of extracellular 2+Mg and in the

presence of ‘1pM TTX.

1) N-methyl-D-aspartate (NMDA): Activation of NMDA sensitive glutamate receptors increased 2[Ca in almost all morphologically identifiable neurons in culture. The 50EC forj NMDA-induced 2Ca + responses was 25 ± 1.6 pM in the absence of extracellular 2Mg (Fig 3.7A). In the presence of 0.8 mM extracellular 2Mg 200 to 400pM NMDA were required to produce an effect comparable to, that of 20 pM NMDA in the absence of extracellular 2Mg • 97 NMDA-evoked changes in 21[Ca were abolished by the classical non- competitive NMDA antagonists, iMK-801 (2 pM) and dextromethorphan (100 pM) in an agonist-dependent manner, as well as by the competitive antagonist, APV (40 pM) (Fig 3.8). Blockade of voltage-gated 2Ca channels (VGCC) using 2 pM nifedipine, resulted in a 40% reduction in the peak 2Ca responses to NMDA indicating the involvement of voltage-gated 2Ca fluxes in the NMDA-induced responses. Changes in extracellular pH )0(pH had dramatic effects on NMDA-induced changes in 21[Ca At 0pH 6.9 in 7C0HC0-buffered BSS without 2Mg 2[Ca responses.] to 2OpM NMDA32were on the average 52 ± 5% lower, than controlj responses at 0pH 7.3 while raising 0pH to 7.9 increased NMDA responses by 32 ± 7% more than the control responses (Figs 3.9, 3.15).

2) 2+Ca responses to other EAA: Transient exposure of hippocampal neurons to alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and (KA) caused a reversible increase in 2+].[Ca In the absence of extracellular 2Mg at 0pH 7.3, 8OpM KA and 4OpM AMPA evoked 2Ca responses comparable in magnitude to 20 pM NMDA. AMPA- and KA-evoked responses were sensitive to CNQX, a competitive AMPA/KA antagonist, and disappeared in the absence of extracellular 2+.Ca

Synthetic (+)quisqualate (QA) was the most potent of all the EAA tested in evoking 21[Ca responses with a Kd of 0.5 ± 0.09 pM. However, in all experiments j performed it was obviously less efficacious than NMDA, AMPA or KA (Fig 3.10). QA evoked responses were dependent on the presence of extracellular 2+Ca and sensitive to 0pH in a manner similar to other EAA. Beyond higher potency and lower efficacy, QA displayed an extreme lack of 98 homogeneity in agonist concentration-dependence of 21+][Ca responses in different cells within the same experiment. While some cells showed the familiar graded peak increases, a large fraction of cells showed ‘step function’- and ‘all or none’- type of concentration dependence (Fig 3.11). It should also be noted that, regardless of the pattern of the cell response to QA, QA-induced 2+Ca responses were abolished in the absence of extracellular 2+.Ca

Glutamate (GLUT) is the primary endogenous agonist for GLUT receptors and in cultured neurons it induced 2+Ca changes that were a composite of the pattern of activation evoked by the excitation of the GLUT receptor subtypes. Its use was generally avoided whenever possible to minimize uncertainty regarding the receptor subtypes involved. However, it was used to study excitotoxicity-associated 2+Ca changes and in 2+Ca buffering experiments (see later) where an examination of neuronal 2+Ca handling, rather than the route of 2+Ca influx, was the main research objective. c. Depolarization-induced 2+Ca fluxes

Increasing extracellular K+ to 50 mM for 20 seconds depolarized neurons and resulted in the activation of several types of voltage-gated 2+Ca fluxes and a detectable rise in intraneuronal 2[Ca +1. A variety of selective 2+Ca channel blockers were used to identify subtypes of VGCC in our cultured neurons.

1) Contribution of endogenous GLUT: In the presence of glutamate receptor blockade using AP5 (40 pM) and CNQX (20 pM), the depolarization induced neuronal 2+Ca signal was reduced by 5 ± 2% of the control value indicating only a minor contribution of depolarization-induced release of endogenous GLUT to the overall 2+Ca change recorded during exposure to 0high-K (HK) medium. Nonetheless, all voltage-gated 2Ca channel (VGCC) 99

blockage studies were conducted in the presence of a combination of APV and CNQX.

2) Subtypes of VGCC: Nifedipine, a highly selective L-type 2Ca channel blocker, caused a concentration-dependent reduction in HK-induced rises in 21[Ca with an IC50 of 11 ± 2 nM. The maximum effect of nifedipine was attained at a concentration of 1-5 pM which resulted in more than 80% reduction in HK-induced 2Ca responses. At these high nifedipine concentrations, reversal of the blockade was very slow (Fig 3.13). The residual nifedipine-resistant 2Ca changes were further reduced by 50% in the presence of 10 pM Conus geographicus snail neurotoxin, omega-Conotoxin

GVIA (CgTx). The remaining nifedipine- and CgTx-resistant component of the HK-induced 2Ca signal were abolished by the application of 1:1000 crude funnel web spider (FWS) venom (Fig 3.14).

In summary, depolarization-induced changes in 2[Ca are predominantly (>80%) due to activation of the nifedipine-sensitive L-type 2Ca channels while the rest is equally contributed by fluxes through CgTx-sensitive N- channels and channels insensitive to both DHP and CgTx but sensitive to the FWS venom. Any involvement of the low-threshold rapidly inactivating T-type 2+Ca channels can be excluded since the extended 20 seconds depolarizing stimuli and the low temporal resolution of the imaging system makes the detection of the rapidly inactivating T-channel contribution to the signal very unlikely.

3) The Effect of extracellular pH on VGCC fluxes: Changes in depolarization-induced 2+Ca responses at different pH values were very similar to those observed with NMDA-induced changes in 2].[Ca In HEPES-buffered 100

BSS, at 0pH 6.9, HK-induced 2Ca responses were 39 ± 1% lower than the control value at pH 7.4 while raising 0pH to 7.9 was associated with increased 2Ca response by 49 ± 3%. In 7C0HC0-buffered BSS corresponding values were a reduction of 37 ± 321% at 0pH of 6.9 and an increase of 78 ± 3% in 0pH of 7.9. The 50EC for 0pH of HK-induced responses in 7C0HC0 buffered BSS was 7.02 ± 0.05 (Fig 3.15). -32 Sensitivity to extracellular pH was also evident if the dominant nifedipine sensitive component of the 2Ca + flux was inhibited. In the presence of 10 pM nifedipine, residual nifedipine-resistant HK-induced 2+Ca responses showed a 40% reduction and a 25% increase compared to control values as 0pH was changed from 7.3 to 6.9 and 7.5, respectively. d. Effects of the absence of extracellular 2+Ca In addition to 2+Ca fluxes through plasma membrane voltage- and neurotransmitter-gated channels, hippocampal neurons have been reported to possess EAA- and caffeine-releasable intracellular 2+Ca stores (Murphy and Miller, 1989; Harada eta!, 1992, Uneyama eta!, 1993). Several approaches were employed to test the existence of such stores in our preparations of cultured hippocampal neurons. In nominally 2Ca-free BSS, supplemented with 0.8 mM 2Mg and 200 pM EGTA, exposure of hippocampal neurons to HK, NMDA or AMPA was not associated with any detectable increase in 21[Ca (Fig 3.16). In other experiments NMDA and EAA] quisqualate, two reported to induce release of

2Ca + from intracellular 2Ca + stores (Murphy and Miller, 1989; Harada et a!, 1992), applied at concentrations as high as 400pM could not elicit any intracellular 2+Ca release in the absence of extracellular 2+.Ca This 101

refractoriness persisted even when applications of these agonists were performed very shortly after removal of extracellular 2+Ca in an attempt to avoid or minimize the potential depletion of intracellular 2Ca stores (Fig 3.17). Also, the application of the same agonists immediately upon re-introduction of

extracellular Ca evoked increases in 2+ 2[Ca that were not different from those recorded immediately before exposurej to 2Ca +4ree medium (Fig 3.17).

A direct and presumably more specific way of activating metabotropic GLUT receptors is to use isomers of ACPD (1-amino-i ,3-cyclopentane- dicarboxylic acid) as agonists (Manzoni eta!, 1991; Manzoni eta!, 1992). In our hands, three out of the four optical ACPD isomers were capable of evoking rises in 2j:[Ca 1R,3R-, 1S3R- and 1S,3S-ACPD, but only in the presence of extracellular 2Ca Except for 3 cells out of 200 cells tested, these 2Ca responses were. completely abolished by 2pM MK-801. ACPD-induced 2Ca responses were also significantly reduced in the presence of extracellular 2+Mg (Fig 3.18-3.20). In the presence of MK-801 or extracellular 2Mg changes in ACPD-induced 2+Ca responses were similar to changes in NMDA-induced, responses suggesting that active ACPD isomers were activating NMDA channels rather than the metabotropic subtype of GLUT receptors. e. Effects of other 21+Ca release agonists

Caffeine treatment (3-25 mM) of cultured neurons did not evoke any significant change in 2+].[Ca In some cultures less than 10% of the neurons demonstrated a small transient rise in their 2+][Ca on application of caffeine (Fig 3.21A). The more consistent finding was that in the presence of caffeine both NMDA- and depolarization-induced 2Ca responses showed a 25%

the

[Ca 2 j inactivation concentrations

filling

Ca 2 +

agonist

presence

exposure

reduction

peak

Other

of

even

in

used

any

or

of

to

the

in

pharmacological

after

rates

of

releasable

caffeine

their

potential

at

presence

intracellular

up

concentrations

prolonged

of

peak

to

recovery

200

(Fig

intracellular

Ca 2 + 1

values

or

3.21B).

nM,

absence

membrane

20-minute

tools

of

stores.

which

thapsigargin,

of

NMDA-

also

100-300

Ca 2

of

was

Ryanodine,

Ca 2 -ATPase,

extracellular

exposure

consistently

and

pools

readily

pM,

which

HK-evoked

(Shmigol

induced

of

reversed

a

causes

neurons

Ca 2 +,

failed

intracellular

had

etal,

rises

no

to

no

nor

an

on

to

change

demonstrate

effect

in

irreversible

termination

did

1994).

HK

Ca 2

[Ca 2 ].

it

to

on

influence

in

+.release

ensure

At

baseline

resting

the

of

the 102 103

DISCUSSION

SURVIVAL OF HIPPOCAMPAL NEURONS IN CULTURE

Three types of neuronal cultures were used in our experiments: fetal face down (lowest plating density), fetal face-up (medium density) and postnatal face-up (highest density). In all culture types, a remarkable decline in the number of viable neurons was observed as the cultures got older. Face-up cultures, both fetal and postnatal, lost more than 90% of the initially plated neurons after 7 DIV. Maintaining fetal cultures face-down (‘sandwich’ technique), has been in use in our laboratory since 1985, and cultured neurons maintained this way displayed a significantly lower incidence of spontaneous neuronal loss after plating with an approximately 50% survival rate after 7 DIV.

Brewer and Cotman (1989) reported that the use of sandwich culture technique could improve the survival of fetal hippocampal neurons grown in serum-free media, and they attributed the improvement to a lower oxygen tension in the limited diffusion space around the neurons. The authors also argued against a contribution of glial paracrine factors in this limited diffusion space as the cause of enhanced survival. While oxidative stress in face-up cultures may harm neurons, it is our opinion that glial factors may play a significant role in the improved survival under these conditions. In our experiments, survival of face up cultures was critically dependent on higher plating density, including glial plating density, delayed application of antimitotic drugs and the limiting of the culture medium volume/well. All these techniques help reduce volume/density ratio suggesting that some secreted factors are the major determinants of neuronal survival and that these factors may well be glial in origin. 104

In order to define more precisely the potential effect of growth factors we examined the effect of adding bFGF to cultures grown in serum-free conditions.

The inclusion of bFGF in the culture media resulted in a significant enhancement of survival of postnatal neuronal cultures. This effect may be mediated via an action on glia since the presence of bFGF results in a remarkable increase of glial number and size, as well as a dramatic change in morphology.

After 3 DIV bFGF treatment was discontinued and as a result neurons, conditioned with bFGF treatment, displayed an accelerated neuronal loss far exceeding that observed in sister cultures treated with vehicle only. This phenomenon may reflect an apoptotic neuronal death similar to that observed in response to withdrawal of NGF in cultured rat sympathetic neurons (Edwards and Tolkovsky, 1994), DRG neurons (Eichler and Rich, 1989), and bFGF withdrawal in rat ventral mesencephalic dopaminergic neurons (Mayer et a!, 1993)

A very important offshoot of the significant time-dependent neuronal loss in vitro is that the results obtained from cultured neurons represent, at best, the characteristics of a highly selected neuronal population. The sensitivity to growth factors in vitro also indicates that the development of cultured neurons can deviate significantly from their in vivo program in a manner dependent upon the culture conditions. A direct extrapolation of the results obtained with the in vitro preparations to the intact animal should thus be done with extreme caution.

CaBP EXPRESSION IN CULTURED NEURONS

The expression of CaBP in cultured neurons was found to be a function 105 of the culture method and the type of neurons, i.e. fetal or postnatal. All culture types displayed an initial loss of CaBP neurons for the first 3 days in culture. In fetal face-up cultures this trend continued until no CaBP neurons could be detected after 14 DIV. In face-down fetal cultures the number of neurons expressing CaBP then increased and the ratio of CaBP/CaBPN neurons stabilized after 7 DIV. In postnatal cultures, the number of neurons expressing CaBP increased steadily after 3 DIV, in spite of the ongoing time- dependent loss in the total number of viable neurons and by 12 DIV all surviving neurons were CaBP. It would seem, therefore, that all of the postnatal neurons have the capacity to express CaBP, although the factors controlling this expression are not clear. It may be a part of the developmental process in vitro or it may be a function associated with the selected fraction of neurons that survive in vitro over time.

bFGF treatment of postnatal cultures enhanced neuronal survival and increased the expression of CaBP even after the bFGF treatment was stopped. Although the percentage of neurons expressing CaBP varied between batches of cultures, it was always significantly higher in bFGF-treated cultures than in controls, and reached 100% expression in the majority of cultures tested.

While it is possible that the expression of CaBP was a direct effect of bFGF, our data support the possibility that bFGF may be necessary but not sufficient for CaBP expression, and that other secreted factors, possibly of glial origin, may be required. This is particularly evident in fetal neuron cultures in which bFGF-treatment alone produced only a small increase in neuronal CaBP expression whereas adding bFGF in the presence of conditioned medium from postnatal cultures produced a very large increase in the number of neurons expressing CaBP. than observed was although subtype had was capable Ca 2 scattered Miller, the is differentiated cultured et cultured expression state synthesis between turn the of expressed a! glial pattern a effect also completely one Almost release (1 This of step RESPONSES 1982; 993) cells of agonists, differentiation neurons, rat single type capable with a interneurons of result, of function-like responding specific of of hippocampal that all factors in only cultured Miettinen bFGF neuronal of CaBP. expression other postnatal neuron neurons dependent Quis concluded together of in NMDA, which IN is, induction evoking that a EAA. However, neurons. CULTURED receptor sub-population at to and of responses population. and appearance tested, can leads least cultures glutamate neurons seen the AMPA with This on not Freund, that an be of neurons the in to with in at the regardless phenomenon increase On it CaBP achieved the part, and enhanced neonatal to (data all HIPPOCAMPAL is from presence use the Additionally, as rather different 1992). of increasing growth in of kainate, synthesis due with well interest CA3 basis an of not CAl in by antimitotic than to of undifferentiated or [Ca 2 a as shown), survival of factors pyramidal bFGF of subsequent affinities. an their may adult pyramidal with to extracellular to concentrations the our +11 rather action the the note NEURONS reflect culture treatment, rats an in graded data can and leads expression prototypic agents all increase than neurons the on in neurons Quis-induced cause an cultures we the constitutive conditions, vivo, us glial recent to response Ca 2 +. increase an cannot to to presence a does of a effect cells in (Baimbridge limit highly in ionophore conclude of and transition tested Quis [Ca 2 j 1 report which CaBP Quisqualate not distinguish which the in pattern a upon responses were typically few of CaBP reflect number of CaBP in that more that of in the Ray all and 106 107

were also dependent on the presence of extracellular 2+Ca and a ‘pure’ metabotropic response could not be elicited even when selective metabotropic agonists, such as 1S,3R-ACPD and 1R,3S-ACPD, were used. Interestingly 2+Ca responses induced by these ACPD isomers, which are commonly used to test for the presence of metabotropic glutamate receptors in neurons, were in our hands mediated solely by activation of the NMDA receptor-associated ionophore as indicated by the sensitivity of these responses to the non- competitive NMDA antagonists, 2Mg and MK-801.

Depolarization of our cultured neurons was also associated with an increase in ]21[Ca via VGCC. Using blockers of different types of VGCC we determined that the largest fraction of the 2Ca signal was mediated via 2Ca influx through L-type channels, while less than 20% of the signal was equally contributed by channels sensitive to CgTx or to funnel-web spider venom.

Both NMDA- and depolarization-induced 2Ca signals were sensitive to changes in extracellular pH. While increased extracellular acidity resulted in the reduction of NMDA- and depolarization-evoked 2+Ca responses, these responses were progressively potentiated as extracellular alkalinity was increased. These results are in agreements with the reported neuroprotective effect of reducing 0pH and the detrimental effects of high extracellular alkalinity on cultured neurons (Giffard eta!, 1992; Kaku eta!, 1993).

INTRANEURONAL 2Ca STORES AND INTRACELLULAR RELEASE

We conducted several experiments to test for the presence of intraneuronal releasable 2+Ca stores in our cultured neurons. In the absence of extracellular 2+Ca no intracellular release could be demonstrated using NMDA, Quis or tACPD. The latter two agents in particular have been demonstrated by to extracellular small induced neurons between continued by and lack Ca 2 + [Ca 2 j nor established presence such Manzoni many fill the ryanodine filling any On intracellular caffeine-releasable as release, authors recent in [Ca 2 ] 1 (DRG) that the detectable central etal, of of presence a agents small K+induced other basis those within observation could to and prepared 1991; signal (neocortical, release Ca 2 number of types caffeine stores were EAA-releasable of the induce these Manzoni extracellular could stores. endoplasmic from of Ca 2 + Ca 2 + also Ca 2 + must by of results intracellular was any be neurons. Shmigol unsuccessful. hippocampal 5-8 eta!, stores influx from be obtained, [Ca 2 +] 1 capable day we extremely Ca 2 +. intracellular an 1992). reticulum which, whereas eta! conclude postnatal Ca 2 + lP 3 response of particularly and (1994) This sensitive inducing these Attempts For labile stores in (Murphy brain that Ca 2 conclusion rats. example, DRG indicative of authors and our using only stem) intracellular substantial when The stores to neurons dependent and cultured demonstrate a neither central other concluded, and small has preceded of Miller, or intracellular a peripheral that been well differences Ca 2 + large increase neurons thapsigargin neurons upon 1989; emptying supported by the served caffeine store the a either in had high 108 109

blue

deep

microns.

(pyramidal,

Pale

neuronal

staining

with

100

immunostained

field

the

=

blue

field.

GFAP.

for

this

bar

(B)

neurons the

in

deep

culture,

in

and

the

stained

NSE

that

and

Calibration

neurons postnatal

identifiable

(A)

morphologies

Note

fixed

with

nuclei.

were

color).

glial

neuronal

age-matched

of

stained

counter-stained

morphologically

An

cultures

(brown

all

(B)

(CV)

different

staining

cultures

the GFAP

labels

CV

violet

postnatal

Note

which

marker Postnatal

Dlv, multipolar).

cresyl

6

represent

NSE

and

glial

color. 3.1:

After

the

Figure (A)

marker brown bipolar

for represents staining

A

B I- 0

U

0 c3 U,

microscope

represent

fetal

highest squares),

rapid

used.

In

Figure

all

2000

1000

1500

500

face-down

the

initial

0

A

3.2:

percentage

hippocampal

fetal mean

I

loss

Neuronal

field

face-up

of cultures

±

(0.245

standard

neurons

survival

survival

neuronal

(solid

mm had 5

I

that

ieviation rates

). circles)

the

in

culture

Age

slowed

vitro

lowest

of

the

and

varies

(days

of

types

down

3

initial

at postnatal

10

culture

least

with

in

tested,

after

plating

vitro)

1 the

types

2

(solid

4

fields,

different

fetal

DIV

density,

(B). squares),

15

(A).

face-down

HPF

Data

culture

they

Although

points

high

there

had

conditions

(open

power

the

was

the

20

110 a

LI-

(1)

‘ 4 -J

ci)

C) ci) C

(‘3 C) ci)

> (‘3 >

20

40

60

Figure

0 L

0

3.2

B

5

Age

(days

10

in

vitro)

15

20 111

microscope

density

cultures

BSA-treated

unpaired

and

Postnatal Figure

C,)

3

20

10

30

40

0

in

3.3:

after showed

- vitro

t-test)

cultures

bFGF

field

cultures

12

demonstrated

2DIV

neuronal

DIV

(0.245

an

enhances

treated

accelerated

(p

(solid

<

mm 2 ),

survival

with

0.001

circles).

neuronal

a

significantly

n

2.5

neuronal

Days

using

during

ng/ml

12

After

6D!V

survival

unpaired

fields

in

bFGF

loss

of

cessation

vitro

higher

bFGF

in

or

and

administration

vitro.

more.

t-test).

(p

were

(open

of

<

bFGF

0.001

significantly

HPF

circles)

12DIV

treatment, compared

=

after

high

on

2

days

lower

power

DIV

to

these

0,

using

the

in

1 112 ()

02 0

z o_

LL a, Cr)

D

symbols).

more.

neurons synthesis

detectable squares)

number

immunoreactive

culture

In

Figure

40

30

20

50 60

10 70

0

fetal/face-down A

0 3.4:

I

types.

of

detected

after

resulted

CaBP

HPF

but

Survival

an

CaBP’

cells.

initial

at

high

in

cultures

nçurons

and

different

an neurons

5 However,

I

power

drop

increase

number

(open

decreased

in Age

in

microscope

the persisted

vitro

of

in

in

squares)

(days

number

the

CaBP

fetal/face-up ages

10

steadily

absolute

(A).

in

field

is

and

of

cultured

vitro)

presented

CaBP’

The

postnatal

(0.245

until

number

cultures

percentage

no

neurons

mm 2 ),

15 neurons,

in

CaBP

of

cultures

(solid

Fig

CaBP

3.4B

n

varies

of circles)

=

active

neurons

CaBP(H

(solid

12

(same

in

fields

20

different

the

were

or 113 Percentage of CaBP Neurons

0 0 0 0 0 0 0 0 ‘.1 0 CD

-I CD c) w

0,

CD CD

0

C/) 0

0

(1

N) C

Th

+

C-)

0

z

0

C-) CU 0) C)

C) C CU

o C U)

I.

respectively,

and

neurons

(solid

Postnatal

Figure

100

40

20

60

80

0-

3

circles)

in

3.5:

at

vitro

cultures

all

bFGF

using

(p

demonstrated

the

2DV

<

increases

culture

treated

0.05,

unpaired

p

ages

with

neuronal

<

a

t-test).

0.001

significantly

tested

2.5

Days

ngfml

n

CaBP

and

6DIV compared

=

in

p

12

of

vitro

higher expression

<

bFGF

fields

0.001

to

percentage

(open

or

the

after

more.

in

BSA-treated

circles)

vitro.

2,

12DIV

6

of

and

CaBP

on

12 cultures

days

DIV.

0,

1 115

was

more)

However,

conditioned

compared

regardless

neurons

but

enhanced Figure In

12

C)

0 z

4-,

fetal a)

(‘3

C-) ci) 0)

C 0 (0

0) D 0 C

Cl)

significant

highly

produced

3.6:

40

30

10

50 60

70

80

face-down

0

and

combining

in

to

of

significant

In

medium,

the

glia

BSA

whether

fetal

(p<0.05),

a

presence

also

treated

more

neurons,

face-down

bFGF

BCM)

produced

(p<0.001

postnatal

than

increase

controls.

of

treatment

or

bFGF

additive

media

bFGF

cultures,

Treatment

a

neurons

in

modest

treatment

in

both

conditioned

Addition

(bFGF-conditioned

the

with

enhancement

bFGF

cases).

had

percentage

increase

BCM

Group

alone

of

been

effect

media

or

by

produced

in

with

treated

postnatal

in

on

of

CaBP

CaBP

conditioned

medium,

CaBPIH CaBP

FCM

with

expression

expression

9nly

neurons.

(n

expression BSA

=

FCM).

neurons

a

by

10

moderate,

(BSA

postnatal

fields

that

is

when

or 116 117

Figre 3.7: NMDA induces a concentration-dependent increase in intraneuronal [Ca ] In 69 neurons, 2[Ca +1 increased in response to exposure to NMDA (solid triangles) for 25 seconds at different concentratjons (2.5, 5, 10, 20, 40, 80 and 160 pM) in the absence of extracellular Mg + 50(EC 26.3 ± 1.6 pM). The magnitude of the peak response was concentration-dependent and approached a maximum between 80 and 160 pM of NMDA. In B, a 20 pM NMDA-induced response (n=45 neurons) is illustrated to show the extent of normal variability in cell responses by plotting the mean (solid line), mean ± SEM (dotted line) and mean ± standard deviation for the set of neurons tested.

A

1500

1200

C + 900 CD (-) 1. CD

a) 600 C-) CD .4-I

300

0 I I I . I. III 0 500 1000 1500 2000 2500 3000 3500 4000 Time (s)

eq

+

C-)

G) 1

CD C.) CD

400 200 600

800

Figure

0

0

3.7

B

100

Time

(s)

200

300 118 c’J

+ (.) (‘3 C-) 0 C C (‘3 I.

(‘3

antagonist-free

NMDA

inhibition induced

respectively,

In

Figure

31

200

400

600

800

0

neurons,

0

3.8:

antagonists

by

was

25

APV

solid

rapidly

seconds

extracellular

medium.

500

and

bars)

is

Mg 2

quite

and

application

resulted

The

completely

1000

noticeable

abolish

application

reduction

in

Time

of

complete

completely

1500

reversible

NMDA

in

of

of

this

(s)

APV

resting

experiment.

inhibition

(20

NMDA-induced

and

on

2000

pM,

[Ca 2 ]

perfusing

Mg 2

solid

of

the

on

2500

(40

triangles).

the

[Ca

application

Ca 2

neurons

ii

and

responses.

3000

increases

This

1.5

of

with

mM,

the 119 120

Figure 3.9: NMDA-induced 2Ca responses are modulated by changes in the extracellular pH. The mean of 20 pM NMDA-induced 2Ca + responses (solid triangle) in this record (n = 20 neurons) shows that an increase in 0pH to 7.9 (solid horizontal bar) was associated with a 30% increase in the NMDA-induced 2+Ca response compared to responses in a 0pH of 7.3. Lowering 0pH from 7.3 to 6.9 (open horizontal bar) was associated with a 37% decrease of the 2+Ca response.

1000

800

C + 600 C)

Co D 400 C) Co I... C 200 -

A A A

I I I I I 0 I I 0 1000 2000 3000 4000 5000 Time (s) 121

Figure 3.10: Quisqualate is highly potent in inducing intraneuronal 2Ca responses. Exposure of 49 neurons to increasing concentrations of quisqalate (Quis, open squares) resulted in concentration dependent increases in [Ca+] with 50arEC below 1 pM of Quis. In spite of this maximum Quis-induced1 Ca + potency, responses were much lower than those induced by a 20 pM of NMDA (25 seconds exposure, solid triangle) which induced a submaximal response to NMDA (compare to figure 3.7a).

800

600 C +

(‘3 C-) 400 (‘3

ci) ci (‘3 4-, C 200

0 0 500 1000 1500 2000 2500 3000

Time (s) 122

Figure 3.11: Quisqualate-induced 2Ca responses show that QUIS does not have a homogeneous potency on hippocampal neurons.

