Brain ischemia and reperfusion: molecular mechanisms of neuronal injury

33
Journal of the Neurological Sciences 179 (2000) 1–33 www.elsevier.com / locate / jns Review article Brain ischemia and reperfusion: molecular mechanisms of neuronal injury a,b, a a,b a * Blaine C. White , Jonathon M. Sullivan , Donald J. DeGracia , Brian J. O’Neil , e d c a ´ Robert W. Neumar , Lawrence I. Grossman , Jose A. Rafols , Gary S. Krause a Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA b Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA c Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA d Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA e Department of Emergency Medicine, University of Pennsylvania, Philadelphia, PA, USA Received 8 May 2000; received in revised form 10 July 2000; accepted 12 July 2000 Abstract Brain ischemia and reperfusion engage multiple independently-fatal terminal pathways involving loss of membrane integrity in partitioning ions, progressive proteolysis, and inability to check these processes because of loss of general translation competence and reduced survival signal-transduction. Ischemia results in rapid loss of high-energy phosphate compounds and generalized depolarization, which induces release of glutamate and, in selectively vulnerable neurons (SVNs), opening of both voltage-dependent and glutamate- 21 regulated calcium channels. This allows a large increase in cytosolic Ca associated with activation of m-calpain, calcineurin, and phospholipases with consequent proteolysis of calpain substrates (including spectrin and eIF4G), activation of NOS and potentially of 21 Bad, and accumulation of free arachidonic acid, which can induce depletion of Ca from the ER lumen. A kinase that shuts off translation initiation by phosphorylating the a-subunit of eukaryotic initiation factor-2 (eIF2a) is activated either by adenosine degradation 21 products or depletion of ER lumenal Ca . Early during reperfusion, oxidative metabolism of arachidonate causes a burst of excess oxygen radicals, iron is released from storage proteins by superoxide-mediated reduction, and NO is generated. These events result in peroxynitrite generation, inappropriate protein nitrosylation, and lipid peroxidation, which ultrastructurally appears to principally damage the plasmalemma of SVNs. The initial recovery of ATP supports very rapid eIF2a phosphorylation that in SVNs is prolonged and associated with a major reduction in protein synthesis. High catecholamine levels induced by the ischemic episode itself and / or drug administration down-regulate insulin secretion and induce inhibition of growth-factor receptor tyrosine kinase activity, effects associated with down-regulation of survival signal-transduction through the Ras pathway. Caspase activation occurs during the early hours of reperfusion following mitochondrial release of caspase 9 and cytochrome c. The SVNs find themselves with substantial membrane damage, calpain-mediated proteolytic degradation of eIF4G and cytoskeletal proteins, altered translation initiation mechanisms that substantially reduce total protein synthesis and impose major alterations in message selection, down-regulated survival signal- transduction, and caspase activation. This picture argues powerfully that, for therapy of brain ischemia and reperfusion, the concept of single drug intervention (which has characterized the approaches of basic research, the pharmaceutical industry, and clinical trials) cannot be effective. Although rigorous study of multi-drug protocols is very demanding, effective therapy is likely to require (1) peptide growth factors for early activation of survival-signaling pathways and recovery of translation competence, (2) inhibition of lipid peroxidation, (3) inhibition of calpain, and (4) caspase inhibition. Examination of such protocols will require not only characterization of functional and histopathologic outcome, but also study of biochemical markers of the injury processes to establish the role of each drug. 2000 Elsevier Science B.V. All rights reserved. Keywords: Brain ischemia; Lipid peroxidation; Translation initiation; Apoptosis; Growth factors; Signal transduction *Corresponding author. Department of Emergency Medicine, 6G University Health Care Center, 4201 St. Antoine, Detroit, MI 48201, USA. Tel.: 11-313-577-5738; fax: 11-313-577-4131. E-mail address: [email protected] (B.C. White). 0022-510X / 00 / $ – see front matter 2000 Elsevier Science B.V. All rights reserved. PII: S0022-510X(00)00386-5

Transcript of Brain ischemia and reperfusion: molecular mechanisms of neuronal injury

Journal of the Neurological Sciences 179 (2000) 1–33www.elsevier.com/ locate / jns

Review article

Brain ischemia and reperfusion: molecular mechanisms of neuronal injury

a,b , a a,b a*Blaine C. White , Jonathon M. Sullivan , Donald J. DeGracia , Brian J. O’Neil ,e d c a´Robert W. Neumar , Lawrence I. Grossman , Jose A. Rafols , Gary S. Krause

aDepartment of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USAbDepartment of Physiology, Wayne State University School of Medicine, Detroit, MI, USA

cDepartment of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USAdCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA

eDepartment of Emergency Medicine, University of Pennsylvania, Philadelphia, PA, USA

Received 8 May 2000; received in revised form 10 July 2000; accepted 12 July 2000

Abstract

Brain ischemia and reperfusion engage multiple independently-fatal terminal pathways involving loss of membrane integrity inpartitioning ions, progressive proteolysis, and inability to check these processes because of loss of general translation competence andreduced survival signal-transduction. Ischemia results in rapid loss of high-energy phosphate compounds and generalized depolarization,which induces release of glutamate and, in selectively vulnerable neurons (SVNs), opening of both voltage-dependent and glutamate-

21regulated calcium channels. This allows a large increase in cytosolic Ca associated with activation of m-calpain, calcineurin, andphospholipases with consequent proteolysis of calpain substrates (including spectrin and eIF4G), activation of NOS and potentially of

21Bad, and accumulation of free arachidonic acid, which can induce depletion of Ca from the ER lumen. A kinase that shuts offtranslation initiation by phosphorylating the a-subunit of eukaryotic initiation factor-2 (eIF2a) is activated either by adenosine degradation

21products or depletion of ER lumenal Ca . Early during reperfusion, oxidative metabolism of arachidonate causes a burst of excessoxygen radicals, iron is released from storage proteins by superoxide-mediated reduction, and NO is generated. These events result inperoxynitrite generation, inappropriate protein nitrosylation, and lipid peroxidation, which ultrastructurally appears to principally damagethe plasmalemma of SVNs. The initial recovery of ATP supports very rapid eIF2a phosphorylation that in SVNs is prolonged andassociated with a major reduction in protein synthesis. High catecholamine levels induced by the ischemic episode itself and/or drugadministration down-regulate insulin secretion and induce inhibition of growth-factor receptor tyrosine kinase activity, effects associatedwith down-regulation of survival signal-transduction through the Ras pathway. Caspase activation occurs during the early hours ofreperfusion following mitochondrial release of caspase 9 and cytochrome c. The SVNs find themselves with substantial membranedamage, calpain-mediated proteolytic degradation of eIF4G and cytoskeletal proteins, altered translation initiation mechanisms thatsubstantially reduce total protein synthesis and impose major alterations in message selection, down-regulated survival signal-transduction, and caspase activation. This picture argues powerfully that, for therapy of brain ischemia and reperfusion, the concept ofsingle drug intervention (which has characterized the approaches of basic research, the pharmaceutical industry, and clinical trials) cannotbe effective. Although rigorous study of multi-drug protocols is very demanding, effective therapy is likely to require (1) peptide growthfactors for early activation of survival-signaling pathways and recovery of translation competence, (2) inhibition of lipid peroxidation, (3)inhibition of calpain, and (4) caspase inhibition. Examination of such protocols will require not only characterization of functional andhistopathologic outcome, but also study of biochemical markers of the injury processes to establish the role of each drug. 2000Elsevier Science B.V. All rights reserved.

Keywords: Brain ischemia; Lipid peroxidation; Translation initiation; Apoptosis; Growth factors; Signal transduction

*Corresponding author. Department of Emergency Medicine, 6G University Health Care Center, 4201 St. Antoine, Detroit, MI 48201, USA. Tel.:11-313-577-5738; fax: 11-313-577-4131.

E-mail address: [email protected] (B.C. White).

0022-510X/00/$ – see front matter 2000 Elsevier Science B.V. All rights reserved.PI I : S0022-510X( 00 )00386-5

2 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

1. Introduction sufficiently understand the mechanisms involved in neuro-nal injury and repair to design clinically effective therapy.

Stroke is the leading cause of serious, long-term dis- There are as yet no clinically effective therapeutic proto-ability in the United States [1], with about 600,000 people cols for amelioration of brain damage by ischemia andsuffering a new or recurrent stroke each year. Three reperfusion. In the 1980s clinical trials of barbiturate-million Americans are currently permanently disabled induced coma [3] or calcium antagonists [4] failed tobecause of ischemic stroke, and 31% of stroke survivors reduce neurologic damage caused by cardiac arrest andneed help caring for themselves, 20% need help walking, resuscitation. More recently, both the radical scavenger71% have an impaired vocational capacity when examined tirilazad [5] and the glutamate receptor antagonist selfotelan average of 7 years later, and 16% have to be in- [6] have been found ineffective in clinical treatment ofstitutionalized. The direct and indirect cost of stroke in stroke. Thus the theoretical syntheses that led to these trials1998 is estimated at $43.3 billion [1]. In addition to this, were inadequate. Here we will examine new informationcardiopulmonary resuscitation for victims of cardiac arrest, regarding neuronal injury (Fig. 1) and repair, indicatingboth within and outside of the hospital, succeeds in that:restoring spontaneous circulation in about 70,000 patientsa year in the United States. At least 60% of these patientssubsequently die in the hospital as a result of extensive 1. Many phenomena observed during brain ischemia andbrain damage; only 3–10% of resuscitated patients are reperfusion can be accounted for by damage tofinally able to resume their former lifestyles [2]. Thus, membrane lipids, specifically by lipolysis during is-brain injury by transient complete global brain ischemia chemia and by radical-mediated peroxidation of poly-(cardiac arrest) and regional incomplete brain ischemia unsaturated fatty acids (PUFAs) during reperfusion.(ischemic stroke) afflicts a very large number of patients 2. Protein synthesis in vulnerable brain neurons is rapidlywith death or permanent disability. and persistently inhibited at the level of translation

In order to reduce this neurologic morbidity, we must initiation during post-ischemic reperfusion.

21Fig. 1. Ischemic energy depletion leads to increased neuronal cytosolic [Ca ] through pump failure and depolarization- and neurotransmitter-opened21channels. Enzymes activated by cytosolic Ca overload include the protease m-calpain, the phosphatase calcineurin, and phospholipase A2; phospholipase

C is activated directly by depolarization. Cytoskeletal proteins and eIF4G (docks capped-mRNAs for translation) are degraded by m-calpain.Dephosphorylation by calcineurin activates NOS and releases Bad from sequestering proteins. Phospholipases liberate free fatty acids, particularly

21arachidonate, from membranes. Arachidonate causes depletion of ER Ca , inducing the ER unfolded protein response that includes activation of a eIF2a

kinase. Reperfusion allows superoxide generation during arachidonate metabolism and NOS production of NO, which together cause membrane damage bylipid peroxidation. Adrenergic signaling through PKA leads to inhibition of protein phosphatase 1. Together eIF2a kinase up-regulation and PP1down-regulation result in a rapid |20-fold increase in eIF2a(P), which inhibits initiator methionine introduction for peptide synthesis. CHOP transcriptionand caspase 3 activation are induced by eIF2a(P) through unknown mechanisms. Lipid peroxidation inhibits anti-apoptotic growth factor survivalsignaling. Dephosphorylated Bad sequesters Bcl-2, with consequent liberation of Bax, which induces mitochondrial release of cytochrome c and caspase 9to interact with APAF1 for activation of caspase 3 and apoptosis. Thus, the neurons die from radical and proteolytic injury to which they are unable torespond because of failed growth factor signaling and loss of translational competence.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 3

3. Apoptotic mechanisms are engaged in vulnerable ‘charge’ recovers rapidly and approaches normal within 15neurons during early reperfusion, and these mecha- min of reperfusion [8,9].nisms include modifications of translation initiation,activation of proteolysis, and activation of endonu-cleases. 2.2. Reperfusion injury and selective vulnerability

4. These events not only exacerbate damage to criticalmolecules during reperfusion but also impair the Negovskii [10] proposed that the extent of tissue injurycompetence of cellular repair processes. observed following ischemia might largely reflect damage

5. Neuronal competence for antioxidant defense, transla- incurred during reperfusion. Morphologic studies providedtion initiation, inhibition of apoptosis, and repair of direct support for this hypothesis [11,12] and also iden-damaged cellular organelles is regulated at a fun- tified SVNs (hippocampal hilar and CA1 pyramidal neu-damental level by signal transduction mechanisms rons and cortical pyramidal neurons in layers 3 and 5).involving growth factors. Based on conventional staining, reperfusion damage in the

SVNs is observed in two phases [13]. Cytosolic mi-crovacuolation is noted within the first 15 min of reperfu-Because multiple independently lethal mechanismssion, although a substantial degree of normalization is(radical damage, loss of translation competence, proteolyticobserved by 1 h of reperfusion. Morphologic evidence ofactivation, and induction of apoptosis) are involved inprogressive damage to these neurons is then seen duringpost-ischemic neuronal death, single drug intervention willthe following 6 h, and by 48 to 72 h the neurons havebe ineffective. Thus we must undertake a complex ex-disintegrated.perimental effort that will involve both classical characteri-

zation of neuronal and functional loss as well as assays ofbiochemical markers reflecting the success or failure of

2.3. Post-ischemic brain hypoperfusioninterventions against the specific injury mechanisms inorder to develop and validate an effective multi-drug

Capillary hypoperfusion was first shown qualitativelytherapeutic protocol.[14], and subsequent quantitative studies demonstrated thatcortex capillary perfusion was only 30% normal by 90 minof reperfusion following 10 to 20 min of global ischemia[7]. This phenomenon occurred without a change in2. Historical observations of major phenomena inintracranial pressure and was originally taken to representbrain ischemia and reperfusion‘secondary ischemia,’ thought to lead to failure of high-energy metabolism and neuronal death during reperfusion.Four major observations have provided the foundationHowever, by the mid-1980s it was clear that calciumfor investigation of brain injury by ischemia and reperfu-antagonists [7], superoxide dismutase and deferoxaminesion [7]: (1) rapid loss of high-energy phosphate com-[15], or U74006F [16] (a lipid peroxidation chain ter-pounds during ischemia followed by their recovery withinminator [17]) inhibited post-ischemic brain hypoperfusion,the first 15 min of reperfusion; (2) morphological evidencebut had little effect on neurologic outcome. Moreover,that most structural damage occurs during reperfusion,tissue high-energy charge, which approaches zero within 4especially in selectively vulnerable zones; (3) progressivemin of complete ischemia [7], recovers rapidly duringbrain hypoperfusion during post-ischemic reperfusion; andreperfusion following ischemia, and this recovery is appar-(4) prolonged suppression of protein synthesis in selective-ently not adversely affected by the post-ischemic hypo-ly vulnerable neurons (SVNs).perfusion [18–20]. This evidence argues that post-ischemichypoperfusion is an epiphenomenon of more fundamental

2.1. High-energy phosphate compounds biochemical events and is not likely to be itself involved inneuronal death that occurs during reperfusion.

