BCL-xL Regulates Synaptic Plasticity

Embed Size (px)

Citation preview

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    1/15208

    Elizabeth Jonas

    Yale University School of Medicine, Section of Endocrinology, Department of Internal Medicine,

    333 Cedar Street, PO Box 208020, New Haven, CT 06520-8020

    Mitochondria are the predominant organelle within many presynaptic terminals. During times of high synapticactivity, they affect intracellular calcium homeostasis and provide the energy needed for synaptic vesiclerecycling and for the continued operation of membrane ion pumps. Recent discoveries have altered our ideas

    about the role of mitochondria in the synapse. Mitochondrial localization, morphology, and docking at synaptic sitesmay indeed alter the kinetics of transmitter release and calcium homeostasis in the presynaptic terminal. In addition,the mitochondrial ion channel BCL-xL, known as a protector against programmed cell death, regulates mitochondrialmembrane conductance and bioenergetics in the synapse and can thereby alter synaptic transmitter release and therecycling of pools of synaptic vesicles. BCL-xL, therefore, not only affects the life and death of the cell soma, but its

    actions in the synapse may underlie the regulation of basic synaptic processes that subtend learning, memory andsynaptic development.

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    2/15 2August 2006

    Volume 6, Issue 4

    Introduction

    Transmission of signals through the nervous system requires cell-

    to-cell communication via neuronal synapses. The basic features

    of chemical synaptic transmission include close apposition of two

    nerve cells and release of a chemical neurotransmitter by one cell

    into the synaptic cleft between two neurons (1). After release, neu-

    rotransmitter influences responses of the second neuron via recep-

    tors on the postsynaptic cell (2). Neurotransmitter is packaged into

    small vesicles within the presynaptic terminal, and collections of

    vesicles wait for a calcium signal produced by calcium entry into

    the presynaptic cell during action potential firing (3). Elevation of

    intracellular calcium during synaptic activity enhances the probabil-

    ity of vesicle fusion. Many of the features of synaptic transmission

    can be enhanced over the short and long term (4). These include

    changes in presynaptic calcium levels, changes in vesicle numbersand probability of release, and alterations in postsynaptic receptor

    numbers and function. Such changes lead to short- and long-term

    modifications in synaptic strength and account in part for plasticity

    of synaptic activity. Many of these phenomena require energy, and,

    therefore, may be regulated by mitochondria as will be described in

    this review. Mitochondria also buffer and re-release calcium inside

    the synapse, altering the time course and amplitude of the change

    in calcium concentration during vesicle fusion and recycling (5).

    Unexpectedly, the BCL-2 family proteins that are known to regulate

    apoptosis through their actions at mitochondrial membranes have

    been newly identified as regulators of synaptic activity. Thus, the

    actions of BCL-xLa BCL-2 family memberat mitochondria posi-

    tion it to influence learning, memory, and alterations in behavior.

    Mitochondria Regulate SynapticTransmission

    Mitochondria are known to be important for synaptic transmission

    and are the predominant organelle within presynaptic terminals that

    release neurotransmitter at high rates (6).Mitochondria provide ener-gy in the form of ATP and buffer calcium at these active synapses.

    Some synaptic mitochondria may buffer calcium even at the expense

    of ATP production. Indeed, different types of neuronal synapses con-

    tain different numbers of mitochondria with slightly different proper-

    ties, depending on whether the main function of the mitochondriais to provide energy or buffer calcium. At some synapses, oxidative

    metabolism by mitochondria is crucial to successful neurotransmis-

    sion, which can be altered considerablyfor example, by the rapid

    onset of synaptic fatigueif mitochondrial function is eliminated (7).

    Moreover, mitochondrial bioenergetics are altered acutely in synapses

    that have undergone conditioning, providing for enhanced oxidative

    competence (7). Therefore, an interaction may exist between neuro-

    nal plasticity and mitochondrial plasticity(8). In this review, we will

    focus on the role of mitochondria in synaptic transmission and syn-

    aptic plasticity and consider possible ways in which the mitochondri-

    al protein BCL-xL brings about changes in mitochondrial properties

    that may influence these important synaptic events.

    Mitochondria Alter Calcium Homeostasis

    During Synaptic Events

    Synaptic transmission depends on mitochondria not only for energy

    production but also for maintaining calcium homeostasis within the

    presynaptic terminal (915). During synaptic events, calcium influx

    through voltage-gated channels and the release of calcium from

    intracellular stores produce elevations of cytosolic calcium that are

    BCL-xL at the Synapse

    Ca2+

    Postsynapticpotentials

    Stimulus

    tetanus postetanicpotentiation

    residualcalcium

    no potentiation

    Control No mitochondrialCa2+ uptake

    A B

    C Posttetanic potentiation

    Ca2+

    2. After thetetanus,calcium hasaccumulatedinside themitochondrialmatrix.

    1. Repeatedactionpotentials(tetanus)

    3. Upon repeated stimulation,the extra calcium provided bymitochondria is available forvesicle fusion.

    Figure 1. Synaptic potentiation requires mitochondria.A. A diagram of

    changes in postsynaptic potentials and presynaptic calcium levels during

    and after tetanic stimulation to the presynaptic cell. B. In the absence of

    mitochondria, the prolonged tail of residual calcium observed after the teta-

    nus is not apparent and posttetanic potentiation is inhibited. C. Re-release of

    calcium from mitochondria inside the presynaptic terminal controls short-term

    synaptic plasticity.

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    3/15210

    crucial for synaptic vesicle fusion (16). In the crayfish neuromuscular

    junction, fast synaptic transmission is dependent on elevated calcium

    levels inside the presynaptic terminal (10). During a high frequency

    train of stimuli (tetanus), the amplitude of the response of the post-

    synaptic cell to neurotransmitter release gradually increases, and even

    after the tetanus has ceased, the ability of the synapse to release neu-

    rotransmitter is increased for up to several minutes (Figure 1A).

    At the very least, although there may be other factors, the abili-

    ty of the synapse to increase the amount of neurotransmitter released

    is based on: 1) the ability of the pool of releasable neurotransmitter-

    containing vesicles to change size, 2) a change in the probability of

    individual vesicle fusion, or 3) a change the amount of calcium avail-

    able for release per vesicle (15). Different synapses may have different

    degrees of potentiation or depression of release of neurotransmitter,

    both during and after the tetanus, depending on their particular

    attributes. It has been argued that synapses with a high probability ofinitial release will depress subsequent release, because they deplete

    their vesicle pools more rapidly, whereas synapses with a low prob-

    ability of release will augment release upon increased stimulation,

    because these synapses contain abundant vesicles that, during base-

    line stimulation are released less frequently(16).

    Although the causes of a change in release probability are

    complex, both the level of cytosolic calcium and the proximity of

    sites of calcium influx into the cytosol to sites of vesicle fusion par-

    ticipate in enhancing the probability of fusion events (17, 18). The

    level of residual calcium during frequent synaptic activity can also

    play a role in recovery from vesicle depletion (19, 20). In many syn-

    apses, tetanic stimulation causes depression of synaptic responses,

    whereas, as discussed above, in the crayfish neuromuscular junc-tion, neurotransmitter release is enhanced during the tetanus, in

    part because vesicles may reaccumulate rapidly even during frequent

    events. Other synapses have different responses to tetanic stimula-

    tion. For example, the squid giant presynaptic terminal and the large

    mammalian central nervous system auditory relay synapse (the calyx

    of Held) of the medial nucleus of the trapezoid body (MNTB, Box

    1) manifest synaptic depression during repetitive stimulation (19,

    21). The depression at these synapses is most likely mediated by a

    high probability of release of vesicles from multiple sites (i.e., active

    zones) as well as by elevated calcium in the terminals. Under experi-

    mental conditions in the squid presynaptic terminal, if extracellular

    calcium concentration is decreased, then synaptic potentiation canbe elicited (21), as in the crayfish synapse.

    Mitochondria participate in shaping the time course and

    amplitude of neurotransmitter release from presynaptic nerve end-

    ings after the invasion of the endings by action potentials. In the

    example of the crayfish neuromuscular junction, eliminating the

    ability of mitochondria to sequester calcium during influx through

    voltage-gated calcium channels leads to a higher rise in intracellular

    calcium inside the presynaptic terminal during a tetanus but also to

    prevention of the normal potentiation of neurotransmitter release

    after the tetanus (Figure 1B) (10). The findings demonstrate that

    mitochondria are important for the persistent elevation in intracel-

    lular calcium (residual calcium) normally found in the presynaptic

    terminal after it has fired action potentials at a high rate. After mito-

    chondria sequester calcium, they act as a source of persistent release

    of calcium from the matrix into the cytosol (Figure 1C). In bullfrog

    sympathetic neurons, mitochondria also slow the rise in intracellular

    calcium that occurs during a depolarizing stimulus by removing cal-

    cium from the cytosol, and slowing the recovery of normal calcium

    levels after the stimulus (9). At these synapses, mitochondria act as a

    high capacity buffer of cytosolic calcium and also re-release calcium

    rapidly in response to a calcium load in the mitochondrial matrix. At

    the synapse of the MNTB, however, mitochondria play a slightly dif-

    ferent role; the effect of mitochondrial calcium sequestration here is

    to speed the recovery from synaptic depression (20).

    Mitochondrial Presence At Presynaptic Sites

    Regulates Intense Synaptic Activity

    We have so far suggested that mitochondria play an important role

    in regulating neurotransmission in several well-studied models.

    Another invertebrate model, that of the Drosophila melanogaster

    neuromuscular junction, provides an ideal system for studying

    mutations that affect mitochondria and neurotransmission. A

    genetic screening technique for mutations that affect synaptic trans-

    mission in the Drosophila visual system has led to the fascinating

    Review

    Box 1. Characteristics of the MNTB

    The medial nucleus of the trapezoid body (MNTB)

    is located in the auditory brainstem of mammals.It participates in neural pathways that compute thedirection of sounds in space by comparing the tim-ing and the intensity of signals that arrive at the twoears. To ensure the accuracy of this information,MNTB neurons, and certain other neurons in thesepathways, are capable of firing action potentialsat very high rates (600 Hz or more). Such ratesare about one order of magnitude faster than mosttypical neurons (115118). Moreover a very largepresynaptic terminal, termed the calyx of Held,envelops the soma of an MNTB neuron and pro-

    vides the very strong and secure excitatory inputto these cells. These and other features ensure thatMNTB neurons fire with very high temporal preci-sion and allow them to lock their action potentialsto rapidly changing features of sound stimuli (119,120). The high energy demands of high frequencyactivity in both the presynaptic terminals and thepostsynaptic cells are associated with mitochondrialspecializations, such as the tethering of presynapticmitochondria directly to the active zones where neu-rotransmitter is released (36).