A characteristic feature of the Quis concentration response curve was the extreme heterogeneity of its potency on different neurons. This is illustrated in this graph where 3 neurons, from the same experiment, were exposed to increasing concentrations of Quis (solid triangles; concentrations from left to right: 0.1, 0.2, 0.5, 2, 5 and 20 pM). While the overall 50EC of Quis in this experiment was 0.65 ± 0.17 pM, individual cells showed wide patterns of response including a steep ‘all-or-none’ response pattern (e.g., middle panel).

400 -

A 200

0

400

B 200 -

0 A A A A A A

400 -

C 200

0 0 500 1000 1500 2000 2500

Time (s)

C

with

cells.

extracellular

concentration:

Unlike

homogeneous

Figure

B

A

parallel

A

Quis

3.12:

representative

(Fig

Mg 2 i

(or

NMDA-induced

1000

1

1000

1500

than

1000

500

1

500

500

2.5,

500

500

proportional),

0

3.11),

0

0

0

Quis-induced

5,

displayed

10,

NMDA-induced

sample

20

750

Ca 2 +

and

a

but

of

fairly

responses.

3

not

4OpM

responses

neurons

Time

homogeneous

equal,

Ca 2

1500

from

(s)

show

increases

responses

in

left

neurons

a

to

2250

potency

graded

right

in

(solid

are

[Ca

in

rsponse

more

on

the

triangle;

+11.

3000

all

absence

responding

pattern

NMDA

of 123 124

Figure 3.13: Nifedipine inhibits depolarization-induced 2Ca responses In 41 neurons, exposure to high 0K+ (HK; 50 mM) for 20 seconds resulted in an increase in the mean t][Ca which was completely reversible upon restoration of low mM)2 K+ Exposure of (3.5 0 neurons to nifedipine (solid horizontal bars) at concentration. of 0.05, 1 and 5 pM caused an increasing inhibition of the HK-induced responses (80% reduction at 5 pM nifedipine compared to control responses). Reversibility of inhibition at this concentration was slow and incomplete 1 hour after cessation of exposure to nifedipine.

800

600

+

C-) 400 0.05gM 1.0gM 5.0gM 0 C)

200

0 I I I I 0 2000 4000 6000 8000 10000

Time (s) ______

125

Figure 3.14: Multiple types of VGCC contribute to HK-induced 2Ca responses in cultured hippocampal neurons.

In the presence of APV and CNQX (40 and 20 pM, respectively, solid bar) HK induced 2+Ca responses were not significantly changed. Under these conditions, nifedipine (10pM, top open bar) caused more than 80% reduction in the HK-induced responses. The nifedipine resistant component of the response was further reduced by 50% in the presence of CgTx (middle open bar). The remaining nifedipine/CgTx resistant response was nearly abolished by 1:1000 crude funnel web spider (FWS) venom. Graph represents the mean response in 9 neurons.

500 APV 4OpM + CNQX 42OuM

400 I Nifedipine 10pM

C -

300 - omega-CgTx 5pM C-)

D FWS Venom j5 200 - 1:1000

100-

0 I I I 0 2250 4500 6750 9000 Time (s) 126

Figure 3.15: HK- and NMDA-induced 2Ca responses are modulated by changes in extracellular pH. Changing 0pH from 6.7 to 7.9 resulted in a 72% and 99% enhancement in the magnitude of HK-induced response (solid triangles) and NMDA-induced responses (open triangles), respectively (n=59). The two pH ramps indicate the change in 0pH which occurred in steps of 0.2 pH units, and the horizontal segments indicate the control 0pH of 7.3. For both HK and NMDA-induced responses, extreme 0pH effects were rapidly and completely reversible.

800

600

7.3 400

ci) C.) CD 200 I AAAAAAAAA A I I 2zD 0 0 2000 4000 6000 8000 10000 12000

Time (s) 127

Figre 3.16: HK-induced and EAA-induced Ca responses are abolished in + 2 Ca free media.

In 55 neurons, exposure to HK (open triangles), 200 pM NMDA (solid triangles) and 40 pM AMPA (solid diamonds) resulted in 2Ca + responses that were completely abolished if the exposure to HK or EAA was performed in the absence of extracellular 2+Ca (solid bars).

750

500 +

CD C)

CD

ci) C) 250

0 0 3000 6000 9000

Time (s) 128

Figure 3.17: The effects of Ca-free medium on NMDA-induced Ca responses. 2 2 Exposure of 61 neurons to 200 pM NMD for 25 seconds (open triangles in the presence of 0.8 mM extracellular Mg + resulted in an increase in [Ca +], that was totally abolished in nominally Ca-free perfusion (solid horizontal bar) even when NMDA was applied within2 5-7 seconds of perfusion with 2Ca free medium. Within 5 seconds of resumption of perfusion with standard perfusion medium, ANM (200 pM, 25 seconds) induced a response that was identical to the pre-’Ca9 +.4ree perfusion.

400

300 C +

(‘3 C) 200 (13 D

G) C-) (‘3 .4-’ C 100

0 0 300 600 900 1200 Time (s) 129

Figure 3.18: The effect of extracellular 2+Mg on the calcium responses to NMDA and ACPD isomers. In 47 neurons, switching from Mg-free to 0.8 mFMg-containing perfusion medium resulted in a 2depression of the Ca + responses induced by the transient application of NMDA (200pM, solid triangles), 1S,3R-ACPD (1 mM, open squares), 1S,3S-ACPD (1 mM, rosses) and 1R,3R-ACPD (100pM, solid squares). Removing extracellular Mg + was2 associated with fast recovery of the responses. The rank order of responses to different agonists wa retained under both perfusion conditions. Percentages of depression by Mg’ (±SEM) were 68.1 ± 3.05%, 75.6 ± 2.90%, 71.9 ± 2.66 and 66.7 ± 3.26, respectively.

800

700

600 C + 500 C’4 CD Ci 400 CD D a) C.) 300 CD 4-I C 200

100

0 0 1000 2000 3000 4000 5000 6000 Time (s) 130

Figure 3.19: MK8O1 inhibited ACPD induced rises in neuronal free calcium. In 45 neurons, the addition o2pM MK-801 to Mg-free perfusion medium abolished the increase of [Ca induced by NMDA2 (100 pM, solid triangles), 1R,3R-ACPD (100pM, open squares), 1S,3S-ACPD (1 mM, crosses) as well as 1S,3R-ACPD (1 mM, solid squares). The figure represents the mean [Ca of 38 cells. The effect of MK8O1 was not reversible even after prolonged]2 (>1 hr) perfusion with MK-801 free mdium. Notice the use-dependent effect of MK 801 on the NMDA-induced Ca responses.

600

500

C 400 + c..J CD ci 300 CD

0a) CD 200 1-’c

100

0 0 500 1000 1500 2000 2500 3000 3500 4000 4500 Time (s) 131

Figure 3.20: An example of a single cell where MK8O1 did not abolish the calcium response to 5 mM 1S,3R-ACPD.

In 191 of 194 neurons, MK8O1 abolished completely both NMDA- and ACPD induced Ca + responses (Fig 3.19). In the remaining 3 cells (only 1 cell is shown), [Ca increased in response to the application of 1S,3R-ACPD despite the2 presence of 2 pM MK8O1. These responses were, however, much reduced comparedj to the control responses (Numbers below the symbols represent the agonist concentration in millimolars).

400

A NMDA 350 D 1S,3R-ACPD

• 1R,3S-ACPD 300 0 1S,3S-ACPD c • 1R,3R-ACPD + 250 C”

C-) 200 D 2pM MK8O1

AL1O • L1 AALI .2 1 1 1 1 5 5 .2 .2 5 1 1 I 0 I I 0 20 40 60 80 100 Time (mm) 132

Figure 3.21: Caffeine induces only a small increase in intraneuronal 2+].[Ca In 56 neurons, exposure to 10 mM of caffeine (solid horizontal bar) resulted in a transient small increase in +][Ca that was not potentiated when the exposure to caffeine 21 highK+ was repeated after a depolarizing stimulus using (open triangle)(A). In (B), 38 cells were exposed o 20 pM NMDA for 025 seconds (open triangles). The NMDA-induced Ca + response was reduced, in a reversible manner, in the presence of 10 mM caffeine (solid horizontal bar).

A

600

500

C 400 + C.,’

C-) 300 D

U) C-) ( 200 C

100

0 0 500 1000 1500 2000

Time (s) 133

Figure 3.21 B

600

C 400 + c’4 CU C-)

(U D a) C) CU 200 C

0 0 500 1000 1500 2000 2500

Time (s) 134

CHAPTER 4

THE EFFECTS OF ARTIFICIAL BUFFERS ON NEURONAL 2Ca + RESPONSES adjusted DMSO-treated before The duration, 33°C employed minutes vigorously ester used neurons derivative, THE neuronal experiments the VGCC Statistical paired derivative, intraneuronal different first LOADING in (buffer-AM), (20 Since Loading This any t-test or order at with to protocol cells cultures. NMDA or a significance mixed room measurements dimethyl-BAPTA the on chapter final the 60 to presented treatment to the Ca 2 were control the neurons PROCEDURE highest compare pM, 20 achieve temperature buffer-AM receptors in required used kinetics prepared mM will balanced washed buffering 60 group of a concentration in groups with buffer-AM examine minutes, different the final the this were concentration of and concentration from (DM8) in was the & pre- salt Ca 2 (20 effects concentration capacity, chapter CONTROLS: were BSS measured made. artificial the pM, 33°C). a included and + solution levels stock was 20 without responses performed consequences of present post-loading 15 were mM using achieved the of of solution Ca 2 minutes, After using of (BSS). with enhanced stock the buffers the performed of 20 in BAPTA induced 20 corresponding buffer the loading using fura-2 Ca 2 the or solution by was Two pM values. RT). 60 DMSO was of buffer-treated incubating neuronal an and BAPTA pM of buffer prepared increasing microfluorimetry. on by loading for The estimated unpaired the in its for fetal activation concentration the Comparisons DMSO second for buffer-AM dimethyl 60 or acetoxy-methyl Ca 2 + required protocols face-down cultured in 20-40 its minutes the t-test. DMSO, group using and dimethyl of buffering. protocol minutes either for across a were (e.g., All at a 1 5 the 135 136

0.1 % DMSO if the highest buffer-AM concentration was 20 pM). A second control group was treated with half-BAPTA-AM (hBAPTA-AM), a control compound with a BAPTA nucleus modified to eliminate its 2Ca binding capacity. Neither DMSO nor hBAPTA treatment resulted in any significant change in the kinetics of the 2+Ca responses when compared to the controls. For the rest of this chapter, all the comparisons reported use the hBAPTA group for reference unless comparing the BAPTA-treated group to the DMB treated group.

As a result of the increased intracellular 2+Ca buffering capacity after loading neurons with BAPTA or DMB, in situ calibration using the 2Ca ionophore, Br-A23187, becomes increasingly difficult. For the purpose of this section, the results are presented as background-corrected but uncalibrated ratios. Statistical comparisons were done on experiments performed under identical optical and gain settings. Under such conditions a larger ratio value represents a larger absolute concentration of 2+Ca in spite of the non-linear relationship between the fluorescence ratio and the corresponding 2+Ca concentration. Reference to 2+Ca ‘concentration’ in this section implicitly means 2+Ca concentration as estimated by the background-corrected fluorescence ratio’.

I. BAPTA EFFECTS ON SHORT 2Ca TRANSIENTS

a) Changes in Kinetic parameters: Loading neurons with BAPTA-AM (20 pM, 15 minutes, RT) resulted in significant changes in the magnitude and time course of the 21[Ca responses induced by brief exposure of neurons to NMDA (20 pM,j 25 seconds) or to high extracellular K (HK; 50 mM, 20 seconds). Both treatments resulted in nearly equal 2+Ca responses and the 137 changes recorded in these responses after buffer loading were similar in both the depolarization-induced and NMDA-induced 2Ca transients. The kinetic parameters used in the analysis are illustrated graphically in Fig 2.6.

In 14 experiments using NMDA, baseline 2+Ca levels were similar in the control and BAPTA-loaded cultures. Peak 2+Ca levels were, however, significantly lower in the BAPTA-loaded neurons (74.3 ±8.7% of the control values, p < .001). Changes in 21[Ca were slower in the BAPTA-loaded group ] with in compared to the control group the rate of the increase 21[Ca being of ] 52.6±13.1% control while the rate of recovery was only 23.7±9.4% of the control value (p < 0.001 in both cases).

BAPTA-loading resulted in 2Ca responses that not only had smaller peaks and slower rates of rise and recovery, but were also of longer duration than the control responses. The mid-span width was 160.1 ± 28.6% of control while the low-span was 222.4±69.2% of the control values (p < 0.001). With smaller peaks and more prolonged responses, the aspect ratio of the 2+Ca transient was also significantly reduced in the BAPTA-treated neurons (43.4±13.6%, p < 0.001). An illustration of the effects of BAPTA-loading on transient 2+Ca responses is shown in Fig 4.1.

b) Kinetic differences between the effects of loading with BAPTA and

DMB: In 10 experiments, fetal neurons were loaded with DMB, which has higher affinity for 2Ca than that of BAPTA. Using the same loading protocol as above (20 pM, 15 minutes, RT), these DMB-loaded neurons displayed changes in the kinetics of 2+Ca responses that were similar to those observed in BAPTA-loaded neurons both in the percentage of change from control values and in the statistical significance levels (see Table 4.1) with two exceptions.

show

BAPTA-AM.

medium.

those

recovered

experiments

loading

medium

experiments,

[Ca 2 +]

the

using

after

order

from

Although

were

were

control

II.

significantly

neurons

Both

AM

BASELINE

loading

those

any

induced

then

fura-2

to

attempted. the

a)

In

with

in

treatment

esters

at

compare

[Ca 2 ],

(p

initial

Simultaneous

mid-span

change

the

quickly

the

room

compared

in

<

respectively).

fluorimetry.

DMB-AM

In

with

shorter

exposure

responses

the

neurons

by

Ca 2

0.05

was

experiments,

addition,

temperature

increased

depolarization

to

in

Ca 2 +

control

The

the

with

performed and

and

the

pre-exposure

CHANGES

in

between

Ca 2

and

effects

(Fig

of

hBAPTA

DMB-loacled

recording

low-span

background

responses

in p

raw

cultures,

neurons

hBAPTA-AM

<

the

during

4.2).

In

buffers.

the

fluorescence

0.005

was

all

of

at

coverslips

buffer-loaded

DURING

or

was

cases,

loading

buffer

room

from

Similar

parameters

exposure

levels

associated

to

by

within

additional

respectively)

neurons

fluorescence

20

performed

For

NMDA

temperature

cell-free

loading

Ca 2 +

(10

pM

on

of

THE

changes

that

these

the

data

neurons

perfusion

to

out

BAPTA-AM

compared

neurons

receptor

experiments

with

of

same

BAPTA

responses

received

buffer-AM

on

areas

experiments,

from

of

before

the

(Table

during

were

an

neuronal

10

set

while

with

Ca 2 +

cells

LOADING

were

on

with

increase

activation,

and

different

any

of

to

also

4.1).

in

were

the

the

BAPTA,

ester:

monitoring

neurons

that were BAPTA-loaded

responses

4

the

the

kinetically

Ca 2 +

Ca 2 +

observed

out

coverslip

application

incubation

as

in

ester-free

responded

perfusion

performed

PROCEDURE

In

treatments.

of

the

and steep

DMB

measurements

responses

13

before

4

mean [Ca 2 +] 1

were

[Ca 2 j 1

distinct

out

during

did

or

as

of with

and

of the

to

not

in

15 138 blocker Ca 2 presence to Ca 2 , the required. than induce response Since to Cells responses widely perfusion [Ca 2 i cultures associated the altered (Fig responding exposure examine the presence beginning 4.2). a that the levels ci hi buffer-AM BAPTA-AM result Ca 2 + but (Church background during that The Cell-to-cell of to association showed Second, did to to the with the cells loperamide, specific indicating BAPTA-AM of mechanism of influx NMDA responded not or magnitude exposure mechanism(s) a the et an (Fig after application non-specific respond a we (2 al, via increase lipophilic fluorescence close variability: channel and 4.3A) experiments) between 1994), the that used the BAPTA-AM to to by HK of of association to termination activation Ca 2 BAPTA-AM the did BAPTA-AM loperamide, an in treatments the activation (lower either AM to involved. perturbation such [Ca 2 increase Within AM not test individual flux (Fig ester, did stimuli, indicate compounds, +1 panel, did cell-to-cell the of 4.3B). through to of cultures not in exposure suggested loading-induced additional specific in not a (top First, route the exposure a potent Fig [Ca 2 +] cause neuronal of possibly set any cause magnitude and The the in 4.3A,B). the of that of variability fluorescence Ca 2 + the individual with the middle broad any cell use 3 that experiments plasma an as to showed experiments, glial response absence Ca 2 AM change well increase membrane of an conductances the spectrum of increases panels, DMSO cells, increase and ester membrane as peak buffer-AM cell flux. an of data in intensity to peak in never responses were elevation suggestive the alone [Ca 2 +] 1 extracellular Fig buffer-AM permeability [Ca 2 In even Ca 2 + in in from Ca 2 + the baseline 4.3A,B). performed [Ca 2 ] 1 [Ca 2 ] 1 : responded was rather might was ‘1. shifts in non- channel of varied Fig not of 139 at in

which

ensure

conditions

changes.

level.

prolonged

the

the

BAPTA

for

Ill. concentration

the

HK-

4.5B).

during

displayed

independent:

prevented

4.4

[Ca 2 ]

(25

neurons

slower

absence

BUFFER

results

pM)

or

shows

neurons

Changes

In

d)

a

its

NMDA-induced

or

Thus,

to

plateau the

to

Buffering

To

loading

the

large

DMB

of

or

rate

of

baseline

that

the

to

EFFECTS

of

face

extended

test

interrupted,

experiments

the In

usual

was

reach

were

the

of

in

perfusion

increases and

during

all

Ca 2 +

of

the

were

buffering

the

rise

and

increased

buffer.

the

levels.

kinetic

briefly

an

exposed

a

effects

latter

ON

in

plateau

Ca 2 +

level

Ca 2 +

experiments

a

buffer-AM

enhanced

independent

[Ca 2 j

BAPTA-AM

medium

in

subsequent

EXTENDED

using

exposed

changes

Smaller

action

may

was

[Ca 2 ]

responses

of

during

challenge,

to

Ca 2 +

enhanced

250

be hBAPTA-AM

reached.

rather

buffering

resulted

loading-induced

of

peak

of

where

(25

the

associated

may

phenomena.

pM

level

BAPTA

induced

NMDA-

Ca 2

critical

than

were

several

seconds)

loading

Ca 2 +

glutamate

be

after

neuronal

in

direct

For

capacity

RESPONSES

responsible

actual

and

a

observed

Ca 2

importance

or

where

values

these

fast

with

a

experiments

procedure

HK-induced

the

BAPTA-AM

Ca 2 +

to

This

[Ca 2

for

depression

buffering

and

response,

for

NMDA

BAPTA

no

experiments,

Ca 2

in

longer

was

although

load

complete

Ca 2 +,

kinetic

for

neurons

in

(Fig

increases

responses

cytopathological or

also

vivo

loading

is

were

Ca 2 +

capacity

responses

intervals

adding

HK

of

longer

the

4.5A).

changes

supported

baseline

where

loaded

the

recovery

may

conducted

buffer

time

responses

(Figs

are

plateau

than

loperamide

observed

under

to

reflect

required

were

in

with

Ca 2 +

4.4,

the

in

of

by

140 in 141

loading was also increased in order to test the effects of a much higher buffering capacity on the plateau 2Ca levels under high 2Ca flux conditions.

Several cultures were tested for the consequences of loading neurons with BAPTA-AM or DMB-AM (20 or 6OjiM, 60 minutes, 33°C) on 2Ca responses induced by short exposures to NMDA (20 pM, 25 seconds) compared to short (25 seconds) and long (3-4 minute) exposure to glutamate (250pM). The resulting effects on 2Ca responses are shown in Fig 4.6(A-E). Control cultures were prepared by treating them with DMSO (0.3%) or hBAPTA-AM (60 pM) under conditions identical to those used for loading the buffer-AM (60 minutes, 33°C).

In the hBAPTA-AM loaded neurons (Fig 4.6A), responses to NMDA (20 pM, 25 seconds), glutamate (250 pM, 25 seconds) and to glutamate (250 pM, 4 minutes) were identical to the responses obtained from the 0.03% DMSO treated group and from ‘naive cultures (data not shown). Except in neurons loaded using 2OpM of DMB-AMt (Fig NMDA-induced increases in 4.6D), 2[Ca were severely reduced in both the BAPTA-loaded groups (20 and 60 pM)i (Fig 4.6CE) and in the 60 pM DMB-loaded group (60 minutes) (Fig 4.6B) compared to the hBAPTA-loaded group (Fig 4.6A). This buffering effect on the

NMDA-induced responses was more evident than that observed in neurons loaded using the ‘20 pM, 15 minutes, RT’ protocol, and the larger effect confirmed that the buffering capacity was much larger in neurons loaded using the ‘20 or 60 pM, 60 minutes, 33°C’ protocol. Like their effect on NMDA induced 2Ca responses, BAPTA and DMB loading (20 pM, 60 minutes, 33°C) induced a marked attenuation of 2+Ca responses that were induced by a short (25 seconds) exposure of neurons to glutamate (Figs 4.6D, 4.6E) glutamate continued recovery minutes evident DMSO glutamate 40 cultures acids, DMB-AM exposure are EXPOSURE IV. slower recovery during Ca 2 + exposure cells transient pM, exposed BAPTA compared In for Under peak In (0.3%), the in with at after were the began contrast phases to (60 responses, the extended to presence resulted exposure. 8-minute increase responses EFFECTS 250 control to 250 complete excitotoxic pM, the buffer-loaded initially graded glutamate, immediately, to pM being pM termination to 60 control in of intervals. and intervals cultures, the glutamate the glutamate a minutes, challenged ON during glutamate. recovery concentration-dependent particularly rapid recovery situations markedly magnitude Ca 2 groups. and groups rise for and of extended i.e. We 33°C). probably on was to exposure LEVELS 25 in with it phases cultures therefore diminished On baseline neurons in prolonged compared In characteristically [Ca 2 seconds, of relatively vivo, spite termination 3 the In Ca 2 + concentrations other 11 DURING of the to peak treated such of loaded Ca 2 + which the tested NMDA. followed (Fig flux to absence NMDA- this unchanged endogenous extended Ca 2 + responses the as EXCITOTOXIC levels 4.6). situations, of relative declined with with the cerebral control exposure displayed Exposure and response of by effects of hBAPTA-AM either usually extracellular responses a in lack glutamate-induced NMDA: to excitatory only 30-minute groups, ischemia, the the NMDA BA of to of a to within to a buffer GLUTAMATE fast Ca 2 + PTA-AM a glutamate, effect 250 little 2.5, a 30-minute with were 3-4 (60 were initial amino neurons Mg 2 , pM 3 loaded in changes 10 on minute pM) the the or much and the 142 or 143

component that achieved more than 80% of the total recovery within 10 minutes followed by a slower component that completed the restoration of j2[Ca to pre-exposure levels within an additional 20-30 minutes (Fig 4.7A,B). In the BAPTA-loaded neurons (Fig 4.7C), NMDA-induced 2Ca responses were significantly diminished to the extent that exposure to 2.5 pM NMDA was not accompanied by any detectable rise in 2+][Ca at all. However, exposure to 10 and 4OpM NMDA were associated with an increase in 21[Ca but responses to both ] concentrations were blunted and prolonged compared to those in the control cultures. The incomplete recovery of 2[Ca between consecutive NMDA challenges, separated by 8 minute intervals,i resulted in a staircase appearance of the 2+],[Ca presumably due to the accumulation of the residual 2+.Ca Responses induced by a 30-minute exposure to glutamate were, however, not reduced in peak magnitude in spite of the considerably slower rate of increase in 21+][Ca compared to control cultures. The small recovery component normally occurring during exposure to glutamate was characteristically lost in the BAPTA-loaded neurons and actually replaced by a slow and steady increase in 21+][Ca After resuming perfusion with glutamate- free BSS, recovery of 21Ca .was so delayed that 40 minutes later barely 40% of recovery was achieved. In the DM8-treated cultures, 2+Ca responses followed a pattern similar to that observed in BAPTA-loaded cultures with two differences. First, NMDA-induced responses (at 10 and 40 pM) were less diminished than in the BAPTA-loaded cultures although recovery was still incomplete at the 8-minute response spacing that was used. Second, the rate of increase in 2[Ca + ] on exposure to 250 pM glutamate was faster than in the BAPTA-loaded neurons (Fig 4.7D). 144 DISCUSSION

The present experiments were designed to gain some insight into the potential effects of artificial buffers, with well established physico-chemical properties, upon stimulated increases in 21+][Ca in neurons. The experiments were facilitated by the use of cultured neurons (making direct measurements of 2[Ca with fura2 technically straightforward) and the use of membrane permeantj derivatives of artificial 2+Ca buffers. Effects on 2[Ca +11during the loading procedure: Exposure of neurons to the acetoxymethyl esters of BAPTA, DMB or hBAPTA (but not to DMSO) was, unexpectedly, associated with a rapid and largely persistent increase in baseline 2[Ca +1 that recovered completely when the exposure to AM compounds was terminated. This effect was not observed in the absence of 20+][Ca nor in cells that did not respond to NMDA or HK before the buffer loading. The addition of loperamide, which is a voltage-gated 2Ca channel blocker, in the continued presence of BAPTA-AM, resulted in complete reversal of the increase in 21[Ca induced by BAPTA-AM. We concluded from these findings that 2Ca j influx through specific 2Ca channels, most likely VGCC, was the primary event responsible for the increase in 2[Ca that was induced by the AM ] three derivatives tested. Such an effect on 21[Ca during buffer loading is interesting since several studies have reported ani increase in the amplitude and duration of EPSP in rat hippocampal slices and in acetyl choline release in frog neuromuscular junction observed during exposure to BAPTA-AM and DMB-AM respectively (Niesen et a!, 1991; Robitaille and Charlton, 1992). The authors of these studies suggested that the buffer-AM effect was mediated via inhibition of -2Ca dependent potassium conductance secondary to enhanced 2+Ca buffering. To 145 the contrary, our results suggest that the increase in 2[Ca during loading may be independent of 2+Ca buffering for several reasons.i First, the effect on 2[Ca appeared very rapidly reaching a plateau that lasted for the duration of exposurej to the AM compounds with a similarly rapid recovery following their removal. Unlike this square wave response, the increase in buffering capacity should assume a ramp-like waveform that reaches a peak when the buffer-AM is completely hydrolyzed, probably several minutes after termination of exposure to the buffer-AM. Second, exposure to hBAPTA-AM resulted in an increased 2[Ca despite the fact that the hydrolysis products of hBAPTA-AM have no Caj buffering ability. Thus, the observed increase in may be 2 2[Ca a direct effect of the BAPTA nucleus AM ] and/or the esters with the activation of VGCC, directly or secondary to perturbation of the membrane lipids by the lipophilic esters, being a possible mechanism. Effect on resting 21[Ca Cytoplasmic BAPTA or DMB had minimal effect on the resting ]2[Ca :]suggesting that enhancing 2Ca buffering capacity did not affect the cellular mechanisms responsible for setting 2+][Ca at rest (e.g., 2Ca +..ATPase and basal ‘leak’ influx). The only exception to this was observed with the maximum loading regimen (60 pM BAPTA-AM at 33°C) where a very small increase in resting i21[Ca was observed (0.1-0.2 ratio units). Effects on transient 2+Ca responses: When neurons were exposed to NMDA (20 pM for 25 seconds) or high extracellular K (HK, 50mM for 20 seconds), 21+][Ca increased rapidly to a plateau and then recovered completely and rapidly following wash out. Loading neurons with BAPTA or DMB resulted in changes consistent with the effects of increased neuronal 2+Ca buffering capacity, such as lower peak 2+Ca responses and a damping of onset and recovery kinetics. However, these changes did not result in a ‘miniature 146

response’ as the recovery was so slow and prolonged that the duration of the

recovery phases and the responses in general were considerably longer-lasting in the buffer-loaded compared to control neurons. The prolongation of the recovery phase may have major functional consequences on the net biological effect(s) of the 2Ca buffer. Examining the 2[Ca responses shown in Fig. 4.1 raises the possibility that in buffer-loaded neurons, while Ca-mediated processes with low affinity may be reduced, or even not evoked2 at all, other ‘high-affinity’ processes may be stimulated for longer period of time when compared to control neurons exposed to the same stimulus (e.g., 20 pM NMDA for 25 seconds).