The brain’s ATP supply is dependent on continuousperfusion and approaches zero within about 4 min ofcomplete ischemia [7]. This energy depletion is associated 2.4. Inhibition of brain protein synthesis duringwith depolarization and loss of the normal 10,000/1 reperfusion

21partition of Ca between the extracellular fluid and thecytosol. Depolarization and the increase in intracellular Protein synthesis in vulnerable neurons is substantially

21 21Ca (Ca ) sets off a chain of events including lipolysis suppressed during post-ischemic reperfusion [18,21,22].i

during ischemia followed by free fatty acid metabolism Protein synthesis is a fundamental cellular activity, and a? 2and generation of superoxide radical ( O ) during reperfu- substantial part of our discussion below will focus on the2

sion [7]. During post-ischemic reperfusion, even after mechanisms involved in inhibition of protein synthesisischemic periods up to 1 h, the high-energy phosphate during reperfusion.

4 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

3. Classical mechanisms implicated in selective to salvage neurons following complete global ischemia.1vulnerability Furthermore, H (accumulated during ischemia) inhibits

the NMDA receptor [32], and there is little evidence forTwo major hypotheses emerged during the 1980s from persistently increased extracellular glutamate during re-

efforts to explain the phenomenon of selective vulnerabili- perfusion. These observations raise the question of whetherty. One is the excitotoxic neurotransmitter hypothesis ‘excitotoxicity’ at NMDA receptors is causally related todirected largely at events during ischemia, and the other is morphologic evidence of damage progression during re-the free radical hypothesis directed largely at events during perfusion; there is better evidence that AMPA receptorreperfusion. blockade is neuroprotective following complete global

ischemia [33]. In any case, glutamate excitotoxicity seemsonly to provide special mechanisms in SVNs for ischemia-

213.1. Excitotoxic amino acid neurotransmitters induced Ca overloading, which occurs anyway due tothe large transmembrane ionic gradient and ATP depletion-

The first of these suggests that SVN cell bodies and induced failure of the calcium pumps. Perhaps the mostdendrites receive projections that release large amounts of important thing about this hypothesis is that it representsamino acid neurotransmitter (i.e. glutamate) during is- an early insight that cell signaling mechanisms are in-chemia-induced depolarization [23]. There are two classes volved in neuronal death.of glutamate receptors, the ionotropic receptors, which arethe ligand-gated ion channels, and the metabotropic gluta- 3.2. Lipolysis, free radicals, and lipid peroxidationmate receptors (mGluR), which are coupled to cellulareffectors via GTP-binding proteins. On SVNs there are Free fatty acids (FFAs), and in particular free arach-two subtypes of glutamate ionotropic receptors that are idonic acid, are released during brain ischemia as adistinctively activated by either N-methyl-D-aspartate consequence of the activity of both phospholipase C(NMDA) or a-amino-3-hydroxy-5-methyl-4-isoxazole pro- (activated by depolarization) and phospholipase A (acti-2

21pionic acid (AMPA). NMDA receptor activation opens a vated by increased Ca ) [35,36], and elevated concen-i21 21Ca channel, allowing Ca influx, and AMPA receptor trations of these FFAs persist into the early recirculation

1 1activation opens a Na channel, allowing Na influx. The phase [34,37–42]. The rate of lipolysis during ischemia in1 1increased intracellular Na concentration (Na ) is thought the selectively vulnerable zones is significantly greateri

21to induce reversal of normal Ca extrusion through a 3 than in other areas of the brain [42]. Reperfusion-initiated1 21Na /Ca antiporter, thereby again resulting in increased metabolism of free arachidonic acid may be the major21 ? 2Ca , which is thought to exert cytotoxic effects, as we source of O . Cyclooxygenase catalyzes the addition ofi 2

will discuss below. The mGluRs are coupled to multiple two molecules of O to an unsaturated fatty acid, like2

second messenger systems, including those involved in arachidonic acid, and produces prostaglandin PGG, whichactivation of phosphoinositide hydrolysis, phospholipase D is rapidly peroxidized to PGH with concomitant release of

? 2activation, regulation of cAMP formation, and ion (calcium O [7].2

and potassium) channel modulation. The mGluR agonists The morphological progression of injury during reperfu-have two unusual effects, (1) reduction of transmission at sion [10–12] led to the hypothesis [43] that acceleratedexcitatory amino acid (glutamate) and inhibitory synapses structural damage during reperfusion is a consequence ofand of inhibitory post-synaptic potentials (probably via excessive generation of oxygen radicals followed by lipiddecreased GABA release) in the striatum and hippocam- peroxidation. The free radical hypothesis suggests that thepus, and (2) in the hippocampal CA region induction of SVNs are especially prone to radical-induced damage1

long-term synaptic potentiation, thought to be mediated by (specifically lipid peroxidation) during reperfusion becauseprotein kinase C-induced increases in NMDA receptor (1) they are deficient in glutathione peroxidase [44] andcurrents [24]. Intraocular injection of metabotropic re- (2) they are surrounded by iron-laden supporting cells thatceptor agonists protected against retinal damage induced release this iron during ischemia and reperfusion [45,46].by NMDA, but intra-hippocampal injections of mGluR The changes in the conformation of the fatty acidsagonists caused massive hippocampal damage [25,26]. resulting from lipid peroxidation would then alter the

Both glutamate release [23] and massive increases in permeability and fluidity of the membrane. In addition, and21Ca [27] (from |70 nM to|30 mM) occur during probably more importantly, membranes contain receptors,i

21complete global ischemia, and the Ca influx itself ion channels, and other proteins whose functions are likelystimulates glutamate release from presynaptic vesicles [7]. to be compromised by alterations in the lipid membrane.Re-uptake of glutamate is inhibited by arachidonic acid Lipid peroxidation is a set of radical-mediated chemical[28] or products of lipid peroxidation [29]. Although chain reactions whereby the double bonds in the polyun-magnesium, which inhibits synaptic transmitter release and saturated fatty acid (PUFA) side chains are rearrangedis a natural calcium antagonist, inhibits cell death in vitro [47,48]. In the presence of a transition metal (such as[30,31], NMDA receptor antagonists have generally failed ferrous iron), lipid peroxidation chain reactions can expand

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 5

geometrically [49]. The brain glia have abundant stores of ates oxygen radicals that result in cell death due tooxidized (ferric) iron [50], mostly in ferritin and transferrin subsequent lipid peroxidation, we have found that arach-

? 2[51]. Although O is not itself a potent oxidizer, it does idonate-induced cell death does not require radical mecha-2

promote reduction of ferric and release of ferrous iron nisms [77]. In neuronally-differentiated NB-104 cells, cellfrom ferritin [52], and iron delocalization into low-molecu- death caused by cumene hydroperoxide (a source oflar-weight species is seen in the brain during post-ischemic alkoxyl radical) is completely inhibited by either radicalreperfusion [45]. Peroxynitrite, formed by the reaction of scavengers or iron chelation, but cell death induced by? 2O with NO [produced by neuronal constitutive (nNOS) arachidonate is not inhibited by these strategies. The2

or inducible (iNOS) nitric oxide synthetase], is implicated radical-independent cell death induced by arachidonate is21as the lipid peroxidation-initiating radical species during associated with a prompt increase in cytosolic Ca

reperfusion [53,54]. Peroxynitrite-mediated nitrosylation of derived from both intracellular stores and the extracellulartyrosine residues is also seen in reperfused SVNs [55]. fluid; cumene hydroperoxide does not induce early release

21Membrane lipids are extensively peroxidized by iron- of intracellular Ca stores. These results show that,dependent radical reactions during reperfusion [46,56–60]. although radical damage can be lethal to this neuronal cellFormation of PBN (a radical spin trap) adducts peaks at 5 line, there is also a non-radical mechanism of arach-min reperfusion following 10 or 20 min of carotid occlu- idonate-induced toxicity associated with early release ofsion in rats, and total PBN adduct increases with increasing calcium from intracellular stores, an issue further consid-ischemic time [59]. Lipid peroxidation generates PUFA ered below.hydroperoxides and alcohols that are subsequently de-graded into several aldehydic products, including malon- 3.3. What is wrong with the excitotoxic neurotransmitterdialdehyde [61] and 4-hydroxynonenal [62]. Such lipid and free radical hypotheses?peroxidation products are seen by 15–30 min reperfusion[59,63], persist for up to 72 h [58], and are associated with We note that, because they are largely concerned withsubstantial loss of PUFAs, development of gaps in the two different phases of injury (i.e., ischemia vs. reperfu-plasmalemma, and failure of ion partitioning [12]. These sion), the glutamate and free radical hypotheses are notmarkers of progressive membrane damage are substantially mutually exclusive. There is evidence for the occurrence ofinhibited by pharmacologic iron chelators [64]. Three calcium entry mediated by both energy depletion anddifferent histochemical techniques have shown localization glutamate receptor activation during ischemia. Further-of lipid peroxidation to SVNs during reperfusion [65–67]; more, substantial evidence exists for membrane damagethe lipid peroxidation products appear to accumulate in the caused by both calcium activation of lipases and freeGolgi apparatus, an important site of membrane recycling radical generation during reperfusion. Unfortunately treat-[68,69]. ments directed only at these injury mechanisms do not

There is also evidence of continuing lipolysis during work.reperfusion [70]. Enzymatic lipolysis and lipid peroxida- Attempts at improving post-ischemic neurologic out-tion may act synergistically in membrane destruction come by blocking NMDA receptors have been frustratingduring reperfusion. For instance, erythrocytes damaged by [6,78], even though there is convincing evidence linkinglipid peroxidation are lysed more rapidly by phospholipase NMDA receptor excitotoxicity mechanisms to both mem-

21than are normal membranes [71,72]. Thus, fatty acids that brane damage by Ca -mediated mechanisms and radicalhave been peroxidized are better substrates for phospholip- induction [79]. Likewise, experimental therapeutic inter-ase. Moreover, some of the products of lipid peroxidation ventions that substantially inhibit lipid peroxidation duringstimulate the activity of phospholipase [73,74], and the reperfusion have had marginal effects on neurologicactivities of the lipid repair enzymes lysophosphatidylcho- outcome or morphometric evidence of cell death. Bio-line acyltransferase and fatty acyl CoA synthetase are chemical markers of damage by lipid peroxidation aresubstantially inhibited during reperfusion [75]. In addition, inhibited by deferoxamine [64]; however, deferoxaminelipid hydroperoxides interfere with the reacylation (salvage failed to substantially improve survival of SVNs [80].pathway) of neuronal phospholipids [76]. Similarly, treatment with deferoxamine combined with

Lipid peroxidation also appears to be involved in the superoxide dismutase [15] did not demonstrate majorgenesis of the post-ischemic hypoperfusion phenomenon, improvements in neurologic outcome, although a study ofwhich is inhibited by superoxide dismutase and deferox- hetastarch-complexed deferoxamine suggested a marginalamine [15] or U74006F [16], a lipid peroxidation chain improvement [81]. A report of improved neurologic out-terminator [17]. Thus, there is now evidence implicating come in post-arrest dogs treated with U74006F [82], amembrane damage by reperfusion-induced lipid peroxida- 21-aminosteroid that acts as a radical chain reactiontion in both histological damage to neurons and in the terminator, could not be confirmed [83]. Similarly, wegenesis of the post-ischemic hypoperfusion syndrome. were unable to demonstrate any improvement in neuro-

However, although this discussion suggests that during logic outcome with ethylmethylhydroxypyridone (EMHP)reperfusion oxidative metabolism of arachidonate gener- [84], a small lipophilic iron chelator.