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    4/15 2August 2006

    Volume 6, Issue 4

    finding that multiple genes for mitochondrial targeting are necessary

    for normal synaptic transmission at the neuromuscular junction

    (2224). The first mutated mitochondrial targeting protein to be

    identified in this screen was Milton (22). Milton binds to kinesin

    heavy chain, linking mitochondria to microtubules for transport

    into synaptic endings (25). Animals lacking Milton have abnormal

    on- and off-transients on electroretinograms, indicating a defect

    in synaptic transmission to second-order neurons, not in photo-

    transduction itself(22). Immunoblots and immunocytochemistry

    performed with Milton-specific antibodies demonstrated that Milton

    localizes to axonal endings and synaptic sites and is co-localized

    with mitochondria and with kinesin heavy chain. The mutant pho-

    toreceptors contain abundant somatic mitochondria but completely

    lack synaptic mitochondria. In other ways synaptic morphology is

    fairly normal. For example, neurotransmitter-containing vesicles are

    targeted normally to synapses, as evidenced by the presence of syn-aptic vesicles at active zones, but the density of synaptic vesicles is

    slightly reduced, suggesting that the lack of mitochondrial targeting

    influences the establishment or maintenance of vesicle pools in the

    presynaptic terminal.

    Two recent studies have shed further light on the role of mito-

    chondria in vesicle pool dynamics. A genetic screening of Drosophila

    yielded two other mutants for synaptic transmission, one of which

    is a mutation in the GTPase dMiro, a protein that participates in the

    anterograde transport of mitochondria to presynaptic terminals (24,

    25). As observed with the Milton mutation, the dMiro-mutated flies

    lack mitochondria in the presynaptic terminals of neuromuscular

    junctions. The flies exhibit defects in locomotion and die prema-

    turely. It is fascinating to note that in these dMiro mutants the mito-chondria line up in regular rows in the soma and cannot be escorted

    out to the neuritic processes. The result is a defect in synaptic bou-

    ton shape and size and an absence of the normal microtubule loops

    that form in mature synapses. During high frequency activity at these

    terminals, there is a slight increase in levels of intracellular calcium

    compared to controls, and a more rapid fatigue of neurotransmitter

    release. Calcium is rapidly cleared, however, from the terminals after

    stimulation has ceased, and this clearance is no different from that

    of controls. Another striking finding in these synapses is the desyn-

    chronization of neurotransmitter release, such that activity causes a

    barrage of miniature excitatory postsynaptic currents (EPSCs) after

    the stimuli have ceased. These minis are unlikely to be related tocalcium homeostasis, which appears to be normal after the stimuli,

    but may be related to inadequate or delayed functioning of vesicle

    mobilization inside the presynaptic terminal.

    Mitochondrial ATP Production Regulates Normal

    Functioning Of Synaptic Vesicle Pools

    Many studies in synaptic physiology have contributed to the idea

    that distinct pools of vesicles have different probabilities of release,

    thoroughly reviewed in Rizzoli and Betz (26). Several different

    nomenclatures have been employed to describe the pools, but one

    will be used here (26). The readily releasable pool is defined as the

    vesicles that are immediately available for release, or docked at the

    active zone. In hippocampal synapses, for example, there appear to

    be approximately 510 vesicles that are docked at each active zone,

    but a single brief stimulus (such as an action potential) may release

    only one vesicle. The recycling pool is defined as the pool of vesicles

    that continue to release and reaccumulate during moderate or

    physiological stimulation. This pool contains 520% of all vesicles,

    but these estimates vary in different synapses. The reserve pool is

    defined as those vesicles that only release upon extremely frequent

    stimulation. The reserve pool of vesicles makes up about 8090% of

    the vesicles in most terminals.

    Experiments on the temperature-sensitive Drosophila shi-

    bire mutant (27) demonstrated that the reserve pool of vesicles is

    normally mobilized only after the recycling pool is depleted. This

    mutant exhibits defective endocytosis at high temperatures, leadingto an inability of vesicles to re-accumulate after exocytosis. In con-

    ditions of mild or moderate stimulation, which would not usually

    mobilize the reserve pool in controls, the reserve pool is neverthe-

    less mobilized at the high temperatures in the mutant, suggesting

    that the reserve pool must be used under circumstances where the

    recycling pool has been depleted. The recycling pool therefore may

    contain vesicles that are privileged for release, either by their interac-

    tion with specific cytoskeletal elements, or their location, or both

    (28). Surprisingly, however, as observed in synapses where vesicles

    were fluorescently labeled and then photoconverted for electron

    microscopy, the recycling pool is not located adjacent to the active

    zone. Rather, the vesicles of the recycling pool are distributed widely

    throughout the vesicle cluster(28).ATP is required for a myriad of cellular processes, and certain

    steps in synaptic vesicle mobilization, release, and recycling, could

    be compromised by the lack of locally and rapidly generated ATP.

    Specific enzyme-dependent steps in synaptic transmission include

    refilling single vesicles with neurotransmitter(29),membrane fis-

    sion during endocytosis (30), and coated pit formation (31, 32).

    Using whole-terminal capacitance measurements of goldfish retinal

    bipolar neurons, Heidelberger showed that ATP was required for fast

    compensatory membrane retrieval after exocytosis because dialysis

    of a non-hydrolyzable form of ATP into the terminal completely and

    rapidly inhibited endocytosis (30).

    Recent evidence suggests that ATP is required for normalfunctioning of vesicle pools (23). Studies of another mutation in

    Drosophila that prevents normal synaptic transmission suggest that

    ATP is needed for mobilizing the reserve pool. In this set of experi-

    mental findings, homozygous mutations in the eye for a gene that

    encodes the dynamin GTPase family mitochondrial fission protein

    DRP1 (dynamin-related protein 1) caused abnormal synaptic trans-

    mission, as evidenced by abnormal electroretinograms. In the mutat-

    ed flies, mitochondrial movement into presynaptic sites at the photo-

    receptor synapses was absent, but presynaptic morphology appeared

    normal in other ways. Photoreceptor somata contained numerous

    mitochondria that were functional. In the neuromuscular junction,

    BCL-xL at the Synapse

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    5/15212

    however, mitochondria were conspicuously absent, and resting cal-

    cium levels were twice as high as those observed in controls.

    Synaptic transmission at the neuromuscular junction failed dur-

    ing intense stimulation, and this effect was temperature-dependent,

    suggesting that transmission is normally mediated by a metabolic

    change within the synapses. In addition, the effect on failure of syn-

    aptic transmission during intense stimulation was partially rescued

    by perfusion of ATP into the synapse. Verstrecken et al. reasoned

    that during intense stimulation, mobilization of vesicles from the

    reserve pool might require local ATP synthesis. Relying on a previ-

    ous finding that the recycling pool of vesicles refills constantly dur-

    ing stimulation, but that the reserve pool fills only after stimulation

    has ceased (27), Verstrecken and colleagues were able to use the

    styryl dye FM1-43 to differentiate between effects of the mutation

    on the two different pools (Figure 2A). FM1-43 is taken up into

    synaptic vesicles during vesicle recycling, where it fluorescently

    labels collections of vesicles. By stimulating the nerve in a way that

    produced exocytosis of vesicles from, and endocytosis of vesicles to,

    the recycling pool alone, the authors demonstrated that there were

    no differences in the properties of the recycling vesicle pool between

    the mutants and the controls (Figure 2A, B).

    Although recycling pool endocytosisexocytosis kinetics

    appeared to be normal, the endocytosisexocytosis kinetics of the

    reserve pool were not. The authors determined that the difference

    in the mutants was in the ability of the reserve pool to take up dye

    (Figure 2B). By adding the dye to the bath after strong depletion ofall pools, and letting the cells re-accumulate their vesicle pools in

    the presence of dye, they discovered that the size of the filled pool

    in controls was much larger than that of the mutants, and, when

    they unloaded only the recycling pool of vesicles with a brief stimu-

    lus, dye remained in the controls, but not in the mutant synapses,

    suggesting that the mutant synapses contained a poorly functioning

    reserve pool. The mutants could be rescued by overexpression of

    the normal DRP1 protein, or by perfusion of ATP into the synapse.

    Furthermore, the authors found that control reserve pools could be

    functionally altered by treatment of synapses with inhibitors of mito-

    chondrial function.

    Additional experiments enabled the authors to conclude that

    the defect in the drp1 mutants was in mobilization of vesicles fromthe reserve pool, not in the size of the reserve pool. They deter-

    mined that an ATP-sensitive site was an intracellular motor that

    moved vesicles from pool to pool in an energy-dependent manner.

    The ATP sensitive motor turned out to be the myosin light chain

    because: 1) inhibitors of the mysosin light chain kinase caused the

    same defect in reserve pool cycling in controls as that seen in the

    mutants, and 2) in the presence of the myosin light chain kinase

    inhibitor, the reserve pool defect could no longer be rescued by per-

    fusion of ATP into the synapse.

    It is clear that mitochondria need to be targeted to the syn-

    apse for synaptic transmission to function normally during intense

    stimulation. Many questions remain, however. For example, what isthe mechanism of mitochondrial targeting to the synapse? When a

    new synaptic connection is made, what is the role of mitochondria?

    Does mitochondrial fission help target mitochondria to new synap-

    tic sites? What is the signal that a mitochondrion is needed? How

    does the release of ATP from mitochondria increase at the time it is

    needed during intense stimulation? What are other ATP-dependent

    steps in vesicle pool management?