The duration of 2+Ca responses was assessed using the mid-span and low-span kinetic parameters, both of which were significantly prolonged after neurons were loaded with BAPTA or DMB. Moreover, mid- and low-span intervals were the only parameters that displayed significant difference between the two buffer-loaded groups used (Table 4.1) with BAPTA loading being associated with longer 2Ca responses than the DMB loading. A possible explanation for this difference may be that DMB has a higher affinity for 2Ca than that of BAPTA, and at resting 2j,[Ca as well as any other 2],[Ca the fractional saturation of DMB will be larger than for BAPTA. Thus the ‘effective’ buffering capacity available for the stimulated 2Ca influx may be more for BAPTA-loaded neurons than for DMB-loaded neurons when the total buffer concentration is the same (as might be reasonably assumed when neurons are loaded under identical conditions). As soon as the available buffering capacity is saturated by 2Ca influx, the kinetics of 2Ca change should be similar to those of control value, i.e., faster. This possibility is supported by the conclusions of Augustine eta! (1991) concerning the differences in effect of 147

dibromoBAPTA, BAPTA and DMB in squid giant presynaptic terminals on neurotransmitter release.

Effect of 2Ca buffers on prolonged 2Ca responses: The ability of artificial 2+Ca buffers to reduce the amplitude of 2+Ca responses during situations of excessive stimulation is an essential requirement for neuroprotection under the excitotoxicity/calcium hypothesis of neuronal death.

Our results indicate that while both BAPTA and DMB were capable of reducing peak responses induced by transient stimulation, peak 2+Ca levels during prolonged exposure to NMDA or glutamate were hardly changed, even though the rate of rise of 2[Ca to those peaks was markedly reduced in buffer- loaded neurons, andj more so in BAPTA-loaded than in DMB-loaded neurons. An equal peak 2[Ca in the presence of 2Ca buffers also indicates that the net 2Ca influxi was larger in the presence of intracellular BAPTA or DMB. A larger net influx of 2+Ca could be a result of reduced 2Ca +.mediated inactivation of either VGCC (Kohr and Mody, 1991) and/or NMDA channels (Legendre eta!, 1993). Recovery of ]2[Ca to pre-stimulation levels should, understandably, be extremely slow in the presence of additional intracellular buffers as a result of the larger net 2+Ca influx during stimulation, and the competition between the recovery mechanisms (e.g., 2+Ca pumps and exchangers) and the high affinity 2Ca binding sites of the added buffer. Larger 2+Ca influxes and slower recoveries are probably the basis for the ‘staircase’ appearance of the 2+Ca profile observed in buffer-loaded neurons during repeated stimulation. The buffer in this situation may be acting as an integrator of the calcium signal (i.e., a capacitance effect). The repeated stimuli that normally leave no residual effect on j21[Ca can thus temporally 148

summate in the presence of the buffer giving rise to a high and prolonged 2Ca signal with potentially harmful consequences.

EFFECTS OF BAPTA UNRELATED TO BUFFERING ACTIVITY

A very important but largely ignored aspect of the effects of artificial 2+Ca buffers on cells are the non-buffering properties of these buffers. A large number of studies have utilized BAPTA and its analogues to analyse the role of 2Ca in cell function and a sensitivity of a biological process to buffer loading has generally been accepted as evidence that this function is 2Ca +.dependent. While this conclusion may be true in a large number of cases, since 2+Ca buffering is undoubtedly an effect of many BAPTA-like compounds, an increasing body of evidence, including our own observation that BAPTA-AM evokes a direct increase in 21[Ca suggests that some of the BAPTA effects are directly mediated by BAPTA,j rather than secondary to 2Ca chelation.

Several studies have implicated BAPTA and many of its analogues, including Quin2, in enhancing the synthesis and release of different prostaglandins and arachidonic acid (Raspe et a!, 1989; Van der Zee et a!, 1989; Boeynaems et a!, 1993). BAPTA loading has also been associated with severe depletion of cellular ATP (Tojyo and Matsumoto, 1990), and inhibition both of protein kinase-C (Dieter et a!, 1993) and phospholipase-D activation (Coorssen and Haslam, 1993). Other non-buffering effects of BAPTA have been reported including direct blocking of muscarinic inhibition of 2Ca currents (Beech eta!, 1991) and competitive inhibition of 31P binding to its receptor (Richardson and Taylor, 1993). For a highly charged molecule such as

BAPTA, diverse direct interactions should not come as a surprise and these

induced

resting the

lower small

effect

pronounced

increased

influxes)

peak

actual stimuli

difference

state

be that

the

the

response.

with

effects

interactions

more

rate

slow

rates

the

A

Ca 2 +

To

BAPTA

Ca 2 +

conditions

affinity

on

duration

noticeable

[Ca 2 ] 1 , in

of

Ca 2

of

summarize,

balance

precise

will

the

rate

the

of

Ca 2

in BAPTA

change

During

responses

influx

on

increase

should

the

have

or

rate

of

presence

of

loads

Ca 2

of

DMB

and

buffering

BAPTA

to

is

rise

magnitude

between

of

than

difference

the

and

longer

a

in

not

state

+.mediated

be

thus

(Augustine

major

change

we

in

[Ca 2 ] 1

and

increases

stimulus.

during

its

altered

considered

on

of

[Ca 2 j 1

for

conclude

that

stimulation

more

recovery

derivatives.

capacity

increased

Ca 2 +

processes

impact

Ca 2

of

in

was

short

than

the

by

associated

free

[Ca 2 ] 1 .

the

processes reduces

etal,

This

influx

observed

the

changes

which

that

on

was

as

since

and

of

neuronal

BAPTA

Ca 2 +

the

associated

the

differential

presence

possibilities

1991).

Ca 2 +

and

loading

DMB,

long

the

makes

BAPTA

the

peak

cell

Ca 2

that

buffering

in

extrusion

between

is

attainable

responses.

stimuli

Ca 2

effect

Ca 2 +

behavior

which available

+

[Ca 2 i 1

cultured

are

of

the

with

influx

was

effect

when

BAPTA-like

normally

buffering

of

responses

BAPTA

(or

capacity

may

BAPTA

mechanisms

more

larger

the

peak

is

more

in

small

for

of

hippocampal

evaluating

During

not

the

be

buffer

Ca 2

buffering

effective

less

Ca 2 +

of

associated

than

attributed

and

capacity

reduced,

and

presence

reflect

buffers.

to

the

short

saturated

buffers

will

short

DMB

large

it

influx.

under

Ca 2

the

reflects

the

stimulus

in

be

neurons

stimuli,

and

indicating

in

and

of

to

biological

slowing Ca 2 +

It

with

more

on

steady-

their

may

the

slows

at

long

the

the 149 150

Figure 4.1: The effects of BAPTA loading on short-duration 2Ca transients. The mean 2Ca + response of 53 neurons exposed to 20 pM NMDA for 25 seconds (solid horizontal bar) is illustrated (solid line) compared to an NMDA induced 2+Ca response after the same neurons were loaded with BAPTA by exposure to BAPTA-AM (20 pM, 15 minutes, RT)(dashed line). The mot noticeable changes were the reduction in th peak magnitude of the Ca + response, the slower rate of increase in [Ca] and the slow and extended recovery phase. The time axis of the second response has been shifted to facilitate the comparison of the two responses.

2.0

0 4-, CD

ci) C.) 1.5 C ci) C-) Cl) ci) 0 U 0 OD 1.0 (Y) (1 C)

0.5 0 100 200 300 Time (s) 151

Figure 4.2: Changes in 21[Ca during exposure to BAPTA-AM. Fifty-four neurons were jexposed to 0.1% DMSO (open horizontal bar) and later to 20 pM BAPTA-AM (solid horizontal bar) at room temperature. Exposure to BAPTA-AM, but not to DMSO, resulted in a marked increase in the mean [Ca that was reversed at the end of exposure to BAPTA-AM. The peak magnitude2j, f both NMDA- (open triangles) and HK depolarization-induced (solid triangle) Ca responses was reduced after loading the neurons with the buffer, BAPTA.

3.0

0 2.5

CD

0) C.) 2.0 C U) C-) Cl) ci) 0 1.5 U 0 ()03 1.0

C) C) 0.5

0.0 0 1000 2000 3000 4000 5000 6000 7000 Time (s) 152

Figure 4.3: Heterogeneity of the responses to BAPTA-AM exposure. A) Intracellular 2Ca changes in cells during exposure to BAPTA-AM (solid bar) varied from one cell to another as shown in representative cells in 3A. B) Raw fluorescence values at 334 nm and 380 nm (dotted and solid lines respectively) of cells shown in 4.1A indicate that neither addition nor cessation of exposure to BAPTA-AM (20 pM) was associated with detectable shifts in the raw fluorescence intensity suggestive of altered background fluorescence in the presence of BAPTA-AM, regardless of the individual cell response to the AM- ester. The response to BAPTA-AM expsure in individual cells was apparently correlated with the magnitude of the3Ca + response in these cells to HK (open triangles).

3.0

A 2.0 0 4J (‘3

1.0

0.0 3.0

2.0 - 0 4-, (‘3

1.0

0.0 - 3.0 -

2.0 - 0

(‘3 cr

1

A A 0.0 0 500 1000 1500 2000 2500 3000

Time (s) 4

153

Figure 4.3 B

200

0U) 150 C U) C) U) 100 U) 0 D U- 50

0 200

U) 150 cC-) 0U) C’) 100 U) 01_ 4A4w D V\.’ k—’f?r\. U- 50

A A 0 200

a) 150 C-) C U) C) U) 100 .,... U) 0

U- 50 -

A A 0 0 500 1000 1500 2000 2500 3000

Time (s) 154

Figure 4.4: Loperamide reverses BAPTA-AM induced increases in 21[Ca In 50 neurons exposed to 2OpM BAPTA-AM (solid horizontal bar), the [Ca in . was abruptly increased and remained high the presence of BAPTA-AM.J21 Addition of loperamide (25 pM, open horizontal bar) to the bathing medium in the presence of BAPTA-AM resulted in an immediate terminatin of the BAPTA AM induced 2+Ca change. A subsequent, NMDA-induced Ca + response (open triangle) had a smaller peak after BAPTA exposure compared to the pre exposure response.

2.0

0 4-’ 03 1.5 a) C) C a) C) Cl) a) 0 1.0 U 0 a) (Y) 0.5 C’, C’,

0.0 0 1000 2000 3000

Time (s)

to

(solid increase

exposure

depolarization-induced

AM

bar)

A)

independent Figure (I) C-, 0 0 C.)

C’,

BAPTA-AM

The

(20

2.0 while

1.5

2.5

triangles)

A 4.5:

0

mean

pM,

in to

I

reductions

[Ca

Ca 2

BAPTA-AM.

open

phenomena.

[Ca 2 +]

I 2O

1000

was j

I

buffering

pM, horizontal

while

observed

in

was

(solid solid

2000

the

its

B)

increased

and

NMDA-induce

buffering

horizontal

In

triangles)

bar) after

I

this buffer-AM

3000 and

I

Time

experiment

the

in

AAA

to

effect

Ca

bar)

52

buffer

BAPTA-AM

(s) neurons

4000 induced

+

was

(open on

I

changes

loading.

(mean

the

not

triangle)

during

increases HK-induced

5000

associated

(20

were

of

pM,

31 exposure

I

and

observed neurons)

I

6000 solid

in

HK

wit

Ca

[Ca 2 i 1

horizontal to

+

an

only

exposure response

7000

hBAPTA

are

after 155 156

Figure 4.5 B

2.5

0 4-’ 2.0

G) C-) C 1.5 ci U) ci) 0

LL 0 1.0 OD C,,

C,, C,, 0.5

0.0 0 500 1000 1500 2000 2500 Time (s) 157

Figre 4.6(A-E): The effect of buffering on short- and extended-duration + 2Ca Ca responses.

Neurons were treated for 60 minutes at 33°C with 60 pM of the AM ester of hBAPTA-AM (78 neurons, Fig 6A), DMB (72 neurons, Fig 6B) or BAPTA (40 neurons, Fig 6C). Figures 6D and 6E display results of neurons loaded for 60 minutes at 33°C with 20 4uM of the AM ester of DMB (49 neurons) or BAPTA (66 neurons) respectively. 2+Ca responses were evoked by NMDA (20 jiM, 25 seconds: open triangles) or 250 pM glutamate for 3-4 minutes (solid bar) or for 25 seconds (solid triangles). In comparison to control response in hBAPTA-Ioaded neurons, transient 2+Ca respones in buffer-loaded neurons were much more reduced than the prolonged Ca+ responses to a 3-4 minute glutamate exposure. BAPTA was more effective than DMB at the same loading concentration in reducing the peak response to extended glutamate application and in slowing the rates of 2+Ca increase and recovery. The higher loading concentration (60 pM) of each buffer was more effective than the lower (20 pM) concentration of the same buffer under the same loading conditions of time and temperature.

3.0 A

0 2.5 4-,

0 C-) 2.0 C a) Ci U) a) 01. 1.5 D U 0 1.0 C’)

C’) C’) 0.5

0.0 0 500 1000 1500 2000 2500

Time Cs)

Figure

C

B

4.6

Cr)

CO 0

Cr) U

(Y)

Cr)

Cr) )

. 4 -J

cf 0

U

CI)

U)

0 ‘-

C)

a) C

c. a)

(‘3

CI)

D Cd)

ci)

C

C.)

3

C13

B,C

0.0

0.5

0.0

0.5

1.0

2.0

15

2.5

3.0

1.0

2.0

1.5

2.5

3.0

0

0

500

500

1000

1000

Time

Time

(s)

(s)

1500

1500

2000

2000

158

2500 2500 Figure 4.6 D,E 159 3.0

2.5

CU

D 2.0 c C) Cl) 1.5 U 0 a 1.0 C)

(Y) CY, 0.5

0.0 0 500 1000 1500 2000 2500

Time (s) 3.0

E 2.5 4-, CU

G) cC-) C) C’) 1.5 :3 U- 0 OD 1.0 c)

0) 0.5

0.0 0 500 1000 1500 2000 2500

Time (s) 160

Figure 4.7(A-D): The effect of 2+Ca buffers on ‘excitotoxic’ exposure to glutamate.

Neurons were treated for 60 minutes at 33°C with 60 pM of the AM ester of hBAPTA (48 neurons, Fig 7A), BAPTA (62 neurons, Fig 7C) or DMB (69 neurons, Fig 7D). Figure 7B displays results of neurons treated for 60 minutes at 33°C with 0.03% DMSO (47 neurons). 2Ca responses were evoked by NMDA (2.5, 10 and 40 pM, 25 seconds: open triangles) or 250pM glutamate for 30 minutes (solid bars). In the control groups (A, B) 8-minute intervals between consecutive NMDA responses were adequate for complete recovery of [Ca+] to its resting pie-exposure level. After the 30-minute glutamate exposure, recovery was relatively rapid and almost complete within the experiment time. In the buffer-loaded neurons (C, 0) short NMDA-induced 2+Ca changes were extensively reduced (in the BAPTA-loaded more han in the DMB-loaded neurons). However, recovery of intracellular Ca+ w,s incomplete between stimuli and residual Ca accumulated giving th Cat+ tracings a step-ladder 21 + appearance. A 3O-minue glutamate-induced Ca response showed a slower +]j rise and recovery in [Ca in the buffer-loaded groups than in the control groups, and in the BAPTA-loaded neurons than in the DMB-loaded neurons. However, the peak of the response was not lower in magnitude in the buffer- loaded neurons compared to the control neurons.

A

3.0

0 2.5

(V

a) U 2.0 C a) C-) C,, a) 0 1.5 D LJ 0 10 C)

C’) C’) 0.5

00 0 1000 2000 3000 4000 5000 6000 Time (s)

Figure

D

C

B

4.7

B,C,D

C’) C,)

C’) 0

U C,,

co C,,

C,) C,) C,,

Q3

0 C,,

U 0

LL

0

U)

D 0, C-) 0)

0) C

C) Co

0)

D

0 0

0 0) 0) 0

C o C)

0)

C

0

C

0.0

05

2.5

1.0

1.0

15

2.0

1.5

2.5

0

0

1000

1000

1000

2000

2000

2000

Time

Time

Time

3000

3000

3000

(s)

(s)

s)

4000

4000

4000

5000

5000

5000

6000

6000

6000 161

Aspect

Low-Span

Mid-Span

SlopeDn

SlopeUp

Peak

Parameter

buffer-loading

both

any This

in of

in

capacity Table

NMDA-induced

the

different

the

of

Ratio

table

buffer

4.1:

individual

values

the

(Ratio/s)

(Ratio/s)

(s)

by

(s)

lists

listed

The

experiments

groups

loading

of

(p

and

perentage

Ca

that

experiment.

parameters

<

were compares

+

0.001).

neurons

parameter

BAPTA-loaded

responses

222.4

160.1

Mean

43.4

23.7

52.6

74.3

(%

Notes:

after

significantly

of

changes

was with * * * *

*

control)

All

loading

the

NS:

p-Values

All

Means

before

‘n’

in

the

cultures

after

values

pre-loading

calculated

neurons

effects

BAPTA

measurements represents

69.19

13.61

28.58

neurons

not

13.07

9.43

8.74

S.D.

(n=14)

in

changes

different with

loading

are

the

the

statistically

of

compare

using

of

ranging

or

calculated

the

the

kinetic

neurons

increased

loaded

value. from

DMB.

an

with

corresponding

in

0MB-loaded

buffer,

unpaired from

150.8

133.4

experiment

Mean

39.9

27.5

47.9

66.9

the

(%

are

BAPTA-

from

significant

the

buffer,

parameters

of with

were The

from

the

based

listed

values

control)

40

expressed

neuronal

t-test.

value

control

compared

the

BAPTA

and

to

loaded

parameters

on with

90.

parameter

(n=1O)

18.28

23.14

20.52

11.49

19.26

10.88

percentage

S.D.

of

DMB-loaded

ratios.

of

of

a

that

buffering

values

or

with

transient

number

the

as

to

with

percentages

parameter

its

‘mean’

the

<0.005 and

<

before

NS

of

NS

NS

NS DMB.

0.02

p

buffer

in

of 162 163

CHAPTER 5

CALBINDIN-D28K EXPRESSION AND NEURONA L 2Ca + RESPONSES 164

The objective of the experiments described in this chapter is to compare EAA- and depolarization-induced 2Ca responses in CaBP(H and CaBPN hippocampal neurons using fetal face-down and postnatal cultures. Our working hypothesis was that CaBP neurons, much like BAPTA-loaded neurons, would display 2+Ca responses with lower peaks and slower recovery rates when compared to responses in CaBPN neurons. All statistical comparisons were performed using unpaired t-test.

I. THE ASSOCIATION BETWEENCaBP EXPRESSION AND RESTING INTRANEURONAL j2[Ca Basal ‘resting’ levels of intracellular 2+Ca were found to be significantly higher in CaBP neurons (96.9 ± 6.63 nM, n=82 neurons) when compared to CaBPN neurons (75.3 ± 1.14 nM, n=1004) with a difference of 21.6 nM (p <0.002).

II. THE ASSOCIATION BETWEEN CaBP EXPRESSION AND TRANSIENT INCREASES IN j21[Ca IN CULTURED FETAL HIPPOCAMPAL NEURONS a. NMDA-induced 2Ca transients:

In 25 different experiments, exposure to NMDA (20 pM, 25 seconds) resulted in an increase in 2[Ca to 383.8 ± 20.8 nM in CaBP neurons (n=82) which was significantlyj larger (p < 0.001) than the peak 2Ca concentration in CaBPN neurons (303.7 ± 4.84 nM, n=1004).

The absolute increase (Basal-to-Peak) was also significantly larger in

CaBP than in CaBPN neurons (286.9 ± 20.69 nM, and 228.4 ± 4.79 nM respectively, p < 0.01). The duration of the 2Ca transient was longer as seen by measurement of the mid-span and low-span parameters which were 16

neurons were

neurons

resting

duration

the

peak

fetal

resting

CaBPN

closely

parameter

studied

individual

and

different

well

Similarly,

the

in

seconds

respectively

CaBP

application

basal-to-peak

their

Interesting

It

as

culture

A

still Ca 2 +

is

‘basal’

typical

Ca 2 +

reflected

neurons

during

parameters,

displayed

while

of

also

and

in

larger

statistics).

the

experiments.

was

the

response

and

the

were

(both

concentration

29.1

of

rates

the

Ca 2

experiment

the

of

transient

found

two

for

variants

CaBPN

observed

significance

the

the

exposed

changes

peak,

a

recovery

p

seconds

the

of

lower

populations

though

level

frequency and

<

It

agonist

to

change

CaBP

must

0.001

neurons

Table

basal-to-peak

are

was

be

the

was

in

is

peak

to so

larger

in

longer

phase

measured

shown

the

illustrated

to

be

basal-to-peak

longer

than

20

that

in

the

).

5.3

not

with

Ca 2 +

note

population

stressed

(See

same

(5.14

[Ca 2 j

pM

Widening

in

CaBP

lists

of

higher in

the

that

in

in

the

which

that

NMDA the

the

Table

change

Fig

and

experiment.

from

aspect the

±

in

the

of

CaBP(+)

during

CaBP

that

Ca 2 +

0.48

Fig

in

in

5.3A

the

CaBP

the

frequency

(Fig

neurons the

of

change

5.1

the

the

4

for

5.1

the

on the

ratio

CaBPN

out

width

and

differences

transients,

and

5.2).

for

whole

NMDA

CaBP 25

exposure

where

population

significant

transients

than

of

full of

4.97

were

seconds.

neurons.

3B.

(n=

the

of

with

the

neurons

pool

listing

receptor

in

the

85

3)

25

In

than larger,

±

CaBPN

transient

were

which

were

to

Fig

was of

0.14,

Ca 2

neurons

experiments

was

as

differences

The of

NMDA

in

neurons

(n=82).

5.3A

compared

and

activation,

all

higher

not

the

observed

proportional

neurons

a

p=0.7).

transient

mean

was

parameters

particular

of

the significantly

CaBPN

CaBP

than

tested,

an

before

similar

The

of

in

the

to

8-DIV

in

the

the

as

the

165

the to 166

CaBPN neurons yet the recovery was slower in spite of the reduced peak

height. Fig 5.3B illustrates how a moderately larger peak response in the CaBP neurons was associated with much longer recovery where mid- and low-span parameters were almost double those of the CaBPN neurons in the same field.

Another illustration of the difference in the recovery process is evident in the extent of recovery of intracellular 2+Ca after NMDA receptor stimulation of the CaBP neurons. Over the observation period (6-8 mm), CaBPN neurons tended to have rapid and complete recovery to pre-stimulation levels of 2+]i[Ca whereas CaBP neurons displayed a residual increase in intracellular 2+Ca over pre-NMDA exposure levels (Fig 5.1, 2 and 3).

CaBP(H neurons were also over-represented in the few neurons that

‘collapsed’ on challenging them with NMDA (15 out of 23 neurons in the 25 experiments conducted using NMDA). CaBP immunoreactivity was also more frequently found in neurons that had extremely high 2Ca levels at the beginning of the experiment (10 out of 19 neurons). Such neurons were excluded from the statistical analysis since kinetic parameters could not be measured. b. Depolarization-induced 2+Ca transients:

In another set of 18 experiments with a total of 742 neurons, of which 59 were CaBP(+), depolarization-induced 2+Ca responses were evoked by exposure of neurons to high extracellular concentrations of K+ (50 mM). Similar statistical analyses were conducted and the results were very similar to those obtained from NMDA-induced 2+Ca responses. Only the parameters that were significantly different with NMDA stimulation were also significantly the when exposure were although that ‘narrower’ tested exposed is experiments, cultured whether NMDA to expected EXPOSURE two Ill. values CaBP(H different almost that postnatal THE were stimulation In On The much compared was spite or as of exposure neurons the to ASSOCIATION the and double in [Ca 2 ] results if HK qualitatively BAPTA, well responses faster 613 the CaBP the OF of kinetic magnitudes CaBP 8-day CaBPN exposure as these NMDA CULTURED case of to procedures that reported at (compare functioned the in to which which postnatal old trends a the NMDA, of neuronal observed with kinetic statistical more neurons similar (20 cultures (see depolarization-induced postnatal BETWEEN of reduced 80 in high pM, observed tables POSTNATAL advanced the (Fig chapter the differences CaBP were postnatal intracellularly to populations displayed in 25 aspect transients significance of previous those 5.4, fetal the 5.1 cultures. later seconds). 4 CaBP 4). neurons, in day-postnatal Tables stage magnitude and recorded cultures ratios neurons fetal found between However, many HIPPOCAMPAL EXPRESSION section were 5.4). were as of This cultured of 5.2 and The a that to development. Ca 2 of the of displayed Ca 2 + greater from very and be of resulted were faster the postnatal the total were differences it hippocampal the CaBP, transients. was 5.3). neurons comparable cultured characteristic same buffer AND not number and peak recovery observed NEURONS increases in of what and in the taller TRANSIENT interest in Ca 2 a would In fetal vitro between a frequency of rates fetal a neurons would Absolute manner to among and series in neurons response neurons in age. trends, TO pre exist the to neurons, of [Ca 2 ] 1 test be NMDA the change of the were similar in that 14 167 to that was recovery aspect neurons 2.61 of neurons. same significantly versus 37.72 proportion rate neurons, had statistics were population larger CaBPN p difference comparison =0.479). the in significantly complete of The and Also also population. recovery in 13 ratio ± 15.06 increase neurons (p was the net during 2.82 for 1.17 With out in larger to was < (695.94 of larger between However, contrast CaBP postnatal result the incomplete 0.01). of to ± the seconds nM/s larger that process, was the 0.56 (70.65 in pre-exposure higher 14 increase The Ca 2 (p CaBP was experiments, increase the ± in 53.25 Another peak with < fetal population nM/s) rate peak cultures. the 17.46 respectively) rates that 0.01), resting ± were transients over in fetal CaBP CaBPN [Ca 2 i 1 2.50 of ± neurons [Ca 2 ] 1 from the mid-span and of nM, recovery prominent the only 3.00 levels was CaBP [Ca 2 ] 1 change, nM peak (939.95 recovery that recovery same p neurons and slightly was and nM/s not and in < while (Fig than and in and response 0.001) only of was markedly time fetal significantly 72.74 neurons, compared finding the 5.5). in ± the [Ca 2 ], basal-to-peak phases in low-span of longer (p CaBP slightly 43.05 almost CaBPN course. CaBPN 1 [Ca 2 j 1 same < and 5 Table ± in to out 0.001), different in postnatal 1 of the the though to NMDA nM) .08 identical parameters larger the neurons. of parameters, larger neurons. neurons the 5.4 in the postnatal basal-to-peak 25 nM compared the CaBP parameters Ca 2 + an lists postnatal mid-span, was statistically fetal (18.25 in respectively CaBP increase CaBP in the compared parameter A significantly transients. CaBP(H in cultures cultures as neurons notable CaBP the to ± CaBPN indicators of the changes neurons the that neurons 1.08 CaBPN the to with and tested, mean CaBP was is (by The in 168 169

IV. THE ASSOCIATION BETWEEN CaBP EXPRESSION AND PROLONGED 2Ca RESPONSES

In a smaller series of experiments, extended exposure of cultured fetal hippocampal neurons to NMDA (20 pM, 2 or 5 mm) or glutamate (500 pM, 5 mm) resulted in a prolongation in the 2+Ca response. Under these conditions, CaBP neurons demonstrated larger peak responses and slower rates of recovery than their CaBP counterparts (Fig 5.6A,B).