6 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

The failure of therapeutic approaches directed at bloc- calpain is found primarily in CA1 and CA3 pyramidal cellsand in dentate gyrus granule cells, and the highest con-kade of excitotoxic neurotransmitters or free radical mech-centration of calpastatin is in ischemia-resistant pyramidalanisms indicates that these hypotheses do not yet adequate-cells of the CA3 region [100]. Calpastatin immunoreactivi-ly encompass the underlying mechanisms involved inty is present in both neurons and glia with diffuse stainingdeath of vulnerable neurons. We will suggest that otherof the cytosol and more pronounced staining of the cellimportant mechanisms involve terminal differentiation inmembrane [101].neurons, calpain-mediated proteolysis, fundamental altera-

The calpain system appears to regulate neurite out-tions in the systems that regulate protein synthesis, apo-growth, long-term potentiation, and synaptic remodeling inptotic mechanisms, and the effects of growth-factor signalthe brain [102,103]. These processes all involve alterationstransduction on many of these phenomena.of the plasmalemma and suggest that under physiologicconditions calpain activity is tightly regulated by local

21 21alterations of Ca concentration while the general [Ca ]4. Neurons and terminal differentiation i21remains normal. In contrast, the general increase in [Ca ]i

Regulation of the cell cycle may affect resistance to and that occurs during brain ischemia is likely to overwhelmrepair of membrane damage and be important in surviving endogenous regulatory systems and thus result inischemia and reperfusion. Non-replicating cells, such as pathologic calpain activity. Observations of proteolyticthose in the glomeruli, the myocardium, and the central degradation of calpain substrates were utilized to indirectlynervous system, are quite sensitive to damage by ischemia infer pathologic calpain activation in the brain in situationsand reperfusion. The linkage of DNA replication and including excitatory amino acid toxicity [104], ischemicmembrane synthesis follows from the requirement for both neuronal injury [105–110], Alzheimer’s disease [111], andfor cellular reproduction. In prokaryotes, experimental apoptosis [112].manipulations that specifically inhibit lipid synthesis block Both calpain isoforms share a common 30 kDa regula-DNA replication [85–87], and blocked DNA replication is tory subunit but have unique 80 kDa catalytic subunitsassociated with down-regulation of phospholipid synthesis with only 50% sequence homology [113,114]. Both the 80[88]. In human promyelocytes, sterol and phospholipid kDa and 30 kDa subunits of m-calpain and m-calpain (in

21synthesis are markedly decreased following induction of the presence of appropriate [Ca ]) undergo auto-myeloid differentiation [89], and there is a 90% decrease in proteolysis, which significantly reduces the calcium re-neuronal cholesterol synthesis following completion of quirement for subsequent proteolytic activity [97,115,116].differentiation in fetal rats [90]. Tumor cells, which have This autoproteolysis reduces the mass of the m-calpain 80their replication and anti-apoptotic signaling machinery kDa subunit first to 78 kDa and then to 76 kDa [117]. Inswitched on, are not easily killed by lipid peroxidation the presence of adequate calcium concentrations in vitro,[91], and the degree of differentiation is related to suscep- calpains will autoproteolytically degrade into inactive

21tibility to radical oxidant-induced death in many cell lines fragments. The [Ca ] required for autoproteolysis of m-[92]. Thus it is not surprising that there is a substantial calpain is about 10 mM but for m-calpain is about 500 mM,

21enhancement of radical-induced death in Neuroblastoma and for both m-calpain and m-calpain the [Ca ] required21B104 (NB104) cells after neuronal differentiation, and for autoproteolysis is slightly higher than the [Ca ]

differentiation-enhanced ultrastructural damage specifically required for subsequent proteolytic activity. Therefore,includes altered Golgi morphology similar to that observed autoproteolysis is universally accompanied by activation ofin vulnerable neurons of reperfused brains [93]. proteolytic activity [115,118,119]. Phospholipids, particu-

larly phosphatidylinositol (PI), lower the calcium require-ment for autoproteolysis of both m-calpain and m-calpain

5. Ischemia-induced calpain activation by five- to 10-fold [120]. Polyphosphatidylinositol 4,5-bisphosphate (PIP ) allows m-calpain to undergo auto-2

Calpains (EC 3.4.22.17) are a family of non-lysosomal proteolysis at calcium concentrations #1 mM [120].21neutral cysteine proteases [94,95] whose proteolytic activi- The differences in [Ca ] required for autoproteolytic

ty is absolutely dependent on calcium but is also regulated activation of the two isoforms predict autoproteolysis ofby phospholipids, by a specific endogenous inhibitor m-calpain but not m-calpain during brain ischemia, and the(calpastatin), and by a specific activator protein [96]. immunohistochemical localization of m-calpain predictsCalpains and calpastatin have been found in all vertebrate ischemia-induced activity of this protease may be mosttissues studied [97]. The two ubiquitous calpain isoforms important in the SVNs. In the section below on the effectsare m-calpain (calpain-I, CANP-I) and m-calpain (calpain- of ischemia and reperfusion on protein synthesis, we willII, CANP-II). Immunohistochemical studies have demon- see autoproteolytic activation of m-calpain during brainstrated that m-calpain is concentrated in neurons (soma and ischemia [121] and evidence that m-calpain degrades some

7dendrites) compared to glia, while m-calpain is more of the proteins involved in introduction of m G-cappedconcentrated in glia [98–100]. In the hippocampus, m- mRNAs for translation initiation. In the section below on

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 7

apoptosis, we will see evidence for early activation of ischemic preconditioning involves activation of Ras-me-caspase proteases by brain ischemia and reperfusion, and diated signaling.there is now evidence that m-calpain is involved in Although the patterns of post-ischemic protein synthesisactivation of caspase 7 by proteolytic processing of its suppression in the brain are well described, the mecha-proenzyme [122]. These findings show that the increased nisms that cause it were completely unknown until recent-

21[Ca ] associated with brain ischemia activates a calcium- ly. The integrity of DNA [134–136], transcription machin-i

dependent protease as well as phospholipases, and activa- ery [137–142], mRNA processing and transport [143–tion of calpain appears to be involved in both alterations of 145], and translational competence of purified ribosomestranslation initiation mechanisms and activation of apo- [146] are preserved in early reperfusion, and there isptotic pathways. adequate recovery of high-energy phosphates to support

peptide synthesis [147]; thus, these components are notresponsible for the inhibition of protein synthesis. Al-though there has not been a systematic study of the effects

6. Alterations in systems regulating protein synthesis of brain ischemia and reperfusion on intracellular levels ofall amino acids, the levels of those that have been

6.1. General considerations examined are not substantially reduced [148]. Additionalinvestigation is in order here because levels of aminoacyl-

Post-ischemic suppression of protein synthesis was first tRNAs do fall to |60–70% of control values during earlyreported in 1971 by Kleihues and Hossman [123], who reperfusion [127], and we will see below that unchargedobserved a 30% decrease in labeled amino acid incorpora- tRNAs can be involved in kinase activation directed attion after 4 h reperfusion following 30 min of ischemia. In inhibition of translation initiation by modification ofthe SVNs the post-ischemic suppression of brain protein eukaryotic initiation factors (eIFs).synthesis is prompt, severe, and prolonged. Cooper et al. Several lines of evidence are consistent with the hypoth-

14measured [ C]-phenylalanine incorporation by in vitro esis that protein synthesis is suppressed by inhibition oftranslation utilizing the post-mitochondrial supernatant translation initiation during early reperfusion. Ribosomalfrom rat brain homogenates [124]. Incorporation was sedimentation profiles obtained from reperfused brainsnormal in the homogenates derived from rats after 15 min show a preponderance of monomeric ribosomal subunitsof ischemia, but had fallen 80% after 15 min of reperfu- [18,123], suggesting a block in the formation of thesion. Nowak et al. found that in vitro amino acid incorpo- initiation complex [149]. In the study by Cooper et al. of inration by the post-mitochondrial fraction prepared from vitro translation by brain homogenates, the addition of agerbil brain homogenates was normal after 30 min of chain initiation inhibitor (polyinosinic acid) decreasedischemia without reperfusion [125]. However, after 10 min protein synthesis 63% in samples from control animals asof recirculation, in vitro protein synthesis was decreased expected [21]. However, polyinosinic acid had no effect on90% but recovered to 60% of baseline by 6 h of reperfu- in vitro translation with the postmitochondrial supernatantsion. These studies showed that as little as 3 min of obtained after 15 min of reperfusion, indicating thatischemia triggers suppression of protein synthesis during translation initiation was already maximally inhibited. Wereperfusion. The degree of suppression was dependent on extended this observation by showing that global brainthe duration of the ischemia for ischemic periods of less ischemia (10 min cardiac arrest) and 90 min reperfusionthan 5 min; longer ischemic times did not result in any reduce by 90% the rate of in vitro-initiated translation byadditional translation suppression but delayed translational brain homogenates [150].recovery in the surviving areas [22].

Translation competence in the post-ischemic brain is not 6.2. Formation of the translation initiation complexregionally homogeneous; the cortex, hippocampal CA1and hilus, and caudate show severe and prolonged suppres- The assembly of the initiation complex is an intricatesion of protein synthesis, whereas the hippocampal dentate process involving over 140 proteins, including at least ninegyrus and brainstem structures are less affected [126–131]. initiation factors in the eukaryotic cell [151]. Formation ofIn several of these studies the CA1 zone, which is most the initiation complex requires free 40S ribosomalsusceptible to neuronal death following ischemia [132], subunits; however, under normal physiologic conditions,never recovered protein synthesis, indicating that a pro- formation of inactive 80S ribosomes is favored. Binding oflonged deficit in post-ischemic protein synthesis correlates eIF6 to the 60S subunit and of eIF3 plus eIF4C to the 40Swith selective vulnerability. Furuta et al. showed that subunit act to keep the subunits disassociated. The eIF4improved neuronal survival following a substantial is- complex introduces mRNAs onto the small ribosomalchemic insult could be induced by brief periods of prior subunit. The eIF4 complex includesischemia, and that this tolerance was associated with earlyrecovery of protein synthesis in the hippocampal CA1

neurons [133]. We will see below that this phenomenon of 1. eIF4G, which provides docking sites for the other eIF4

8 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

components and for attaching the eIF4 complex to ribosome by an eIF4E-independent mechanism favored byeIF3 on the small ribosomal subunit, proteolytic fragments of eIF4G, and several viruses (in-

72. eIF4E, which binds m G-capped mRNAs, and cluding that for polio) utilize proteolysis of eIF4G and3. eIF4A, which is a helicase that unwinds the mRNA IRES-containing messages to favor translation of viral

near the 59 cap structure. proteins. Similar fragmentation of eIF4G occurs in stressedcells undergoing apoptosis [164], and this proteolysis of

Next, methionine attached to its special initiator tRNA eIF4G has the potential to affect selection of proapoptotic(Met-tRNA ) joins an eIF2-GTP complex to form the messages.i

‘ternary complex’ (Met-tRNA -eIF2-GTP) which then Our studies of brain ischemia and reperfusion have nowi

binds to the free 40S subunit. The 48S ribosomal complex identified two important changes in eIFs: (1) there is(including the 40S ribosomal subunit, the ternary complex, substantial proteolytic degradation of eIF4G mediated byand the mRNA-eIF4 complex) then ‘scans’ the mRNA in m-calpain during both ischemia [165] and reperfusionthe 59 to 39 direction to locate the AUG start codon. Once [150], and (2) there is a rapid |20-fold increase inthe appropriate match is made, eIF5 triggers the hydrolysis eIF2a(P) during reperfusion [150]. Autoproteolytic activa-of the eIF2-bound GTP and release of an eIF2-GDP tion of m-calpain occurs during ischemia in associationcomplex, and eIF4D then brings about a conformational with degradation of eIF4G that is blocked by calpainchange such that the 60S subunit joins the 48S subunit to inhibitors [165].complete the 80S initiation complex. In non-ischemic brain homogenates |1% of total eIF2a

The overall rate of translation initiation is regulated by is eIF2a(P); however, after a 10 min cardiac arrest and 90phosphorylation of serine-51 on the a-subunit of eIF2 min reperfusion (but not after ischemia alone), eIF2a(P) is[eIF2a(P)] [151]. The eIF2-GDP complex released during increased to |23% of total eIF2a [150,166]. This alterationinitiation must be recycled to eIF2-GTP; this is accom- occurs rapidly; at 10 min reperfusion eIF2a(P) is alreadyplished by exchange of the GDP for a new GTP. This increased |18-fold, and there is prominent eIF2a(P)transaction is catalyzed by the guanine nucleotide ex- immunostaining in the cytoplasm of neurons in both thechange enzyme eIF2B. Three key facts about this exchange hippocampus and cortex [166]. At 1 h reperfusion,reaction provide the basis for regulation of the overall rate eIF2a(P) remains only in SVNs [166] and is now promi-of translation by the amount of eIF2a(P): nent both in their cytoplasm on ribosomes and in their

nuclei in association with both the nucleolus and chromatin[167]. The nuclear translocation probably reflects the

1. GDP cannot be released from a-subunit-phos- nuclear localization motif (RRRIR) immediately adjacentphorylated eIF2 by eIF2B. to the ser-51 phosphorylation site on eIF2a, and a-subunit

2. a-Subunit-phosphorylated eIF2 has a 150-fold greater phosphorylated (but not unmodified) eIF2 binds DNA inaffinity for eIF2B than does unphosphorylated eIF2 electromobility shift assays [168] and may have important[152]. consequences for transcription. Immunohistochemical

3. In the brain, eIF2 is present in about a five-fold excess staining of eIF2a(P) and inhibition of in vivo protein35over eIF2B [153,154]. synthesis (by autoradiography of S-labeled amino acid

incorporation) co-map in the reperfused brain [169], andThus, a-subunit-phosphorylated eIF2 is a competitive insulin can induce dephosphorylation of eIF2a and full

inhibitor of eIF2B, and when .20% of eIF2 contains a recovery of protein synthesis in SVNs by 90 min reperfu-phosphorylated a-subunit, regeneration of the eIF2-GTP sion after a 10 min cardiac arrest [169]. These resultscomplex and delivery of the first amino acid for all new provide a mechanistic basis for the inhibition of proteinprotein synthesis is severely inhibited. synthesis in SVNs during reperfusion, and together with

The issue of message selection is complex and not other evidence of a role for eIF2a(P) in the mechanisms ofcompletely understood. Normally, the cell appears to apoptosis (discussed below) suggest that the inhibition ofregulate which mRNAs bind to the ribosome by modu- protein synthesis in SVNs during reperfusion may be a

7lating phosphorylation of the m G-cap-binding protein fundamental phenomenon in the causal pathway leading toeIF4E [155]. Availability of eIF4E is limited in cells [156], the death of vulnerable neurons.and the selection of mRNAs for translation (such as thosefor protooncogenes) can be modulated by phosphorylation 6.3. Implications of eIF modifications induced byof eIF4E [157]. eIF4E is phosphorylated on serine-53 in ischemia and reperfusion for message selectionresponse to several growth factors including insulin [158],PDGF [159], NGF [160] and epidermal growth factor Both the proteolytic degradation of eIF4G and the[161]. Multiple sites on eIF4G are phosphorylated, and this phosphorylation of eIF2a are likely to have importantis thought to induce increased binding of eIF4E [162] and implications for message selection for residual proteinenhanced protein synthesis [163]. Uncapped mRNAs con- synthesis in SVNs. The response of translation initiation totaining internal ribosome entry sites (IRES) can enter the heat shock and to reperfusion in SVNs shares some