    Review

    PostPre

    Synaptic depletion After synaptic depletion,only the recycling and readilyreleasable pools label with dye

    PrePost

    PostPre

    Synaptic depletion After synaptic depletionall three pools label with dye

    PrePost

    PostPre

    Recycling Pool

    Reserve Pool

    Readily releasable pool

    Calcium channel

    During moderate stimulation,recycling and readily releasablepools lable with dye

    PrePost

    A Wild-type synapse

    B Drp1 mutant synapse

    Figure 2. Drosophila drp1 mutation prevents mobilization of neu-

    rotransmitter-containing vesicles from the reserve pool.A. Labeling of

    distinct pools of synaptic vesicles with FM 1-43 in the wild-type synapse is

    achieved with different stimulation paradigms. B. In the mutant synapse, lack

    of mobilization of reserve pool vesicles prevents dye uptake into the reserve

    pool during stimulation as compared to control in A. See text for details.

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    6/15 2August 2006

    Volume 6, Issue 4

    Axonal Targeting ofMitochondria and Their Docking

    The axonal transport of mitochondria may be important for target-

    ing of mitochondria to sites of presynaptic activity. Mitochondria

    appear to move along the axon via cytoskeletal motors and can

    move in both directions along the axon, as well as remain stationary

    for prolonged periods of time when they are presumably docked at

    a site where they are needed (33). Mitochondria in cortical neurons

    in culture respond to application of the neurotransmitter glutamate

    by ceasing all movement and changing morphology, suggesting that

    neuronal activity and elevation of cytosolic calcium concentrations

    may play a role in mitochondrial docking, as well as in cessation

    of movement during excitotoxicity(34). Docking can also occur in

    response to changes that produce axonal growth or in response to

    intracellular signaling pathways stimulated by the binding of growthfactors extracellularly(35). The anterograde movement of mito-

    chondria employs microtubules and kinesin motors, and it appears

    that different organelles may utilize different adapter proteins to

    link them to microtubules (35). As stated above, Milton and dMiro

    are proteins that bind mitochondria to what is likely to be a large

    microtubule-based complex of proteinsalso including the protein

    syntabulininvolved in movement (25). After traveling along the

    microtubule, mitochondria arrive at the synapse, where they trans-

    fer from microtubules to an actin-based complex that docks the

    mitochondrion. This complex most likely includes other membrane

    anchoring proteins as well as actin, but most of those proteins have

    not yet been identified (33). As seen in electron micrographs, the

    brainstem auditory synapse of the MNTB (the calyx of Held), whichis specialized to release neurotransmitter at extremely high frequency

    and fidelity, contains a mitochondrial adherens complex. The

    complex is a collection of filaments that tethers mitochondria very

    closely to the synapse in a regulated fashion, orienting the matrix

    cristae perpendicular to the active zone (36). It is likely that the

    organization of mitochondria within this specialized synapse enables

    the mitochondria to carry out precisely timed ATP release and cal-

    cium buffering. In hippocampal neurons, which have considerably

    different synaptic organization than that observed in the calyx of

    Held, it appears that mitochondria are mostly untethered and that

    they sometimes move and sometimes remain stationary. When hip-

    pocampal neurons are stimulated by local application of growth fac-tors to points on the axon, mitochondria move preferentially to the

    stimulated site, presumably mimicking the in vivo situation where

    mitochondria might be targeted rapidly during growth or plasticity

    (37).At synaptic sites, mitochondria bind actin, under the control of

    phosphatidylinositol-3 kinase (PI3K) (35, 38).

    Function of DRP1 inSynaptic Targeting and Localization

    Fusion and fission of mitochondria are dynamic processes that occur

    within many cell types (39). Whether mitochondria exist as an inter-

    connected network or as individual, discrete organelles most likely

    depends on the requirements of the individual cell type. The equilib-

    rium between fusion of individual mitochondria and fission of mito-

    chondria into two or several individual mitochondria is a complex

    and highly regulated process involving the replication and segregation

    of mitochondrial DNA (40). The proteins that control mitochondrial

    fission in mammals include Drp1 (4143) and Fis1 (44). Proteins that

    control fusion include OPA1 (for Optic Atrophy Type 1, a dynamin-

    related GTPase) (39), and Mitofusin 1 and 2 (45, 46). During apop-

    tosis, mitochondria fragment under the control of the mitochondrial

    fission proteins (47, 48), and this fragmentation and some of the

    features of cell death can be prevented (48, 49) by overexpression of

    Drp1K38A, a dominant negative mutant of Drp1 (41).

    In neurons, a putative function of mitochondrial fission is pre-

    sumed to create more mitochondria during growth, and particularly

    to target mitochondria to nascent synapses during development ortimes of synaptic plasticity. In a study of the role of mitochondrial

    targeting and fission in the postsynaptic compartment of hippocam-

    pal neurons in culture, Li et al. (50) determined that 89% of the

    total cellular mitochondria were found within or close to dendritic

    protrusions (the site of contact with the presynaptic cell), and that

    the time of greatest co-localization of mitochondria with dendritic

    spines was during active phases of synaptic development. At these

    developmental stages, in resting cells, approximately 10% of den-

    dritic spines contained mitochondria. After repetitive depolarization

    of the neurons, however, mitochondria changed shape from elon-

    gated structures to aggregated clusters and 21% redistributed rapidly

    to dendritic spines (at three hours after stimulation), suggesting that

    acute alterations in mitochondrial morphology could play a role insynaptic plasticity. When a stimulating electrode was placed on the

    cell, mitochondria were found to be more likely to change shape the

    closer they were to the site of stimulation, and the morphological

    changes of the mitochondria were prevented by inhibition of NMDA

    receptors, suggesting that the changes in mitochondrial shape and

    location were correlated with synaptic excitation. The changes in

    mitochondrial morphology could also be brought on by overex-

    pression of Drp1 and inhibited by overexpression of Drp1K38A.

    Accordingly, the number of synapses was increased in neurons

    overexpressing Drp 1. In contrast, the number of synapses was

    decreased in controls overexpressing the dominant negative mutant

    of Drp1K38A, indicating that Drp1 was both required and limitingfor the development and plasticity of spines and synapses. Li and

    colleagues also studied the effect of activity on mitochondrial fission

    and fusion by time-lapse microscopy. They found that a decrease in

    neuronal activity in neurons treated with tetrodotoxin (TTX) (which

    prevents action potential firing) increased the rate of fusion over fis-

    sion, whereas increased activity in the setting of neuronal depolariza-

    tion caused an increase in fission over fusion, presumably to make

    new mitochondria that would be available for new or increasingly

    active synaptic sites.

    BCL-xL at the Synapse

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    7/15214

    Mitochondrial Ion Channels

    Specific targeting of mitochondria is thus required for normal

    synaptic transmission at high frequencies. The regulated targeting

    of mitochondria to sites of high energy demand suggests that the

    mechanisms of ATP production and release by mitochondria could

    very well be regulated during frequent synaptic events. Mitochondria

    are suggested to release ATP via the voltage-dependent ion channel

    (VDAC), the most ubiquitous protein in mitochondrial outer mem-

    branes. VDAC is the major pathway for the release of metabolites

    across the mitochondrial outer membrane, and its regulation is

    important for normal cell function as well as for cell death (51, 52).

    It is predicted that during synaptic events (such as synaptic plastic-

    ity), regulation of the opening of VDAC in the outer mitochondrial

    membrane could occur. Another prediction is that there is likely to

    be a second messenger that signals the opening of VDAC duringsynaptic events.

    The first evidence that mitochondrial ion channel activity could

    be regulated during synaptic events came from studies of mito-

    chondrial membrane conductance during synaptic transmission in

    an intact presynaptic terminal, that of the squid stellate ganglion.

    Through the use of a double-barreled patch pipette (53), recordings

    were made both at rest and during and after intense synaptic stimu-

    lation (11).

    In control recordings within the resting squid presynaptic termi-

    nal, the most frequent mitochondrial ion channel activity was small,

    with a conductance of less than 50 pS, but other conductances were

    occasionally seen. In contrast, during frequent electrical stimulation

    of the squid presynaptic nerve, there occurred a marked increasein activity and conductance of mitochondrial membrane patches

    within the presynaptic terminal (11). With a delay of less than one

    second after the onset of nerve stimulation, mitochondrial membrane

    conductance increased by as much as sixty-fold, a change that per-

    sisted for approximately a minute after the stimulus. The delay and

    persistence of the mitochondrial membrane activity after stimulation

    implied that the mitochondrial outer membrane channel activity was

    not simultaneous with the opening of plasma membrane channels

    and suggested that the increase depended on an intracellular second

    messenger. Such a messenger could be calcium, which remains ele-

    vated in the squid terminal for approximately one minute after stimu-

    lation, just as in the crayfish neuromuscular junction and superiorcervical ganglion (9, 10, 21). In keeping with these reports, in a calci-

    um-deficient bathing medium, there was no change in mitochondrial

    conductance in response to stimulation of the presynaptic terminal,

    demonstrating that the evoked mitochondrial membrane chan-

    nel activity was dependent on calcium influx (11). Mitochondrial

    membrane channel activity was also found to be dependent on an

    intact mitochondrial membrane potential. Uncoupling mitochondria

    with FCCP (carbonyl cyanidep-trifluoromethoxyphenylhydrazone),

    completely eliminated the increase in conductance recorded dur-

    ing and after nerve stimulation. The acute changes in mitochondrial

    membrane conductance were also correlated with synaptic plasticity,

    because FCCP application also eliminated short term potentiation of

    the synapse following nerve stimulation.

    The possible candidate channels that could be activated on

    mitochondrial membranes during high frequency activity of the

    synapse include the channels known to be most abundant in the

    outer membrane of adult mitochondria in healthy resting neurons

    such as VDAC (51). The opening of VDAC is most likely very tightly

    regulated. Kinnally and Tedeschi (54) have pointed out that there

    are several hundred VDAC channels in a patch that has a diameter

    of 0.5 m, assuming a random distribution of channels. If even one

    channel were open, the patch resistance of a resting mitochondrial

    membrane would be 1.7 giga-ohms (G) for a channel with a

    conductance of 650 pS, yet studies have demonstrated the ability

    to obtain patch resistances of up to 10 G(11, 54). Regulation of

    VDAC may influence important functions of the synapse such as

    learning and memory, because knock out mice lacking two of thethree known mammalian isoforms of VDAC display abnormalities

    consistent with the absence of long term potentiation, the elec-

    trophysiological correlate of learning found in hippocampal slice

    recordings (55). Regulation of VDAC opening influences the flux

    of ATP and other metabolites across the outer mitochondrial mem-

    brane (56) and, therefore, could be the conduit for the synchronous

    release of ATP or calcium during high frequency synaptic events.