DISCUSSION FETAL NEURONS: In fetal cultures, resting pre-stimulation 2[Ca and peak 21[Ca during NMDA-induced responses were significantly higherj in CaBP jneurons than in CaBP neurons. Larger peak responses were not simply a reflection of the higher pre-stimulation ]2[Ca since the basal-to-peak change in 21[Ca was also significantly larger than in the in ] CaBP neurons. Also, some experiments where pre-stimulation 2+][Ca was similar in both populations, the response amplitudes were still larger in the CaBP neurons.

Larger response peaks in CaBP neurons were associated, in fetal cultures, with a proportional prolongation in the recovery of 2[Ca +j, compared to CaBPN neurons at both the mid-span and low-span levels. While longer span values are to be expected in association with larger 2+Ca responses, prolonged recovery may not be entirely due to larger peak responses. In some experiments where the peak of the 2+Ca response was lower (or marginally larger) in CaBP neurons, the duration of 2+Ca responses was still more prolonged than that of the CaBPN neurons. Slower recoveries of 2+][Ca to pre-stimulation values were also reflected in an incomplete recovery of the 170 mean 21[Ca of CaBP(H neurons that was observed in 15 out of 25 experiments,j while incomplete recovery was observed in the mean 2[Ca +1 of the CaBPN neurons in only 3 out of 25 experiments. The differences observed between 2+Ca responses to transient NMDA exposure in CaBP and CaBPN neurons were also observed when 2+Ca responses were induced by depolarization using high extracellular K+ and when exposure to NMDA or glutamate was extended for several minutes.

These results lead us to conclude that the development of CaBP immunoreactivity in fetal hippocampal neurons is associated with a change in EAA- and depolarization-induced 2+Ca responses characterized by larger magnitudes and longer durations. It is also associated with a tendency for an elevated basal 2+][Ca and incomplete recovery following a 2+Ca load.

In general, larger peaks and longer-lasting responses could result from larger 2+Ca influx, greater intracellular 2+Ca release, less efficient recovery mechanisms or a combination of these factors. The larger 2+Ca influx could be a result of an increased 2+Ca channel density and/or altered channel kinetics (e.g., larger unitary conductance levels, slower inactivation or desensitization, enhanced sensitivity to agonist or to depolarization). Similar factors could contribute to a larger intracellular release of 2+.Ca Impaired recovery may result from impaired extrusion/sequestration mechanisms or as a result of competition between recovery mechanisms and a high capacity intracellular calcium buffer.

While our experiments cannot precisely identify which of these mechanisms are responsible for the changes observed in 2+Ca responses in CaBP fetal neurons, it is unlikely that a single mechanism underlies all the 171 changes observed (i.e., elevated basal 2i,[Ca larger peak responses and slower recovery) since they were not all present in every experiment. For example, slow and prolonged recovery was pronounced even when mean response amplitudes were lower in CaBP neurons than in CaBPN neurons. Very similar changes in both NMDA- and depolarization-induced 2Ca responses also suggest that the difference between the two neuronal populations may be more downstream to 2+Ca influxes, although a parallel change in agonist- and voltage-induced fluxes in CaBP neurons cannot be excluded.

Changes in 2+Ca responses associated with the onset of expression of CaBP in fetal neurons may be required for developmental purposes (e.g., synaptogenesis), but the same changes may also heighten the vulnerability of CaBP neurons to 2Ca + mediated injury. This issue is addressed in more detail in chapter 6 but a coincidental finding in the present set of experiments indicates that this may actually be the case since CaBP neurons were significantly over represented in neurons that had ‘de-regulated’ 2+Ca levels before exposure to NMDA or depolarization, and in those that collapsed, i.e. showed no sign of recovery, after stimulation and for the duration of post- stimulation period.

It is important to stress that statistical comparison of two populations can be complicated and biased when one of these populations has a much lower frequency of incidence than the other. In our case, CaBP neurons constitute approximately 8% of the total neurons in fetal cultures. A few extreme responses can cause a significant bias in the calculated parameters, and subsequently skew the results of significance tests. In order to avoid this situation the results of our experiments were obtained by pooling the results of 172

all the CaBP neurons before performing statistical comparisons to the pooled results of the CaBPN neurons. However, we contend that the consistent and

reproducible trends observed in the large number of experiments performed lend more support to the validity of the observations than does the actual numerical results of the analysis although in our results complete agreement exists between the two approaches (Table 5.3).

POSTNATAL NEURONS:

Fetal neurons are in a phase of extensive developmental change and may not be representative of neurons in adult animals. For this reason, postnatal cultures were also studied to test whether the trends observed in fetal cultures would persist during later stages of development. Compared to fetal neurons, postnatal neurons had larger peak responses regardless of their CaBP immunoreactivity. Despite these larger 2+Ca responses, recovery in postnatal neurons was surprisingly fast with mid- and low-span values that were 40 to

50% shorter in postnatal neurons for responses that were more than double the magnitude of fetal response, indicating a significant increase in the efficiency of recovery mechanisms relative to the NMDA-induced 2+Ca load. Also, it is interesting to speculate that larger 2+Ca responses are associated with larger 2+Ca influxes that may have saturated cellular 2+Ca buffers and resulted in the faster initial recovery of the 2+Ca responses observed in postnatal neurons.

Within the postnatal cultures, CaBP neurons displayed significantly larger peak 2+Ca responses compared to CaBPN neurons although basal 2+][Ca was similar in the two groups. Mid-span and low-span parameters were marginally longer in CaBP neurons but not in proportion to the increase 173

in peak levels since the rates of increase and recovery were significantly faster in the CaBP neurons. Larger peaks and faster rates of increase and recovery caused 2+Ca responses in CaBP(H neurons to look more ‘pointed’, a feature that was numerically expressed as a significantly larger aspect ratio.

It is clear from our data that the presence of CaBP is associated with greater differences in resting and stimulated 2+][Ca responses in fetal neurons compared to postnatal neurons. However, we cannot conclude that CaBP is the cause of these differences. Further, we have made no attempt to quantify the amounts of CaBP expressed in neurons derived from these two sources, mostly due to the capricious nature of immunohistochemistry as a detection method for this protein. The determination of neurons as being simply “positive” or “negative” on the basis of immunohistochemistry may well mask the possibility of large absolute differences in concentrations of CaBP in fetal and postnatal neurons which could account for the differences observed.

Although CaBP has been the focus of a large number of studies, only a few of these have been aimed at identifying the functional correlates of the expression of this protein. In 1991, Kohr eta/studied the high voltage activating (HVA) 2Ca current in acutely dissociated adult hippocampal dentate granule (DG) neurons prepared from normal and kindled rats using the whole cell recording technique. Kindling, while inducing seizure activity in rats, causes a selective loss of CaBP from DG cells (Miller and Baimbridge, 1983). Comparing CaBP-deficient DG neurons from kindled rats to normal CaBP DG neurons from non-kindled animals, indicated that HVA (L-type) ‘Ca displayed marked inactivation in CaBP-depleted cells when compared to normal, CaBP

DG cells, which showed very little inactivation. This difference in inactivation was significantly diminished by the inclusion of the fast 2Ca buffer BAPTA in 174

the recording pipette indicating that the difference in inactivation was the result of a reduced 2+Ca buffering capacity, presumed to be due to the loss of CaBP. Peak ‘Ca was similar in both cell types and their respective I-V relationships were identical. The authors concluded that CaBP may have a physiological role as an intracellular 2+Ca buffer in the immediate submembraneous zone with important functional consequences (Kohr eta!, 1991; Kohr and Mody, 1991).

Mattson eta! (1991) used primary cultures of E18 fetal hippocampal rat neurons, plated at very low density (50 neurons/mm and maintained in media containing 55 mM d-glucose, 20 mM KCI2 and 10% fetal bovine serum, to study neuronal ) vulnerability to excitotoxic injury and calcium responses in relation to CaBP expression. Their results showed that CaBP neurons are better capable of handling 2Ca loads induced by exposure to glutamate or to the 2Ca ionophore Br-A23187 (4 neurons). Pre-stimulation }2[Ca and peak 2+Ca responses were similar in CaBP and CaBPN neurons but the extent and rate of recovery was significantly faster in CaBP neurons. The use of 2+Ca ionophore, presumably inducing equal 2+Ca loads in different neurons, led the authors to conclude that the difference between the two populations was not a result of different glutamate receptor density. However the assumption of an equal 2Ca load induced by A23187 is based on the assumption of a uniform neuronal surface/volume ratio which may not be valid for cultured neurons with different CaBP immunoreactivities since, in our own experience, Ca8P neurons were generally larger than CaBPN neurons (data not shown).

A novel and potentially very useful approach to the study of the functional role of CaBP was employed by Liedo eta! (1992) where they used a retrovirus to transfect 3GH pituitary cell line with the cDNA of CaBP. The recombinant 175

cell line displayed reduced magnitudes of both L- and T-type ‘Ca upon current- evoked depolarization. Inactivation of both L- and T-currents was also

enhanced in the cell line expressing CaBP (a result entirely opposite to that of Kohr and Mody, 1991) but peak 2Ca responses to depolarization were decreased. However, in the recombinant cells resting 2[Ca was increased, which in itself could provide an explanation for the enhanced inactivation observed in these cells. Analysis of the decay time of 2[Ca transients indicated a ] the loss of fast decay component evident in the wild type cell line, while the slow component of decay was not significantly altered. These authors also increased intracellular CaBP content by including it in the patch pipette. Using this loading method, increased CaBP content of the cell was not associated with reduced 2+Ca current amplitudes and resting intracellular 2+Ca was not significantly changed. Peak 2[Ca responses to depolarization were, however, reduced and the fast decay component was lost while the slow component was slightly prolonged.

A more recent study also utilized patch pipettes for loading cultured postnatal dorsal root ganglion (DRG) neurons with CaBP (Chard eta!, 1993). Introduction of CaBP had no effect on the inactivation or rundown of calcium currents. Basal 2+][Ca was unaltered while depolarization-induced changes in 2+][Ca were reduced in magnitude and the rates of increase and decay of ]21[Ca were markedly diminished in the CaBP-Ioaded cells. These results are therefore consistent with a fast 2Ca buffering action of CaBP in DRG neurons.

Direct comparisons between the results of the above-mentioned studies and our own are extremely difficult since diverse preparations and experimental paradigms were employed. A particularly important experimental difference is the time scale of the stimuli used to induce 2+Ca responses. While

their

of

conditions

The

(1991) capacity,

recovery

results while

loss

with

following

neurons.

‘acute’

results

currents

and

taken

saturation

milliseconds

using

electrophysiological

CaBP

common

conceivably

of

individual

However,

the

In

not

20

into

were

but a

showing

loading

spite

will

effects

fast in

of

fast

second

stimulation

producing

The

were

of

account.

their

CaBP

[Ca 2 +]j

depend

obtained

in

of

feature

recovery

cytoplasmic

Ca 2

elevation

kinetic

this

of

duration,

this

very

sample

of

larger

extended

stimuli

wild

transfecting

conclusion

after

buffering

cautionary

identical

on DG

measurements

different

of

was

by

properties,

Ca 2

component

type

the

size

our

or

cells,

or

our

Chard

transmembrane

Ca 2

quite

recovery

resting

longer.

full

results

GH 3

was

responses

fluorescent

effects

action

resulting

from

cannot

profile

remark,

et GH 3

evident

binding

cell

quite

in

a!

[Ca 2 +]

and

and

Thus

ours.

were

any

of

cells

of

to

(1993)

with

of

be

small

[Ca 2 +]i,

a

in

CaBP.

these

transfection,

the

in

in

particular capacity

Ca 2

slightly

[Ca 2 +]

total

conducted

Ca 2 +

reconciled

with

larger

CaBP-containing

our both

It

and

lack

is

in

and

studies

fetal

Ca 2

understandable

acutely-loaded

extended

CaBP

regulatory

fetal

flux,

is

Ca 2

of

extended

measurements

their

during

consistent

cell

inactivation

neurons

with

and

using

loads

consistent

was

(Liedo

seems

type.

neuronal

influx

longer

recovery

postnatal

that

associated

mechanisms,

stimuli

and

recovery.

patch

et

and

to

(Kohr

with

DRG

of

a!,

that

the

stimuli

of

be

culture

with

were

in

Mattson

pipettes,

1992).

of

Ca 2

the

phase

a

eta!,

CaBP

agreement

possibility

the

neurons.

delayed

tens

a

with

Similar

conducted

present

should

high

effects

and

1991),

of

The

et

the

a!

176

of be 177

Another difficult observation to explain is the larger amplitude of 2Ca responses in association with a 2+Ca buffer such as CaBP. While expression of CaBP is probably not the only distinguishing feature between CaBP and CaBPN neurons, and thus other mechanisms might be responsible for larger 2Ca responses, it is possible that the relief of Ca-dependent inactivation of Ca channels in immediate 2+ the submembraneous2 zone (Kohr eta!, 1991) per se can cause an increase of 2+Ca influx that outweighs the enhanced buffering capacity associated with CaBP expression. Another possible explanation is that a 2Ca buffering action of CaBP causes ‘uncoupling’ of physiologically paired messages. For example, altering the time relationship between changes in 21[Ca and the production of 31P or AA might result in a change in the temporalj and/or spatial relationship between inhibitory and stimulatory effects of these second messenger pairs with the subsequent enhancement (or inhibition) of other 2+Ca process such as release, uptake or extrusion.

It should be made clear that none of the studies discussed here, including our own, can establish any causal, rather than associative or coincidental, relationship between altered 2+Ca responses on one hand, and CaBP expression on the other. The only exception could have been the transfection experiments (Lledo et a!, 1992) although the use of wild type cell line as a control, rather than a retrovirus-infected cell line (without CaBP-cDNA or with a

‘dummy’ cDNA) precludes establishing a cause-effect relationship. This lack of an appropriate control group makes it difficult to evaluate the conclusions of that study.

While our results are not consistent with the hypothesis that CaBP is exclusively functioning as a simple 2Ca buffer, similar to BAPTA, prolonged 2+Ca responses and delayed recovery in fetal neurons indicate that buffering 178

2+Ca may be one function of CaBP in neurons. This conclusion is supported by the results of Kohr et al (1991), where BAPTA could reverse the enhanced inactivation of 2Ca currents observed in DG cells that lost their CaBP, and by the results of Lledo etal(1992) and Chard etal(1993), where peak 2Ca responses and rates of change in 2+][Ca were reduced whenever cellular CaBP content was increased. Larger 2+Ca responses in both fetal and postnatal CaBP neurons most probably reflect other functional consequences of CaBP expression, although they may as well reflect a different aspect of the phenotype development in which CaBP expression may be coincidental or a secondary phenomenon. 179

Figurç 5.1: NMDA-induced 2+Ca responses are larger and longer-lasting in CaBPIH fetal hippocampal neurons. Eighty-seven cultured fetal hippocampal neurons wer simultaneously exposed to NMDA (20 pM) for 25 seconds. Three were CaBP’ and displayed a higher initial base line +][Ca a larger peak response and a slower recovery (solid line) when compared21 the mean of 84 CaBPN neurons line). The (dotted recovery of the ,former was also incomplete during the interval studied.

600

CaBP, n=3 500 J

:3 400 C) 0(‘5 1 (‘5 300 :3 0G) (‘5 -4-, C 200 u) 1ci) U-

CaBP n=84

0 100 200 300 400 500 600 Time (s)

transient

recovery intracellular

Of

neurons Figure

U-

0 a) 1) (ES C) ci) I

C) E

C

79

neurons

5.2:

even

was

and

0

Larger

Ca 2 +

extended

complete.

in

exposed

the

levels

and

100

absence

longer-lasting

recovery

to

were

NMDA,

of

200

similar

initial

(solid

5

CaBP

Ca 2 +

to

Time

high

line,

CaRPN

300

resting

responses

mean

(s)

neurons

neurons

of

Ca 2 +

400

5

are

responded

neurons),

(dotted

levels.

observed

500

line)

although

with

in

and

CaBP

a

600

larger

initial 180 181

Figure 5.3: Comparison of the mean 2Ca response in CaBPU and CaBP neurons following exposure to 20 pM of NMDA for 25 seconds. A) Although the peak of the mean 2Ca response is smaller in CaBP neurons (n=5, solid line), recovery parameters, i.e. mid- and low-span, are longer than those in CaBPN neurons (n=32, dotted line).

B) In this experiment, a small increase in the peak response in CaBP neurons (n=2) was associated with an increase in the mid- and low-span parameters (37.3 to 57.5 seconds and 71 .2 to 99.0 sends respectively) and a slower and incomplete recovery compared to CaBP neurons (n=32).

A

250-

C 200

- 150 :3 0a) 100 LCaBP+,n=5 ci) ci) U- 50

NMDA

I 0 I 500 550 600 650 700 750 600 Time (s) CaBP ______

182

Figure 5.3 B

700•

600-

—500C E D 400 CaBP+,n= - I -j300o - C 200 ci)

U— 1•

100 . .. ,n=32 NMDA I 0 I 0 100 150 200 250 300 Time (s)

CaBP the

B) A)

responses

(dotted is

Figure LL 0 -4-,

1 I 0

a) ci) D ctS 0 C 1 E

C

slower

In

Five

aence a

-,

A

5.4:

different

neurons.

100 CaBP

line)

in

and

Depolprization-induced

CaBPIH

of

in

150

slow

the

raised

experiment,

neurons

same

200

incomplete

compared

initial

field.

(solid

250

[Ca]

larger

line)

to

recovery

300

CaBP

increases

and

in

had

Time

5

longer-lating

350

CaBP’

higher

when

fetal

(s)

in

400

neurons.

compared

[Ca 2 +1 1

resting

neurons

450

responses

[Ca 2 i 1 ,

are

to

compared

500

larger

79

larger

CaBP’

were

550

and

to

evident

recovery

Qeak

69

600

neurons

183 in 184

Figure 5.4 B

350

300 CaBP+n=5

-250C E 200

150 1 0 4-, .... C ; 100 - - 1) - - .. - IL LCaBP , nt=69 50

0 I I 500 550 600 650 700 750 800 850 900 Time (s) 185

Figure 5.5: Mean [Ca in cultured 4-day postnatal hippocampal neurons (8- DIV) exposed to 202j, pM NMDA for 25 seconds is larger in CaBP neurons than in CaBP neurons.

Within the postnatal cultures, CaBP neurqns (solid line) showed larger pea responses and faster rates of change in [Ca] compared to postnatal CaBP neurons (dotted line). Recovery was marginally delayed but was complete in almost all cases.

C E D 0 0

C”

0 C” 4-,

a) U

0 100 200 300 400 500 600 Time (s) The CaBPN for the shared The similarities NMDA glutamate Figure U- 0 C 1 ctS U) 0 U) U) 1 0 D E C 2 mean mean prolonged mm. A 5.6: for a neurons, larger of peak results shorter to In Extended 54 the (B) Ca 2 + peak [Ca 2 J 1 CaBPN and 74 pattern in times larger neurons [Ca responses recovery exposure neurons in Figs frequently +] 3 Ca CaBP’ and were 5.1-5.4). was + of in (dotted responses a fetal CaBP exposed slower much observed neurons Time hippocampal line) In slower. incomplete (A), neurons in (s) to when in was CaBP 500 3 C 8 BP(H CaBP larger pM exposed (solid neurons recovery neurons. glutamate than neurons line) neurons to to that retained 20 of NMDA for exposed pM [Ca 2 +] of (solid 5 71 NMDA or mm. many line) than to 186 187

Figure 5.6 B

500

450

400

350

300 C-) 250- Z3 200 CaBP, flt3 I

50 CaBP n=7rJ

0 0 500 1000 1500 2000 2500 3000 3500 4000 Time (s)

Low-Span

SlopeDown

Mid-Span SlopeUp(nM/s)

Basal-to-Peak

Peak

Parameter

Basal

Table

25 responses

seconds

Ca2+

Ca2+

5.1:

(s)

(s)

(nM)

(nM/s) (nM)

-

in

Comparison

(nM)

(NS fetal

=

CaBP

111.7

228.4

303.6

Mean

55.3

11.2

75.3

2.8

not

CaBP(-)

statistically uing

+

151.93

153.37

57.09

21.51

36.21

3.18

9.32

S.D.

and

n=1004

unpair

CaBP

S.E.M.

0.68

1.80 0.10

0.29

4.79

4.84

significant). 1.14

-

neurons

t-test)

142.5

287.0

383.8

Mean

71.3

12.8

96.9

2.4

of

CaBP(+)

exposed

the

187.37

188.02

76.26

36.02

10.33

60.06

2.21

S.D.

kinetics

n=82

to

S.E.M.

20.69

20.76

20

8.42

3.98

0.24

1.14

6.63

of

pM

the

NMDA

Ca 2

Mean

30.86

16.00

58.59

21.58

80.18

-0.41

value

1.55

Difference

for

<0.001

<0.001

<0.01

188

<0.01

<0.01

(NS)

(NS) p 189

Table 5.2: Comparison (uing unpairec t-test) of the kinetics of the CaBP+1 2Ca responses in fetal and CaBP’ neurons depolarized by exposure to 50 mM 0K for 20 seconds (NS = not statistically significant).

CaBP(-) n=683 CaBP(+) n=59 Mean Difference

Parameter Mean S.D. S.E.M. Mean S.D. S.E.M. value p

Basal Ca2+ (nM) 78.6 38.96 1.49 99.8 65.81 8.57 21.23 <0.05

Peak Ca2+ (nM) 301.6 119.58 4.58 358.6 146.16 19.03 56.98 <0.01

Basal-to-Peak (nM) 223.0 121.25 4.64 258.7 140.27 18.26 35.74 <0.05

SlopeUp (nM/s) 12.9 9.28 0.36 15.0 11.04 1.44 2.14 (NS)

SlopeDown (nM/s) 2.8 2.43 0.09 2.3 2.15 0.28 -0.58 (NS)

Mid-Span (s) 49.2 29.65 1.13 65.1 36.80 4.79 15.83 <0.01

Low-Span (s) 97.0 50.61 1.94 140.6 82.97 10.80 43.66 <0.001

Low-Span SlopeDown

Mid-Span

SlopeUp

Basal-to-Peak

Peak

Basal

Parameter

that

mean

CaBP’

occurrence the

pooled

This

Table

NMDA-

(e.g.,

Ca2+

Ca2+

CaBP’

significant

table

values

larger

5.3:

(nM/s)

(s)

çlata

(s)

or

(nM)

(nM/s)

neurons

(nM)

lists,

HK-induced

(nM)

neurons

is

from

peak

of

listed

p-values

the

for

in

all

response

each

parameter

the

next

CaBP(+)

of

the

NMDA

(25

Ca

the

NMDA

neurons

reflected

experiments)

kjnetic

to

same

22

19

17

21

19

19

exposure

or

8 the

>

response

CaBP(-)

longer

under

group).

p-value

parameter,

experiment.

(e.g.,

the

study

mid-span)

trend

76%

88%

32%

68%

76%

76%

84% was

1004

obtained

The

%

in

I

the

in

associated

purpose

<0.001

<0.001

<0.01

<0.01

<0.01

CaBP’’

the

(NS)

(NS)

The

the

p

number

in

frpm

CaBP

the

relationship

percentage

CaBP(+)

High

of

neurons

(18

CaBP

t-test

with

of

this

experiments)

K+

experiments

and

14

13

12

13

13

14

5

a

listing comparisons

>

exposure

versus

larger

between

CaBP(-)

of

CaBP

neurons

such

is

parameter

to 82

neurons.

72%

78%

28%

67%

72%

72%

78%

where

the

than

show

%

of

f

the

190

<0.001

in

<0.01

<0.05

<0.01

<0.05

(NS)

(NS) p 191

Table 5.4: Comparison (using unpaired t-test) of the kinetics of the 2Ca responses in cultured 4-day postnatal CaBP and CaBP’ neurons exposed to 20 pM NMDA for 25 seconds (NS = not statistically significant).

CaBP(-) n=533 CaBP(+) n=80 Mean Difference

Parameter Mean S.D. S.E.M. Mean S.D. S.E.M. value p

Basal Ca2+ (nM) 72.7 24.93 1.08 70.7 22.37 2.50 -2.09 (NS)

Peak Ca2+ (nM) 695.9 403.21 17.46 940.0 385.02 43.05 244.01 <0.001

Basal-to-Peak (nM) 623.2 402.30 17.43 869.3 384.05 42.94 246.10 <0.001

SlopeUp (nM/s) 37.7 27.01 1.17 53.2 26.82 3.00 15.53 <0.001

SlopeDown (nM/s) 15.1 12.94 0.56 18.3 9.69 1.08 3.20 <0.01

Mid-Span (a) 36.7 8.84 0.38 39.3 6.31 0.71 2.61 <0.02

Low-Span (s) 56.6 18.11 0.78 59.4 11.89 1.33 2.82 (NS)

Aspect Ratio 18.9 16.37 0.71 23.1 11.81 1.32 4.16 <0.01 192

CHAPTER 6

EXCITOTOXICITY IN HIPPOCAMPAL NEURONAL CULTURES temperature. identifiable glutamate resulted resulted assessed (± their protocol a. from I. vacuolation minimize assessment injury neuronal The results on exposure EXCITOTOXICITY SE) Fetal glutamate-induced influence sensitivity cultures included Fetal Although The of that in in at Cultures: variability injury, after 68.6 of EC 50 a neuronal neurons objective room cultures were of only. reduced cultured tested In of a the swelling to ± of all viability the further temperature different comparable 23.1 excitotoxins cytoplasm Criteria 192 the as were injury absence which after of IN sensitivity hippocampal a neuronal results this pM ± and ‘NAIVE’ 24 result assessment killed 33 7 culture which used had hour and chapter to loss of pM. and to (RT) reported of 10 by at been injury for the extracellular to those incubation NEURONAL was of the conditions a loss DIV. using in glutamate neurons In morphological phase glutamate addition is maintained using the culture both concentration-dependent will to of obtained below a presence present neuritic also 30-minute brightness groups, fluorescent to in of and Mg 2 +, age CULTURES toxicity are HEPES-buffered high concentration be Mg 2 by face-down in treatment the processes. examined. based identification of all concentrations morphological vitro, exposure glutamate results of as 1 morphologically to vital jiM the indicated on the results TTX. with stains the were neuronal of incubation of In to BSS the with 3 of latter exposure order Ca 2 glutamate were examined mM Viability assessment by yielded neuronal of effects at an the soma, glutamate. method to (Fig room obtained buffers EC 50 medium higher 6.1). was of for 193 of of 194

During exposure to glutamate, neurons appeared swollen with vacuolated cytoplasm and their somata lost their phase brightness. These changes were observed even at concentrations that were not subsequently associated with wide-spread neuronal damage. In the majority of neurons, the early morphological changes associated with cell injury were completely reversed 4-6 hours after termination of exposure to glutamate (even at high concentrations), and the neurons regained their phase bright appearance. However, this morphological recovery was only temporary and many neurons were assessed as being dead 24 hours after the initial insult.