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 9

common elements, including phosphorylation of eIF2a overall protein synthesis in SVNs during reperfusion and[170,171]. However, the response to heat shock is not suggestions that administration of cyclohexamide (ancompletely analogous to that seen after ischemia; although inhibitor of peptide elongation) reduces what is thought todephosphorylation of eIF4E is seen in heat shock [172– be apoptotic degeneration of SVNs [184,185].174], we did not find dephosphorylation of eIF4E inducedby brain ischemia and reperfusion [150]. Joshi-Barve et al. 6.4. eIF2a kinases and phosphataseshave shown that antisense-induced depletion of eIF4E andeIF4G substantially inhibits protein synthesis, but about 20 There are four mammalian eIF2a kinases: (1) GCN2proteins, including HSP-70, continue to be synthesized in [186], (2) the hemin-regulated inhibitor in reticulocytesthis situation [175]. In the situation of brain ischemia and (HRI) [187], (3) ‘RNA-activated’ protein kinase (PKR)reperfusion, proteolytic loss of eIF4 components is there- [187], and (4) PERK (PKR-like ER kinase) [188]. We dofore inadequate to explain the blocked translation of HSP- not yet know which one is responsible for phosphorylation70 [176,177] in SVNs, which in fact is almost certainly a of eIF2a during brain reperfusion.consequence of the large increase in eIF2a(P). GCN2 was first identified in yeast, where it is activated

The translation of all messages is not uniformly in- by amino acid starvation [186]. Recently, GCN2 analogueshibited by increased concentrations of eIF2a(P). An have been identified in the CNS of Drosophila [189] andimportant example of this is the case of the message for mice [190]. Three mammalian GCN2 isoforms are gener-GCN4 in yeast [178]. GCN2 is the yeast eIF2a kinase and ated by alternative splicing of the sequence for the N-is activated by uncharged tRNAs caused by amino acid terminal region [190], and all three isoforms are strongly(specifically histidine) deprivation. GCN4 is a transcription expressed in the mouse brain [190]. GCN2 appears to befactor for a family of enzymes for de novo amino acid constituitively associated with and down-regulated bysynthesis. Activation of GCN2 is followed by increased HSP90 [191], and geldanamycin, which has been shown toeIF2a(P) and enhanced translation of message for GCN4 protect neurons from glutamate-induced oxidative damage[178]. The message for GCN4 is polycistronic with a long and apoptosis [192], stabilizes the association of GCN259 leader containing three open reading frames (ORFs) and HSP90 [191]. There are at least three potentialfollowed by the actual code for GCN4 in the 4th and most ischemia and reperfusion-induced activation mechanisms39 ORF. In the absence of eIF2a(P), the ORF nearest the for GCN2. Although there are no published observations59 leader is translated and GCN4 is not synthesized. of the effects of brain ischemia and reperfusion on levelsHowever, in the situation of the marked reduction in the of aminoacylated histidine tRNA, histidine loss occurs inavailability of eIF2-GTP complexes imposed by eIF2a(P), association with brain ischemia [193,194], and histidinethe ribosome is less likely to be able to initiate translation administration offers some neuroprotection [195]. Second,at the upstream ORFs and continues scanning [178]. The a cytoplasmic unfolded protein response, consistent withprobability of a ternary complex binding to the small HSP transcription, could induce dissociation of HSP90ribosomal subunit is enhanced over longer scanning times, from GCN2 and activation of the enzyme. Finally, Rolfesand thus GCN4 gets translated because initiation does not and Hinnebusch [196] found that GCN2 is also activatedoccur on the ORFs nearer the 59 end of the message [178]. by low adenine levels, known to occur in response to brain

These considerations suggest that translation of proteins ischemia and reperfusion [197]; however, their work didencoded by 39-distal ORFs on mRNAs with IRES and long not rule out the simple possibility that, in their model,59 leaders containing other ORFs might actually be favored purine deficiency reduced tRNA charging as a conse-in the circumstance of degradation of eIF4G and increased quence of ATP deficiency.eIF2a(P). Several examples of such mRNAs are relevant HRI activity is stimulated by heavy metals (e.g., iron)to the problems associated with brain ischemia and re- [198], oxidized glutathione, and polyunsaturated fatty acidsperfusion. The mRNA for eIF4G itself has a long 59 leader and lipoperoxides [199]. These activation mechanismsand contains multiple ORFs [179] and an IRES [180], and suggests a potential causal link between radical-mediatedtransfer of this 59 leader onto reporter genes can increase iron delocalization, lipid peroxidation, and the reperfusion-their translation over 100-fold [179]. Thus, the levels of dependent suppression of protein synthesis. However, PaleIF4G should be able to recover during reperfusion. Other et al. could not find HRI in rabbit brain sections [200], andimportant mRNAs that have recently been found to also we have also found little HRI in brain homogenates byhave these characteristics include those for apoptosis-ac- Western blotting. Furthermore, HRI-knockout mice (kindlytivating factor-1 (APAF-1) [181], vascular endothelial provided by J.J. Chen, Harvard-MIT, Cambridge, MA,growth factor [182], and fibroblast growth factor-2 [183]. USA) show no reduction in brain eIF2a phosphorylationApoptosis, in particular, is a crucial area in which these during reperfusion (DeGracia et al., unpublished data).issues are likely to be important. The above considerations PKR is widely distributed in various tissues, includingsuggest that careful examination of the products of residual the brain [201]. PKR was first identified as a double-translation in SVNs during reperfusion may resolve the stranded RNA-activated eIF2a kinase [202]. Its majorapparent conflict between the biomolecular inhibition of function is thought to be to oppose viral infection [203],

10 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

although recent observations indicate a broader role for antisense strategies in conjunction with rat cardiac arrestPKR in cellular events [203,204]. PKR is increased in and resuscitation.differentiated cells [204], and a number of endogenous The greatly enhanced serine-51 phosphorylation oninhibitors and activators of PKR have been identified eIF2a during brain reperfusion could also reflect inhibition[205]. Over expression of either inactive mutants of PKR of the phosphatase activity against eIF2a(P). Althoughor endogenous inhibitors of the enzyme result in tumori- Kimball et al. found decreased eIF2a(P) phosphatasegenesis, suggesting that PKR may be a tumor suppressor activity in the liver after amino acid deprivation [221], we[205]. Furthermore, activation of PKR has been correlated did not observe diminished phosphatase activity againstwith changes in gene expression; it is particularly interest- eIF2a(P) in reperfused brain homogenates [211]. How-ing that activated PKR appears to be required for induced ever, the issue of phosphatase activity is certain to betranscription of the gene for the FAS receptor involved in complicated by subcellular targeting issues.apoptosis [206,207]. PKR is found tightly associated with Ser-Thr protein phosphatases are grouped into fourribosomes [187] and on the rough ER [208] with a small major classes based on their catalytic subunits and respon-fraction present in the nucleolus [209]. PKR transcription siveness to various inhibitors [222]. PP1 and PP2A de-is induced by interferon, and PKR activity is antagonized phosphorylate eIF2a(P) in vitro, but neither PP2B norby Ras-activated cell signaling processes [210], such as PP2C do so [223]. This is also the case in cells [224]those known to be under the control of the insulin receptor. where addition of okadaic acid [222] (an inhibitor of PP1

2 / 2However, we have shown that PKR mice show no and PP2A) results in shutdown of protein synthesis.reduction in the accumulation of eIF2a(P) induced by Furthermore, PP1, but not PP2A, is specifically inhibitedbrain ischemia and reperfusion [211]. by two peptides known as inhibitors 1 and 2 (I-1 and I-2)

PERK is an eIF2a kinase that responds to ER stress [222], and in the reticulocyte lysate addition of either I-1[188]. Many nascent peptides during their synthesis enter or I-2 leads to increased eIF2a(P) and inhibition ofthe ER to undergo processing. ER-resident chaperones translation [225]. Thus, PP1 is the dominant phosphatase(such as GRP78) and protein isomerases facilitate proper for eIF2a(P) [226]. There are four PP1 isoforms (a, g1,folding of the new peptides, and ER glycosylases carry out g2, d), with PP1 a, g1, and d present in the brain [227],post-translational processing on many new proteins [212]. and PP1 a in particular present in the neuronal perikaryalSeveral of these ER protein processing reactions require cytoplasm [228,229].

21physiologic ER Ca concentration (similar to that in PP1 activity is extensively regulated by signal transduc-21extracellular fluid), and depletion of Ca from the ER tion and molecular targeting mechanisms [230]. PP1

leads to an unfolded protein response (UPR) that in higher enzyme complexes consist of a 35 kDa catalytic unit (noweukaryotes includes a rapid suppression of protein syn- itself called PP1 [230] but previously designated PP1c)thesis [213–217] mediated by increased phosphorylation of combined with a wide variety of targeting and regulatoryeIF2a [218–220] that does not require PKR [188]. In the subunits. A bimetal center (Fe /Zn or Fe/Mn) is present inwell characterized yeast ER UPR [212], ER lumenal the PP1 active site and appears crucial for both bindingunfolded proteins are detected by IRE-1, followed by and catalytic removal of the target phosphate [230]. Thereactivation of its cytosolic RNAse. The IRE-1 RNAse is compelling evidence that PP1 functions only whenprocessing of Hac1 mRNA allows translation of this associated with one of |30 different targeting subunitstranscription factor for the ER chaperones and isomerases. (including the ribosome L5 protein), and known targetingPERK is an evolutionary recombinant protein with homol- subunits indicate PP1 regulation of glycogen metabolism,ogy to the IRE-1 unfolded protein detector in its N- protein synthesis, and cell cycle machinery [230]. The PP1terminal ER-resident domain and homology to GCN2 in its targeting issue can be crucial in preventing accumulationcytosolic eIF2a kinase domain [212]. PERK activation of eIF2a(P); although herpes virus activates PKR, ainvolves autophosphorylation, and the active form displays virally-expressed PP1 targeting unit with homology toretarded electrophoretic mobility [188]. PERK is present GADD34 causes an |10,000-fold increase in PP1 activityand activated in response to thapsigargin (a selective against eIF2a(P) and thus prevents PKR-induced shut-off

21inhibitor of the ER Ca pump [188]) or arachidonate in of translation initiation [231]. PP1 can also inactivate PKRneuronally-differentiated NB104 cells (O’Neil et al., un- [232] and by inference could be involved in down-regula-published data, PERK antibody kindly provided by David tion of the activity of other eIF2a kinases.Ron), and arachidonate induces both release of intracellular In general, signal transduction mechanisms do not

21Ca stores and phosphorylation of eIF2a in these cells induce post-translational modification of PP1 itself but[77]. Unfortunately, in our hands Westerns of brain rather regulate phosphorylation of PP1 targeting subunitshomogenates have substantially more ‘noise’ around and/or inhibitors [222,230,233]. In this regard PP1 andPERK than is present with homogenates from NB104 PP2A often appear to be counter-regulatory in signalingcells; this has obstructed identification of any PERK mechanisms in which growth factors and adrenergicmobility shift induced during brain reperfusion. We are agonists exert opposite effects. For example, in differen-therefore currently evaluating the role of PERK utilizing tiated myocytes insulin induces rapid doubling of PP1

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 11

activity against glycogen synthase through a site-specific different absolute concentrations of p67, and cells withphosphorylation on the appropriate targeting subunit, high p67 content exhibit enhanced resistance to viral-whereas PP2A dephosphorylates this targeting subunit site induced eIF2a phosphorylation [248]. Viral-induced phos-and thereby inactivates PP1 dephosphorylation of glycogen phorylation of eIF2a and shut-off of host-cell proteinsynthase [234]. PP2A itself is activated through the cAMP- synthesis is associated with rapid activation of a 60 kDadependent protein kinase A (PKA) but deactivated by an enzyme that deglycosylates p67 [247], which makes p67insulin receptor-induced tyrosine phosphorylation [235]. unable to prevent phosphorylation of eIF2a but does notRecently, two peptide inhibitors of PP2A have been lead to its degradation [245]. These findings suggested thatidentified and shown to enhance the activity of PP1 against p67 deglycosylation during ischemia and reperfusion and/a number of substrates in a manner dependent on a or the level of expression of p67 by various groups of

21physiologic concentration of Mn , and these investigators neurons could affect their susceptibility to eIF2a phos-have noted the potential of this system to account for phorylation. However, we have recently found that neitherseveral examples of PP1 and PP2A counter-regulation the glycosylation state of p67 nor its brain immuno-[236]. localization are affected by ischemia and reperfusion

Of the several known PP1 inhibitors [237], one [249]. Although altered p67 glycosylation does not appear(DARPP-32) is found primarily in basal ganglia rather to be in the causal pathway leading to phosphorylation ofthan in vulnerable hippocampal or cortical neurons, nearly eIF2a during post-ischemic reperfusion, the persistence ofnothing is known about the intriguing ribosome inhibitor glycosylated p67 may be important in limiting the extentof PP1 (RIPP-1), and NIPP-1 is in the nucleus and of eIF2a phosphorylation (which in our hands neverunlikely to be involved in any regulation of cytoplasmic exceeds 23% of total eIF2a) and supporting the potentialeIF2a(P). As we noted above, I-2 can inhibit PP1 activity, for recovery of translation competence in vulnerablebut emerging evidence suggests that I-2 may be more neurons.important as a folding chaperone than as an inhibitor ofPP1 [230,237]. Thus, here we focus on I-1, which is an 6.6. The importance of recovery of protein synthesis18.7 kDa cytosolic peptide that, when phosphorylated on during early reperfusionthreonine 35 by PKA, competitively inhibits PP1 with a Ki

of 1.6 nM [237,238]. The average concentration of I-1 (1.6 Late in reperfusion, other fundamental mechanisms maymM in skeletal muscle) is about twice that of all forms of serve to perpetuate the suppression of protein synthesis. ByPP1 combined. Epinephrine, isoproterenol, and dopamine 8 h of reperfusion following a 15 min cardiac arrest ininduce PKA activation of I-1; this effect is opposed by dogs, ionic partitioning is lost, associated with a concomi-insulin [234,239], which along with other growth factors tant 30% decrease of lipid double bond content andinduces increased PP1 activity [240]. I-1 is found mostly ultrastructural evidence of large breaches in the plas-in the pyramidal neurons in the hippocampus and layers malemma of SVNs [12]. The loss of ionic gradients of