    The opening of VDAC is also modulated by the presence of NADH

    on the outside of the outer membrane (5759), suggesting that

    the metabolic state of the neuron might determine whether VDAC

    remains closed or opens.

    In the squid presynaptic terminal, the activation of mitochon-

    drial channel activity during synaptic transmission is calcium sensi-tive. Although the recordings were most likely obtained on outer

    mitochondrial membranes, the only known calcium-sensitive (as

    opposed to calcium conducting) channel is an inner membrane

    channel that is activated by elevated calcium concentrations within

    the mitochondrial matrix. Thus, Ca2+-dependent responses during

    synaptic stimulation could represent opening of an inner membrane

    channel whose activity might be linked to the opening of VDAC

    in the outer membrane (60). A channel spanning two membranes

    could permit the efflux of calcium (as well as ATP and other ions

    and metabolites) from the matrix into the cytosol during synaptic

    potentiation (9, 10).

    BCL-xL Is Expressed inAdult Nervous System

    Another important set of proteins expressed in the mitochondrial

    outer membrane that could be regulated during synaptic events is

    that of the BCL-2 family. The properties of BCL-xL and other BCL-2

    family members position them to regulate the processes of synaptic

    transmission, synaptic plasticity, and synaptic development. BCL-xL

    is highly expressed in the mammalian nervous system both during

    development and in adults (6164), and is localized at least par-

    tially to mitochondria (65).During synaptic development, levels of

    Review

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    8/15 2August 2006

    Volume 6, Issue 4

    BCL-xL rise in the brain (66)with a similar time course to that thatgoverns the increase in size of presynaptic vesicle clusters (67, 68).

    In adult brain, only a few BCL-2 proteins continue to be expressed

    at high levels including the pro-apoptotic protein BID and the anti-

    apoptotic protein BCL-xL (66). BCL-2 family proteins both cause

    and prevent cell death, but their precise mechanism of action is still

    incompletely understood. Properties of these molecules have been

    widely studied in the hopes of increasing understanding of the com-

    plex set of cellular behaviors that occurs during cell death. Some of

    the characteristics of the molecules that have been uncovered have

    shed light on their possible function in the synapse. BCL-2 proteins

    may either protect the synapse from untimely elimination or con-

    tribute to its elimination either during development of redundant

    synapses or in pathological states such as ischemia and neurode-

    generative diseases. Several of the known properties of BCL-xL may

    contribute to mitochondrial function in the synapse.

    BCL-xL Regulates Apoptosis

    Programmed cell death (or apoptosis) plays an important role in

    the development and throughout the life of many organ systems,

    including the nervous system (69).In the nervous system, damaged

    cells or cells not destined for the adult animal are removed (70).

    Failure of the death program can lead to growth and proliferation of

    cancer cells, whereas untimely onset of cell death leads to degenera-

    tive changes such as found in Alzheimer Disease, and amyotrophic

    lateral sclerosis (71). In addition, during some pathological insults

    to the brain such as ischemia or trauma, some cells die immediately,

    whereas others meet their demise by turning on a programmeddeath pathway(72, 73).

    BCL-2 family proteins regulate the permeabilization of mito-

    chondrial membranes, release of cytochrome c, and eventual activa-

    tion of caspases, enzymes that support the breakdown of cellular

    components (7478). Although it is widely held that anti-apoptotic

    proteins protect against cell death, and pro-apoptotic molecules kill

    cells, it is now also firmly acknowledged that anti-apoptotic proteins

    such as BCL-2 and BCL-xL can be transformed into pro-apoptotic

    molecules by activation of endogenous proteases (7982). In addi-

    tion, some pro-apoptotic molecules serve important pro-survival

    functions in neurons and in the synapse (83, 84). In their classical

    role, however, anti-apoptotic molecules such as BCL-xL regulateand prevent cell death in several ways, including binding to pro-

    apoptotic molecules (85), thereby preventing the effects of the pro-

    apoptotic molecules on mitochondrial membrane permeability to

    cytochrome c and other pro-death factors (8688); increasing the

    conductance of the outer mitochondrial membrane to metabolites

    (89); and possibly by directly altering the efficiency of mitochondrial

    metabolism (90, 91).

    In the synapse, the role of both anti-and pro-apoptotic proteins

    is emerging. Evidence is accumulating that mitochondrial ion chan-

    nel activity of the BCL-2 family proteins can strengthen or eliminate

    a synapse during plasticity or degeneration without causing the death

    of the cell soma (9295). Therefore, in addition to their role in con-

    trolling cell death, BCL-2 family proteins regulate aspects of synaptic

    physiology even when cell death is not occurring (83, 93, 94).

    BCL-xL Is an Ion Channel That

    Regulates Conductance of the

    Mitochondrial Outer Membrane

    BCL-2 family proteins conduct ions when reconstituted into artificial

    lipid bilayers (86, 9698). The three-dimensional structure of BCL-

    xL consists of two central hydrophobic helices surrounded by five

    amphipathic helices (99).The structure is similar to that of pore-

    forming bacterial toxins. In lipid vesicles or planar lipid bilayers, the

    induction of ion channel activity by BCL-xL is related to its known

    ability to target to, and insert into, lipid membranes. In these arti-

    ficial membranes, the channel is cation selective at neutral pH, anddisplays multiple conductances, with a prominent conductance

    of 276 pS, and several smaller conductance levels. Some of the

    small conductance channels appear to display typical single chan-

    nel behavior, whereas the larger conductances have more complex

    behavior, indicating that multiple proteins could influence the activ-

    ity of, or constitute, the channel.

    A key feature of the ion channel activity of BCL-xL is that it can

    induce metabolite exchange across mitochondrial membranes (89,

    100). In particular, it performs this function in mitochondria from

    cells that have been exposed to apoptotic stimuli, such as growth

    factor deprivation. In this pathological setting, BCL-xL appears to

    protect cells from death by maintaining VDAC in its open configura-

    tion despite the pro-apoptotic effect of an early loss of permeabilityto metabolic substrates.

    A surprising dichotomy of the effects of anti-apoptotic molecules

    is that they enhance the release of ATP and phosphocreatine from

    mitochondria (101), but prevent the release of cytochrome c(87), and

    large fluorescent moieties from artificial lipid vesicles (86). How this

    works is not completely understood, but one possibility is that bind-

    ing of BCL-xL and BCL-2 to pro-apoptotic molecules may alter the

    death promoting functions of the pro-apoptotic molecules (85, 88,

    102). Therefore, both the channel activity of BCL-xL and its ability

    to alter the activities of pro-death molecules through protein-protein

    interactions may comprise the anti-apoptotic functions of BCL-xL.

    BCL-xL Produces Mitochondrial Ion Channel

    Activity within the Presynaptic Terminal

    Whether the ion channel activity of BCL-xL might participate in

    synaptic plasticity and development apart from its role in protection

    from cell death is now being explored. As a preview to experiments in

    mammalian neurons, we have studied the effects of BCL-xL on syn-

    aptic plasticity in the squid giant presynaptic terminal, a high fidelity,

    exctitatory, axo-axonal synapse that is critical for the animals escape

    behavior(93). Squid stellate ganglia are immunoreactive by light

    microscopy for BCL-xL in the large presynaptic terminal fingers. As is

    BCL-xL at the Synapse

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    9/15216

    typical for mitochondrial staining in large neurons and axons (7, 8),

    staining is found throughout the axoplasm, but the density of immu-

    noreactivity is greatest close to the plasma membrane, particularly

    that apposed to the postsynaptic axon. At higher power, striations

    in BCL-xL staining likely correspond to the location of the spine-like

    postsynaptic structures that represent contact points between the pre-

    synaptic terminal and the postsynaptic axon (103, 104). Observation

    at still higher power reveals a punctate cytoplasmic pattern that co-

    localizes BCL-xL with a mitochondria-specific dye.

    Further evidence for the mitochondrial localization of BCL-xL

    in squid was obtained by preparation of a purified mitochondrial

    fraction from the stellate ganglion (105). Immunoblot analysis of

    this fraction revealed the presence of squid BCL-xL that co-migrated

    with recombinant human BCL-xL. Also detected in these fractions

    was the mitochondrial outer membrane protein VDAC1.

    Full-length recombinant human BCL-xL protein produces char-acteristic channel activity with multiple conductances when applied

    by patch pipette to mitochondrial patches within the living presyn-

    aptic terminal (93). Unitary openings of the channel correspond to

    conductances between 100 pS and 760 pS, and a series of rapid

    voltage steps to successive potentials reveals current-voltage relations

    that are linear or very slightly outwardly rectifying.

    BCL-xL Enhances Synaptic Transmission

    Because BCL-xL induces mitochondrial ion channel activity and

    induces a change in mitochondrial membrane conductance within

    the squid presynaptic terminal, BCL-xL might influence the release

    of calcium or metabolites into the cytosol that could in turnregulate synaptic responses. In support of this hypothesis, injec-

    tion of recombinant BCL-xL protein into the presynaptic terminal

    enhances the rate of rise of postsynaptic responses, resulting in

    an earlier latency for evoked action potentials in the postsynaptic

    cell as compared to the latency recorded in control synapses (93).

    Interestingly, injected BCL-xL protein produced potentiation of

    synaptic transmitter release in both healthy synapses and in those in

    which transmission had run down (i.e., decreased) to the point that

    the postsynaptic potential no longer triggered postsynaptic action

    potentials. Under these conditions, injection of BCL-xL protein into

    the terminal enhanced the amplitude of the postsynaptic potential,

    restored suprathreshold responses, and effectively brought the syn-apse back to life.