Face-up fetal cultures were exposed to excitotoxic glutamate concentrations using the extended exposure protocol, i.e. for 24 hours at 34°C in their N2 culture medium containing 2+.Mg Under these circumstances the 50EC of glutamate in the face-up cultures was 72 ± 11 .6 jiM but 45-50% of the fetal neurons were found to be resistant to glutamate toxicity up to a concentration of 1 mM. This is interesting in view of the observation that, in the vast majority of morphologically identifiable neurons, glutamate exposure evoked an increase in neuronal 2+].[Ca b. Postnatal Cultures:

Postnatal cultures displayed a significantly higher sensitivity to the neurotoxic effects of glutamate when compared to either type of fetal cultures. Using the extended exposure protocol, approximately 90% of the neurons were killed using a glutamate concentration of 1 mM. The 50EC for glutamate toxicity was 20.4 ± 9.7 pM, the lowest amongst the 50EC values in the three culture types tested. For example, only 68 ± 5.3% of the neurons survived an exposure to glutamate at a concentration of 12.5 pM (Fig 6.2).

and

slow,

over following

which

wash-out

population

glutamate

neurons d.

test).

8%

compared pM)

under

loperamide

higher than

using

varying

neurons

c.

[Ca 2 +] 1

Effects

magnitude

survival

the

increased

10%

and

unpaired

Using

our

was

Glutamate

survival

degrees

was

following

using

its

of

in

experimental

(3

to

to

under

maintained

of

changes

(25

the

fura-2

wash-out. rate

many

mM)

no-antagonist

determined

73

Antagonists:

the

neuronal

of

rate

t-test).

pM),

glutamate

the

with study

±

receptor

this

at

4-6

cases

extended

microfluorimetry,

8%

of

under

excitotoxic

were

room

early

p<0.05

hours

84

throughout

showed

A

conditions,

and

In

survival

complete,

at combination

±

antagonists

most

much

temperature, excitotoxic

(Fig

glutamate-treated

post-insult

regular

55

exposure

although

9%.

and

6.3A). a

±

neurons, effects

in

less

rapid

the

The

10%,

the

intervals

p

although

recovery

the

effective

for

conditions:

exposure

protocol.

and

of

recovery

VGCC

presence

The

increase

of

and

respectively

[Ca 2 ]

both

this

individual

glutamate

VGCC

average

during

for

they

respectively,

of

cultures

blockers,

was

than antagonists

[Ca 2 j 1

varied

a

to

to

of

MK8O1

of

further both

blockers

then

glutamate

a

a 3

neurons identified

glutamate

[Ca 2 j 1

high

as

30-minute

mM

(p<0.001

(28

widely.

improved

nifedipine

to

followed

tested

(4pM)

18-24

glutamate

steady-state

using

pre-insult

± achieved

ameliorated

of

the

and

fetal

11

antagonists

the

in

exposure

and

%

hours

unpaired in

time

survival

by

postnatal

for

(2

face-down

both

and

neuronal

a

from

pM)

a

values

CNQX

some

course

phase

slightly

after

value

to

35

cases,

and

when

t

to less

±

time

(40

195 of 196

Subsequent cell death also occurred after widely varying time intervals in different neurons. The final cell collapse was indicated by a very high fluorescence ratio followed shortly by loss of fluorescence intensity resulting from leakage of fura-2 out of the cell through the injured neuronal membrane.

Several examples of the responses of individual neurons are shown in Fig 6.4. Also, neurons with higher peak responses to glutamate generally demonstrated an earlier collapse in the post-insult phase (Fig 6.5). When glutamate exposure and 2+][Ca measurements were conducted at 34°C similar findings, but with an accelerated time course, were observed and neuronal loss was complete within 18 hours after glutamate exposure compared to >24 hours at room temperature.

A small number of cells (<5%) that responded to glutamate with an increase in 2+][Ca displayed a very early and usually complete recovery of 21+][Ca in the continued presence of glutamate. These cells remained viable for the duration of the experiment and it is likely that they were morphologically ‘neuron-like’ glia demonstrating a transient increase in 2+][Ca as a result of metabotropic glutamate receptor activation (Holzwarth et a!, 1994).

II. EXCITOTOXICITY IN NEURONS LOADED WITH BAPTA AND ITS ANALOGUES

The loading of neurons with the artificial 2Ca buffers BAPTA and di methyl-BAPTA (DMB) for excitotoxicity experiments was achieved by incubating the cultures with the cell permeant acetoxy-methyl ester, BAPTA AM or DMB-AM. The objective was to test the hypothesis that increasing neuronal 2+Ca buffering capacity would confer a degree of neuroprotection against excitotoxic insults believed to be mediated largely via 2Ca-dependent 197

processes. DMSO and the non-Ca-binding molecule, half-BAPTA (hBAPTA), were employed as controls for2 the vehicle and the BAPTA ‘nucleus’ respectively. The concentrations of the DMSO and hBAPTA-AM were selected to match the highest BAPTA or DM8 concentrations used. a. Fetal Cultures:

Fetal cultures which have been maintained face-down were turned face up and loaded for 30 minutes with 60 pM of BAPTA-AM or DMB-AM at RT, before an exposure to 500 pM glutamate (30 minutes, RT). This treatment resulted in a significantly greater neuronal loss detected 8 hours later when compared to control cultures (39.1 ± 8.2% and 25.8 ± 13.6% survival for DMB- and BAPTA-loaded neurons respectively compared to 87.1 ± 2.7% and 86.6 ± 2.1% neuronal survival in the DMSO- and hBAPTA-treated groups). Reducing the concentration of buffer-AM used for loading from 60 pM to 20 jiM reduced, but did not abolish, the enhanced neuronal damage in buffer- loaded neurons (78.5 ± 5.5% and 79.7 ± 4.3% respectively, Fig 6.6) when compared to that seen in the control groups.

A complete concentration response curve for glutamate toxicity was determined for both control and 20 pM BAPTA-loaded neurons. At all concentrations of glutamate tested, toxicity determined 24 hours after exposure was greater in the BAPTA-loaded cultures compared to the control cultures (Fig 6.7). The estimated 50EC for glutamate was reduced from 68.6 ± 23.1 pM in the control group to 16.7 ± 4.9 pM in the BAPTA-loaded group. b. Postnatal Cultures: Using the extended glutamate exposure scheme, postnatal cultures loaded using 20 pM BAPTA-AM or DM8-AM showed the same trend of 198

heightened glutamate toxicity in the buffer-loaded neurons when compared to the DMSO and hBAPTA control groups (Fig 6.8). c. Correlation with 2+Ca changes in BAPTA-loaded neurons: Using fura-2 microfluorimetry, the 21[Ca of neurons loaded with BAPTA was determined prior to, during and followingi a 30-minute exposure to glutamate (3 mM) at room temperature. The rates of increase and recovery of intracellular 2+Ca levels were much slower in the buffer-loaded neurons although the plateau of 21[Ca achieved was not significantly different when compared to the control jcultures. Fewer cells were capable of near-complete restoration of their pre-stimulation 2[Ca after wash out of the glutamate (28% and 73% of cells 6 hours afteri the glutamate exposure in the BAPTA loaded and control groups respectively), and neuronal collapse occurred earlier

(Fig 6.3B); consistent with the observed increase in glutamate toxicity in buffer-loaded neurons reported above.

Ill. THE ASSOCIATION BETWEEN CaBP IMMUNOREACTIVITY AND

GLUTAMATE NEUROTOXICITY a. Fetal Cultures:

The low incidence of CaBP neurons in fetal neuronal cultures grown in serum-free conditions made it impractical to study excitotoxicity in CaBP neuronal sub-population in fetal cultures. b. Postnatal Cultures:

The relationship between CaBP immunoreactivity in postnatal cultures and the survival rate of these neurons under excitotoxic conditions was studied

found

the

of

CaBP

VI.

neuronal

subject

100%

number

higher

concentrations

glutamate-treated

population.

6.9B

count

significant

populations

toxicity

CaBP

population.

glutamate

by

bFGF

BSA-treated

comparing

EFFECTS

show

to

In

at

of

Another

glutamate

to

of

treatment

(50

neurons view

be

cultures

each

the

neurons

high

viable

concentrations

with

a

significantly

pM

is

The

At

cultured

greater

OF

of

concentration

to

the

way

sampling

of

all

p<0.001

control

and

the

difference

to

neurons

calculate

concentration

bFGF

with

was

cultures

12.5,

survival

on

of

glutamate

of

200

finding

decline

the

neuronal

neurons

less

2.5-5

discerning

TREATMENT

cultures

50

more

errors.

pM

glutamate

was

in

normalized

the

was

of

than

and

between

that

both

of

ng/ml in

glutamate)

CaBP

toxic

toxicity

small

(see

ratio

responses

the

glutamate

statistically

cannot

200

that

(Fig

the

the

cases

bFGF

CaBP

chapter

in

of

concentrations

pM.

and

treatment

the

6.10),

ON

of

difference

to

the

and

CaBP

discussed

be

CaBPN

(Fig

the

the

GLUTAMATE ratio

statistical

resulted

to

Analysis

used.

bFGF-treated

reliably

CaBPN

significant

3),

with

population

differences

control

excitotoxic

6.9A).

in

of

and

neurons.

neurons

in

untreated

statistically

The

above,

in

4-day

performed

subpopulations

behavior

of

parameters

tested,

the

cell

CaBP

results

groups

at

TOXICITY

relative

increased

cultures

were

count

to

postnatal

insults.

we

glutamate

In

expression

cultures

the

the

between

the

studied

significant

presented

since

treated

statistically

of

total

are

survival

to

mid-range

compared

Glutamate

the

sensitivity

the

hippocampal

at

therefore

the

and

neuronal

the

with

different

respective

the

in

CaBPN

total

in

of

in

almost

effects

two

Fig

the

of

to

was

of 199 200 differences more obvious in the mid-range of the neurotoxic response (p

DISCUSSION

The experiments presented above were conducted to study the effect(s) of different concentrations of glutamate on the survival of cultured hippocampal neurons under a variety of conditions. Under all the conditions tested, glutamate induced a concentration-dependent increase in the frequency of neuronal death. However, the sensitivity of neurons to glutamate toxicity varied depending on the type and conditions of the cultures. In view of the relationship between glutamate-induced increases in 21+][Ca and neuronal death, we further examined the potential neuroprotective effect of the presence of CaBP or artificial 2Ca buffers.

EXCITOTOXICITY IN DIFFERENT CULTURE TYPES

All fetal neurons maintained face-down were vulnerable to a 30-minute exposure to glutamate at room temperature. A total loss of neurons could be induced by exposure to 1 mM glutamate. Inclusion of 0.8 mM 2Mg in the incubation medium reduced the sensitivity to glutamate and increased the estimated 50EC from 68 to 192 pM. A notable phenomenon was the complete reversal of the widespread glutamate-induced morphological changes (swelling, loss of phase brightness) shortly after the exposure to glutamate was terminated. Within 20-24 hours, these changes together with vacuolation and breakdown of neurites reappeared in neurons that were considered dead.

Fetal and postnatal cultures maintained face-up were exposed (always in the presence of 2Mg to glutamate at 34°C for 24 hours before neuronal survival was assessed.i Two important differences were observed between the fetal and postnatal neurons. First, the neurotoxic 50EC of glutamate was significantly lower for postnatal neurons than for fetal neurons. Second, 202

almost half the fetal neurons were resistant to glutamate toxicity up to a

concentration of 1 mM while nearly all postnatal neurons were vulnerable.

Our cultured neurons therefore displayed a similar sensitivity to the toxic effects of glutamate previously reported for hippocampal (Rothman, 1985; Michaels and Rothman, 1990) and cortical neurons (Choi, 1985). The shift of the glutamate concentration response curve to the right in the presence of 2+Mg is also in agreement with a neuroprotective effect of 2+Mg (Rothman, 1983; Choi, 1987b; Garthwaite and Garthwaite, 1987).

EXCITOTOXICITY IN BAPTA- AND DMB-LOADED NEURONS

In both fetal (face-down) and postnatal (face-up) cultures, a marked increase in the sensitivity of neurons to glutamate toxicity was observed after neurons had been loaded with either BAPTA or DMB but not with the control compound hBAPTA. In fetal cultures increased 2+Ca buffering was achieved by the use of either 20 or 60 pM of the buffer-AM during the loading process.

The use of a higher buffer-AM concentration was associated with a higher incidence of neuronal loss following a subsequent exposure to glutamate. In fetal and postnatal neurons, no differences were observed between the effects of loading neurons with BAPTA or DMB. A full concentration response curve was constructed for glutamate toxicity in neurons loaded using 20 pM BAPTA

AM and a shift to the left was observed in the BAPTA-loaded compared to control neurons, with the 50EC reduced from 68.6 pM to 16.7 pM. Measurements of 21+][Ca in fetal ‘face-down’ neurons loaded with BAPTA were performed during a 30-minute exposure to 3 mM glutamate at room temperature and for up to 24 hours after the exposure. Similar to our finding in chapter 5, the rates of rise and recovery of 2+][Ca were much slower in the

adding

on

activation

result

inactivation

properties

compounds.

in

concentrations

in

with

DMSO

effect

neuronal

concentrations

an

with

BAPTA who

detected

2

resting

in

BAPTA

BAPTA-

from

buffer-loaded

cellular

control

exposure

Several

BAPTA-buffers.

any

reported

In

in

was

to

or

individual

general,

or [Ca 2 +]

loaded

an

loss

the of

earlier

with

of

of

energy

neurons.

even

and

not

increase

of

several

Ca 2

possibilities

to

BAPTA

potential

(>

that

VGCC

hBAPTA-AM

DMB-loaded

seen

of

of

neurons

500

in

fura2)

neurons

as

our

+.dependent

80%)

neurons,

stores.

formaldehyde

the

the

loading

a

BAPTA-buffer

pM

in

under

results

Fewer

and

rapid

and

Additionally,

buffer-AM

buffer-loaded

the

Ca 2 -induced

was

independent

although

glutamate

were

exist

DMB

NMDA

Delayed

total

cultured

the

increase

presumably

neurons

associated

neurons. are

eliminated

K+

to

a

could

conditions

in

Ca 2 +

during

consequence

channels,

(100

explain

the

channels.

agreement

derivatives

for

and

our

fetal

in

were

neurons,

of

lead

peak

cell

For

influx

pM)

the

5

with

the

the

use

extended

through

any

minutes.

hippocampal

the

to

injury,

used

capable

example,

mean

possibility Ca 2 +

were

de-esterification

and

of

effects

enhanced

and

a

enhanced

Any

with

and

control

(e.g.,

of

reduction

for

the

injured

[Ca 2 +1 1

may

associated

thus

the

responses

recovery

or

neuronal

of

Moreover, those

the

of

reduced

the

BAPTA,

all

accumulation

recovering

that

play

groups

impose

neurons

glutamate.

neuronal

loading

glutamate

plasma

of

Ca 2

in

and

of

the

these

a

processes,

the

collapse

Dubinsky

of

with were

likelihood

less

DMB,

by

treated

high

a

chelating

effects

the

of

Ca 2 -dependent (after

membranes.

further

loss

the

to

effects

significant

extensive

our

no

neurotoxicity

The

AM

of pre-exposure

dibromo

loss

was

following

lower

15-17

with

neurons

(1993a)

toxic

observed

while

of

latter

stress

would

of

the

either

than

fura

DIV)

role 203 direct arachidonic relative effects Taylor, (Nistri an resulting Berlin, example, regulation chelation was Also, similar procedure contribute during mobile Ca 2 + Ca 2 + neurons, glutamate in the impaired not It Several the effects and influx chelation case is 1992), increase exposure of and 1993). energy significantly in also that Ca 2 + survival and BAPTA (Olson Cherubini, not to exposures a acid high-affinity refilling of and other reduction of the important 24 an could, associated very deficit chelation in loading effects release of extrusion/sequestration efflux eta!, hours enhanced on of [Ca 2 i studies neurons prolonged of neurons other directly worse 1990), used during 1P 3 -sensitive of 1990), of in may cells to (Van BAPTA has with the face-up glutathione was have cellular consider in glutamate in to be and protein or with loaded der our been cellular excitotoxic control an enhanced BAPTA-AM observed a indirectly, reported or following Zee experiments derangement inhibition BAPTA. fetal functions. DMB associated intracellular the kinase-C with capacity et cultures toxicity (Brodie neurons possibility a!, molecules. as during a glutamate-induced either events contribute excitotoxic or multitude The of 1 a 989) activation DMB-AM result and For protein with in (30 to not of former Ca 2 and buffer exposure buffer-loaded or handle example, the have Reed, of minutes exposed a during of The postnatal to of direct loss synthesis normal stores situations, the (using will most all effects cell (Dieter oxidative 1991) increase to of been presence the for accentuate non-chelating ATP to injury. neuronal hBAPTA-AM, cell (Richardson likely coupling 20 neurons). face-down glutamate. extended neurons of potentially et (Preston reported depletion volume pM while in a!, Ca 2 + stress, does For of [Ca 2 j, buffer-AM) 1 loss. between 993) the highly since the not and Other to and fetal and a be and 204 a functional fluorescent treatment ionophore, responses exposed been delayed prolonged resistance adult to inclusion free increased et BAPTA increased Deyoetal, loading exacerbate Plasma latter confer a!, radical-mediated The observed Some hippocampal 1 two 990) resulted membrane in the to of presence enhanced of peroxide using collapse Ca 2 + electrical of and hepatocytes effects A23187 BAPTA ischemic studies the 1991). BAPTA loss and frog the loss in energy BAPTA in buffering of ventral in spinal hippocampal could slices, the (assessed of injury could analog have electrophysiological rat of in survival stimulation. (2pM) insults cell an membrane solutions thymocytes reduction deficit as motoneurons (Ueda enhance root) reported artificial has be Scharfman injury capacity. a and or 3 result under alleviated also by in days and 10mM and slices Ca 2 induced filling response the of A potential) the Ca 2 protective been the of a Shah, after similar before DNA loss variety For prepared and probe, increased activity + with glutamate the Ca 2 -mediated when associated integrity buffer in irradiation example, damage of Schwartzkroin 1992). intracellular to stabilizing (Tsubokawa the renal of glutamate-induced of Quin2, effects cytoplasmic a of dentate from in acetoxy conditions. prolonged lipid NMDA tubular (Kudo (e.g., neurons in buffering with (Story was gerbil hepatoma associated peroxidation effect recording cell hilar decreased eta!, eta!, methyl channels cells BAPTA (1989) also buffering exposure has et brains injury For neurons of of a!, 1990). found by 1992). been BAPTA cell Ca 2 + ester depolarizing example, with and 1 observed electrodes exposure that (Choi, which 992). (Carpenter input line capacity to suggested to during Quin2 of an using Finally, had Pre the has delay (Dypbukt the 1990). may Also, in that been Ca 2 + to also was the 205 in

postnatal

EXCITOTOXICITY other

Ca 2 +

neurons

buffers

effects

rapid,

the

BAPTA,

toxicity.

neuronal

neurons end

BAPTA

after

neurons,

[Ca 2 i neurons

artificial

experiments

significant

a

recent

pre-exposure

of

In

the

It

agents

buffers

and

glutamate

experiments

of

may

is

is

they these

displayed

Ca 2 +

and

(Kudo

loss

study

termination

neurons

In a

interesting

artificial

associated

cell

neuroprotection

30-minute

contrast,

well

such

and

a

may

are

during

neurons

death

concentration-dependent

buffers

eta!,

by

depend

those

level

unlikely

exposure,

AND

were

as

well

Tymianski

Ca 2 early

using

45-minute

was

to

of

glutamate.

with

1990;

in

exposure

yet

in

be

had

CaBP

of

more compare

exposure

the

and

buffers,

upon

cultured

prevented.

to

the

toxic

Dubinsky

prolongation

cell

a

in

Tymianski

continued

and

marked

be

eta!

post-exposure

vulnerable

EXPRESSION

24-hour

cultured

death

the

exposure

‘universally’

in

to

a

the

suggest

to

hippocampal

themselves,

substantial

(1993b),

neuronal

glutamate

(1993a)

glutamate.

recovery

was

[Ca 2 j 1

These

presence

excitotoxicity

eta!,

explants

of

to

cell

to

an

that

the

glutamate

BAPTA

populations

glutamate.

recovery

neuroprotective.

differences,

death

1993b).

early

with

responses

results

enhancement

of

[Ca 2 i

the

or

neurons

of

of

Prior

[Ca 2 +]j

potentiate

those

event

biological

mouse

which

glutamate,

loading

in

protocol,

of

toxicity

loading

In

recovery

a

and

observed

Even

in

examined

[Ca 2

cultured

with

to

and

sustained

occurs

spinal

cultured

a

was

of

the

the

effects

the

plateau

substantial

when

of

than

cultured

Indeed,

the

glutamate

effect

found

toxic

phase

which neurons

neurons.

only

opposite

hippocampal

in

CaBP

and

CaBPU

increase

spinal

loaded

of

the

close

effects hours

in

of

these

to

that

at

spinal

was

present

some

with

the confer

with

to

in

206 of 207 neurons at all concentrations of glutamate tested. CaBP neurons were eliminated from the glutamate-treated cultures faster than CaBPN neurons such that the ratio of neurons expressing CaBP to the total neurons surviving the insult was reduced as the glutamate concentration was increased.

Few studies have focused on the relationship between CaBP expression and neuronal vulnerability to excitotoxic stimuli in vitro. Mattson eta! (1991), using fetal hippocampal neurons, reported that CaBP neurons were more resistant to both glutamate-induced and 2Ca ionophore-induced injury than CaBPN neurons. Also CaBP neurons were more capable of regulating their 2+][Ca in the face of glutamate or 2+Ca ionophore exposure. An earlier preliminary report from our laboratory (Baimbridge and Kao, 1988) suggested that fetal CaBP hippocampal neurons were more resistant to glutamate toxicity. The cultures used in the latter study were, however, of the fetal face down variety maintained in serum-containing culture medium and had a higher incidence of CaBP expression than the cultures used in the experiments described here. In contrast with these results, Mockel and Fischer (1994) could not find any selective sparing of CaBP neurons in fetal hippocampal neurons using short exposure to glutamate, long exposure to NMDA or kainate, or to a combined glucose/oxygen deprivation. Our results, however, indicate that CaBP expression is associated with enhanced vulnerability to glutamate induced neurotoxicity.

Conflicting results regarding the role of CaBP as a neuroprotectant in vitro are also observed in studies using in vivo models of ischemia and epilepsy

(Sloviter, 1989; Freund eta!, 1990), as well as in studies on post-mortem human brains from patients with neurodegenerative disorders (lchimiya et a!,

1988; lacopino and Christakos, 1990b; Yamada eta!, 1990). Such a widely

cultured TNF

Similar

during that

hippocampal

as

neuroprotection

(reviewed

of

survival

cultured

days

glutamate

the

percentage

the

EFFECT

expression

multitude

biological

emphasizes

varied

BSA-treated

bFGF,

effect

cell

TNFa

treatment

A

Postnatal

after

glucose

observations

association

large

type

rate

hippocampal

OF

neurons

nerve

in of

systems.

and

of

toxicity.

the

may

of

BFGF

the

Mattson

number

of

neurons

developmental

and

bFGF

end

neurons

TNFB

were

deprivation

neurons

control

neurons

growth

against

difficulty

therefore

were

on

ON

of

treatment between

The

were

associated

Since

other

as

of

et

treated

neurons

the

GLUTAMATE

cultures.

CaBP.

expressing

factor

studies

well

treated

ischemic/hypoxic

al,

in

excitotoxic

bFGF

of

also

have

developmental

bFGF-treated

injury.

CaBP

1993a).

cell

CaBP

as

determining

with

on

(NGF)

exposed

reported

NGF treatment

with

have

with

different

changes

the

expression

At

expression

Attenuated

excitotoxic

CaBP

TOXICITY

exposure

all

were sensitivity

and

bFGF

enhanced For

recently

to

glutamate

by

cultures

example, (see

effects

tumor

cause/effect

rather

changes

protocol.

and

glutamate.

capable

displayed

Mattson

is

chapter

and

reported

was

concentrations

[Ca 2

excitotoxic

likely

survival

of

necrosis

than

(on

was

concentrations

neuronal

postnatal

Cheng

of

taking

performed

etal

to

By

vulnerability)

a

an

stabilizing

3).

significantly

relationships

A

changes

significant

that

be

that

of

isolated

factors

recent

(1989)

eta!

place

We

neuronal

one

these

vulnerability

growth

neurons

time,

of

after

therefore

part

(1994)

as

study

simultaneously.

[Ca 2 i

(TNF)

glutamate,

neurons.

event,

using

tested,

increase

depending

over

a

less

6

of

in

factors

injuries

result

to

DIV,

from

complex

a

reported

confer

bFGF

than

70%

such

tested

in

the

i.e.

fetal

of in

such

the

and

that

in

of

208

the on 3 209 same group (Mattson et a!, 1993b) reported that bFGF treatment in cultured fetal hippocampal neurons reduced the expression of NMDA receptors and that this may account for its neuroprotective effect since 2+Ca influx through NMDA-receptor operated channels would be reduced. However, the experimental protocols in these experiments were very different from our own. For example, while the studies mentioned above exposed the neurons to excitotoxic insults in the presence of the respective growth factors, our results were obtained after 6 DIV from neurons that were treated with bFGF only for the first 3 DIV. In chapter 3 (Fig 3.3), we show that bFGF-treated neurons undergo an accelerated neuronal loss and are more vulnerable to glutamate toxicity after bFGF treatment is terminated and when most neurons have been induced to express the relatively long-lived CaBP. It is possible that the increased neuronal vulnerability after termination of bFGF treatment may reflect the presence of CaBP, which may out-last the other effects of bFGF treatment, or even a ‘rebound’ hyper-expression of NMDA receptors.

On the basis of all the available evidence it would seem that the presence of certain growth factors can indeed protect neurons against excitotoxic injury. However, neurons which become dependent on the presence of bFGF may be more vulnerable to excitotoxicity following its removal. The outcome of an excitotoxic incident can thus be determined by the abundance of growth factors relative to the excitotoxic stress on neurons, a factor that may have strong impact on the design of therapeutic approaches to the control of acute and chronic neurological disorders of excitotoxic origin.

standard

using absence

when

the

(solid

Exposure

extracellular

Figure

D

>

o

presence

unpaired

bars)

assessed

6.1:

100

120

20

40

of

60

80

deviation

0

of

extracellular

reduces

Glutmate

fetal

Mg

of

t-test).

24

Control 1

.

neurons

pM

of

hours

neuronj

at

Glutamate

TTX)

toxicity

Mg’

least

later.

3OpM

to

induced

glutamate

damage

12

in

(open

Inclusion

fields

fetal

‘I

OOpM

a

bars).

when

Concentration

concentrq,tion-dependent

face-down

from

for

of

300pM

30

compared

Mg+

at

Data

minutes

least

neurons:

points

in

4

1

the

mM

coverslips

to

at

represent

that

extracellular

room

protective

3mM

induced

temperature

neuronal

(**

the

p<0.01

effect

medium

in

mean

the

loss

of

(in 210 ± 211

Figure 6.2: Glutamate toxicity in postnatal culture.

Exposure of postnatal neurons to glutamate (24 hours, 34°C) in DMEM-N2 culture medium induced a concentration dependent neuronal loss (assessed at the end of the 24-hour period). Data points represent the mean ± standard deviation of at least 10 fields from at least 3 coverslips.

140

120

100

- 80 > > 60 C,) 40

20

0 Control 12.5pM 5OpM 200pM 800pM 3.2mM

Glutamate Concentration

earlier.