1 1 21 21II–III and V–VI in rat cerebral cortex [241,242], and I-1 is Na , K , Ca and Mg are incompatible with transla-activated in hippocampal neurons by glutamate-induced tional competence [250–252] and cell viability. Accord-long-term potentiation [237]. Taken together these findings ingly, effective therapy will probably have to allowsuggest investigation for threonine 35 phosphorylation on recovery of protein synthesis before 4–8 h reperfusion inI-1 during brain reperfusion and raise the possibility that order to enable the vulnerable neurons to respond in waysinsulin’s post-ischemic neuroprotective effects (that are that will prevent the later deterioration.independent of any hypoglycemia [243]) and its ability to As one would expect, there is evidence that an irrevers-induce dephosphorylation of eIF2a(P) and restoration of ible suppression of protein synthesis represents a lethaltranslation competence could involve dephosphorylation of cellular lesion, and inability to express proteins assures cellI-1. death. Furuta et al. have demonstrated that recovery of

protein synthesis is probably a prerequisite to neuronal6.5. Regulation of the availability of eIF2 to eIF2a salvage [133], and Hossmann’s group has very recentlykinases demonstrated in a focal ischemia model that early failure

of protein synthesis (at 1 h ischemia) occurs in a penumbr-A final mechanism that might be involved in eIF2a al zone before ATP depletion and precisely demarcates the

phosphorylation during post-ischemic brain reperfusion is ultimate extent of the expanding lesion [253]. Someregulation of the availability of eIF2 as substrate for eIF2a investigators have reported a reduction in delayed neuronalkinases. Gupta and collaborators [244–247] have identified death with the administration of known inhibitors ofa peptide with M 67 kDa (p67) that, when glycosylated, protein synthesis such as cylcohexamide [184,185]; how-r

binds to eIF2 and protects eIF2a from phosphorylation, ever, it is not possible to conclude from these experimentsand they have shown that preincubation of brain homoge- that the inhibition of protein synthesis that develops as anates with antibody against p67 enhances subsequent in consequence of ischemia and reperfusion is directedvitro phosphorylation of eIF2a. Different cell lines contain toward neuron sparing. Indeed, these investigators did not

12 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

examine the effect of their intervention on protein syn- into their specialized positions, the excess cells are elimi-thesis by either autoradiography of tissue sections or nated by apoptosis. These genetically programmed pro-translation by brain homogenates. It is likely that identifi- cesses during development have provided a reproduciblecation and molecular elimination of the kinase responsible phenomena, and developmental apoptosis has been exten-for eIF2a phosphorylation during reperfusion will allow sively studied in Caenorhabitis elegans. Embryonic de-these issues to be settled by evaluation of neuronal survival velopment of this nematode produces exactly 1090 cells,when the reperfusion-associated inhibition of protein syn- of which 131 undergo apoptosis [258]. Studies of apop-thesis is prevented. tosis in this model reveal five groups of genes involved

with this process [255]: those triggering cell death, thoseinvolved in the cell death process itself (including ced-3

7. Apoptosis and ced-4, that is, C. elegans death gene), those required] ] ]

for engulfment of the dying cell, those implicated in final7.1. General considerations disposal of the cellular corpse, and a single gene (ced-9 )

involved in suppression of cell death. Mammalian genesApoptosis is a process of self-destructive cell death that have been identified with sequence homology to three of

involves activation of mechanisms encoded in the genomes those in the C. elegans system [255]; there are genes withof all higher eukaryotes [254]. Appreciation of the impor- homology to ced-3 [ICE-protease (interleukin 1b

]tance of apoptosis in mammalian systems has only de- converting enzyme and the more general family of caspase

] ]veloped during the last 5–7 years [255], and some of the proteases)], ced-4 (apoptosis-activating factor 1, APAF-1),details of the process remain poorly understood. Neverthe- and ced-9 (the Bcl-2 family).less, it has become clear that the process of apoptosis isintimately involved in several acute disease states (includ- 7.3. Receptor-mediated caspase activationing injury mechanisms involved in ischemia and reperfu-sion of organs), and mutational events leading to inhibition In higher eukaryotes several genes with homology toof apoptosis are involved in the development of many ced-3 have been identified. These include genes for thecancers. Furthermore, some prototypes of therapeutic FAS (also called Apo-1), TNFR (tumor necrosis factorapproaches to these processes are now emerging. Thus receptor), and TRAIL-R (tumor necrosis factor-relatedhere we will examine the signaling pathways and the apoptosis-inducing ligand receptor), plasma membranespecific execution mechanisms of apoptosis. receptors that activate intracellular signaling pathways

In early work, DNA fragmentation into size ‘ladders’ leading to apoptosis by activation of caspase proteasesobserved after electrophoresis was thought to be the [259,260]. These receptors include a cytoplasmic region ofhallmark of apoptosis. These ‘ladders’ were generated |70 amino acids, called the ‘death domain,’ that is abecause DNAse activity produced random double strand protein–protein interaction zone shared by many of thecuts in the internucleosomal regions. Increased intracellu- proteins in this system [255]. Ligand binding to these

21lar Ca appears to be involved in activation of this receptors leads to recruitment of adapter-linkers (FADD,DNAse(s) [256]. Subsequently it was recognized that there CRADD, TRADD, etc.) that then recruit upstream caspaseswere additional morphologic alterations characteristic of to the activation complex by association of death domainsapoptosis; these include chromatin condensation, extensive [261]. Caspase 8 (Mach1, FLICE) is an upstream caspasealterations in the microtubular array, fragmentation of the that is recruited to the receptor–adapter complex and thencell into ‘apoptotic bodies,’ and detachment of the cell cleaves other pro-caspases into the active proteases [262].from neighbors or culture substrate [254,255]. The altera- An important function of the apoptotic system is totions in a specific instance of apoptosis are somewhat provide a viral defense in which infected cells are inducedvariable, and it has now become clear that the cytoplasmic to die, and several viruses encode death-domain-containingalterations of apoptosis (and indeed cell death) can occur proteins that inhibit caspase-mediated apoptosis. Thiswithout the appearance of DNA fragmentation [257]. This inhibition is achieved by either binding caspase 8 (cowpoxvariability in the phenomenology of apoptosis raised protein CrmA [262,263] and herpes virus protein E8 [264])problems with respect to assays that allow its confident and blocking both its ability to associate with the receptor–recognition. adapter complex and to activate downstream caspases

[262] or by binding linker-adapters (molluscum con-7.2. Insights from apoptosis during embryonic tagiosum virus MC159 protein [264]) and inhibiting theirdevelopment of C. elegans function.

The relevance of this membrane receptor signalingApoptosis is an important process in embryonic de- system to reperfusion has recently been shown. FAS

velopment. Many more cells are generated than are finally protein is increased |130-fold in post-ischemic myocytesneeded, particularly in highly differentiated organs such as at 2 h reperfusion following myocardial ischemia [265],the kidney, heart, and brain [255]. As these cells migrate and there is now compelling evidence for the involvement

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 13

of apoptosis in the post-ischemic death of cardiac an amino-terminal-truncated IkBa lacking Ser-32 [273].myocytes [266]. The situation with regard to apoptosis This truncated IkBa cannot be phosphorylated on theinduction by ligand activation of FAS, TNFR, or TRAIL-R missing Ser-32, is not ubiquinated and degraded, andduring brain reperfusion is less clear. Although there is statically binds and inhibits NF-kB, which is then unablelittle FAS mRNA in normal brains and Matsuyama et al. to provide prosurvival transcriptional support. Althoughfound FAS mRNA induced by 6 h reperfusion following we found no studies of NF-kB activity or localization30 min ischemia in mice [267], this group’s mapping by in during the early minutes of brain ischemia and reperfusion,situ hybridization did not observe FAS expression by it has been shown that IkB is degraded and nuclear NF-kBvulnerable neurons during reperfusion. However, Martin- activity is increased within minutes of cardiac ischemiaVillalba et al. [268] have elegantly demonstrated co-locali- [281], and neuronal nuclear localization of NF-kB haszation of N-terminal phosphorylation of the transcription been observed at 24 and 72 h brain reperfusion [282,283].factor JUN (which can be induced by signaling from these NF-kB activity is lost in the ischemic core in strokereceptors [260]) and production of FAS ligand in neurons models, and inhibition of NF-kB activation considerablyadjacent to the ischemic core and provided evidence that enhances the sensitivity of neuronal cell lines to apoptosisFAS ligand can kill neuronal cell lines, an effect that can induced by TNFa [284]. Furthermore, although knockoutbe overcome by expression of a dominant negative mutant of the p50 component of NF-kB is not lethal, these miceof FADD [269]. Brain localization of TRAIL-R by im- are deficient in transcription factor binding to the NF-kBmunohistochemistry or in situ hybridization has not been consensus sequence and display substantially enhanced inreported, but brain injection of TRAIL is not neurotoxic, vivo injury of SVNs by excitotoxic mechanisms [285].although it can eradicate malignant glioma [270]. The The presence and role of death-inducing plasmalemmalpicture with respect to TNFR is similarly complicated; ligand receptors in brain ischemia and reperfusion requiresChopp’s group showed appearance of TNFR as quickly as further clarification, and the possibility remains that a1 h after MCA occlusion in mice, but the protein was receptor-activated caspase apoptotic response is not en-localized to microvessels [271]. Moreover, TNFR knock- gaged as a primary mechanism of death in the vulnerableout mice showed exacerbated brain damage in response to neurons. There are, however, intracellular mechanisms thatfocal ischemia [272], possibly because TNFR activation can engage the caspases independent of the receptor-also leads to activation of the transcription factor NF-kB activated mechanisms.[206], which can counter-regulate apoptotic mechanisms.

NF-kB is a transcription factor comprised of 50 and 65 7.4. Intracellular mechanisms of apoptosiskDa subunits and is normally associated with its inhibitorIkB, which binds and sequesters NF-kB in the cytoplasm 7.4.1. Non-receptor-mediated caspase activation[273]. Ligand-induced association of TNFR with tumor The caspase system is more complicated than just thenecrosis factor receptor-associated factor 2 (TRAF2) [274] well characterized FAS, FADD/CRADD, caspase 8 mech-induces activation of the IkB kinase complex, which anism reviewed above. Currently about 14 caspases havephosphorylates IkBa on Ser-32 and Ser-36 [273]. Phos- been identified and can be divided into ‘initiators’ andphorylation of IkBa on these sites is followed by its ‘effectors.’ The interactions with caspase-recruiting adap-ubiquination on Lys-21 and -22 and its degradation by the ters by long pro-domains in initiator caspases 8 and 9proteosome. This liberation of NF-kB from IkB allows activate the proteolytic function of these caspases, whichnuclear translocation of NF-kB, which acts as a transcrip- in turn activate other effector caspases by proteolysistion factor to induce expression of genes promoting cell [286]. This type of associative activation mechanism cansurvival, such as ‘cellular inhibitors of apoptosis’ 1 and 2 be set off inside the cell without involvement of plas-(c-IAP1 and c-IAP2) [275,276], A20 [277] (an anti-apo- malemmal receptors. For example, the prototypic caspaseptotic zinc-finger protein whose mechanism of action CED-3 can be activated by direct association with CED-4remains unknown [278]), and manganese-superoxide dis- [287], which undergoes nuclear localization and inducesmutase [279]. We note that because pro-survival NF-kB cell death that requires function of CED-4’s nucleotide-signaling is induced from TNFR and requires transcription binding P-loop [288]. Co-expression of CED-9 with CED-and translation, the use of translation inhibitors has been 4 blocks the nuclear localization and lethality of CED-4required in many studies of TNFR-induced apoptosis. The [288] by a direct interaction between these two proteinsc-IAPs appear to block both activation of caspase 8 at the [289].ligand receptor complex and the activity of proteolytically- In mammalian cells the homologue of CED-4 (apop-processed caspases [276], and vector-directed overexpres- tosis-activating factor 1, APAF-1 [290]) and its associativesion of the X-linked IAP blocks both caspase 3 activation initiator caspase (caspase 9) are normally in separateand death of hippocampal SVNs during brain reperfusion compartments; APAF-1 is cytosolic, while in SVNs both

2 / 2[280]. NF-kB p65 embryos die from massive apoptosis cytochrome c and caspase 9 reside in the mitochondrial[273]. Furthermore, during apoptosis caspase activity intermembranous space [291]. In the presence of dATPappears to lead to inactivation of NF-kB by generation of and cytochrome c, APAF-1 undergoes self-oligomerization

14 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

and activates caspase 9, which then activates caspase 3 and localization of specific caspases during post-ischemic[286]. Cytochrome c and caspase 9 are coordinately brain reperfusion.released in response to mitochondria undergoing mem-brane permeability transition or interaction with the pro- 7.4.2. Bcl-2, Bax, and Badapoptotic Bcl-2 homologue Bax (discussed below) [291]. In the C. elegans system, the protein encoded by ced-9SVN mitochondria release both cytochrome c [292,293] inhibits apoptosis. The human genes encoding CED-9-and caspase 9 [291] within 1–2 h following ischemia but related proteins include Bcl-2, Bax, Bad, Bcl-x, mcl-2, Bak,do not appear to undergo membrane permeability transition and Bak-2 [255]. In terms of the ‘viruses vs. eukaryotes[12,291,294]. Proteolytically-activated caspase 3 can be wars,’ it is interesting that several mammalian viruses,seen as early as 1 h reperfusion [295]. After release of including the Epstein–Barr virus (mononucleosis andcaspase 9 from SVN mitochondria, it is substantially Burkitt’s lymphoma), carry genes encoding Bcl-2 homo-translocated into SVN nuclei [291], consistent with other logues that inhibit apoptosis [315]. There is no evidenceobservations of an ‘apoptosome’ complex containing that Bcl-2 directly inhibits caspases, but it does appear to