    The time course of enhanced postsynaptic responses after

    injection of BCL-xL is longer than the changes produced by opening

    of mitochondrial ion channels during short-term synaptic plasticity.

    Enhancement of transmission lasts as long as forty-five minutes in

    some cells, with an average peak response of twenty minutes after

    injection, suggesting that perhaps endogenous BCL-xL participates

    in longer lasting changes in synaptic function. The initial record-

    ings of mitochondrial ion channels during synaptic transmission in

    response to tetanic stimulation (11) suggested that the activation

    of a calcium-dependent conductance of the outer mitochondrial

    membrane regulates short term synaptic changes. Although that

    conductance is clearly activated during normal physiological behav-

    iors of the synapse, its identity is not clear. Activity of BCL-xL could

    contribute to such changes in permeability of the outer membrane.

    BCL-xL Enhances Recovery

    from Synaptic Depression

    In addition to its ability to stimulate neurotransmitter release in an

    infrequently active synapse (0.03 Hz), injection into the synapse of

    recombinant BCL-xL protein also enhanced transmitter release from

    presynaptic terminals stimulated at 2 Hz, a higher frequency that

    normally produces a significant degree of synaptic depression (21).

    This finding suggested that, just as calcium buffering by mitochon-

    dria alters the recovery from depression in the MNTB (20), BCL-xL

    may counteract the effects of synaptic depression on the readilyreleasable pool (93).

    Experiments to test the role of BCL-xL in management of ves-

    icle pools demonstrated that recovery of vesicle pools after synaptic

    depression is, indeed, regulated by BCL-xL. Different stimulus para-

    digms were employed in order to study the effects of BCL-xL on the

    kinetics of different vesicle pools. In the first paradigm, stimulation

    was carried out at 2 Hz before and after the tetanus (Figure 3A). As

    reported previously(21),during this basal, high rate of stimulation,

    synaptic depression occurs as the readily releasable pool is depleted.

    After the depletion, a more reluctantly releasable set of vesicles is

    usedthat of the recycling pool. During the recovery phase follow-

    ing the administration of a tetanus given against the background of

    continuous 2 Hz stimulation, vesicles do not re-populate all pools,but re-populate only the recycling pool. The time course of the

    recovery of the recycling pool is rapid, and is not affected by previ-

    ous injection of BCL-xL.

    In the second paradigm, recovery from tetanic stimulation

    was measured during very infrequent basal stimulation (Figure

    3B). Under these conditions, full recovery of all pools occurs, as

    evidenced by the ability of the synapse to release as fully after the

    tetanus as it does during the control period at the beginning of the

    experiment. Nevertheless, the time course of recovery of synaptic

    responses following the tetanus is slower than it is at 2 Hz, sug-

    gesting that, when all the pools are re-populated, the most readily

    releasablethe first pool to be released at the onset of stimula-tionre-populates quite slowly(17, 18).The amount of recovery to

    this pool measured within thirty seconds after the end of the admin-

    istered tetanus, however, was significantly enhanced by BCL-xL

    injection when compared to recovery measured in controls. Thus,

    although synaptic depression during a tetanus is unaffected by BCL-

    xL, a slow component of the time course of recovery of the total

    vesicle pool is sensitive to the actions of BCL-xL, and the pool that

    is affected may be the most readily releasable pool. BCL-xL therefore

    appears to enhance the ability of a subset of neurotransmitter-con-

    taining vesicles to become available for release.

    Review

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    10/15 2August 2006

    Volume 6, Issue 4

    BCL-xL Effects on Synaptic Transmission

    Do Not Require Calcium Buffering

    Calcium release from mitochondria is known to participate in syn-

    aptic plasticity; specifically, re-release of calcium from mitochondria

    following the initial buffering of calcium entering the presynaptic

    terminal is responsible for the long tail of residual calcium that

    causes posttetanic potentiation at many synapses (Figure 1) (9, 10,

    11). At the squid giant synapse in physiological solutions, how-

    ever, the calcium that enters the terminal during repeated action

    potentials produces strong synaptic depression, thought to result

    from depletion of synaptic vesicles (21). Thus, it is unlikely that the

    enhancement of transmission by BCL-xL, particularly at higher stim-

    ulus frequencies (e.g. 2 Hz), results from further elevation of calcium

    levels alone in the presynaptic terminal.

    To examine the potential role of mitochondrial calcium flux in

    the enhancement of synaptic transmission during BCL-xL injection,

    neurons were treated with ruthenium red, an agent that is taken up

    by neurons and inhibits uptake of calcium into mitochondria (106,

    107). Ruthenium red blocks short term synaptic potentiation that

    is dependent upon mitochondrial calcium handling in the synapse.

    Even under these experimental conditions, however, BCL-xL poten-

    tiates transmitter release, suggesting that the actions of BCL-xL in

    squid presynaptic terminal do not require calcium uptake by mito-

    chondria, and further suggesting that BCL-xL might regulate the

    local production or release of ATP.

    BCL-xL at the Synapse

    Post

    Pre

    Recycling Pool

    Reserve Pool

    Readily releasable pool

    A

    B

    Readily releasable poolRecycling pool

    Reserve pool

    Time (min)

    PSP

    (mV/ms)

    Control

    50 Hz 50 Hz

    BCL-xL injection

    BCL-xL

    Figure 3. BCL-xL enhances recovery of vesicles to the readily releasable pool. A. During 2 Hz stimulation of the squid synapse, the readily releasable pool

    remains depleted, and vesicles recycle and re-release from the recycling pool. B. After BCL-xL protein injection into the presynaptic terminal, the postsynaptic

    potential is enhanced, but after a tetanus, there is no effect on recovery of the recycling or reserve pools. C. Between stimuli at 0.033 Hz, full recovery of all

    pools occurs. D. Injection of BCL-xL protein into the presynaptic terminal speeds recovery of the readily releasable pool of neurotransmitter. See text for details.

    Readily releasable pool

    Recycling pool

    Reserve pool

    Time (min)

    PSP

    (mV/ms)Control

    50 HzBCL-xL injection

    BCL-xL

    Post

    Pre

    Recycling Pool

    Reserve Pool

    Readily releasable pool

    C

    D

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    11/15218

    BCL-xL Controls Mitochondrial Bioenergetics

    Mitochondria require substrates such as the end products of glycoly-

    sis in order to carry out oxidation. Oxidation of substrates hyperpo-

    larizes the mitochondrial membrane potential for the purpose of ATP

    production. In growing or proliferating cells, growth factors induce

    cells to increase nutrient uptake from the environment in order to

    supply the proper amount of substrate for mitochondrial metabolism

    (91, 108). Nutrients provide energy sources and building blocks for

    cell growth (91). In the setting of growth factor withdrawal, signals

    within the cell are activated that can lead to a decrease in the abil-

    ity of cells to use glycolytic or oxidative substrates. The decline in

    substrate use eventually causes mitochondrial membrane depolariza-

    tion. The delicate balance between pro- and anti-apoptotic BCL-2

    family proteins appears necessary for the regulation of mitochondrial

    metabolism at times of deprivation and controls the onset of theeventual release of pro-apoptogenic factors such as cytochrome c into

    the cytosol (76). Although overexpression of anti-apoptotic BCL-2 pro-

    teins such as BCL-xL protects cells from death, in cells that express

    BCL-xL at normal physiological levels, growth factor withdrawal and

    metabolic decline can still cause pro-apoptotic proteins to override

    the protective effects of BCL-xL (91). Whether BCL-2 family proteins

    participate directly in changes in mitochondrial metabolism in healthy

    cells is being explored.

    Limitation of nutrient stores or oxygen causes the decline in

    ATP/ADP ratio in the cell cytoplasm. Evidence suggests that, in this

    setting, BCL-xL acts downstream of metabolic changes in the cell

    to increase the release of ATP into the cytososl (91). When cells

    deprived of growth factors were made to overexpress BCL-xL veryearly in apoptosis, their ability to condense the mitochondrial matrix

    in response to ADP could be restored, suggesting that, within twelve

    hours of growth factor deprivation in the absence of BCL-xL, the

    cause of the change in cellular metabolism is the reversible inability of

    mitochondria to translocate ADP and ATP across the outer membrane

    (101). BCL-xL can reverse the pathological situation by activating the

    opening of VDAC (89). If, despite the protective actions of BCL-xL,

    the apoptotic program progresses, the eventual release of cytochrome

    c will occur, and indeed may mark the time of irreversibility of the

    apoptotic event.

    Effects of BCL-xL on Synaptic Transmission

    Mimicked by Synaptic Perfusion of ATP

    If BCL-xL regulates the flux of metabolites across the outer mitochon-

    drial membrane (89, 100), then this property may enhance neuro-

    transmission in the physiological setting. The evidence to support this

    hypothesis came from studies of the effect of ATP injection into the

    synapse on the degree of synaptic responses (23, 93).Direct microin-

    jection of ATP into the synapse produced a similar degree and time

    course of enhancement of synaptic transmission as did the effects of

    BCL-xL injection (93), and, in fact, occluded the effects of injection of

    BCL-xL, suggesting that the two agents acted via the same mechanism.

    Pro-apoptotic Proteolytic Cleavage Fragment of

    BCL-xL Causes Synaptic Decline

    Growth factor or oxygen withdrawal causes a decline in ATP/ADP

    ratio in the cell (101). Therefore, it may follow that processes that

    use a lot of energy such as synaptic vesicle recycling and membrane

    pumps that maintain ionic homeostasis within the cell are at risk.

    Mitochondria from growth-factor deprived cells have lost their abil-

    ity to condense their matrix in response to ADP, and this sign of

    dysfunction is accompanied by a loss of ability to make ATP during

    respiration (101).

    After the changes in mitochondrial respiration occur, if nutrient

    or substrate deprivation continues, then apoptotic events at the cell

    soma may become irreversible. If this occurs in a neuronal synapse,

    that synapse could be marked for elimination. At this time, a set of

    changes occurs in the mitochondrial outer membrane that negativelyaffects synaptic function (75, 105, 109, 110). Under pro-apoptotic

    conditions, BCL-2 family proteins activate large channel activity that

    participates in the release of cytochrome c, either in the absence

    of any change to the properties of the inner membrane (112) or, as

    may occur during ischemia, accompanying induction of permeability

    transition of the inner mitochondrial membrane (75).