A)

recovery

experimental

delayed

individual

15 minutes,

pre-stimulation

Figure

B

A

Q

In

4-., Q

minutes,

o

(13 4-I

(‘3

o

fura-2

6.3:

gradual

0.0 0.5

2.0 2.5

3.0 3.5

1.0

1.5 0.0 0.5

1.0 2.0

2.5

3.0

1.5

3.5

of

RT,

neurons

Changes

mean

loaded RT) - - - - -

-

conditions,

1

0

0

pM

levels

increase similr

______

[Ca

at

TTX)

fetal

in

different

within

+j

[Ca 2 j 1

changes

face-down

______

but resulted

in

was

mean

2

using

time

BA

hours

not

5

5

during

in

PTA

in

[Ca.+]

as

BAPTA-loaded

[Ca 2 j

points

neurons,

an

fter

complete

group

Time

glutamate

increase

represents

glutamate

(n=61

were

(67 glutamate

10

(hours)

10

and

in

observed

toxicity.

neu

neurons).

neurons

[Ca+J,

final

was

the

ro

ns)

exposure

collapse

progressive

washed

that

(20

(67

B)

pM

neurons)

15

15

Under

recovered

occurred

(3

off.

BAPTA-AM,

mM,

collapse

similar

A

but

30

to

212

of

20 20

the

7

334/380

3 terminal

different

[Ca 2 ]

Figure

ratio

mM,

dead

6.4:

units).

30

in

increase

times

cell.

ratio

minutes,

individual

Time

to

All

of

a

course

in

cell

panels

value

34°C)

[Ca 2 ] 1 ,

fetal

collapse.

of

of

have

neurons

displayed

glutamate-induced

0

is

is

the

widely

the

Note,

treated

same

result

widely

in

variable

x-axis

panel

with

of

varied

complete

neuronal

D,

in

(0

glutamate

different

that

to

patterns

20

the

leakage

death,

hours)

(solid

rapid

cells.

of

‘recovery’

as

and

of

horizontal

drop

assessed

fura2

y-axis

in

the

from

and

(0

bars,

by

213 to

+

significant

the

neurons)

bar). At

25

hours

peak

U Figure

C-) C CD CD CD

C

D

room

seconds,

first

1000

1200

Ca

400

200 Dotted

600 800

(not

625:

80

0

temperature, had

+

0

recovery

shown)

minutes

responses.

Neurons

solid

line

smaller

represents

triangles)

after

at

of

that

Ca

[Ca 2 +] 1

a

the

he

corresponding collapse

+

20

experiment

responses

and

end

the

in

glut.mate

[Ca1 of

neurons

early

Time

glutamate

to

is

time

in

changes

shown.

the

that

40

response

(3

(mm)

(n=

initial

mM,

exposure

had

13

in

near

exposure

30

to

neurons

neurons).

minutes,

glutamate

complete

(solid

60

to

that

line,

NMDA

Note

solid

have

recovery

did n

=

that

horizontal

not

higher

35

(20

show

only

80

pM,

6 214

unpaired solid

either

loaded

indicated.

Neurons

Figure

(I)

.?

>

bars)

10o

120

DMSO-

20 40

60

80

6.6: using

0

were

t-test).

resulted

A

BAPTA

60

subsequent

or

treated

pM

0.3%

DMSO hBAPTA-treated —

1

T

and

in

BAPTA-

neuronal

with

DMB

30

6OpM

hBAPTA

I

DMSO

minutes

or

promote

loss

Treatment 0MB-AM

cultures

or

2OpM

BAPTA

that

Buffer-AM

exposure

glutamate

was

when

(*

6OpM

BAPTA

Group

p<0.05, significantly

*

to compared,

***

for toxicity

500

30

2OflM

DMB

I

jiM

minutes

in

higher

glutamate

8

p<0.OO1

fetal

hours

U

6OpM

DMB

Glutamate

at neurons.

*

in

RT

500pM

Control

later, cultures

-

(at

using

as

RT,

to 215

p<0.001

coverslips

treated

deviation toxicity

At

(20

Figure

all pM,

a ir

> 0

C’,

concentrations

6.7:

with

to

30

20

of

40

30

10

50

70

80 using

90

when

the

0

BAPTA

minutes

neuronal

DMSO

left.

unpaired assessed

Control

shifts

(open

at

of

survival

RT,

glutamate

Glutamate

t-test).

the

bars). 24

3OpM

solid

concentration-response

hours

of

bars)

a

Data

tested, minimum

100pM

after

was

points

Concentration

the

survival worse

300pM

of

glutamate

represent

1

2

compared

fields

of

BAPTA-loaded curve

1mM

the

exposure

or

from

mean

of

to glutamate

3mM

control at (***

±

least

standard

neurons

cultures

4 216

coverslips

represent

hours,

34°C)

Postnatal

Figure

(I)

>

> Co

100

120

20

40

60

80

were

6.8:

34°C)

0

cultures

the

(*

BAPTA more

*

mean

compared

p

<0.01,

Control

sensitive

loaded

and

±

standard

DMB * to

* using

*

the

Glutamate

to

p

promote

the

<0.001).

DMSO

BAPTA-AM

deviation

neurotoxic

or

glutamate

1

2.5pM

concentration

hBAPTA-treated

of

or

at

effect

DMB-AM

least

toxicity

of

1

2

glutamate

fields

(20

in

cultures.

postnatal

4 uM,

5OpM

from

30

exposure

at

Data

minutes,

neurons.

least

points

(24

4 217 218

Figure 6.9: CaBP postnatal neurons are more sensitive to glutamate toxicity. A) When postnatal neurons used for glutamate toxicity experiments (24 hours, 34°C; Fig 6.2) were stained for CaBP reconstructed glutamate concentration response curves showed that p(+l neurons (open bars) were much more sensitive to glutamate than CaBPN nyrons (solid bars). B) As more CaBP neurons were lost compared to CaBP’’ neurons, the percentage of CaBP neurons decreased with increasing concentration of glutamate. The total number of neurons at higher glutamate concentrations (inset) is small and the ratio estimation may vary considerably as a result of sampling error (* * p<0.01, p<0.001 using unpaired t-test).

140 A 120

100

CD 80 > > I 60 C’) 40

20

0 Control 12.5pM 5OpM 200pM 800pM 3.2mM Glutamate Concentration

B 40 +a Control OD Co 30 C) 12. 5pM 0 0) 0) 20 Co 4-. C 0) IC-) 0) 10 a- 200pM 3200pM 0

0.245

least

using concentrations

bars),

bars)

exposed

Postnatal

neurons.

Figure (I)

-

U

0

>

D

>

G)

0

C’)

12

starting

unpa,red

bFGF-treated

mm.

6.10:

fields

to

40

20

30

10

60

70

80

neurons

0

different

Effect

on

from

t-test).

tested.

the

treated

Control

of

at

neurons

concentrations

sixth

bFGF

least

HPF

Data

Glutamate

with

DIV.

3

is

treatment

were

1

points

2.5pM

coverslips

high

bFGF

When

more

power

represent

of

(2.5

on

compared

glutamate

5OpM

sensitive

Concentration

(*

glutamate

microscope nglml)

p<0.05,

the

200pM

at

to

to

for

mean

0,

BSA-treated

glutamate

toxicity

24

**

field,

1

±

p<0.Ol,

hours

and

standard

800pM

approximately

in

3

toxicity at

postnatal

DIV

cultures

34°C

were

error

p<0.OO1

at

(solid

all

of

(open

at 219 220

CHAPTER 7

GENERAL DISCUSSION AND CONCLUSIONS 221 The potential neuroprotective role of artificial 2Ca buffers and -2Ca binding proteins of the ‘buffer’ variety, such as CaBP, is still a matter of controversy (Mattson eta!, 1991; Dubinsky, 1993a; Tymiansky eta!, 1993b,

Mockel and Fischer, 1994). The work presented in this thesis was conducted with the objective of testing the hypothesis that an enhanced Ca-buffering capacity in neurons, either by loading with BAPTA-like 2Ca buffers2 or by the natural expression of CaBP, improves the neuronal capacity to handle agonist- or depolarization-induced 2+Ca influx, and thereby confers a degree of resistance to glutamate-induced neurotoxicity. Primary cultures of dissociated hippocampal neurons were used for our experiments, but in view of the diverse tissue culture methods used by our laboratory and those of other authors, it was necessary to characterize in detail many aspects of our hippocampal neuronal cultures relevant to the subsequent interpretation of experimental results.

All types of primary cultures tested displayed both EAA- and depolarization-induced increases in 2[Ca in morphologically-identified In ] neurons. both fetal and postnatal neurons NMDA, AMPA, kainate and glutamate induced increases in 2+][Ca that were sensitive to known EAA antagonists such as APV, 2Mg and MK8O1 for NMDA-evoked responses, and CNQX for AMPA/kainate responses. In the fetal cultures, a detailed characterization of VGCC was conducted and three types of 2+Ca channels; L, N and P, could be identified. By far the largest portion of the 2Ca response resulted from the activation of L-channels while N and P channels were equally responsible for the remainder. We can conclude therefore that our cultured neurons, regardless of culture method, were equipped with the basic voltage and agonist-operated channels required for experiments designed to examine mechanisms postnatal combination Ca 2 + and regardless evoked intracellular sensitive their prolonged signals Ca 2 + a! differences peripheral report small. almost by glutamate-, addition Ca 2 the (1992) intracellular NMDA role +..mediated An Ca 2 + efflux/sequestration stores by Our compared completely in interesting of we Ca 2 + neurons. Shmigol and fetal high (DRG) of content Ca 2 + receptor-associated findings between Ca 2 + NMDA- of can may where the Uneyama neurons an extracellular stores release neurotoxicity eliminate postnatal route be buffers to responses increase eta! lacking a difference central or contrast those part central few in quisqualate-induced of from (1994). compared eta! central of in Ca 2 + minutes mechanisms. any in observed neurons in neurons modulating the K-induced caffeine-sensitive (neocortical, with with our (1993) the believed was significant channels) maturational influx. neurons The cultured density higher those to after observed with displayed in but latter postnatal to DRG either These these of Ca 2 Such respect are peaks were hippocampal contribution of be with neurons. authors Murphy intracellular membrane neurons. entirely central in differences transient stores a responses changes Ca 2 labile influx an neurons. the and small to enhanced + have pattern their and a Calcium to and consistent have stores (Shmigol much caffeine-induced to Moreover, and excitotoxic Ca 2 + Ca 2 + in release Miller observed caffeine-releasable spontaneously between the The may the been of brain were faster efficiency responses Ca 2 responses latter channels Ca 2 + Ca 2 + be (1989), eta!,1994). since with filled stem) due also caffeine- fetal recovery substantial process. signal had responses regulatory the during this to very Harada of [Ca 2 +] 1 and and (VGCC evoked faster lost or a recent the was by the a In 222 et 223

Major differences were also observed between culture types with respect to neuronal survival, the percentage of neurons expressing CaBP, and in their vulnerability to glutamate toxicity. While a time-dependent neuronal loss was a common feature in all cultures, its extent was highly variable, being much greater in both fetal and postnatal cultures that were maintained face-up. Use of the “sandwich technique”, in which the neurons were grown on the face of a glass coverslip in contact with the bottom of the tissue culture dish, proved to be of some benefit. Fetal neurons grown in this way had significantly less initial cell loss and, on average, survived for a longer period. Brewer and

Cotman (1989) attributed this effect to a reduction of oxygen toxicity in the limited diffusional space around the neurons although an equally valid argument is that the restricted ‘extra-neuronal space’ in the sandwich method provides a more suitable volume/density ratio for neurons to respond to secreted trophic

(glial or neuronal) factors with enhanced survival. This argument is supported by our observations that face-up cultures are very sensitive to glial density and that neuronal survival is enhanced by the reduction of the medium volume/density ratio and the inclusion of bFGF in the culture medium.

Regardless of the culture conditions, it is important to note that experiments performed on viable neurons after 7-10 DIV were done on a population that represented a selected fraction of those that were initially plated.

While the total neuronal count declined as a function of time, the number of neurons expressing CaBP increased in both fetal face-down and postnatal face-up cultures. In the former, CaBP immunoreactivity was still limited to a minority of the total population but in the latter all neurons were CaBP after

12 DIV. In contrast, fetal face-up cultures displayed a continuous and disproportionate loss of CaBP neurons so that by 14 DIV CaBP 224 immunoreactivity could not be detected at all although many CaBPN neurons were still viable. The enhanced loss of CaBP neurons from fetal face-up cultures suggests that CaBP neurons may be more vulnerable to environmental stresses than CaBPN neurons. In the postnatal cultures, this may be offset by the presence of secreted trophic factor(s), possibly glial (see later), that were capable of inducing the expression of CaBP as evident by the increase in the number of CaBP neurons and by the enhancement of survival of these neurons. The enhanced, albeit limited, expression of CaBP in fetal neurons maintained face-down, and the improved survival of CaBP neurons contrast sharply with the accelerated loss of CaBP neurons in fetal face-up cultures, and lend more support to our suggestion that the major benefits of the sandwich technique can be attributed to an improved volume/density ratio which increases the effectiveness of glial control over the culture environment.

In postnatal cultured neurons, bFGF treatment was associated with a significant enhancement of neuronal survival and a remarkable increase in CaBP expression, whereas in fetal neurons a significant effect was only observed when bFGF treatment was supplemented with culture medium harvested from postnatal cultures. Considering the potential mechanism(s) by which bFGF increases neuronal survival and CaBP content, it is unlikely that a direct effect on neurons is the only mechanism. Certainly, bFGF influenced the number and morphology of the glial cells and it is quite possible that factors released by glia, as a result of the action of bFGF, could be the major means of enhancing neuronal survival and CaBP content. Indeed, postnatal cultures plated face-up and at high density contained many more glial cells and, even in the absence of exogenous bFGF, most if not all neurons eventually expressed CaBP. It is likely therefore that glial-neuronal interactions are an important factor in survival and 225

differentiation of neurons in culture, and that the observed effects of bFGF are, at least in part, mediated via glial release of neurotrophic factors. We cannot

however rule out the possibility that such factors could play a permissive role in association with a direct action of bFGF on neurons.

A very interesting observation was made when bFGF treatment of postnatal cultures was discontinued. The result was an enhanced loss of viable neurons that may be due to the phenomenon of apoptosis reported previously in response to the withdrawal of NGF from cultures of peripheral neurons (Eichler and Rich, 1989; Edwards and Tolkovsky, 1994) or bFGF from cultures of midbrain neurons (Mayer eta!, 1993). It would appear that treatment of neurons with these growth factors results in an absolute requirement for their continued presence and a rapid induction of a programmed series of events leading to cell death if these growth factors are removed.

Vulnerability of hippocampal neurons in vitro to glutamate-induced toxicity was another major aspect of variability between the types of cultures tested.

In cultures maintained face-uD, postnatal neurons were much more sensitive to glutamate toxicity than fetal neurons. It is interesting to speculate that this heightened sensitivity in postnatal neurons is attributable to their larger depolarization- or NMDA-induced 2+Ca responses. Our earlier suggestion that these neurons may also have more efficient 2+Ca recovery mechanisms does not contradict this speculation since the contribution of 2+Ca recovery mechanisms would be more evident during a transient 2+Ca load than during an extended 20-24 hour exposure of glutamate. An interesting feature observed in fetal face-up neurons was that nearly half the neurons were resistant to glutamate toxicity, although all fetal face-up neurons responded to NMDA and glutamate exposures with an increase in their .j21[Ca It is possible depolarizing glutamate-induced glutamate-induced Ca 2 + gated from agreement receptors (Rothman, excitotoxic Also, reducing 34°C, temperature, glutamate excitotoxicity, In of maintained glutamate conducted, that initially process fetal different the activation Regardless in influx, Ca 2 MK8O1 face-down plated. postnatal which, resistance glutamate-induced is significantly was 1985; with process face-uD, glutamate had central monovalent influx and the Ca 2 + (a larger of the by In resulted of VGCC presence non-competitive Choi, neurons, NMDA cannot fetal Ca 2 toxicity neurons, the the of to widely being however influx when these the exposure age reduced glutamate 1987b). neurons blockers cationic fluxes in receptor-associated be largely induction in of accepted seems in during the compared toxicity. using neurons ruled postnatal Mg 2 + such vitro elimination excitotoxicity (and protocols maintained However, fluxes had mediated exposure NMDA to a a out when direct of 30-minute 20-24 in toxicity) notion be is some These to since glutamate-induced the neurons. the and a excitotoxicity both antagonist) comparison major employed of medium by hour a result neuroprotective protocol that face-down, subsequent two Mg 2 small most more Ca 2 + and an fetal glutamate mediator exposure Ca 2 results The increase of resulted contribution than likely during also and channels the used in was may involvement influx these experiments the postnatal reduced highly activation are 90% reflects exposure of for excitotoxicity to the in and not in estimated effect consistent the [Ca 2 glutamate through two a the and of most be after selective higher from neuronal the this the induction +1 models. valid neurons is against of of at effective exposure were in neurons voltage resulting type VGCC VGCC NMDA room EC 50 . EC 50 with because at survival injury. of of for the in 226 in to 227

associated with the activation of both NMDA and non-NMDA type of glutamate receptors.

It is clear from our results that in cultured hippocampal neurons several important properties (e.g., 21+][Ca regulation mechanisms, CaBP expression and survival in vitro) are sensitive to a multitude of factors in the culture environment. These include the culture type (fetal vs postnatal), culture age (Dlv), orientation state (face-up vs face-down), the number of glial cells and growth factor availability. Other factors such as the plating density and the serum content of the culture medium are also major determinants of the neuronal culture properties but they have not been the object of a detailed assessment in our work. The sensitivity of vital neuronal characteristics to culture conditions complicates attempts to compare results obtained from different laboratories and precludes generalization and extrapolation of results to other experimental models using different species for cultured neuron preparation or using whole animals.

Having established some of the basic properties of our cultured neuron preparation our next aim was to determine the effect of artificial 2+Ca buffers and the presence of CaBP on increases in 2[Ca +1 induced either transiently or in a manner which would, depending upon the degree of stimulation, lead to cell death by a process of excitotoxicity.

During short exposures to NMDA or high K-induced depolarization, changes in 21[Ca in BAPTA- or DMB-loaded neurons displayed reduced peak responses andj slower rates of increase and recovery compared to control neurons confirming the role of BAPTA (and DMB) as effective and fast intracellular 2+Ca buffers. Characteristically, the recovery phase was slow and

regulation

postnatal

compared

postnatal

associated

and

interesting

stimuli

of

however,

that

an

relative

used

the

effects

buffer,

expressing

seem

responses

comparison

even

change

responses

neurons,

neurons

prolonged

CaBP

increase

neuronal

postnatal

may

in

larger

therefore

are

the

of

activity

our

in

was

after

recoveries

CaBP

have

that

both

to

is

CaBP

of

[Ca 2 +]

to

in

with

when

causing

expression

CaBP.

experiments,

in

than

markedly these

postnatal

phenotype.

quite

neurons

a

note

CaBP

the

the

during

resulted

fetal

very

of

that

prolonged

may

CaBPN

compared

density

stimulus

neurons

Ca 2 +

different

that

were

evident While

[Ca 2 1

and

supports

short

there

have

electrophysiological

loaded

different

CaBPN

of

neurons

larger

from

similar

postnatal

recovery

control

BAPTA

or

CaBP

It

of

nature

are

and,

therefore

(Chard

displayed

has

to

responses

to

is

by

membrane

other

with

our

peak

major

possible,

linger

their

neurons.

a

in

is

to

although

been

in

values

most

higher

(tens

earlier

fetal

possibly

mechanisms.

many

etal,

CaBPN

CaBP

a

coincidental

Ca 2 +

CaBPN

BAPTA-like

differences

at

been

discontinued.

fast

are

likely

and

of

a

in

however,

Ca 2 +

net

1993;

cases,

conclusion

This

in

higher

responses

and

milliseconds),

experiments

masked

control

postnatal

reconstructed

just

postnatal

neurons

fetal

counterparts

influx

acts

difference

complete

channels

Lledo

one

incomplete

changes

concentration

between

buffer

neurons

It

as

values

that

of

by

must

aspect

that

Unlike

had

neurons.

were

a

eta!,

Ca 2 +

neurons.

the

fast

the

where

compared

or

recoveries

a

between

be

higher

in

neuronal

in

while

in

these

the

relatively

observed

Ca 2 +

potential

of

a

high

1993). BAPTA-loaded

these

recoveries,

Fig

in

fetal

mentioned,

reduction

a

kinetics

depolarizing

CaBP

For

in

the

7.1.

larger

peak

responses

affinity

buffering

neurons,

buffer-loaded

neurons

to

fetal

Ca 2 +

ease

rates

when

It

long

in

buffering

neurons

It

Ca 2 +

is

change

of

both

would

in

while

neurons

and

also

Ca 2

of

stimuli

of

Ca 2

the

such

effect

and

were fetal

+

228 in 229

During prolonged exposure to glutamate, BAPTA- and DMB-loaded neurons demonstrated slower rates of change in 2+][Ca compared to control neurons indicating that sufficient buffer was available in the cytoplasm to slow down changes in 2+][Ca even during these ‘unphysiological’ conditions of stimulation. Unlike the situation during shorter exposures, peak 2+Ca responses were not reduced in buffer-loaded neurons, leading us to conclude that active 2+Ca regulatory mechanisms determine the peak 2+Ca responses during longer stimuli without a. contribution from the passive intracellular buffering systems. This result is particularly interesting when considering the potential use of BAPTA-like buffers to protect against Ca-mediated neuronal injury believed to play a major role in the in vivo excitotoxic2 circumstances which are almost exclusively of a prolonged nature (e.g., minutes of ischemia). In a small set of experiments, fetal CaBP neurons exposed for several minutes to glutamate or NMDA displayed the same characteristic features observed in shorter stimuli, i.e., higher peak and delayed recovery.

In view of the effects of buffer loading or CaBP expression on the peak of 2+Ca responses during prolonged stimuli, and the prolonged and slow recovery process under the same circumstances, it is probably not surprising that glutamate-toxicity was enhanced in both fetal and postnatal neuronal cultures loaded with either BAPTA or DMB, as well as in postnatal CaBP neurons compared to the corresponding control cultures. In buffer-loaded neurons, an increased 2+Ca load, inferred from the unchanged peak 21+][Ca in the presence of a higher intraneuronal buffering capacity, would impose a greater demand on cellular energy stores in order to restore pre-stimulation 2[Ca and is likely a major factor underlying the enhanced cell death in buffer-loaded neurons. The increase in ]2[Ca in neurons during the exposure to BAPTA-AM or DMB-AM 230 is unlikely to be contributing to the enhanced glutamate toxicity observed in buffer loaded neurons since control cultures exposed to hBAPTA-AM displayed a similar increase in 2+]j[Ca without a subsequent enhancement of glutamate- induced excitotoxicity. The contribution of other ‘non-chelating’ effects of BAPTA or DMB to enhanced glutamate toxicity cannot, however, be excluded. With respect to the increased vulnerability of neurons which express CaBP we can only conclude that there is a clear association between these factors but a causal relationship cannot be established on the basis of either our experiments or, indeed, those of Mattson eta! (1991).

In the light of our findings it would be appropriate to return to the question, should 2Ca buffering be neuroprotective? The hypothesis that an enhanced 2+Ca buffering capacity of neurons will protect them against 2+Ca mediated neuronal injury is based on the assumption that the peak concentration of 2[Ca during excitotoxicity is the sole determinant of the cell injury, and that 2Caj buffering will reduce this peak and thereby reduce or eliminate the cytotoxic consequences. While our findings refute the assumption of a reduced peak response, other flaws are still inherent in the hypothesis. The most obvious of these are, first, the possibility of an impairment of 2Ca +mediated activation of 2Ca +recovery (i.e., direct or indirect negative feed back loops) as a result of reduced peak 2+Ca levels and, second, the fact that the presence of an artificially-high cytoplasmic 2Ca buffering capacity will slow down and prolong the 2[Ca ‘11recovery phase. More subtle flaws become obvious when the high temporo-spatial organization of the 2Ca signal as well as the speed and mobility of the exogenous and endogenous 2+Ca buffers are considered. Disruption of the spatial properties of the 2+Ca signal (such as microdomains and 2+Ca waves) by a fast and 231

mobile buffer can be harmful (by increasing stray signalling and by disrupting 2Ca tmediated physiological processes). Also, the loss of the temporal precision of a 2+Ca signal can lead to a distortion of the appropriate time relationship between the 2+Ca signal and other 2Ca +..activated second messenger systems such as 31P and DAG. As a result of this complexity, the effects of additional 2Ca buffering on a particular biological system are essentially unDredictable and probably very cell-specific (depending on such factors as endogenous buffers mobility and affinity, artificial buffer immobilization as well as the nature of the time/space organization of the 2+Ca signal).

In attempting to arrive at valid conclusions concerning our data we must be mindful of the pitfalls and limitations of the methods used. Examples have already been cited with respect to the tissue culture methods, and some comments regarding our use of imaging technology are also warranted. Attempts to understand the effects of enhancing the 2Ca buffering capacity in neurons have frequently made use of a number of fluorescent 2+Ca imaging techniques. Unfortunately, measurements and conclusions based on these techniques may not reflect the complexity of the 2Ca signal because of the limited spatial and temporal resolution of imaging systems available at present (msec, pm) compared to the resolution of the 2+Ca signalling process (psec and nm). Furthermore, the introduction of fluorescent 2+Ca probes, particularly if used at high intracellular concentrations, into the cell can distort the 2+Ca signal to be measured since these probes would act as fast, mobile and high-affinity 2Ca buffers with a particularly disruptive effect on the fast, short-range 2+Ca processes such as neurotransmission possibly in a manner similar to that of the parent molecule BAPTA (Adler eta!, 1991). During 2Ca judicious selected selected the ‘higher’ being in loops vulnerability vitro- vivo number neuroprotection modulation All link that [Ca 2 +] Additionally, average where Ca 2 + functionally responses, cell these functional measured models Despite may Another in as of properties channels many animal of have vivo the neuronal physiological much [Ca 2 i use technical be culture of high organisms. is will useless, these highly preclude of of Ca 2 been serious consequences this changes not as models, these the and is depend affinity and (e.g., on population conditions. the function reliable. criticisms limitations at:the localized imaging vital in the we in problem methods parameter successful vulnerability the enhanced in and the Ca 2 + on Ca 2 Neuroprotection have inherent level [Ca 2 +]j neighbourhood by ‘hot’ the The not is and +..mediated in of of should in limited not probes enhancing Extrapolation of can vitro. the cell imaging based in studying spots (such very Ca 2 predictions the cell to attempted vivo of test be connectivity a be neuronal by properties. can neuronal fast. on particular very as in buffering processes treatment technology taken the processes should the the the the [Ca 2 +] of also useful Also, from fact intracellular of to immediate cellular improvement possible into somata saturate, development the measure -which cell thus that capacity) The the all or such and if account may effect and the function, of membrane Ca 2 in be it extensive role in rather our as is is in vitro vicinity be and right evaluated changes Ca 2 + appropriate a of not neuroprotection vitro on of or initiated. measurements buffering in measure in become specific than results questions Ca 2 deterioration the any and reproducible vitro release potential) models of feed in overall attempt the the membranous in buffers to [Ca 2 +] to of capacity. whole changes back view neurites. sites, a the the the are large well in of to of and in in of in and 232 a a

neurons

molecular

changes

knockout,

requires

a

neuronal

restricted

to

neuronal

better

might

can

buffer channels,

effects

mediated

distribution

FUTURE

‘and

attempted

new

asked.

causal

a

associate

capacity

So

The

simple’ pharmacological

also

understanding

mobility

on

as

far,

the

relationship

in Answers

characteristic

death

to

biological

use

cellular

gene

versus

DIRECTIONS

fast

controls be

neuronal

in

could

ability

the

the

hypotheses

to

order

very

of

with

short-range

(e.g.,

in

knockout,

study

target

reporting

aDoroDriately

‘deep’

functions.

excitotoxic

to

be

to

helpful

specific

for

to

techniques

properties.

such

between

of

dextran-conjugation)

a

manipulate

tools.

of

give

selectively

(e.g.,

very

neurons

the

slow

the

about

of

questions

gene

in

insight

processes,

subcellular

spatial

powerful

associative

resistance modulating

functional

Fast

Also,

events.

the

Ca 2 +

designed

such

the

induction

that

Achieving

the

expression

Ca 2 -mediated

buffers

into

in

aspects

in

mediated

can

express

as

expression

tool

vitro

vivo

initiated

domains

the

role

or

neuronal-targeted

buffers

or

the

be

or

and

in

vulnerability

and

models

this

possible

mode

incidental

of

vital

of

transfection.