21APAF-1, cytochrome c, and caspase 9 undergoing nuclear inhibit release of Ca from the endoplasmic reticulumlocalization during apoptosis [296]. (ER) [316], an event that we have already seen can induce

Although caspases are constituitively expressed as phosphorylation of eIF2a and that we will see can beproenzymes [262], ischemia and reperfusion also induce crucial to the activation of intracellular apoptosis mecha-transcription for caspase 3 in vulnerable neurons [297]. In nisms. The Bcl-2 gene encodes two proteins generated byvarious model systems caspase 3 is activated before alternative splicing products at the C-terminus, whichseveral other caspases [298]; indeed, caspase 3 is the affects membrane localization. Bcl-2a is localized to thepredominant enzyme involved in very rapid cleavage of outer nuclear membrane, the ER, and the outer mito-the DNA repair enzymes poly(ADP ribose) polymerase chondrial membrane; Bcl-2b is a cytosolic protein [255].(PARP) [299,300] and DNA-dependent protein kinase Both the Bad and Bax gene products are promoters of[301]. PARP recognizes strand nicks in DNA and marks apoptosis. Release of Bax from binding by Bcl-2 [317]them for repair by generating poly(ADP ribose) chains on allows Bax to oligomerize and form channels in mem-adjacent nuclear proteins [302], up-regulates the tumor branes [318], including the outer mitochondrial membranesuppressor p53 [303], is required for cellular recovery after where it can cause the release of cytochrome c [318] andradical-mediated DNA damage [304], and its activity is caspase 9 [291]. Homozygous deletion of Bax inducesrequired for insertion of retroviral-derived DNA into the substantial attenuation of post-ischemic neuronal deathgenome [305]. Caspase 3 also cleaves actin to produce a [319]. The tumor suppressor transcription factor p53 ischaracteristic 15 kDa peptide [306]. Caspase 3 is clearly activated in response to poly(ADP)-ribosylation of nu-involved in developmental apoptosis and is required for cleosomal proteins and both up-regulates Bax and down-normal brain development in mice [307]. Caspase 3 regulates Bcl-2 transcription [255]. These observationstranscripts are present in neurons of developing rat brains suggest that the ratio of Bcl-2 to Bax is important inbut are greatly down-regulated in the adult brain [308], regulating apoptosis.although some caspase 3 is constituitively expressed in the Bad appears to exert its pro-apoptotic effects by bindingadult brain [309]. There is evidence for active caspase 3 in and sequestering Bcl-2 [320]. The activity of Bad isSVNs within a few hours following cerebral ischemia regulated by phosphorylation; the dephosphorylated form[295,309], and the relatively specific caspase 3 inhibitor binds Bcl-2. Bad is phosphorylated on serines 112 and 136N-benzyloxycarbonyl-Asp-Glu-Val-Asp-fluromethylketone by Akt (PKB), a central element in survival signaling, thatdecreases caspase cleavage products, reduces tissue dam- is activated through the growth-factor-mediated PI-3 ki-age, and improves functional outcome in a stroke model in nase pathway [321]. Phosphorylated Bad is sequestered inrats and mice [310]. the cytoplasm by 14-3-3 proteins [296]. A major effect of

The details of non-receptor-mediated caspase mecha- maintaining Bad in its phosphorylated state appears to benisms involved in an apoptosis occurrence appear to reflect prevention of the release of cytochrome c from mito-the initiating event. For example, down-regulation of the chondria, and Akt is unable to inhibit the mechanismcaspase Nedd2 rescues both sympathetic neurons and responsible for cytochrome c release when Bad ser-112PC12 cells from death caused by growth factor withdrawal and ser-136 are mutated to alanines [322]. Bad is de-

21but does not prevent apoptosis induced by down-regulation phosphorylated by the Ca /calmodulin-activated phos-of superoxide dismutase (SOD) [311]. Like caspase 3, phatase calcineurin [323], which is also implicated in NOSNedd2 is expressed in neural precursor cells, is develop- activation.mentally down-regulated [312], efficiently cleaves PARP[313], and its processing from the proenzyme to the active 7.4.3. PKR and phosphorylation of eIF2a

form in response to growth factor withdrawal is inhibited There is recent evidence directly connecting both PKRin a neural cell line overexpressing Bcl-2 [314]. Further activation and eIF2a phosphorylation to apoptosis. Cellsstudies are needed to clarify the identity, activation timing, from PKR knockout mice are resistant to apoptosis in-

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 15

duced by TNF-a, lipopolysaccharides, or viral RNA [331]. FK506 binding to FKBP12 dissociates FKBP12[206,324]. The presence and function of TNFR was not from both receptors and modestly increases their resting

21affected in the knockouts [206] because TNF-a on these probability of being open for Ca transit [332,333], but21cells induced a normal NF-kB response. Unfortunately, after treatment of the RyR with FK506, actual Ca

these studies did not directly examine caspase activation, movement again is only seen at the four discrete ‘subcon-which should have occurred in response to TNF-a unless ductance’ levels [331]. Thus it appears that the overall

21the PKR knockout blocked signaling for caspase transcrip- effect of FK506 on RyR-mediated ER Ca egress is to21tion or activation. However, the issue of whether eIF2a(P) induce a decrease in ligand-activated Ca conductance.

has a direct role in inducing apoptosis has been examined. In recent follow-up of a seminal 1994 study [334],Expression of a mutant eIF2a substituting alanine (cannot FK506 has been shown to both salvage vulnerable neuronsbe phosphorylated) for the ser-51 phosphorylation site after brain ischemia and also to allow them to synthesizeblocks apoptosis induced by TNF-a [324] or PKR [325] stress response proteins within 3 h of reperfusion [335].and also induces down-regulation of Bax expression [326]. This same study examined neuronal death in the hip-Furthermore, expression of a conditional eIF2a mutant pocampus after 7 days reperfusion and found a 16-foldimitating the negative charge at ser-51-P (by substituting increase in the number of surviving neurons in the CA1 (toaspartate for ser-51) induces immediate apoptosis [324], levels statistically no different from neuron density inand activation of caspase 3 [326]. These results suggest sham ischemic controls) of animals treated with FK506.that PKR induces apoptosis through phosphorylation of Similar results with regard to stress protein synthesis andeIF2a. Furthermore, because eIF2a(P) itself appears suffi- neuronal salvage were obtained with FK506 treatment in acient to signal for apoptosis, it is likely that eIF2a kinases model of ischemic stroke induced by 2 h occlusion of aother than PKR are also involved in apoptotic pathways. middle cerebral artery [336].

7.4.4. ER calcium 7.4.5. Immunophilins21As we noted above, depletion of Ca from the ER Immunophilins are a family of cellular proteins that bind

induces PERK (and perhaps PKR) activation, phosphoryla- immunosuppressant drugs, such as FK506, cyclosporin A,tion of eIF2a, and inhibition of protein synthesis, and there and rapamycin. FK506 is a microbially-derived macrolide

21is ER Ca depletion in SVNs during and after transient antifungal antibiotic used clinically to suppress transplant21ischemia [327]. On the ER two Ca channel-regulating rejection [337] and is a structural homologue of rapamycin

receptors are known, the ryanodine and inositol-triphos- but is structurally unrelated to cyclosporin A [338].phate receptors (RyR and IP-3R). Both receptors are Rapamycin and cyclosporin A are not neuroprotectivepresent in vulnerable CA1 neurons, with IP-3R more during post-ischemic reperfusion [334]. FK506 crosses theprominent in these cells and RyR more prominent in the blood–brain barrier, but cyclosporin A does not [339].vulnerable hilar neurons [328]. There is evidence that Both cyclosporin A and FK506 actually inhibit T-cellarachidonate competes with ryanodine for its receptor apoptosis but appear to exert their immunosuppressant[329]; interpreting this result is complicated because, functions by inhibiting T-cell activation, proliferation, andalthough 30 mM arachidonate did not appear to increase stimulation of interleukin-1 production [338].

21the probability that the receptor-gated Ca channel in Generally, cyclosporin A and FK506 bind differentreconstituted ER vesicles was open, arachidonate did in a proteins, but many or all of these proteins catalyze the

45 21dose-dependent fashion stimulate release of Ca . There cis–trans isomerization of peptidyl-prolyl bonds (rotamaseare no published studies of potential interactions between or PPIase activity) [338,340] and thus frequently functionarachidonate and the IP-3 receptor, although IP-3R-de- as peptide folding isomerases in complexes withficient T cells are resistant to apoptosis induced by ionizing chaperones such as heat shock proteins [341]. Because theradiation, FAS ligand, and glucocorticoids [330]. immunophilins have been largely recognized by their

Both the RyR and IP-3R are formed by a tetramer of binding affinity for cyclosporin A or FK506, nomenclaturetheir respective primary proteins, and one 12 kDa peptide, for them is further divided into two large groups,which binds the immunosuppressant tacrolimus (or FK506, cyclophilins and FK-binding proteins (FKBPs).thus FK-Binding Protein — FKBP12), is associated with Several activities of the immunophilins are of substantial

] ] ]each monomer of both receptors [331,332]. Expression of interest with respect to apoptosis. Cyclophilin A (18 kDa),the cloned RyR in naive Spodoptera frugiperda cells B (21 kDa), and C (22.9 kDa) have been found to be(which also do not normally produce FKBP12) generated DNAses independent of their rotamase activity [342]. Thetetrameric RyR receptors, but these receptors did not show DNAse activities of cyclophilin A and B are stimulated by

21 21 1strong ligand activation of Ca conductance and rather increased Ca and inhibited by K (IC 583 mM). This50

exhibited ‘subconductance’ at four distinct levels, sug- DNAse activity is not inhibited by cyclosporin A and isgesting that each monomer included a channel [331]. In directed against double-strand linear, double-strand super-this system, co-expression of FKBP12 generated channels coiled circular, and single-strand linear DNA withthat exhibited ligand-activated opening to full conductance cyclophilin C.B.A [342]. Furthermore, cyclophilin A is

16 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

extensively homologous to an 18 kDa Ca/Mg nuclease nisms emerged after the development of multicelled eu-[343] (NUC18) that may one of the nucleases [344] karyotes.responsible for nucleosomal DNA fragmentation in apop- This suggests that apoptotic mechanisms first arose as atosis. Recent evidence suggests that NUC18 is activated by primitive immune response to viral infection of single cellsproteolysis of an inactive |60 kDa precursor [345]. This in multicelled organisms. Indeed, from the perspective ofproteolytic activation of NUC18 suggests caspase in- the survival of individual organisms to reproduction,volvement, and indeed caspase activation appears to be suicide of the one cell in a one-celled organism does notupstream of endonuclease activation in apoptosis [346]. make any sense. However, suicide of one infected cell in aAnother endonuclease involved in DNA fragmentation multicelled organism could eliminate the replication ma-during apoptosis is DNA Fragmentation Factor 40 (DFF- chinery the virus would utilize to multiply and so promote40, also known as CAD for caspase-activated DNAse). the survival of the multicelled organism. This reasoningThis endonuclease exists in a complex with an inhibitor leads to the idea that apoptotic mechanisms involving(ICAD) that, when cleaved by caspase 3, releases the inhibition of the initiation of protein synthesis may be veryactive DFF-40 [347]. primitive. Apoptosis-naive organisms like yeast regulate

An |21 kDa cyclophilin homologous to cyclophilin D protein synthesis by ser-phosphorylation of eIF-2a in[348] is an integral part of the mitochondrial permeability- response to environmental signals, such as inadequatetransition pore formed in the mitochondrial membrane amino acids. Thus all that had to happen to block viral

21under the simultaneous influence of matrix Ca overload replication was to make the regulation of the introductionand radical stress [349], and both cyclosporin A and of the first amino acid into any new protein responsive to

21 the presence of viral nucleotides, exactly the situation withFK506 inhibit Ca transit of this pore [350]. The integral21 PKR.involvement of an immunophilin with a Ca transiting

21 This perspective also makes intelligible the developmentpore and the ability of FK506 to inhibit Ca transit of theof caspases with very high rates of proteolysis againstpore is consistent with the evidence above suggesting that

21 DNA repair enzymes and the ability to activate nucleases.FK506 inhibits ligand-activated Ca transit across theThe objectives here were to prevent incorporation of theER.viral genome into that of a host cell, to kill the host cell,Some complexes of immunophilins and cyclosporin Aand to degrade viral polynucleotides. Later, governance ofor FK506 inhibit calcineurin (protein phosphatase-2B) butcell survival during embryonic development could havenot other serine / threonine phosphatases [351]. Calcineurinbeen accomplished by signal-transduction regulation ofdephosphorylates and activates Bad [323] and NO-synthet-this platform of apoptosis. By this interpretation, secondaryase [352], and the putative role of NO in formingsignal transduction mechanisms [including the FAS-peroxynitrite radicals together with the inhibition of NOTNFR/FADD-CRADD/caspase 8 activation system, lipid-synthetase by FK506 were the rationale for examining the

21derivative (arachidonate and IP-3) mediated ER Caeffect of FK506 during brain reperfusion [336]. However,shifts, and p-53 regulation of the Bax–Bcl-2 ratio andcomplex formation with calcineurin is not a generalmitochondrial cytochrome c release [356] in response toproperty of FKBPs [353], and derivatives of FK506 that doDNA damage] feed into the more primitive mechanismsnot lead to inhibition of calcineurin are neurotrophic andfor inhibition of protein synthesis, prevention of DNApromote neuron recovery after injury [354]. Furthermore,repair, and DNA degradation.Toung et al. [355] have shown that the neuroprotective