    In the squid synapse, the effects of hypoxia serve as a model to

    study the role of BCL-xL in neuronal injury(94, 105). The presyn-

    aptic terminal is very sensitive to hypoxia, which attenuates synaptic

    transmission over 1030 minutes (94). Patch clamp recordings of

    mitochondrial membrane channel activity during hypoxia revealed

    large conductance activity not found frequently in controls. The

    channel activity was larger than that induced by pipette-mediatedapplication of BCL-xL protein.

    Injurious stimuli such as hypoxia promote the N-terminal pro-

    teolytic cleavage of BCL-xL to form the killer protein N-BCL-xL,

    which induces cell death and cytochrome c release (79, 80, 81).The

    large conductance channel activity recorded in the outer mitochon-

    drial membranes of hypoxic synaptic terminals therefore could be

    a result of activity of proteolytically altered BCL-xL that has formed

    a new kind of channel activity in the outer mitochondrial mem-

    branes. In support of this, when recombinant N BCL-xL protein

    was added to the patch pipette during mitochondrial recordings

    within the synapse or to recordings of isolated mammalian brain

    mitochondria (111) large conductance channels were induced inmitochondrial outer membranes (105, 111). In addition, the appear-

    ance of the hypoxia-induced channel in squid could be prevented

    by pre-treatment of the synapse with zVAD-fmk, a pan-caspase/cal-

    pain inhibitor that prevents the cleavage of BCL-xL. Immunoblots

    confirmed loss of the BCL-xL protein during hypoxia, and although

    antibodies against the proteolytic cleavage fragment N BCL-xL

    did not function in squid, in mammalian brain, neurons that had

    undergone ischemic injury manifested a high level ofN BCL-xL in

    mitochondria (111). Appearance of the channel associated with N

    BCL-xL during hypoxia most likely arose from specific proteolysis of

    BCL-xL and not from general injury, because levels of VDAC were

    Review

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    12/15 2August 2006

    Volume 6, Issue 4

    preserved in both zVAD-treated and untreated hypoxic synapses,

    whereas only in the hypoxic synapses treated with zVAD were levels

    of BCL-xL preserved (94).

    N BCL-xL produces loss of membrane potential and cyto-

    chrome c release from mammalian mitochondria (80, 113). When

    N BCL-xL protein was injected into the squid presynaptic termi-

    nal, it caused a marked decrease in synaptic responses, the opposite

    effect of that observed with full-length BCL-xL, even though both

    variants of recombinant protein produce channel activity when

    added to the pipette during recordings of mitochondria inside

    the synapse. The time course of rundown of synaptic responses

    matched that of hypoxia, suggesting a correlation between the two

    types of synaptic decline (114). In addition, the data suggested that

    large mitochondrial channel activity such as that recorded in the

    setting of hypoxia or with recombinant N 76 BCL-xL could cause

    the synaptic decline, whereas the smaller conductance changesproduced by full length BCL-xL produce synaptic potentiation (84,

    114). A further understanding of how the different channel activities

    produce differential effects on mitochondrial physiology and how

    those effects, in turn, alter synaptic responses is needed.

    VDAC participates in large conductancemitochondrial membrane activity

    VDAC is a relatively non-selective channel that is believed to be the

    major conductance pathway for metabolites such as ATP, ADP, and

    creatine phosphate across the mitochondrial outer membrane (51,

    89, 100). Although BCL-xL causes channel activity in artificial lipid

    membranes, whether it does so in mitochondrial membranes, orwhether it produces all its effects though its biophysical interactions

    with VDAC is still partly in question. To address this issue more

    fully, we have taken advantage of the evidence that NADH reduces

    the conductance of VDAC in mitochondrial membranes (57, 59) but

    has no effect on the conductance ofN BCL-xL in artificial lipid

    vesicles (105). Therefore, if BCL-xL produces its effects solely by

    interacting with VDAC, we would be able to inhibit those effects by

    application of NADH to mitochondrial membranes and to the syn-

    apse itself. Indeed, the activity of recombinant N BCL-xL is attenu-

    ated by application of NADH to patches of mitochondria inside

    the synapse, and both the channel activity produced by hypoxia on

    mitochondrial membranes and the decline in synaptic responsesproduced by hypoxia were inhibited by application of NADH to the

    patches or injection of NADH into the presynaptic terminal during

    synaptic transmission (94, 111). The findings suggest that, during

    hypoxic-ischemic injury, the activity ofN BCL-xL is produced by

    its interaction with VDAC, further supporting a metabolic role for

    the channel activity in cell death in injured neurons.

    Conclusions and Future Directions

    We have painted a picture of BCL-xL as an important regulator of

    events inside the synapse. It actions position BCL-xL to play an

    important role in protecting synapses from a decline in function

    in the setting of injurious stimuli. Not only may BCL-xL serve as a

    protector, however, it can also become biochemically altered rapidly

    inside the synapse, and thereby hasten synaptic decline. The models

    advanced thus far suggest that the two opposite actions of BCL-xL

    could help balance synaptic function between under-and overactiv-

    ity, to protect against both synaptic degeneration and excitotoxic

    death. Furthermore, a protein so integrally related to mitochondrial

    metabolism inside the synapse could serve as sensor of synaptic

    activity, to provide for acute and long term changes in the metabolic

    properties of the synapse necessary for the changes in synaptic

    efficacy that underlie memory and learning. Amounts of BCL-xL

    increase during periods of synaptogenesis in mammalian brain (66),

    thus, in addition to its actions on increasing the availability of ATP

    acutely for synaptic transmission, BCL-xL may play an important

    role in axonogenesis and synaptogenesis, for example, by alteringthe local production of ATP at the synapse during the formation of

    new vesicle pools, or in targeting and docking mitochondria to syn-

    aptic sites during the process of neuronal maturation (Figure 4).

    BCL-xL is expressed in neurons that are rapidly increasing

    in size and complexity. The events that occur during neuronal

    development require not only protein synthesis, but also an ever

    increasing supply of ATP for energy dependent processes of a neu-

    BCL-xL at the Synapse

    BCL-xL

    BCL-xL

    BCL-xL

    Dendrites

    AxonStage 3 Axonal differentiation

    Stage 1 Lamellipodia form

    Stage 2 Neurons developshort processes

    Stage 4 Synaptophysinrestricted to axon

    After Stage 4 Synaptophysinstaining becomes punctate

    Neurotrophin receptor

    Neurotrophin

    Undifferentiated neuron

    X

    Figure 4. Stages of neuronal growth possibly associated with BCL-xL

    expression. In the absence of BCL-xL, some neuronal processes and syn-

    apses may fail to form or function normally.

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    13/15220

    ron of increased size and complexity (Figure 4). If, during neuronal

    development, BCL-xL increases the efficiency of production of ATP,

    then it could strongly influence the ability of a neuron to develop to

    the point of being able to perform the critical functions of rapid and

    intense release of neurotransmitter that are characteristic of synapses

    in the adult nervous system. Without crucial changes in mitochon-

    drial morphology, metabolism and targeting, synapses may not form,

    mature, or display plasticity, because of the absence of local, care-

    fully regulated availability of metabolites. doi:10.1124/mi.6.4.7

    AcknowledgmentsThe author thanks J.M. Hardwick for comments and discussion,

    L.K. Kaczmarek for comments, discussion, and assistance with fig-

    ure preparation, and J. Eisen for help with manuscript preparation.

    References1. Levitan, I.B. and Kaczmarek, L.K. The Neuron: Cell and Molecular Biology,

    (3rd Edition) Oxford University Press (2002).

    2. Malenka, R.C. and Nicoll, R.A. Long-term potentiationa decade of prog-

    ress? Science285, 18701874 (1999).

    3. Stevens, C.F. Presynaptic Function. Curr. Opin. Neurobiol.14, 341345

    (2004).

    4. Bliss, T.V.P. and Collingridge, G.L. A synaptic model of memory: Long-term

    potentiation in the hippocampus. Nature361, 3139 (1993).

    5. Jacobson, J. Duchen, M.R. Interplay between mitochondria and cellular

    calcium signalling. Mol. Cell. Biochem.256257, 209218 (2004).

    6. Blaustein, M.P., Ratzlaff, R.W., and Kendrick, N.K. The regulation of intra-

    cellular calcium in presynaptic nerve terminals. Ann. N.Y. Acad. Sci. 307,

    195212 (1978).

    7. Nguyen, P.V., Marin, L., and Atwood, H.L. Synaptic physiology and mito-

    chondrial function in crayfish tonic and phasic motor neurons. Journal ofNeurophysiology 78, 281294 (1997).

    8. Nguyen, P.V. and Atwood, H.L. Altered impulse activity modifies syn-

    aptic physiology and mitochondria in crayfish phasic motor neurons. J.

    Neurophysiol.72, 29442955 (1994).

    9. Friel, D.D. and Tsien, R.W. An FCCP-sensitive Ca2+ store in bullfrog sympa-

    thetic neurons and its participation in stimulus-evoked changes in [Ca2+]i. J.

    Neurosci.14, 40074024 (1994).

    10. Tang, Y. and Zucker, R.S. Mitochondrial involvement in post-tetanic potent ia-

    tion of synaptic transmission. Neuron18, 483491 (1997). The first study

    of the role of mitochondria in posttetanic potentiation.

    11. Jonas, E.A., Buchanan, J., and Kaczmarek, L.K. Prolonged activation of

    mitochondrial conductances during synaptic transmission. Science286,

    13471350 (1999).

    12. Nicholls, D.G. and Budd, S.L. Mitochondria and neuronal survival. Physiol.

    Rev.80, 315360 (2000).

    13. David, G. and Barrett, E.F. Stimulat ion-evoked increases in cytosolic [Ca2+]

    in mouse motor nerve terminals are limited by mitochondrial uptake and are

    temperature-dependent. J. Neurosci. 20, 72907296 (2000).