Ca 2

the

CaBP

of

processes.

slow

Ca 2

CaBP

possibly

objective

in

to

CaBP

(site-specific

of

selective

before

of

the

processes

manipulate

can

signal

either CaBP

buffers

mechanisms

action

signalling

in

phenomena.

vicinity

and

be

to

neurons

in

by

will

in

spontaneously

and

a

Altering

mRNA modulation

used

vivo

The any

of

using

can

isolation

disease

that

require

immobilization)

a

may

of

subcellular

and,

particular

use

experiments

drug.

to

have

has

influx

of

buffers

antisense

may

test

the

lead

of

Establishing

eventually,

action

process)

been

from

the

differential

of

CaBPN

trigger

artificial

or

specific

to

or

Ca 2 -

use

that

release

Ca 2 +

other

a

of

are

of 233 recovery observed fetal and fetal is face-up Dlv, in than initial cells, culture CaBP of GENERAL changes under requirement under particularly the culture agonist-induced 4. face-up face-down all 3. that 2. 1. age-dependent and and number these the age viable (>90% Compared After Fetal In of associated in observed the vulnerability conditions. SUMMARY effects primary [Ca 2 fetal (Dlv), conditions for neurons remarkable availability cultures an neurons of and an after +j neurons. neurons initial of orientation to appropriate in cultures in with postnatal peak growth neuronal this 3 fetal both AND postnatal These maintained are to loss is Dlv). of with the expressing glutamate an Ca 2 + initial CaBPN. neurons, fetal Following CONCLUSIONS of growth of factors isolated differentiation include state face-up all loss CaBP hippocampal control loss responses and neurons viable using that (face-up factors. in toxicity postnatal of postnatal CaBP. the event short cultures vivo since CaBP neurons is neurons the is culture significantly that transient much or but to neurons, sandwich vs are In it in neurons cultured a is display postnatal face-down), more are from neurons highly distinct type being vitro, unlikely faster much Ca 2 + likely survival, all (fetal technique CaBP evidence lower does dependent display neurons than culture phenotype. continues neurons, that higher responses, one vs the not in (<50% CaBP postnatal), expression depolarization of fetal by density maintained types satisfy of than display on many this 12 an and expression neurons. a by those tested, DIV. increase the increase variety the by of an 3 of glial DIV) 14 In 234

the

recovery

reach

exposures

buffers

reduced

extracellular

BAPTA

accelerated medium

develop

evident

neurons

additional

effect

postnatal

Ca 2 +

induced that,

when

Postnatal

recovery

6.

5.

with

the

compared

regulatory

on

also

or

by

a

from

rate

With

when

excitotoxicity.

In

times

same

certain

neurons.

dependence

neurons

factors

DMB

to

the

the

vitro,

neuronal

effectively

K,

time

of

glutamate.

transient

postnatal

maturation

bFGF

significant

are

levels

rise

are

to

the

mechanisms

cellular

bFGF

is

necessary

still

are

entirely

fetal

and

This

greater

treatment

effects

loss

as

also

for

greatly

enhances

reduce exposures

cultures.

peak

neurons.

these

effect

elements

However,

is

increase

of

the

more

consistent

with

observed

for

of

fetal

[Ca 2 +]j,

result

sustained

prolonged.

the

of

is

prior

is

the

BAPTA

vulnerable

both

combined

at

non-buffer

Postnatal

neurons

of

rate

in

in

Both

the

full

least

in

loading

cultured

postnatal

an

when

the

with

and

an

of

peak

expression

of

otherwise-small

presence

compared

partially

rise

increased

survival

into

In

these

a

a

neurons

with

to

bFGF

with

prolonged

loaded

[Ca 2 j 1

both

buffering

neurons

of

glutamate-induced

cultures

postnatal

the

[Ca 2 1

results

the

treatment

of

instances

mediated

and

of

to

sensitivity

neurons,

treated

presence

this

responses

acetoxymethyl

the

DMB.

to

expression

action,

recovery.

which

lead

effect

neurons,

induced

either

growth

bFGF

with

by

the

is

us

although

may

of

discontinued.

namely

to

of

an

NMDA

effects.

eventually

to

prolongation

factor

conditioned

bFGF

by

glutamate-

neurotoxicity

bFGF

These

changes

indirect

of

conclude

then

prolonged

esters

CaBP

a

or

their

also

since

on

secrete

This

CaZ+

high

fetal

in

in

of

235

of

an is 236

7. In both fetal and postnatal cultures, enhancing the 2Ca buffering capacity by loading neurons with BAPTA or DMB is associated with a significant increase in their vulnerability to glutamate-induced excitotoxicity.

8. In the presence of extracellular 2+,Ca acetoxymethyl derivatives of BAPTA, DMB or the non-calcium binding molecule hBAPTA evoke a rapid increase in 2[Ca that recovers rapidly and completely on removal of the ester. This jincrease is most likely mediated via the activation of VGCC.

9. In both fetal and postnatal cultures, both NMDA- and high extracellular K+induced increases in 2[Ca +11are significantly larger in CaBP neurons than in CaBP neurons. In fetal neurons, these larger responses are significantly prolonged with a slow and incomplete recovery of 21[Ca In contrast, larger 2+Ca responses in postnatal CaBP neurons are.i associated with much faster and complete recoveries compared to those observed in the CaBP neurons.

10. CaBP postnatal neurons are significantly more vulnerable to glutamate-mediated excitotoxicity when compare to their CaBPN counterparts.

An increased vulnerability of CaBP neurons is also observed in fetal face down cultures that fail to recover after a transient exposure to NMDA or high extracellular K+.

11. An increased intraneuronal 2Ca buffering capacity either by artificial 2+Ca buffers, such as BAPTA or DMB, or in neurons that express CaBP does not confer neuroprotection. Rather, both are associated with an increase in the vulnerability of these neurons to glutamate-induced excitotoxicity in vitro.

ci:

0

and

Figure

0.5

3.0

3.5

2.0-

2.5

4.0

1.0-

1.5-

CaBP(+

0

7.1: imulated

,

in

I

I

1

50

response

Ca 2

to

100 changes

I

a

short-duration

Time

in 150

I

fetal

(s)

neurons:

(25

200 seconds)

I

control,

Control

CaBP(+)

BAPTA-loaded

exposure

250

BAPTA-loaded

to

300

NMDA. 237 238

ABBREVIATIONS

FWS:

EPSP:

FOR:

EthD:

ER:

FBS:

EGTA:

EMEM: EAA:

EC 50 :

DRG: DMSO:

DNA: DMB:

DIV:

DMEM:

DAG: CMF

CV:

DHP: CNQX:

CgTx:

CBP: cAMP:

CaBPN:

CaBP:

CaBP:

Ca 2

BSS:

[Ca 2 +]: BSA: ATP:

bFGF: Ara-C:

BAPTA: APV:

APH:

AMPA:

ADP:

AM:

ACPD:

AR: 5-FDU:

+:

BSS:

Funnel

Excitatory

Free

Fetal

Ethedium

Endoplasmic

Ethyleneglycol-tetraacetic

Eagle’s Excitatory

Ligand

Dimethyl

Dorsal Dulbecco’s

Deoxyribonucleic

Day(s) Cresyl

5,5’-dimethylBAPTA

Diacyl Ca 2 /Mg 2

Dihydro-pyridines 6-cyano-7-nitro-quinoxaline-2,3-dione

Omega-conotoxin

Ca 2 -binding

Cyclic

Not

Calbindin-D28K

Immunoreactive

Buffered

Intracellular Bovine

Ionized Basic Arabinoside-C

Adenosine

a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic

2-Amino-7-phosphonoheptanoate

Adenosine

2-Amino-5-phosphonovalerate Acetoxymethyl

1

Arachidonic

5-fluoro-deoxyuridine

1-amino-i

,2-bis-(O-aminophenoxy)ethane-N,N,N’,N’-tetraacetic

immunoreactive

oxygen

bovine

fibroblast

glycerol

Adenosine

violet

root in

web

concentration

minimal serum

calcium

salt

sulfoxide

vitro

homodimer

post-synaptic

,3-cyclopentane-dicarboxylic

amino

triphosphate

diphosphate

ganglion

modified

spider

ionized

serum

radical

free

reticulum

acid

Nissi

solution

protein

albumin

essential

ester

growth

for

acids

BSS

acid

monophosphate

GVIA

stain

calcium

CaBP

for

Eagle’s

evoking

factor

CaBP

potential

medium

acid

concentration

medium

half

the

maximal

acid

response

acid

acid

possible 239 240

GFAP: GliaI fibrillary acidic protein GIuR: Glutamate receptors GLUT: Glutamate hBAPTA: N-(o-methoxyphenyl)-imino-diacetic acid HBSS: Hank’s BSS HK: High-potassium (50 mM) containing medium HS: Horse serum HVA: High-voltage activated iGluR: lonotropic glutamate receptors IHC: lmmunohistochemistry :31P Inositol triphosphate KA: Kainic acid LDH: Lactate dehydrogenase LGCC: Ligand-gate 2Ca channels LVA: Low-voltage activated mGIuR: Metabotropic glutamate receptors mRNA: Messenger ribonucleic acid nAchR: Nicotinic acetyl choline receptors NBQX: 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo(F)quinoxaline NGF: Nerve growth factor NMDA: N-methyl-D-aspartate NO: Nitric oxide NSE: Neuron-specific enolase NT-3: Neurotrophin-3 PBS: Phosphate Buffered saline PCP: PDL: Poly-D-lysine P1: Phasphatidyl inositol PIP: Phasphatidyl inositol phosphate 2PIP Phasphatidyl inositol diphosphate PK-C:: Protein kinase-C PL-A2: Phospholipase-A2 PV: Parvalbumin QA: ROl: Region of interest SE: Standard error SEM: Standard error of the mean SN: Substantia nigra SR: Sarcoplasmic reticulum TC: Tissue culture TNF: Tumor necrosis factor TTX: Tetrodotoxin VGCC: Voltage-gate 2Ca channels 241

BIBLIOGRAPHY Banker Baker Baker Baimbridge Baimbridge Baimbridge Baimbridge Baimbridge Baimbridge Augustine Auer Andressen Albers Allen Adler Loligo. RN, TJA, Topics PF, culture. PF, with current-evoked D28k. and AM, protein calcium-binding bulb binding Neurosci induced dentate Sci. Epilepsy transmitter hypoglycemia markers 56 inflow synapse. ready calcium GA, GW, Siesjo Umbach DiPolo olfactory GJ, Augustine of lucifer C, Baker KG, KG, 635:363-80. KG, KG, KG, KG Cowan Goldberg for J via Memb Synapse. in the protein Biumke gyrus neurotoxicity Physiol. chelators Adler Brain of Res (1992). Abstr. Peet Parkes Mody Miller Kao the clinical BK J R Na 1 /Ca 2 0 rat. PF secretion nerve yellow JA Neurosci. (1984). WM Suppl (1988). Transp. bulb chick J and Res. and EM, MJ, (1985). depolarization in GJ, JJ MP, I, (1987). I, protein (1988). Brain CO 14:1264. trial 383:369-94. cells. 7:269-77. Calcium-binding the Cello (1977). Miller attenuate kindling. its epilepsy. dye (1982). McLennan Chariton 7, Duffy brain. 126:397-425. Chol (1981). from Axonal cerebellum, role in Res. in pp Biological 22:95-147. coupling exchange 11:1496-507 MR Intracellular following JJ Cell brain rat Calcium Calbindin-D28k 211-20. DW SN, in presynaptic Cell Rat (1985). (1993). 245:223-9 lmmunohistochemical CAl MP neurotransmitter Epilepsia. Tissue seizures Ann calcium Vitamin in ischemia?. (1989). H, Charlton hippocamplal Calcium. but (1991). rat differences Church pyramidal buffering commissural, in Neurol. hippocampal proteins Ca 2 Res. CAl Reduction the not Calcium-binding (Ribac and D N-methyl-D-aspartate nerve 26:460-65. MP squid dependent with J pyramidal protects The 2:65-67 271:181-208. Ann indicator 24:699-707. (1991). in in neuron (1991). CE, between neurons terminals. the axons release the calcium axon. Neurol. of formation amygdala, Gall presence rat dentate against localization cultures. quin-2 calcium-binding neurons Bursting and Alien hippocampal CM, proteins: Nature. at in ischemia, signal homeostasis. 25:398-403. dispersed the Ann axoplasm and glutamate gyrus. Mody intracellular inhibits perforant of squid is for antagonists: response NY Soc calbindin of olfactory 315:755- correlated selective I, calcium- Acad In: giant cell Ca 2 Ed.). of path Curr The 242 to 243

Barakat I, Droz B (1989). Inducing effect of skeletal muscle extract on the appearance of calbindin-immunoreactive dorsal root ganglion cells in culture. Neuroscience. 28:39-47.

Beech DJ, Bernheim L, Mathie A, Hille B (1q91). Intracellular Ca buffers + 2 disrupt muscarinic suppression of Ca current and M current in rat sympathetic neurons. Proc NatI Acad Sci USA. 88:652-56.

Benveniste H, Drejer J, Schousboe A, Diemer NH (1984). Elevation of the extracellular concentrations of glutamate and aspartate in rat hippocampus during transient cerebral ischemia monitored by intracerebral microdialysis. J Neurochem. 43:1369-74.

Berridge MJ (1990). Calcium Oscillations. J Biol Chem. 205:9583-86.

Biscoe TJ, Duchen MR (1990). Responses of the type I cells dissociated from a rabbit carotid body to hypoxia. J Physiol. 428:39-59.

Blaustein MP (1988). Calcium transport and buffering in neurons. Trends Neurosci. 11:438-43.

Bloom S, Davis DL (1972). Calcium as mediator of isoproterenol-induced myocardial necrosis. Am J Path. 69:459-70.

Boeynaems JM, Heilporn S, Broeders F, Braekman JC (1993). Enhancement of the endothelial production of prostacyclin by substituted derivatives of BAPTA-AM. Eur J Pharmacol. 233:13-20.

Braughler JM, Bregenzer JF, Chase RL, Duncan LA, Jacobsen EJ, McCall JM (1987). Novel 21-amino steroids as potent inhibitors of iron-dependent lipid peroxidation. J Biol chem. 262:10430-40.

Bredderman PJ, Wasserman RH (1974). Chemical composition, affinity for calcium and some related properties of the vitamin D dependent calcium- binding protein. Biochemistry. 13:1687-94. Brewer GJ, Cotman CW (1989). Survival and growth of hippocampal neurons in defined medium at low density: advantages of a sandwich culture technique or low oxygen. Brain Res. 494:65-74. Brinley FJ (1980). Regulation of intracellular 2Ca in squid axon. Fedn Proc. 39:2778-82. Brodie AE, Reed DJ (1991). Calcium chelation induced glutathione efflux from tumor cells and prevention by ruthenium red or . Biochem Biophys Res Communic. 176:276-79.

Bronner F, Pansu D, Stein WD (1986). An analysis of intestinal calcium transport across the rat intestine. Am J Physiol. 250:G261-69. Brown DA, Gahwiler BH, Griffith WH, Halliwell JV (1990). Membrane currents in hippocampal neurons. Prog Brain Res. 83:141-60. 244 Buchan A, Pulsinelli WA (1990). Hypothermia but not N-methyl-D-aspartate antagonist, MK-801, attenuated neuronal damage in gerbils subjected to transient global ischemia. J Neurosci. 10:311-16.

Buchan AMJ, Baimbridge KG (1988). Distribution and co-localization of calbindin D28k with VIP and neuropeptide Y but not somatostatin, galanin and substance P in the enteric nervous system of the rat. Peptides. 9:333-38.

Carafoli E (1987). Intracellular calcium homeostasis. Annu Rev Biochem. 56:395-433.

Carpenter-Deyo L, Duimstra JR, Hedstrom 0, Reed DJ (1991). Toxicity to isolated hepatocytes caused by the intracellular calcium indicator, Quin 2. J Pharmacol Exp Therap. 258:739-46.

Celio MR (1990). Calbindin D28k and parvalbumin in the rat nervous system. Neuroscience. 35:375-475. Chad J (1989). Inactivation of calcium channels. Comp Biochem Physiol. 93A:95-1 05.

Chan PH, Fishman RA, Longar S. Chen S. Yu A (1985). Cellular and molecular effects of polyunsaturated fatty acids in brain ischemia and injury. Prog Brain Res. 63:227-35.

Chard P, Bleakman D, Christakos S, Fullmer CS, Miller RJ (1993). Calcium buffering properties of calbindin-D28k and parvalbumin in rat sensory neurones. J Physiol. 472:341-57.

Cheng B, Christakos S, Mattson MP (1994). Tumor necrosis factors protect neurons against metabolic-excitotoxic insults and promote maintainance of calcium homeostasis. Neuron. 12:139-53.

Choi DW (1985). Glutamate neurotoxicity in cortical cell culture is calcium dependent. Neurosci Lett. 58:293-97. Choi DW (1987a). and dextromethorphan attenuate glutamate neurotoxicity. Brain Res. 403:333-36. Choi DW (1987b). Ionic dependence of glutamate neurotoxicity. J Neurosci 7:369-79. Choi DW (1988). Calcium mediated neurotoxicity: relationship to specific channel types and role in ischemic damage. Trends Neurosci. 11:465- 69. Choi DW (1990). Cerebral hypoxia: some new approaches and unanswered questions. J Neurosci. 10:2493-501.

Choi DW (1991). Fast and slow excitotoxicity in cortical cell cultures. In: Excitatory amino acids (Moroni F, Simon RP, Wood JH, Eds.). Raven Press, pp 55-61. 245 Choi DW (1992). Excitotoxic cell death. J Neurobiol. 23:1261-76. Christakos S. Norman AW (1980). Vitamin D-dependent calcium binding protein and its relation to 1,25-dihydroxy-vitamin D receptor localization and concentration. In: Calcium-binding proteins: structure and function (Siegle FL, Carafoli E, Kretsinger RH, MacLennan DH, Wasserman RH, Eds.). Elsevier North Holland, Inc., pp 371-78. Church J, Fletcher EJ, Abdel-Hamid KF, MacDonald JF (1994). Loperamide blocks high voltage-activated Ca + channels and N-methyl-D-aspartate evoked responses in rat and mouse cultured hippocampal pyramidal neurons. Mol Pharmacol. 45:747-57.

Collazo D, Takahashi H, McKay RD (1992). Cellular targets and trophic function of neurotrophin-3 in the developing rat hippocampus. Neuron. 9:643-56.

Coorssen JR, Haslam RJ (1993). GTP gamma S and phorbol ester act synergistically to stimulate both -independent secretion and phospholipase D activity in permeabilizedCa human platelets. Inhibition by BAPTA and analogues. FEBS Lett.2 316:170-74. Corsellis JAN, Bruton CJ (1983). Neuropathology of status epilepticus in humans. Adv Neurol. 34:129-39. Cotman CW, Monaghan DT, Otterson OP, Storm-Mathiesen J (1987). Anatomical organization of excitatory amino acid receptors and their pathways. Trends Neurosci. 10:273-80. Culling CFA (1974). Handbook of histopathological and histochemical techniques (Third edition). Butterworths and Co. Publishers Ltd., pp 446-47.

Curtis DR, Johnston GAR (1974). Amino acid transmitters in the mammalian central nervous system. Ergeb Physiol Biol Chem Exp Pharmakol. 69:98-188.

Dalgarno D, Klevit RE, Levine AB, Williams RJP (1984). The calcium receptor and trigger. Trends Pharmacol Sd. 4:266-71. Davies JD, Francis AA, Jones AW, Watkins JC (1981). 2-Amino-5- phosphonovalerate (2-APV), a potent and selective antagonist of amino acid-induced and synaptic excitation. Neurosci Lett. 21:77-81.

Dechesne CJ, Thomasset M (1988). Calbindin (CaBP 28 kDa) appearance and distribution during development of the mouse inner ear. Brain Res. 468:233-42. Denton RM, McCormack JG (1985). 2Ca transport by mammalian mitochondria and their role in hormone action. Am J Physiol. 249:E543- 54. 246 Denton RM, McCormack JG (1990). 2+Ca as a second messenger within mitochondria in the heart and other tissues. Annu Rev Physiol. 52:45 1- 66.

Deshpande JK, Wieloch T (1986). Flunarizine, a calcium entry blocker, ameliorates ischemic brain damage in the rat. Anesthesiology. 64:215- 24.

Dieter P, Fitzke E, Duyster J (1993). BAPTA induces a decrease of intracellular free calcium and a translocation and inactivation of protein kinase C in macrophages. Biol Chem Hoppe-Seyler. 374:171-74.

DiPolo R, Beauge L (1993). Regulation of Na-Ca exchange: An overview. Ann NY Acad Sci. 279:100-11. Dubinsky JM (1993a). Effects of calcium chelators on the intracellular calcium and excitotoxicity. Neurosci Lett. 150:129-32.

Dubinsky JM (1993b). Examination of the role of calcium in neuronal death. Ann NY Acad Sci 279:34-42. Dubinsky JM (1993c). Intracellular calcium levels during period of delayed excitotoxicity. J Neurosci. 13:623-31. Dubinsky JM, Rothman SM (1991). Intracellular calcium concentration during “chemical hypoxia” and excitotoxic neuronal injury. J Neurosci. 11:2545-51. Dypbukt JM, Thor H, Nicotera P (1990). Intracellular 2Ca chelators prevent DNA damage and protect hepatoma 1C1C7 cells from quinone-induced cell killing. Free Radical Res Communic. 8:347-54.

Edwards SN, Tolkovsky AM (1994). Characterization of apoptosis in cultured rat sympathetic neurons after nerve growth factor withdrawal. J Cell Biol, 124:537-46.

Eichler ME, Dubinsky JM, Rich KM (1992). Relationship of intracellular calcium to dependence on nerve growth factor in dorsal root ganglion neurons in cell culture. J Neurochem. 583:263-69.

Eichler ME, Rich KM (1989). Death of sensory ganglion neurons after acute withdrawal of nerve growth factor in dissociated cell cultures. Brain Res. 482:340-46.

Ellisman MH, Deerinck TJ, Ouyang Y, Beck CF, Tanksley SJ, Walton PD, Airey JA, Sutko JL (1990). Identification and localization of ryanodine binding proteins in the avian central nervous system. Neuron. 5:136-46. Enderlin S, Norman AW, Celio MR (1987). Ontogeny of the calcium-binding protein calbindin D-28k in the rat nervous system. Anat Embryol. 177:15-28.

Feher JJ (1983). Facilitated diffusion by intestinal calcium-binding protein. Am J Physiol. 244:C5303-7. 247 Feher JJ, Fullmer CS, Wasserman RH (1992). Role of facilitated diffusion of calcium by calbindin in intestinal calcium absorption. Am J Physiol. 262:C51 7-26.

Freund TF, Buzsaki G, Leon A, Baimbridge KG, Somogyi P (1990). Relationship of neuronal vulnerability and calcium-binding protein immunoreactivity in ischemia. Exp Brain Res. 83:55-66. Friel DD, Tsien RW (1990). Two pharmacologically distinct 2Ca stores which modify [Ca elevations produced by depolarizations in sympathetic neurons. ]2 Fed Proc. 16:1274. Garthwaite G, Garthwaite J (1987). Receptor-linked ionic channels mediate N methyl-D-aspartate neurotoxicity in rat cerebellar slices. Neurosci Lett. 83:241-46.

Garza R, Bickford-Wimer PC, Hoffer BJ, Freedman R (1987). Heterogeneity of nicotine actions in the rat cerebellum: an in vivo electrophysiologic study. Pharmacol Exp Ther. 240:689-95. Giffard RG. Weiss JH. Choi DW (1992). Extracellular alkalinity exacerbates injury of cultured cortical neurons. Stroke. 23:1817-21.

Gill R, Foster AC, Woodruff GM (1988). MK-801 is neuroprotective in gerbils when administered during the post ischemic period. Neuroscience. 25:847-55.

Goldberg MP, Choi DW (1993). Combined oxygen and glucose deprivation in cortical cell culture: Calcium-dependent and calcium-independent mechanisms of neuronal injury. J Neurosci. 13:3510-24.

Goodman JH, Wasterlain CG, Massarweh WF, Dean E, Sollas AL. Sloviter RS (1993). Calbindin-D28k immunoreactivity and selective vulnerability to ischemia in the dentate gyrus of the developing rat. Brain Res. 606:309-14.

Gray R, Johnston D (1987). Noradrenaline and beta-adrenoceptor agonists increase activity of voltage-dependent calcium channels in hippocampal neurons. Nature. 327:620-22. Grynkievicz G, Poenie M and Tsien RY (1985). A new generation of 2Ca indicators with greatly improved fluorescence properties. J Biol Chem. 260:3440-48.

Guthrie PB, Mattson M, Mills LR, Kater SB (1988). Calcium homeostasis in molluscan and mammalian neurons: Neuron-selective set-point of calcium rest concentrations. Soc Neurosci Abstr. 14:582.

Hagberg H, Lehmann A, Sandberg M, Nystrom B, Jacobson I, Hamberger A (1985). lschemia-induced shift of inhibitory and excitatory amino acids from intra- to extracellular compartments. J Cereb Blood Flow Metab. 5 :41 3-19. 248 Halliwell B, Gutteridge JMC (1985). Oxygen radicals and the nervous system. Trends Neurosci. 8:22-26.

Harada K, Yoshimora T, Nakajima K, Ito H, EbinY, Shingai R (1992). N methyl-D-aspartate increases cytosolic Ca+ via G-proteins in cultured hippocampal neurons. Am J Physiol. 262:C870-75

Hashimoto 5, Bruno B, Lew DP, Pozzan T, Volpe P, Meldolesi J (1988). Immunocytochemistry of calciosomes in liver and pancreas. J Cell Biol. 107:2523-31. Haughiand RP (1992). Handbook of fluorescent probes and research chemicals (1992-94). Catalog of Molecular Probes Company, p 114.

Heizmann CW, Braun K (1992). Changes in -binding proteins in human neurodegenerative disorders. Ca Trends Neurosci.2 15:259-64. Heizmann CW, Hunziker W (1991). Intracellular calcium-binding proteins: more sites than insights. Trends Biochem. 16:98-103.

Hendrickson AE, VanBrederode JF, Mulligan KA, Celio MR (1991). Development of the calcium-binding proteins parvalbumin and calbindin in monkey striate cortex. J Comp Neurol. 307:626-46. Henkart M (1980). Identification and functions of intracellular 2Ca stores in axons and cell bodies of neurons. Fedr Proc. 39:2783-89.

Hogan D, Berman NEJ (1991). Calbindin-D is transiently expressed in pyramidal cells of neonatal kittens in an area dependent pattern. Soc Neurosci Abstr. 17:367.