It is clear that a signal for apoptosis can come from anyeffect of FK506 does not involve in vivo inhibition of NOof several organelles (plasmalemma, mitochondria, ER,production during brain ischemia and early reperfusion.and nucleus), and there is evidence for involvement of atleast the ER and mitochondria in activation of apoptotic

7.5. Putting apoptosis together: an evolutionary biochemistry in association with brain ischemia and re-perspective perfusion. From that basic perspective, the complexity of

the details of this biochemistry arises from the presence ofTwo fundamental observations emerge from the above multiple input pathways and the fact that the fundamental

review. First, all multicellular eukaryotic organisms appear execution mechanisms are connected by molecular sig-to have apoptotic systems, but many single-celled eukary- naling.otes are naive with respect to these mechanisms. Second,viruses have developed a variety of mechanisms thatinhibit eukaryotic apoptosis; these mechanisms include at 8. Immediate early genes, growth factor signalleast inhibition of the formation of the membrane re- transduction, and regulation of translation andceptor–adapter linker system for activation of caspases, apoptosisdirect inhibition of caspases, viral expression of Bcl-2, andinhibition of eIF2a kinases. Because single-celled eukary- 8.1. Immediate early genes and ‘heat shock’ proteinsotes do not have these apoptotic mechanisms, theseobservations show that viral resistance to apoptotic mecha- Immediate early genes [357,358] include proto-on-

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 17

cogenes of the c-fos and c-jun families. These proteins stress-activated protein kinases), which are activated inform heterodimers to constitute a variety of AP-1 tran- response to oligomerization of the adapter TRAF-2 toscription factor complexes that bind specifically to consen- either IRE-1 (after activation by ER stress) or TNFR (aftersus promoter sequences upstream of target genes [359] and activation by ligand binding) [383]. There is some evi-also bind TBP (TATA-binding protein), thus facilitating dence that JNK activation and c-JUN phosphorylation areinitiation of transcription [360]. linked to induction of apoptosis [384], but although

In rats [140] and gerbils [141] c-fos and c-jun transcripts Herdegen et al. [385] observed both phenomena after 24 hare elevated in brain homogenates within 30 min of post- brain reperfusion, neither were apparently present at 3 andischemic reperfusion. However, examination of various 5 h reperfusion. Moreover, DNA binding by these proteinscell types during early reperfusion showed that c-fos is post-translationally modified not only by phosphoryla-mRNA is transcribed in large amounts in the damage- tion but also by redox alterations of specific cysteines inresistant dentate granule cells, whereas only modest and the DNA-binding domain [386]; cysteine oxidation (thatdelayed expression is seen in CA and CA pyramidal does not appear to involve disulfide bond formation)1 3

layers of the hippocampus [361–363]. Immunocytoch- blocks DNA binding, and homologous cysteines and redoxemistry has similarly shown early translation of c-FOS regulation are also present in the NF-kB and CREBprotein found in damage-resistant rat cortical layers II and (cyclic-AMP response-element-binding) transcription fac-VI but delayed translation in the SVNs of layers III–V tors [386]. The enzyme (Ref-1) that reduces this cysteine[364] and of the hippocampal CA1 [365]. Furthermore, turns out to be identical to an abasic endonuclease (APE)antisense-mediated inhibition of c-FOS synthesis enhanced that plays a major role in the repair of radical-inducedthe final extent of tissue damage associated with focal DNA damage [386], and the endonuclease and cysteinebrain ischemia [366]. reductase activities of this protein reside in non-overlap-

There are important interrelations between induction of ping domains [387]. Indeed, while the reductase activity ofc-fos and c-jun and the synthesis of other stress proteins Ref-1 /APE is itself down-regulated by oxidation of this[367]. The highly conserved heat shock proteins (HSPs — protein, the endonuclease activity is not [387]. Thisnamed because of their discovery in cells exposed to heat evidence suggests a mechanism down-regulating the activi-stress) are now known to be chaperones to prevent ty of several important transcription factors during radicalaggregation of incompletely-folded peptides. HSPs facili- stress until there is confidence that the genomic codingtate correct folding of proteins [368–370], dissociate sequences are sound.clathrin baskets in the presence of ATP (which is a The issue of the actual transcriptional activity of AP-1presumed requirement for cell membrane recycling) components is further complicated by recent evidence that[371,372], interact with cellular tumor suppressor p53 CHOP, a transcription factor component that forms mainly[373], protect cultured neurons from glutamate toxicity functionally-negative heterodimers with members of the[374], and can be induced by growth factors [375]. Heat C/EBP family and is implicated in apoptosis inductionshock mRNAs [376] and proteins [377,378] are expressed [388], also forms heterodimers with members of the FOS–in the brain during reperfusion. After an ischemic insult, JUN family [389]. Transcription of CHOP occurs inthe SVNs in the hippocampus and cortex transcribe large response to eIF2a(P), is also induced by conditionalquantities of hsp-70 mRNA, but there is little translation of expression of the asp-51 mutant eIF2a imitating a con-HSP-70 by these neurons compared to those in surviving stituitively phosphorylated protein, and is blocked in cellsareas of the hippocampus (i.e. the dentate gyrus) [379]. containing a homozygous mutation for non-phosphor-The c-fos / c-jun /hsp system is directly activated by ioniz- ylatable eIF2a (R. Kaufman, unpublished data). Thus,ing radiation [380], presumably by hydroxyl radical-me- working out the roles of cFOS and cJUN in the response todiated events; the HSP response is induced in the presence ischemia and reperfusion will require at least characteriza-of free radicals [381], but not by increased intracellular tion of (1) regional translation of the messages for thesecalcium alone [382]. proteins, (2) their regulatory phosphorylation state, (3)

The above results suggest the important association of their heterodimerization partners, and (4) their redox state.early transcription and translation of c-fos, c-jun, and In this complicated regulatory environment, although manyhsp-70 with neuronal survival, but the complexity of the of the proteins whose genes are transcriptionally stimulatedmechanisms by which c-FOS and c-JUN are regulated with by AP-1 fall into families that should favor survival [390]regard to DNA binding preclude direct extension of this (antioxidants, growth factors and their receptors, lipidevidence to inferences regarding their transcriptional ac- management, structural proteins, protease regulation, andtivity. The formation of various heterodimers of the FOS– electrolyte and water management), we do not at allJUN family members undoubtedly affects the pattern of understand how c-FOS and c-JUN affect the fate oftranscriptional activation. Furthermore, post-translational neurons after brain ischemia and reperfusion.modifications of these proteins also affect their DNA-binding activity. N-terminal phosphorylation of c-JUN 8.2. Growth factors and neuronal survival(mainly at ser-73) enhances c-JUN activity and is carriedout by a family of kinases (JNKs, also known as SAPKs or Cell growth, proliferation, and differentiation are reg-

18 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

ulated, in part, by growth factors [391–393]. The brain has activated kinases or dephosphorylated by protein phosphat-receptors for several growth factors, including insulin ases that are also activated [418]. It is the modulation of[394–396], fibroblast growth factor (FGF) [397], and the phosphate content of specific proteins that underlies thenerve growth factor (NGF) [398–400], all of which have cellular regulation noted above. Another germane example

21been found in the selectively vulnerable hippocampal and involves the Ca –calmodulin complex, which activatescerebral cortical neurons. Similarly, transforming growth phospholipases and calcineurin; calmodulin is phos-factor beta (TGF-b) [401], insulin-like growth factor-1 phorylated by the insulin receptor [419,420], thereby(IGF-1) [402–406], and epidermal growth factor (EGF) reducing its ability to bind calcium, a reaction that may[407] have been found in the brain or elsewhere in the modulate insulin’s antilipolytic effects. SVNs, includingcentral nervous system. Examples of growth factor re- specifically the cortical pyramids and those in the CA1

sponses following ischemia are an increase in basic FGF in hippocampus, have both insulin receptors and substantialthe rat hippocampus, caudate, putamen, and cortex [408], levels of phosphotyrosine-containing nuclear proteinsincreased production of mRNAs for brain-derived neuro- [421], consistent with transcriptional regulation throughtrophic factor and NGF in rat dentate gyrus and in the rat growth factor tyrosine kinase systems in these cells.hippocampus CA and CA regions [137], and increased1 2

levels of IGF-1 in the lateral cortex, hippocampus, 8.3. Growth factor signal transduction to the apoptoticstriatum, thalamus and pyriform cortex [409]. mechanisms

The mechanisms by which growth factors influence cellsare not completely understood, although we will address Apoptosis is inhibited by a variety of growth factors,the issue of growth-factor signaling to the apoptotic including insulin, IGF-1, NGF, and others. Here we willmechanisms in more detail below. It is clear that trans- focus on signaling from the insulin receptor (Fig. 2) inmembrane receptor proteins for growth factors charac- order to simplify the discussion without intending toteristically include a ligand-activated tyrosine kinase at the suggest that the insulin signaling pathway is unique in itsinner surface of the plasmalemma, and growth factor anti-apoptotic effects, with the possible exception of itseffects are mediated through receptor kinase-induced activation of farnesyltransferase as noted below.‘phosphorylation cascades’ leading to changes in the The anti-apoptotic activity of insulin requires integrityactivity of numerous proteins affecting transcription, trans- of the tyrosine kinase function of the insulin receptorlation, and structural cellular integrity. Thus, it is not [422,423], functional Ras, functional PI-3 kinasesurprising that the administration of growth factors has [424,425], activation of Akt, and activation of the tran-been shown to improve post-ischemic neuronal survival scription factor NF-kB with subsequent increased expres-[410]. Fibroblast growth factor substantially inhibits gluta- sion of TRAF-1, TRAF-2, manganese-superoxide dismut-mate receptor-mediated neuronal damage [411]. Nerve ase (the mitochondrial SOD), cIAP1, and cIAP2 [276,426].growth factor administered either before or after 5 min of Complexes involving the TRAFs and cIAPs inhibit bothcarotid occlusion in mongolian gerbils significantly re- the upstream caspases (caspases 8 and 9) [276]. The classicduced the loss of hippocampal CA neurons [412]. Treat- insulin receptor tyrosine kinase substrates, IRS-1 and IRS-1

ment with IGF-1 after 2 h reperfusion following an 2, may not be involved in insulin’s anti-apoptotic effects.ischemia-hypoxic insult in rats reduced neuronal loss in the Lee-Kwon et al. [422] showed in Chinese hamster ovarycortex [409], and treatment with insulin, either pre- or cells that there was no loss of insulin receptor anti-post-ischemia in the rat, reduced cortical and striatal apoptotic effects with a deletion mutation of the receptor inneuronal necrosis and reduced infarction in the cerebral the juxtamembrane region required for phosphorylation ofcortex, thalamus, and substantia nigra [243,413,414]. IRS-1 and -2. However, these investigators also found that

Insulin effects in particular are complex and affect the insulin-stimulated activation of farnesyltransferase wasfundamental cell regulatory functions well beyond the required for the anti-apoptotic effect of insulin. Rasmanagement of glucose. For example, insulin receptor- farnesylation is required for its attachment to the plas-mediated protein phosphorylation cascades stimulate tran- malemma, but not for activation of the RAS/Raf /ERKscription of c-fos / c-jun, increase mRNA efflux from the pathway.nucleus, induce the enzymatic systems for de novo Several observations indicate a central role for signalinglipogenesis, activate the mitochondrial pyruvate dehydro- through Ras for growth factor-mediated resistance togenase complex, modify the phosphatidylinositol trans- apoptosis. For SVNs, brief periods of ischemic precondi-cytoplasmic signaling system, and stimulate the heat shock tioning are well known to induce substantial resistance toresponse [375,415,416]. The actions of insulin are me- delayed death after a subsequent otherwise lethal transientdiated by a membrane receptor tyrosine kinase that shares ischemic insult. Recent work in primary cultures ofhomology with other growth factor receptors [417]. The cortical neurons has shown that farnesyltransferase in-initial effect of insulin receptor activation is tyrosine hibitors or a dominant-negative Ras block the protectiveautophosphorylation and activation of the intrinsic tyrosine effects of ischemic preconditioning. In contrast, transfec-kinase. Secondarily, other proteins are phosphorylated by tion with a constituitively active Ras provides substantial

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 19

Fig. 2. Many growth factor proteins activate anti-apoptotic signal transduction, which in all cases appears to require Ras and Akt. There is evidence ofunique signaling from the insulin receptor for the C-terminal farnesylation of Ras, which is required for its membrane localization and anti-apoptoticsignaling. The signaling intermediates through which Ras down-regulates PKR are unknown. Other Ras anti-apoptotic signaling involves its directactivation of PI-3 kinase, which generates 39-phosphorylated phosphoinositides that induce up-regulation of p70(s6K) independent of Akt. Phos-phoinositides also bind to pleckstrin homology domains on both phosphoinositide-dependent kinases (PDKs) and Akt. This induces translocation of Akt tothe plasmalemma and activation of Akt’s kinase function through its phosphorylation by PDKs. Growth factor signaling through Akt-mediatedphosphorylation of eIF4E-BP1 and GSK-3, Ras-dependent down-regulation of PKR, and phosphoinositide-dependent up-regulation of p70(s6k) generates acomprehensive enhancement of the mechanisms of translation initiation. Akt phosphorylation (1) of Bad allows Bcl-2 to inhibit Bax-mediated release ofcaspase 9 and cytochrome c from mitochondria, (2) of caspase 9 inhibits its proteolytic activity, (3) of IkB and NF-kB both frees and activates thepro-survival transcription factor NF-kB, and (4) of FKHR1 causes ejection of this pro-apoptotic transcription factor from the nucleus. Mn-SOD inhibitsradical-mediated mitochondrial damage, and complexes of the TRAFs and cIAPs (cellular inhibitors of apoptosis) inhibit caspases.