    14. Kaftan, E.J., Xu, T., Abercrombie, R.F., and Hille, B. Mitochondria shape

    hormonally induced cytoplasmic calcium oscillations and modulate exocyto-

    sis. J. Biol. Chem.275, 2546525470 (2000).

    15. Atwood, H.L. and Karunanithi, S. Diversification of synaptic strength:

    Presynaptic elements. Nat. Rev. Neurosci.3, 497516 (2002).

    16. Stevens, C.F. Neurotransmitter release at central synapses. Neuron40,

    381388 (2003).

    17. Sakaba, T. and Neher, E. Calmodulin mediates rapid recruitment of fast-

    releasing synaptic vesicles at a calyx-type synapse. Neuron32, 11191131

    (2001). This important study identified different rates of recovery to

    distinct pools of transmitter after stimulation.

    18. Sakaba, T. Stein, A., Jahn, R. and Neher, E. Distinct kinet ic changes in neu-

    rotransmitter release after SNARE protein cleavage. Science309, 491494

    (2005).

    19. Wang, L.Y. and Kaczmarek, L.K. High-frequency firing helps replenish the

    readily releasable pool of synaptic vesicles. Nature394, 384388 (1998).

    20. Billups, B. and Forsythe, I.D. Presynaptic mitochondrial calcium sequestra-

    tion influences transmission at mammalian central synapses. J. Neurosci.

    22, 58405847 (2002). Mitochondrial calcium buffering regulates recov-

    ery from synaptic depression.

    21. Swandulla, D., Hans, M., Zipser, K., and Augustine, G.J. Role of residual

    calcium in synaptic depression and posttetanic potentiation: Fast and slow

    calcium signaling in nerve terminals. Neuron7, 915926 (1991).

    22. Stowers, R.S., Megeath, L.J., Gorska-Andrzejak, J., Meinertzhagen, I.A.,

    and Schwarz, T.L. Axonal transport of mitochondria to synapses depends

    on milton, a novel Drosophila protein. Neuron36, 10631077 (2002).

    23. Verstreken, P., Ly, C.V., Venken, K.J., Koh, T.W., Zhou, Y., and Bellen, H.J.

    Synaptic mitochondria are critical for mobilization of reserve pool vesicles

    at Drosophila neuromuscular junctions. Neuron47, 365378 (2005). The

    mitochondrial fission protein DRP 1 regulates mitochondrial targeting

    to presynaptic sites. A Drosophila mutation lacking the protein dem-onstrated abnormal synaptic responses.

    24. Guo, X., Macleod, G.T., Wellington, A. et al. The GTPase dMiro is required

    for axonal transport of mitochondria to Drosophila synapses. Neuron47,

    379393 (2005).

    25. Glater, E.E., Megeath, L.J., Stowers, R.S., and Schwarz, T.L. Axonal trans-

    port of mitochondria requires Milton to recruit kinesin heavy chain and is

    light chain independent. J. Cell Biol.173, 545557 (2006).

    26. Rizzoli, S.O. and Betz, W.J. Synaptic vesicle pools. Nat. Rev. Neurosci.6,

    5769 (2005). An important review of the diversity among species of

    the size and function of different synaptic pools.

    27. Kuromi, H. and Kidokoro, Y. Selective replenishment of two vesicle pools

    depends on the source of Ca2+ at the Drosophila synapse. Neuron35,

    333343 (2002).

    28. Rizzoli, S.O. and Betz, W.J. The structural organization of the readily releas-

    able pool of synaptic vesicles. Science303, 20372039 (2004).

    29. Takamori, S., Rhee, J.S., Rosenmund, C., and Jahn, R. Identification of avesicular glutamate transporter that defines a glutamatergic phenotype in

    neurons. Nature407, 189194 (2000).

    30. Heidelberger, R. ATP is required at an early step in compensatory endocy-

    tosis in synaptic terminals. J. Neurosci.21, 64676474 (2001).

    31. Smythe, E., Pypaert, M., Lucocq, J., and Warren, G. Formation of coated

    vesicles from coated pits in broken A431 cells. J. Cell Biol.108, 843853

    (1989).

    32. Faundez, V.V. and Kelly, R.B. The AP-3 complex required for endosomal

    synaptic vesicle biogenesis is associated with a casein kinase Ialpha-like

    isoform. Mol. Biol. Cell11, 25912604 (2000).

    33. Hollenbeck, P.J. and Saxton, W.M. The axonal transport of mitochondria. J.

    Cell Sci.118, 54115419 (2005).

    34. Reynolds, I.J. and Rintoul, G.L. Mitochondrial stop and go: Signals that

    regulate organelle movement. Sciences Stke:(251):PE46 (2004).

    35. Chada, S.R. and Hollenbeck, P.J. Nerve growth factor signaling regulatesmotility and docking of axonal mitochondria. Curr. Biol.14, 12721276

    (2004).

    36. Rowland, K.C., Irby, N.K. and Spirou, G.A. Special ized synapse-associated

    structures within the calyx of Held. J. Neurosci.20, 91359144 (2000). The

    identification of a cytoskeletal complex adhering the mitochondrion to

    the presynaptic active zone.

    37. Chada, S.R. and Hollenbeck, P.J. Mitochondrial movement and positioning

    in axons: The role of growth factor signaling. J. Exp. Biol. 206, 19851992

    (2003).

    38. Malaiyandi, L.M., Honick, A.S., Rintoul, G.L., Wang, Q.J., and Reynolds, I.J.

    Zn2+ inhibits mitochondrial movement in neurons by phosphatidylinositol 3-

    kinase activation. J. Neurosci.25, 95079514 (2005). The dependence of

    mitochondrial docking on PI3 kinase.

    39. Bossy-Wetzel, E., Barsoum, M.J., Godzik, A., Schwarzenbacher, R., and

    Review

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    14/15 2August 2006

    Volume 6, Issue 4

    Lipton S.A. Mitochondrial fission in apoptosis, neurodegeneration and

    aging. Curr. Opin. Cell Biol. 15, 706716 (2003).

    40. Nunnari, J., Marshall , W.F., Straight, A., Murray, A., Sedat, J.W., and Walter,

    P. Mitochondrial transmission during mating in Saccharomyces cerevisiaeisdetermined by mitochondrial fusion and fission and the intramitochondrial

    segregation of mitochondrial DNA. Mol. Biol. Cell8, 12331242 (1997).

    41. Smirnova, E., Shurland, D.L., Ryazantsev, S.N., and van der Bliek, A.M. A

    human dynamin-related protein controls the distribution of mitochondria. J.

    Cell Biol.143, 351358 (1998). Dynamin-related GTPase regulates mito-

    chondrial fission.

    42. Otsuga, D., Keegan, B.R., Brisch, E., Thatcher, J.W., Hermann, G.J.,

    Bleazard, W., and Shaw, J.M. The dynamin-related GTPase, Dnm1p, con-

    trols mitochondrial morphology in yeast. J. Cell Biol.143, 333349 (1998).

    43. Bleazard, W., McCaffery, J.M., King, E.J., Bale, S., Mozdy, A., Tieu, Q.,

    Nunnari, J., and Shaw, J.M. The dynamin-related GTPase Dnm1 regulates

    mitochondrial fission in yeast. Nat. Cell Biol.1, 298304 (1999).

    44. Mozdy, A.D., McCaffery, J.M., and Shaw, J.M. Dnm1p GTPase-mediated

    mitochondrial fission is a multi-step process requiring the novel integral

    membrane component Fis1p. J. Cell Biol.151, 367380 (2000).

    45. Legros, F., Lombes, A., Frachon, P., and Rojo, M. Mitochondrial fusionin human cells is efficient, requires the inner membrane potential, and is

    mediated by mitofusins. Mol. Biol. Cell13, 43434354 (2002).

    46. Eura, Y., Ishihara ,N., Yokota, S., and Mihara, K. Two mitofusin proteins,

    mammalian homologues of FZO, with distinct functions are both required

    for mitochondrial fusion. J. Biochem.134, 333344 (2003).

    47. Karbowski, M. and Youle, R.J. Dynamics of mitochondrial morphology in

    healthy cells and during apoptosis. Cell Death Differ.10, 870880 (2003).

    48. Lee, Y.J., Jeong, S.Y., Karbowski, M., Smith ,C.L., and Youle, R.J. Roles of

    the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and

    Opa1 in apoptosis. Mol. Biol. Cell15, 50015011 (2004).

    49. Frank, S., Gaume, B., Bergmann-Leitner, E.S., Leitner, W.W., Robert, E.G.,

    Catez, F., Smith, C.L., and Youle, R.J. The role of dynamin-related protein

    1, a mediator of mitochondrial fission, in apoptosis. Dev. Cell1, 515525

    (2001).

    50. Li, Z., Okamoto, K., Hayashi, Y., and Sheng, M. The importance of dendritic

    mitochondria in the morphogenesis and plasticity of spines and synapses.

    Cell119, 873887 (2004). This study suggests that mitochondrial fis-

    sion and movement into postsynaptic spines is necessary for synap-

    tic plasticity.

    51. Colombini, M., Blachly-Dyson, E. and Forte, M. VDAC, a channel in the

    outer mitochondrial membrane. Ion Channels4, 169202 (1996).

    52. Colombini, M. VDAC: The channel at the interface between mitochondria

    and the cytosol. Mol. Cell. Biochem.256257, 107115 (2004).

    53. Jonas, E.A., Knox, R.J., and Kaczmarek, L.K. Giga-ohm seals on intracellu-

    lar membranes: A technique for studying intracellular ion channels in intact

    cells. Neuron19, 713 (1997).

    54. Tedeschi, H., Kinnally, K.W., and Mannella, C.A. Properties of channels in

    the mitochondrial outer membrane. J. Bioenerg. Biomembr.21, 451459

    (1989).

    55. Weeber, E.J., Levy, M., Sampson, M.J., Anflous, K., Armstrong, D.L.,

    Brown, S.E., Sweatt, J.D., and Craigen, W.J. The role of mitochondrialporins and the permeability transition pore in learning and synaptic plastic-

    ity. J. Biol. Chem. 277, 1889118897 (2002). The first study demonstrat-

    ing that mitochondrial ion channels are necessary for normal synaptic

    plasticity in the hippocampus.