HoIm IE, Geneser FA, Zimmer J, Baimbridge KG (1990). Immunocytochemical demonstration of the calcium-binding protein calbindin-D 28k and parvalbumin in the subiculum, hippocampus and dentate area of the domestic pig. Prog Brain Res. 83:85-97. Holzwarth JA, Gibbons SJ, Brorson JR, Philipson SH, Miller RJ (1994). Glutamate receptor agonists stimulate diverse calcium responses in different types of cultured rat cortical glial cells. J Neurosci. 14: 1879- 91.

Honore T, Davies SN, Drejer J, Fletcher EJ, Jacobsen P, Lodge D, Nielsen F (1988). Quinoxalinedione: potent non-NMDA glutamate . Science. 241 :701-703. Huettner JE, Baughman RW (1986). Primary cultures of identified neurons from the visual cortex of postnatal rats. J Neurosci. 6:3044-60. lacopino AM, Christakos S (1990a). Corticosterone regulates calbindin-D28k mRNA and protein levels in rat hippocampus. J Biol Chem. 265:10177- 80. 249 lacopino AM, Christakos S (1990b). Specific reduction of calcium binding protein (28-kilodatlon calbindin-D) gene expression in aging and neurodegenerative diseases. Proc Nati Acad Sci USA. 87:4078-82. lchimiya Y, Emson PC, Mountjoy CO. Lawson DEM, Heizman CW (1988). Loss of calbindin-28k immunoreactive neurons from the cortex in Alzheimer type dementia. Brain Res. 475:156-59

1mb M, Ozawa S, Tsuzuki K (1990). Permeation of calcium through excitatory amino acid receptor channels in cultured rat hippocampal neurones. J Physiol. 424:151-65. lmredy JP, Yue DT (1994). Mechanism of -sensitive inactivation of L-type 2Ca channels. Neuron. 12:1301-18.2Ca James PH, Pruschy M, Vorherr T, PennistonJT, Carafoli E (1989). Primary structure of the cAMP-dependent phosphorylation site of the plasma membrane calcium pump. Biochemistry. 28:4253-58.

Jande SS, Maler L, Lawson DEM (1981). lmmunohistochemical mapping of vitamin-D dependent calcium-binding protein in brain. Nature. 294:765- 67.

Johansen FE, Jorgensen MB, Diemer NH (1986) . lschemic CA-i pyramidal cell loss is prevented by preischemic colchicine destruction of dentate gyrus granule cells. Brain Res. 377:344-47.

Johnson JW, Ascher P (1987). Glycine potentiates the NMDA response in cultured mouse brain neurons. 325:529-31. Kaku DA. Giffard RG. Choi DW (1993). Neuroprotective effects of glutamate antagonists and extracellular acidity. Science. 260:1 51 6-8.

Kemp JA, Leeson PD (1993). The glycine site of the NMDA receptor - five years on. Trends Pharmacol Sci. 14:20-25. Koh JY, Choi DW (1987). Quantitative determination of glutamate mediated cortical neuronal injury in cell culture by lactate dehydrogenase eflux assay. J Neurosci Methods. 20:83-90. Koh JY, Goldberg MP, Hartly DM, Choi DW (1990). non-NMDA receptor- mediated neurotoxicity in cortical cultures. J Neurosci. 10:693-705. Kohr G, Lambert CE, Mody 1(1991). Calbindin-D28k (CaBP) levels and calcium currents in acutely dissociated epileptic neurons. Exp Brain Res. 85:543-51. Kohr G, Mody 1(1991). Endogenous intracellular calcium buffering and the activation/inactivation of HVA calcium currents in rat dentate gyrus granule cells. J Gen Physiol. 98:941-67.

Kretsinger RH, Nockolds CE (1973). Carp muscle calcium-binding protein. II. Structure determination and general description. J Biol chem. 248:33 13-26.

Lucas

Luebke

Lowenstein

Llinas

Llinas

Llinas

Llinas

Lledo

Lindholm

Leonard

Legendre

Lederer

Lavoie

Kurth

Kudo

microdomains

glutamatergic

PM,

inner of calbindin-D28K

11:895-902. channels

DR, R,

R,

functional

R,

R, Y,

in cell currents transfection

12:174.

promote

Neurotrophin-3

Heisenberg MC,

neuromuscular

channels

Neurosci. excitable

normal

neurons. induced

death

JI,

B,

WJ,

the

the

LP,

Yarom

Sugimori

Sugimori

Sugimori

Takeda

D,

R,

Parent

somata

Newhouse

Somasundaram

Dunlap

Weis

layers

DH,

mammalian

Salpeter

perforant

Castren

Rosendund

due

Niggli

and

and

4 °Ca

Purkinje

by

Y

in

cells:

Miles

significance

Neurology.

K,

A

to

13:674-84.

JH,

of

CP,

in

(1986).

K,

M,

M,

cultured

M parkinsonian

the

of

and

E,

intracellular

(1991).

Yarazaki

synaptic Ca

mammalian

influx

the

Turner E,

(1980).

MM

JP calbindin-D28k

junction

Silver

Hiliman

mRNA

fuzzy

Hadley

induced

Choi

MF,

Tesarollo

path.

high

transmitter

Tsoulfas cell

+

central C,

retina.

(1957).

(1979).

overload.

B,

Hatam

and

Specific

hippocampal

DW

Westbrook

molecular

differentiation. space.

RB

TJ

Dopaminergic

in

Neuron.

39:217.

transmission

of

Morton

RW

DE,

Electrophysiological

K

is

by

resultant

the

(1992b).

calcium

(1993). monkeys.

(1989).

the

nervous

L,

(1990).

Arch

cerebellar

mediated

P,

The

tri-iodo-thyronine

(1990).

F,

Cherksey

Parada

Agonist-induced

rat release.

Kolbeck

blockade

P-type,

Nature.

McCabe

Brain

into

AJ,

weight

Ophthalmol.

toxic

6:627-33

hippocampus

GL

homeostasis.

system. neurons

Multiple

Relationship

neuronal

Quin2

Emson

the

Presynaptic

LF,

in (1993).

by

Sodium-calcium

Res.

Neuroreport. neurons

slices.

voltage-dependent

effect

J

B

R,

J

342:283.

the

T of

alcohols.

GH 3

calcium.

Cell

Theonen

(1992a).

Physiol

Berzaghi

(1991).

protects

the

528:48-54.

hippocampus.

PC,

calcium

by

Trends

injury

Biol.

of

properties

cell

J

expressing

Inactivation

in

58:193-201.

myopathy

low

intracellular

following

Physiol.

sodium

Mason

between

calcium

cerebellar

(Paris).

Neuron.

line

J

H

Soc

Distributi 3 n M in

122:443-50.

threshold

Up

neurons

2:601-4.

Cell

channel

Neurosci.

(1993).

cultured

alters

da

exchange

regulation

Neurosci

WT

L-glutamate

of

Biol.

305:171-95.

P,

concentration

at

focal

86:135-38.

glutamate-

Ca calbindin

of

in

9:943-54.

Neuron.

granule

calcium.

Leingartner

(1992).

calcium

the

against

types

calcium

vitro NMDA

+

cortical

82:811-19.

stimulation

and

15:351-55.

channels

in

Abstr.

of

Purkinje

control

in

cells

cell

on

Stable

J

A,

250 the 251 MacDermott AB, Mayer ML, Westbrook GL, Smith SJ, Barker JL (1986). NMDA-receptor activation increases cytoplasmic calcium concentration in cultures spinal cord neurons. Nature. 321:519-22. Madison D, Fox AP, Tsien RW (1987). Adenosine reduces an inactivating component of calcium current in hippocampal CA3 neurons. Biophys J. 51 :30a.

Manzoni 0, Prezeau L, Rassendren FA, Sladeczek F, Curry K, Bockaert J (1992). Both enantiomers of 1-aminocyclopentyl-1,3-dicarboxylate are full agonists of metabotropic glutamate receptors coupled to phospholipase C. Molec Pharmacol. 42:322-27.

Manzoni OJ, Poulat F, Do E, Sahuquet A, Sassetti I, Bockaert J, Sladeczek FA (1991). Pharmacological characterization of the quisqualate receptor coupled to phospholipaseC (Qp) in striatal neurons. Eur J Pharmacol. 207:231-41.

Mattson MP, Cheng B, Smith-Swintosky VL (1993a). Mechanisms of neurotrophic factor protection against calcium- and free-radical-mediated excitotoxic injury: implications for treating neurodegenerative disorders. Exp Neurol. 124:89-95.

Mattson MP, Keshave NK, Wang H, Cheng B, Michaelis EK (1993b). Basic FGF regulates the expression of a functional 71 kDa NMDA receptor protein that mediates calcium influx and neurotoxicity in hippocampal neurons. J Neurosci. 13:4575-88.

Mattson MP, Murrain M, Guthrie PB, Kater SB (1989). Basic fibroblast growth factor and glutamate: opposing roles in the generation and degeneration of hippocampal neuroarchitecture. J Neurosci. 9:3728-40.

Mattson PM, Rychlik B, Chu C, Christakos S (1991). Evidence for calcium- reducing and excito-protective roles for the calcium-binding protein calbindin-D28k in cultured hippocampal neurons. Neuron. 6:41-51.

Mayer E, Dunnett SB, Pelliterri R, Fawcett JW (1993). Basic fibroblast growth factor promotes the survival of embryonic ventral mesencephalic dopaminergic neurons--I. Effects in vitro. Neuroscience. 56:379-88.

Mayer ML, Westbrook GL (1987). Permeation and block of N-methyl-D- receptor channels by divalent cations in mouse cultured central neurons. J Physiol. 394:501-27.

McBurney RN, Neering IR (1987). Neuronal calcium homeostasis. Trends Neurosci. 10:164-69.

McCormack JG, Denton RM (1990). The regulation of mitochondrial function in mammalian cells by 2Ca ions. Trans Biochem Soc. 16:523-27. McDonald JW, Silverstein FS, Johnston MV (1987). MK-801 protects the neonatal brain from hypoxic-ischemic damage. Eur J Pharmacol. 140:359-61. 252

McGraw CE, Somlyo AU, Blaustein MP (1980). Localization of calcium in presynaptic nerve terminals. J Cell Biol. 85:228-41. Meidrum BS, Garthwaite J (1991). Excitatory amino acid neurotoxicity and neurodegeneration disease. Trends Pharmacol Sci. 12:54-62. Michaels RL, Rothman SM (1990). Glutamate neurotoxicity in vitro: antagonist pharmacology and intracellular calcium concentrations. J Neurosci. 10:283-92. Miettinen R, Freund TF (1992). Convergence and segregation of septal and median raphe inputs onto subsets of hippocampal inhibitory interneurons. Brain Res. 594:263-72.

Miller J, Baimbridge KG (1983). Biochemical and immuno-histochemical correlates of kindling-induced epilepsy: role of calcium binding protein. Brain Res. 278:322-26. Miller RJ (1987). Multiple calcium channels and neuronal function. Science. 235:46-52.

Mills LR (1993). Neuron-specific and state-specific differences in calcium regulation: their role in the development of neuronal architecture. Ann NY Acad Sci. 279:312-24

Mintz IM, Adams ME, Bean BP (1992). P-type calcium channels in rat central and peripheral neurons. Neuron. 9:85-95.

Mintz IM, Venema VJ, Swiderek KM, Lee TD, Bean BP, Adams ME (1992). P type calcium channels blocked by the spider toxin omega-Aga-IVA. Nature. 355:827-29.

Mitchell P (1966). Chemiosmotic coupling in oxidative and photosynthetic phosphorylation. Glynn Research. Bodmin.

Mitchell P. Moyle J (1967). Respiration driven proton translocation in rat live mitochondria. Biochem J. 105:1147-62.

Mitchell P. Moyle J (1969). Estimation of membrane potential and pH difference across the cristae membrane of rat live mitochondria. Eur J Biochem. 7:471-84.

Mockel V, Fisher G (1994). Vulnerability to excitotoxic stimuli of cultured rat hippocampal neurons containing the calcium-binding proteins calretinin and calbindin D28k. Brain Res. 648:109-20. Monaghan DT, Bridges RJ, Cotman CW (1989). The excitatory amino acid receptors: their classes, pharmacology and distinct properties in the function of the central nervous system. Annu Rev Pharmacol. 29:365- 402.

Moncrief ND, Goodman M, Kretsinger RH (1990). Evolution of EF-hand calcium modulated proteins. I. Relationships based on amino acid sequences. J Mol Evol. 30:622-62 253

Morrissey RL, Wasserman RH (1971). Calcium absorption and calcium binding protein in chick on different calcium and phosphate intakes. Am J Physiol. 220:1509-15.

Morrissey RL, Zolock DT, Bikle DD, Empson RN Jr, Bucci TJ (1978). Intestinal response to la, 25-dihydroxycholecaciferol. I. RNA polymerase, alkaline phosphatase, calcium and phosphorous uptake in vitro, and in vivo calcium transport and accumulation. Biochim Biophys Acta. 538:23-33. Murphy SN, Miller RJ (1989). Two distinct quisqualate receptors regulate 2+Ca homeostasis in hippocampal neurons in vitro. Molec Pharmacol 35:671-80.

Nedergaard M (1991). Energy depletion kills neurons independently of cytosolic calcium. J Cereb Blood Flow Metab. 11(suppl 2):S241.

Niesen C, Charlton MP, Carlen PL (1991). Postsynaptic and presynaptic effects of the calcium chelator BAPTA on synaptic transmission in rat hippocampal dentate granule neurons. Brain Res. 555:319-25.

Nishimura M (1986). Factors influencing the increase in spontaneous neurotransmitter release by hypoxia at the mouse neuromuscular junction. J Physiol. 372:303-14. Nistri A, Cherubini E (1990). Inactivation of a slow 2Ca current in CAl neurones of the adult rat hippocampal slice. Neurosci Lett. 111:102-8.

Nowycky MC, Pinter M (1993). Time courses of calcium and calcium-bound buffers following calcium influx in a model cell. Biophys J. 64:77-91.

Olanow CW (1993). A radical hypothesis of neurodegeneration. Trends Neurosci. 16:439-44.

Olney JW (1969). Brain lesion, obesity and other disturbances in mice treated with monosodium glutamate. Science. 164:719-21.

Olney JW (1978). Neurotoxicity of excitatory amino acids. In: Kainic acid as a tool in neurobiology (McGreer EG, Olney JW, McGreer PJ, Eds.). Raven Press, pp 97-171. Olney JW, Ho OL, Rhee V (1971). Cytotoxic effects of acidic and sulfur- containing amino acids on the infant mouse central nervous system. Exp Brain Res. 14:61-76.

Olney JW, Ikonomidou C, Mosinger JL, Frierdich G (1989). MK-801 prevents hypobaric-ischemic neuronal degeneration in infant rat brain. J Neurosci. 9:1701-4.

Olney JW, Rhee V, Ho OL (1974). Kainic acid: a powerful neurotoxic analogue of glutamate. Brain Res. 77:507-12.

Olney JW, Sharpe LG (1969). Brain lesion in an infant Rhesus monkey treated with monosodium glutamate. Science. 166:386-88. 254

Olson JE, Fleischhacker D, Murray WB, Holtzman D (1990). Control of astrocyte volume by intracellular and extracellular Ca GLIA. 3:405- 12. .2 Onodera H, Sato G, Kogure K (1986). Lesions to Schaffer collaterals prevents ischemic death of CAl pyramidal cells. Neurosci Lett. 68:169-74. Palade P (1987). Drug induced 2Ca release from isolated sarcoplasmic reticulum. J Biol Chem. 263:4142-48.

Parmentier M (1989). Structure of the human cDNA and genes coding for calbindin D28k and calretinin. Adv Exp Med Biol. 269:27-34.

Patrick J, Sequela P, Vernino 5, Amador M, Luetje C, Dani JA (1993). Functional diversity of neuronal nicotinic acetylcholine receptors. Prog Brain Res. 98:113-20. Penniston JT (1982). Plasma membrane 2Ca pumping ATPase. Ann NY Acad Sci. 402:296-303.

Petito CK, Feldmann E, Pulsinelli WA, Plum F (1987). Delayed hippocampal damage in humans following cardio-respiratory arrest. Neurology. 37:1281-86.

Preston SF, Berlin RD (1992). An intracellular calcium store regulates protein synthesis in HeLa cells, but it is not the hormone-sensitive store. Cell Calcium. 13:303-12.

Rami A, Brehier A, Thomasset M, Rabie A (1987). The comparative immunocytochemical distribution of 28 kDa cholecalcin (CaBP) in the hippocampus of rat, guinea pig and hedgehog. Brain Res. 422:149-53

Raspe E, Ramboer I, Galand N, Boeynaems JM (1989). Enhanced release of prostacyclin from quin 2-loaded endothelial cells. Eur J Pharmacol. 163:345-51.

Ray J, Peterson DA, Schinstine M, Gage FH (1993). Proliferation, differentiation, and long-term culture of primary hippocampal neurons. Proc Nati Acad Sd USA. 90:3602-6. Richardson A, Taylor CW (1993). Effects of 2Ca chelators on purified inositol 1,4,5-trisphosphate (lnsP3) receptors and lnsP3-stimulated Ca + mobilization. J Biol Chem. 5:11528-33.

Roberts WM (1994). Localization of calcium signal by a mobile calcium buffer in frog saccular hair cell. J Neurosci. 14:3246-62.

Robitaille R, Charlton MP (1992). Presynaptic calcium signals and transmitter release are modulated by calcium-activated potassium channels. J Neurosci. 12:297-305.

Roe MW, Lemasters JJ, Herman B (1990). Assessment of fura-2 for measurement of cytosolic free calcium. Cell Calcium. 11:63-73. 255

Roth J, Baetens D, Norman AW, Garcia-Segura LM (1981). Specific neurons in chick central nervous system stain with an antibody against chick intestinal vitamin-D dependent calcium-binding protein . Brain Res. 222:452-57. Rothman SM (1983). Synaptic activity mediates death of hypoxic neurons. Science. 220:536-37.

Rothman SM (1985). The neurotoxicity of excitatory amino acids is produced by passive chloride influx. J Neurosci. 5:1483-89.

Sala F, Hernandez-Cruz A (1990). Calcium diffusion modeling in a spherical neuron: Relevance of buffering properties. Biophys J. 57:313-24. Satoh T, Ross CA, Villa A, Supattaphone S. Pozzan T, Snyder SH, Meldolesi J (1990). The inositol 1,4,5-triphosphate receptor in cerebellar Purkinje cells: Quantitative immuno-gold labeling reveals concentration in an endoplasmic reticulum sub-compartment. J Cell Biol. 111:61 5-24.

Schanne FAX, Kane AB, Young EE, Farber JL (1979). Calcium dependence of toxic cell death: a final common pathway. Science. 206:700-702. Scharfman HE, Schwartzkrion PA (1989). Protection of dentate hilar cells from prolonged stimulation by intracellular calcium chelation. Science. 246:257-60.

Scheardown MJ, Nielsen ED, Hansen AJ, Jacobsen P. Honore T (1990). 2,3- dihydroxy-6-nitro-7-sulfamoyl-benzo(F)quinoxaline: a neuroprotectant for cerebral ischemia. Science. 247:571-74.

Schneggenburger R, Zhou Z, Konnerth A, Neher E (1993). Fractional contribution of calcium to the cation current through glutamate receptor channels. Neuron. 11:133-43.

Schousboe A (1981). Transport and metabolism of glutamate and GABA in neurons and glial cells. Int Rev Neurobiol. 22:1-45.

Shmigol A, Kirischuk S. Kostyik P. Verkhratsky A (1994). Different properties of caffeine-sensitive Ca stores in peripheral and central mammalian neurons. Pflugers Arch. 426:174-76.

Simon RP, Swan JH, Griffith T, Meidrum BS (1984). Blockage of N-methyl-D aspartate may protect against ischemic damage in the brain. Science. 226:850-52. Sloviter RS (1989). Calcium-binding protein (Calbindin-D28k) and parvalbumin immunocytochemistry: Localization in the rat hippocampus with specific reference to the selective vulnerability of hippocampal neurons to seizure activity. J Comp Neurol. 280:183-96.

Sloviter RS, Nilaver G (1987). Immunocytochemical localization of GABA-, chelecystokinin-, vasoactive intestinal peptide-, and somatostatin-like immunoreactivity in the area dentata and hippocampus of the rat. J Comp Neurol. 256:42-60. 256

Smaliwood JI, Gugi M, Rasmussen H (1988). Regulation of erythrocyte calcium pump activity by protein kinase C. J Biol Chem. 263:2195-202.

Soic M, Dow KE, Riopelle RJ (1993). Rat hippocampal neurons in dissociated culture. In: Tissue culture in neurobiology course handbook (Richardson A, Fedoroff S. Eds.). Network of Centers of Excellence on Neuroregeneration and functional recovery (Canada), pp 11-19. Sommer B, Seeburg PH (1992). Glutamate receptor channels: novel properties and new clones. Trends Pharmacol Sci. 13:291-96.

Spencer R, Charman M, Wilson PW, Lawson DEM (1978). The relationship between vitamin D-stimulated calcium transport and intestinal calcium- binding protein in the chicken. Biochem J. 170:93-101.

Story MD, Stephens LC, Tomasovic SP, Meyn RE (1992). A role for calcium in regulating apoptosis in rat thymocytes irradiated in vitro. Int J Rad Biol. 61:243-51.

Sugiyama H, Ito I, Hirono C (1987). A new type of glutamate receptor linked to inositol phospholipid metabolism. Nature. 325:531-33.

Taglialatela M, Canzoniero LM, Cragoe EJ, Di Renzo G, Annunziato L (1990). Na/Ca exchange activity in central nerve ending Il. Relationship 2between pharmacological blockage by amiloride analogues and dopamine release from tubero-infundibular hypothalamic neurons. Molec Pharmacol. 38:393-400.

Takagi T, Nojiri M, Konishi K, Maruyama K, Nonomura Y (1986). Amino acid sequence of vitamin D-dependent calcium-binding protein from bovine brain. FEBS Lett. 201:41-45.

Takahashi K, Tateishi N, Kaneda M, Akaiki N (1989). Comparison of low- threshold 21Ca currents in the hippocampal CAl neurons among the newborn adult and aged rats. Neurosci Lett. 103:29-33.

Takahashi T, Momlyama A (1993). Different types of calcium channels mediate central synaptic transmission. Nature. 366:156-58.

Tang CM, Presser F, Morad M (1988). Amiloride selectively blocks the low threshold (T) calcium channel. Science. 240:213-15.

Tecoma ES, Monyer H, Goldberg MP, Choi DW (1989). Traumatic neuronal injury in vitro is attenuated by NMDA antagonists. Neuron. 2:1541-45. Thayer SA, Miller RJ (1990). Regulation of the intracellular free calcium concentration in single rat dorsal root ganglion neurones in vitro. J Physiol. 425:85-115. Thayer SA, Perny TM, Miller RJ (1988). Regulation of 2Ca homeostasis in sensory neurons by bradykinin. J Neurosci. 8:4089-97.

Tojyo Y, Matsumoto Y (1990). Inhibitory effects of loading with the calcium chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N’,N’-tetraacetic acid 257

(BAPTA) on amylase release and cellular ATP level in rat parotid cells. Biochemical Pharmacol. 39:1 775-79.

Tsien RW, Lipscombe D, Madison DV, Bley KR, Fox AP (1988). Multiple types of neuronat calcium channels and their selective modulation. Trends Neurosci. 11:431-37.

Tsien RW, Tsien RY (1990). Calcium channel Stores and Oscillations. Annu Rev Cell Biol. 6:715-60.

Tsien RY (1981). A non-disruptive technique for loading calcium buffers and indicators into cells. Nature. 290:527-28.

Tsubokawa H, Oguro K, Robinson HP, Masuzawa T, Kirino T, Kawai N (1992). Abnormal 2+Ca homeostasis before cell death revealed by whole cell recording of ischemic CAl hippocampal neurons. Neuroscience. 49:807-17.

Tymianski M, Chariton MP, Carlen PL, Tator CH (1993a). Source specificity of early calcium neurotoxicity in cultures embryonic spinal neurons. J Neurosci. 13:2085-104.

Tymianski M, Wallace MC, Spigelman I, Uno M, Carlen PL, Tator CH, Chariton MP (1993b). Cell-permeant 2Ca chelators reduce early excitotoxic and ischernic neuronal injury in vitro and in vivo. Neuron. 11:221-35.

Ueda N, Shah SV (1992). Role of intracellular calcium in hydrogen peroxide- induced renal tubular cell injury. Am J Physiol. 263:F214-21.

Uneyama H, Munakata M, Akaike N (1993). Caffeine response in pyramidal neurons freshly dissociated from rat hippocampus. Brain Res. 604:24- 31.

Van der Zee J, Mason RP, Eling TE (1989). The oxidation of the calcium probe quin2 and its analogs by prostaglandin H synthase. Arch Biochem Biophys. 15:64-71.

Varghese 5, Lee S, Huang YC, Christakos S (1988). Analysis of rat vitamin D dependent calbindin-D28k gene expression. J Biol Chem. 263:9776-84 Verma A, Hirsch DJ, Hanley MR, Thastrup 0, Christenesen SB, Snyder SH (1990). lnositol triphosphase and thapsigragin discriminate eñdoplasmic reticulum calcium stores in the rat brain. Biochem Biophys Res Commun. 172:811-16.

Wasserman RH, Fullmer CS (1982). Vitamin D-induced calcium binding proteins. In: Calcium and cell function, Vol 2. (Cheung WY, Ed.). pp 175-2 16.

Wasserman RH, Taylor, AM (1966). Vitamin D3-induced calcium-binding protein in chick intestinal mucosa. Science. 152:791-93. Wieloch T (1985). Hypoglycemia-induced neuronal damage prevented by an N methyl-D-aspartate antagonist. Science. 230:681-83. 258

Wieloch T, Gustaffsson I, Westberg E (1988). MK-801 does not protect against brain damage in a rat model of cerebral ischemia. Neurochem mt. 12:24. Williams RJP (1986). The physics and chemistry of calcium-binding proteins. In: Calcium and the cell (Ciba Foundation Symposium 122). Wiley, Chichester, pp 145-161,

Williams S. Johnston D (1989). Long-term potentiation of hippocampal mossy fiber synapses is blocked by postsynaptic injection of calcium chelators. Neuron. 3:583-88.

Wilson PW, Hardin M, Lawson DEM (1985). Putative amino acid sequence of chick calcium binding protein deduced from complementary DNA sequence. Nuci Acids Res. 13:8867-81.

Wong LA, Gallagher JP (1989). A direct nicotinic receptor mediated inhibition recorded intracellularly in vitro. Nature. 341:439-42

Wroblewski F, LaDue JS (1955). Lactic dehydrogenase activity in blood. Proc Soc Exp Biol Med. 90:210-13. Wroblewski JT, Danysz W (1989). Modulation of glutamate receptors: molecular mechanisms and functional applications. Annu Rev Pharmacol. 29:441-74.

Xie X, Berger TW, Barrionuevo G (1992). Isolated NMDA receptor-mediated synaptic responses express both LTP and LTD. J Neurophysiol. 67:1009-13.

Yamada T, McGeer PL, Baimbridge KG, McGeer EG (1990). Relative sparing in Parkinson disease of substantia nigra dopamine neurons containing calbindin-D28k.s Brain Res. 526:303-7. Zhang tJF, Randall AD, Ellinor PT, Home WA, Sather WA, Tanabe T, Schwarz TL, Tsien RW (1993). Distinctive pharmacology and kinetics of cloned neuronal 2+Ca channels and their possible counterparts in mammalian CNS neurons. Neuropharmacology. 32:1075-88.

Zhou Z, Neher E (1993). Mobile and immobile calcium buffers in bovine adrenal chromaffin cells. J Physiol. 469:245-73.