    56. Rostovtseva, T. and Colombini, M. VDAC channels mediate and gate

    the flow of ATP: Implications for the regulation of mitochondrial function.

    Biophys. J.72, 19541962 (1997).

    57. Lee, A.C., Zizi, M., and Colombini, M. Beta-NADH decreases the perme-

    ability of the mitochondrial outer membrane to ADP by a factor of 6. J. Biol.

    Chem.269, 3097430980 (1994).

    58. Hodge, T. and Colombini, M. Regulation of metabolite flux through voltage-

    gating of VDAC channels. J. Membr. Biol.157, 271279 (1997).

    59. Zizi, M., Forte, M., Blachly-Dyson, E. and Colombini, M. NADH regulates

    the gating of VDAC, the mitochondrial outer membrane channel. J. Biol.

    Chem. 259, 16141616 (1994).

    60. Halestrap, A. Biochemistry: A pore way to die.Nature434, 578579 (2005).

    61. Krajewski, S., Krajewska, M., Shabaik, A., Wang, H.G., Irie, S., Fong, L.,

    and Reed, J.C. Immunohistochemical analysis of in vivo patterns of bcl-x

    expression. Cancer Res.54, 55015507 (1994).

    62. Frankowski, H., Missotten, M., Fernandez, P.A., Martinou, I., Michel, P.,

    Sadoul, R., and Martinou, J.C. Function and expression of the Bcl-x gene

    in the developing and adult nervous system. NeuroReport6, 19171921

    (1995).

    63. Gonzlez-Garcia, M., Garcia, I., Ding, L., OShea, S., Boise, L.H.,

    Thompson, C.B., and Nunez, G. Bcl-x is expressed in embryonic and post-

    natal neural tissues and functions to prevent neuronal cell death. Proc. Natl.

    Acad. Sci. U.S.A.92, 43044309 (1995).

    64. Blmer, U., Kafri, T., Randolph-Moore, L., Verma, I.M., and Gage, F.H. Bcl-

    xL protects adult septal cholinergic neurons from axotomized cell death.

    Proc. Natl. Acad. Sci. U.S.A.95, 26032608 (1998).

    65. Kaufmann, T., Schlipf, S., Sanz, J., Neubert, K., Stein, R. and Borner, C.

    Characterization of the signal that directs Bcl-x(L), but not Bcl-2, to the

    mitochondrial outer membrane. J. Cell Biol.160, 5364 (2003).

    66. Krajewska, M., Mai, J.K., Zapata, J.M., Ashwell, K.W., Schendel, S.L.,Reed, J.C., and Krajewski, S. Dynamics of expression of apoptosis-regula-

    tory proteins Bid, Bcl-2, Bcl-X, Bax and Bak during development of murine

    nervous system. Cell Death Differ. 9, 145157 (2002).

    67. Fletcher, T.L., Cameron, P., De Camilli, P., and Banker, G. The distribution

    of synapsin I and synaptophysin in hippocampal neurons developing in cul-

    ture. J. Neurosci.11, 16171626 (1991).

    68. Verderio, C., Coco, S., Pravettoni, E., Bacci, A., and Matteoli, M.

    Synaptogenesis in hippocampal cultures. Cell. Mol. Life Sci.55, 14481462

    (1999).

    69. Lindsten, T., Ross, A.J., King, A. et al. The combined functions of proapop-

    totic Bcl-2 family members bak and bax are essential for normal develop-

    ment of multiple tissues. Mol. Cell6, 13891399 (2000).

    70. Kroemer, G. and Reed, J.C. Mitochondrial control of cell death. Nat. Med. 6,

    513519 (2000).

    71. Yuan, J. and Yankner, B.A. Apoptosis in the nervous system. Nature407,

    802809 (2000).

    72. Banasiak, K.J., Xia, Y., and Haddad, G.G. Mechanisms underlying hypoxia-

    induced neuronal apoptosis. Prog. Neurobiol.62, 215249 (2000).

    73. Zukin, R.S., Jover, T., Yokota, H., Calderone, A., Simionescu, M., and Lau,

    C.-Y. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal

    Death. In: Stroke: Pathophysiology, Diagnosis, and Management(Mohr

    JP, Choi DW, Grotta JC, Weir B, Wolf PA, eds) Philadelphia: Churchill

    Livingstone, pp. 829854 (2004).

    74. Cory, S., Huang, D.C., and Adams, J.M. The Bcl-2 family: Roles in cell sur-

    vival and oncogenesis. Oncogene22, 85908607 (2003).

    75. Green, D.R. and Kroemer, G. The Pathophysiology of Mitochondrial Cell

    Death. Science305, 626629 (2004).

    76. Vander Heiden, M.G. and Thompson, C.B. Bcl-2 proteins: Regulators of

    apoptosis or of mitochondrial homeostasis? Nat. Cell Biol.1, E209E216

    (1999).

    77. Dejean, L.M., Martinez-Caballero, S., Guo, L. et al. Oligomeric Bax is acomponent of the putative cytochrome c release channel MAC, mitochon-

    drial apoptosis-induced channel. Mol. Biol. Cell16, 24242432 (2005).

    78. Polster, B.M. and Fiskum, G. Mitochondrial mechanisms of neural cell apop-

    tosis. J. Neurochem.90, 12811289 (2004).

    79. Cheng, E.H., Kirsch, D.G., Clem, R.J., Ravi, R., Kastan, M.B., Bedi, A.,

    Ueno, K., and Hardwick, J.M. Conversion of Bcl-2 to a Bax-like death effec-

    tor by caspases. Science278, 19661968 (1997).

    80. Clem, R.J., Cheng, E.H.Y., Karp, C.L. et al. Modulation of cell death by Bcl-

    xL through caspase interaction. Proc. Natl. Acad. Sci.U.S.A.95, 554559

    (1998).

    81. Fujita, N., Nagahashi, A., Nagashima, K., Rokudai, S., and Tsuruo, T.

    Acceleration of apoptotic cell death after the cleavage of Bcl-xL protein by

    caspase-3-like proteases. Oncogene17, 12951304 (1998).

    82. Nakagawa, T. and Yuan, J. Cross-talk between two cysteine protease

    BCL-xL at the Synapse

  • 8/7/2019 BCL-xL Regulates Synaptic Plasticity

    15/15

    families: Activation of caspase-12 by calpain in apoptosis. J. Cell. Biol. 150,

    887894 (2000).

    83. Fannjiang, Y., Kim, C.H., Huganir, R.L. et al. BAK alters neuronal excitability

    and can switch from anti- to pro-death function during postnatal develop-

    ment. Dev. Cell4, 575585 (2003).84. Jonas, E.A. Hardwick, J.M., and Kaczmarek, L.K. Actions of BAX on mito-

    chondrial channel activity and on synaptic transmission. Antioxid. Redox

    Signal. 7, 10921100 (2005).

    85. Oltersdorf, T., Elmore, S.W., Shoemaker, A.R. et al. An inhibitor of Bcl-2

    family proteins induces regression of solid tumours. Nature435, 677681

    (2005).

    86. Antonsson, B., Conti, F., Ciavatta, A. et al. Inhibition of Bax channel-forming

    activity by Bcl-2. Science277, 370372 (1997).

    87. Kluck, R.M., Bossy-Wetzel, E., Green, D.R., and Newmeyer, D.D. The

    release of cytochrome cfrom mitochondria: A primary site for Bcl-2 regula-

    tion of apoptosis. Science275, 11321136 (1997).

    88. Cheng, E.H., Wei, M.C., Weiler, S., Flavell, R.A., Mak, T.W., Lindsten, T.,

    and Korsmeyer, S.J. BCL-2, BCL-X(L) sequester BH3 domain-only mol-

    ecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol.

    Cell8, 705711 (2001).

    89. Vander Heiden, M.G., Li, X.X., Gottleib, E., Hill, R.B., Thompson, C.B., and

    Colombini, M. Bcl-xL promotes the open configuration of the voltage-depen-

    dent anion channel and metabolite passage through the outer mitochondrial

    membrane. J. Biol. Chem. 276, 1941419419 (2001). BCL-xL regulates

    the opening of VDAC.

    90. Plas, D.R., Talapatra, S., Edinger, A.L., Rathmell, J.C. and Thompson, C.B.

    Akt and Bcl-xL promote growth factor-independent survival through distinct

    effects on mitochondrial physiology. J. Biol. Chem.276, 1204112048

    (2001).

    91. Plas, D.R. and Thompson, C.B. Cell metabolism in the regulation of pro-

    grammed cell death. Trends Endocrinol. Metab.13, 7578 (2002).

    92. Chapman, P.F., White, G.L., Jones, M.W. et al. Impaired synaptic plastic-

    ity and learning in aged amyloid precursor protein transgenic mice. Nat.

    Neurosci.2, 271276 (1999).

    93. Jonas, E.A., Hoit, D., Hickman, J.A. et al. Modulation of synaptic trans-

    mission by the BCL-2 family protein BCL-xL. J. Neurosci.23, 84238431(2003).

    94. Jonas, E.A., Hickman, J.A., Hardwick, J.M., and Kaczmarek, L.K. Exposure

    to hypoxia rapidly induces mitochondrial channel activity within a living syn-

    apse. J. Biol. Chem. 280, 44914497 (2005).

    95. Li, J.Y., Plomann, M., and Brundin, P. Huntingtons disease: A synaptopa-

    thy? Trends Mol. Med.9, 414420 (2003).

    96. Schendel, S.L., Montal, M. and Reed, J.C. Bcl-2 family proteins as ion-

    channels. Cell Death Differ. 5, 372380 (1998).

    97. Minn, A.J., Velez, P., Schendel, S.L., Liang, H., Muchmore, S.W., Fesik,

    S.W., Fill, M., and Thompson, C.B. Bcl-xL forms an ion channel in synthetic

    lipid membranes. Nature385, 353357 (1997).

    98. Schlesinger, P.H., Gross, A., Yin, X.M., Yamamoto, K., Saito, M., Waksman,

    G. and Korsmeyer, S.J. Comparison of the ion channel characteristics of

    pro-apoptotic BAX and antiapopto