protection from a subsequent ischemic insult, even without release of cytochrome c and caspase 3 activation [431];2 / 2ischemic preconditioning [427]. Furthermore, the precondi- furthermore, the Rce1 mutation is lethal in mice [429].

tioning effect may also involve NO-dependent Ras activa- We have already seen that Ras signaling down-regulatestion [427] by cys-118 nitrosylation [428]. PKR [210,432], which is involved in both phosphorylation

Ras belongs to a family of proteins that prior to post- of eIF2a and induction of FAS transcription; the signalingtranslational processing have at their C-termini a CAAX pathway from Ras to down-regulation of this eIF2a kinasemotif, in which the cysteine is usually followed by two also appears not to involve Raf /ERK [432]. It is probablyaliphatic amino acids [429]. These proteins undergo three to be expected that Ras signaling will be found to similarlysteps of post-translational processing of their C-termini down-regulate the other eIF2a kinases. Ras-mediated[429] in which (1) the cysteine is modified with a lipid- signaling also up-regulates translation through PI-3 kinase-derived thiofarnesyl or thiogeranylgeranyl group, (2) the dependent modulation of the phosphorylation states ofRce1 endoprotease removes the AAX moiety, and (3) p70(s6k), eIF4E-BP1 [433], and eIF2B [434].methylation then generates the final S-farnesylcysteine PI-3 kinase can be activated by association with IRS-1methyl ester at the C-terminus. In the case of Ras, all three but is also activated by a direct association with Ras and isof these steps are essential to its appropriate localization at an upstream effector for activation of p70(s6k) [435],the plasmalemma, where it produces its effects in response protein phosphatase 1 [436] [dephosphorylates eIF2a(P)],to activation of growth-factor receptors. In this regard, it is Akt [321,437], and NF-kB [438,439]. Over-expression ofvery interesting that insulin (specifically via the insulin PI-3 kinase protects sympathetic neurons from apoptosisreceptor), but not IGF-1, epidermal growth factor, or induced by withdrawal of the insulin-analogue NGF [440].platelet-derived growth factor, induces the phosphorylation Ras can directly activate the PI-3 kinase /Akt pathwayand activation of farnesyltransferase [430]. Thus, insulin independent of the association of PI-3 kinase with IRS-1,appears to have an essential ‘priming’ action for activation and specific mutations on Ras can separate its activation ofof the Ras signaling pathways by other growth factors, and the PI-3 kinase system from activation of the Raf /ERKwe note that farnesyltransferase inhibitors can induce system [441], which are thus seen to be separate signaling

20 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

pathways. Active PI-3 kinase principally generates phos- [451] have shown that growth factor up-regulation ofphatidylinositol 3,4 bisphosphate (PI3,4P) and phospha- protein synthesis in neurons involves this system.tidylinositol 3,4,5 trisphosphate (PI3,4,5P) [296], which Thus, anti-apoptotic growth factor signaling involveswe will see are involved in activation of Akt. The PI-3 activation of a Ras /PI-3 kinase /Akt pathway leading to (1)kinase-dependent pathway to activation of p70(s6k) is not general up-regulation of translation initiation systemsclear [442], but activation of this enzyme is abrogated by probably essential for completion of the NF-kB transcrip-expression of a dominant-negative Ras [435]. We note that tional response, (2) maintenance of Bad in its phos-p70(s6k) phosphorylates the ribosomal S6 protein and phorylated state with consequent preserved Bcl-2-mediatedstimulates translation of ribosomal proteins and translation inhibition of Bax-induced channels in the ER and mito-elongation factors whose transcripts contain chondria, (3) Akt-mediated inhibitory phosphorylation of59oligopyramidine tracts [433]. caspase-9, (4) Akt-mediated inhibitory phosphorylation of

The three Akt isoforms are the protein products of the pro-apoptotic transcription factors, and (5) up-regulation ofcellular homolog of the transforming oncogene from the transcription by NF-kB for proteins that enhance mito-AKT8 retrovirus [443]. PI-3 kinase products bind to a chondrial radical resistance and repress the activity of bothpleckstrin homology domain near the Akt N-terminus and upstream caspases. This is a comprehensive and powerfulcause translocation of Akt to the plasmalemma [296], anti-apoptotic system, and priming of this pathway bywhere it is activated by phosphorylation of thr-308 and insulin-induced activation of farnesyltransferase may beser-473 by PI-3 kinase-dependent kinases (PDKs) that essential for anti-apoptotic signaling by other growthappear also to be activated by binding phosphorylated factors. The fact that Akt is caspase degraded [452]phosphoinositides in their pleckstrin homology domains suggests that growth factor activation of this system may[296]. have some urgency during early reperfusion.

Active Akt has several important anti-apoptotic func-tions (Fig. 2). As we saw in the discussion of apoptosis,phosphorylation of Bad by Akt prevents Bad from recruit- 9. Therapeutic considerationsing Bcl-2 from Bax and thus prevents Bax from generatingchannels in ER and mitochondrial membranes. Akt also 9.1. Resuscitationinhibits caspase 9 proteolytic activity by phosphorylating iton ser-196, and expression of ala-196 mutant caspase 9 The data regarding the involvement of insulin in surviv-induces Akt-resistant apoptosis [444]. Activated Akt-1 and al signaling for neurons, as well as other cells, raise-2 can undergo nuclear translocation and phosphorylate the concern about the use of large doses of epinephrine inpro-apoptotic transcription factor FKHR1 [445], thus in- resuscitation of patients from cardiac arrest. Catechol-ducing nuclear export of FKHR1 to the cytoplasm, where amines, such as epinephrine, cause prompt and prolongedit is sequestered by 14-3-3 proteins [446]. Akt also inhibition of insulin secretion by the pancreas [453],phosphorylates and activates IKKa [447] (one of the activate cAMP-induced phosphorylation of the insulinkinases that phosphorylates IkB to induce release of NF- receptor thereby reducing its tyrosine kinase activity [454],kB) and is further involved in anti-apoptotic transactiva- and promote iron-mediated lipid peroxidation [455,456]tion of NF-kB-dependent transcription through phos- and neuronal death [457]. Further examination of thephorylation of the NF-kB p65 subunit [448]. We note that effects of high catecholamine levels on brain growth factorAPAF1 contains a consensus sequence for Akt phosphoryl- levels and receptor responsiveness needs to be conductedation [296] and could also be counter-regulated by this in the setting of cardiac arrest and resuscitation.enzyme. Akt also up-regulates translation by increasing theavailability of eIF4E and inducing activation of eIF2B. As 9.2. Summary of molecular injury mechanismsreviewed above in the discussion of translation initiation,

7eIF4E binds m G-capped mRNAs for delivery to the Brain ischemia (Fig. 1) results in rapid loss of high-ribosome initiation complex. There is a cytoplasmic eIF4E energy phosphate compounds and generalized depolariza-binding protein (eIF4E-BP1 or PHAS-1) that, when de- tion, and adenosine degradation products have the potentialphosphorylated, binds eIF4E and thus inhibits translation to induce the dissociation of GCN2 from HSP 90 withof capped messages. The PI-3 kinase [449]–Akt [442] consequent activation of this eIF2a kinase. SVNs aresystem induces phosphorylation of eIF4E-BP1 and en- characterized by their dendritic fields receiving largehanced translation of capped messages. Akt also induces a amounts of glutamate released by the general depolariza-more general enhancement of translation through up-regu- tion during ischemia. Under these circumstances, bothlation of eIF2B, which regenerates the eIF2-GTP complex voltage-dependent calcium channels and glutamate-reg-

21required for methionine initiation of all new peptides. ulated calcium channels allow a large increase in [Ca ],i21Glycogen synthase kinase-3 (GSK-3) phosphorylates and and this is accompanied by depletion of Ca from the ER

21inhibits not only glycogen synthase but also eIF2B [434]. lumen. These compartmental Ca shifts are associatedAkt directly inactivates GSK-3 [450], and Quevedo et al. with activation of m-calpain, calcineurin, and phospholip-

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 21

ases with consequent proteolysis of calpain substrates peroxidation progress unimpeded while awaiting full ex-(including eIF4G), activation of NOS and potentially of pression of the molecular response to growth factorBad, and accumulation of free arachidonic acid. signaling. Indeed, radical stress can itself induce a 90%

Early during reperfusion, oxidative metabolism of arach- reduction in insulin-induced phosphorylation of ser-473 onidonate is associated with a burst of excess oxygen radical Akt [458].production, iron is released from storage proteins by These considerations argue for a multi-drug therapeuticsuperoxide-mediated reduction, and NO is generated by approach that will probably have to include (1) peptideNOS. These events result in peroxynitrite generation, growth factors for early induction of survival signal-trans-inappropriate protein nitrosylation, and lipid peroxidation, duction and recovery of translational competence, (2)which is substantially localized to the SVNs and in inhibition of calpain, (3) inhibition of radical damage, andultrastructural studies appears to principally damage the (4) caspase inhibition. Rigorous studies to establish suchplasmalemma. The initial recovery of ATP supports very an approach will require evaluation of neurological andrapid phosphorylation of eIF2a, and substantially in- histopathological outcome as well as of molecular end-creased levels of eIF2a(P) persist in SVNs for at least points associated with each intervention component toseveral hours. High catecholamine levels induced by the establish the necessary role of each drug. Such a multi-ischemic episode itself and by catecholamine administra- factorial therapeutic approach to the molecular injurytion down-regulate insulin secretion and induce inhibition mechanisms now appears essential for the development ofof growth-factor receptor tyrosine kinase activity, and substantially improved clinical treatment for brain is-these effects are expected to be associated with down- chemia and reperfusion.regulation of survival signal-transduction through the Raspathway. Release of mitochondrial cytochrome c andcaspase activation occur during the early hours of reperfu- Acknowledgementssion. Thus the SVNs find themselves with substantialmembrane damage, calpain-mediated proteolytic degrada- Supported in part by National Institute of Health Grantstion of eIF4G and cytoskeletal proteins, altered translation NS33196 (Drs. Krause, White, DeGracia, Grossman, andinitiation mechanisms that substantially reduce total pro- Rafols), NS02008 (Dr. Sullivan), NS01832 (Dr. Neumar),tein synthesis and impose major alterations in message GM48517 (Dr. Grossman), and NS39860 (Dr. Rafols). Dr.selection, down-regulated survival signal-transduction, and DeGracia is also supported in part by a grant from thecaspase activation. These circumstances predict indepen- American Heart Association, and Dr. O’Neil is supporteddently fatal pathways involving loss of membrane integrity in part by a grant from the Emergency Medicine Founda-for partitioning ions, progressive proteolysis by calpain tion.and caspases, and inability to check these processesbecause of loss of general translation competence andsurvival signal-transduction.

References

9.3. Single drug intervention will never work[1] Direct costs were calculated by Thomas A. Hodgson, chief econom-

ist and acting director, Division of Health and Utilization Analysis,This picture argues powerfully against the concept ofOAEHP, NCHS, CDC. Estimates of indirect costs were made by

single drug intervention for therapy for brain ischemia and Thomas J. Thom, statistician in the Division of Epidemiology andreperfusion that has characterized the research approaches Clinical Applications, NHLBI.

[2] Krause GS, Kumar K, White BC, Aust SD, Wiegenstein JG.of basic science laboratories, the pharmaceutical industry,Ischemia, resuscitation, and reperfusion: mechanisms of tissue injuryand clinical trials. Antioxidants and NOS inhibitors canand prospects for protection. Am Heart J 1986;111:768–80.reduce the rate of membrane damage, but there is no

[3] Brain Resuscitation Clinical Trial I Study Group. Randomizedreason to believe that they induce survival signal-transduc- clinical study of thiopental loading in comatose survivors of cardiaction, inhibit the activity of calpain or caspases, or induce arrest. New Engl J Med 1986;314:397–401.rapid recovery of translational competence. Similarly, [4] Brain Resuscitation Clinical Trial II Study Group. A randomized

clinical study of a calcium-entry blocker (lidoflazine) in the treat-caspase inhibitors may block the progression of apoptoticment of comatose survivors of cardiac arrest. New Engl J Medmechanisms, but again there is no reason to expect that1991;324:1225–31.

they will inhibit either lipid peroxidation or calpain activity [5] The RANTTAS Investigators. A randomized trial of trilazador induce rapid recovery of translational competence. mesylate in patients with acute stroke. Stroke 1996;27:1453–8.Peptide growth factors can induce early recovery of [6] Davis SM, Lees KR, Albers GW, Diener HC, Markabi S, Karlsson

G et al. Selfotel in acute ischemic stroke: possible neurotoxic effectstranslational competence, induce survival signal-transduc-of an NMDA antagonist. Stroke 2000;31:347–54.tion, and could affect transcription for physiological in-

[7] Krause GS, White BC, Aust SD, Nayini NR, Kumar K. Brain cellhibitors of calpain and radical damage; however, because death following ischemia and reperfusion: a proposed biochemicalsignal-transduction, transcription, and translation take time, sequence. Crit Care Med 1988;16:714–26.it is probably foolish to let calpain remain active and lipid [8] Hossman KA, Sakaki S, Kimoto K. Cerebral uptake of glucose and

22 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

oxygen in the cat brain after prolonged ischemia. Stroke [31] Drejer J, Benveniste H, Diemer NH. Schousboe A: cellular origin of1976;7:301–5. ischemic-induced glutamate release from brain tissue in vivo and in

[9] Siesjo BK, Ljunggren B. Cerebral energy reserves after prolonged vitro. J Neurochem 1985;45:145–51.hypoxia and ischemia. Arch Neurol 1973;29:400–7. [32] Traynelis SF, Cull-Candy SG. Proton inhibition of N-methyl-D-

[10] Negovskii VA. Resuscitation and artificial hypothermia, New York: aspartate receptors in cerebellar neurons. Nature 1990;345:347–50.Consultants Bureau, 1962. ˚[33] Nellgard B, Wieloch T. Postischemic blockade of AMPA but not

[11] Jenkins LW, Povlishock JT, Lewelt W, Miller JD, Becker DP. The NMDA receptors mitigates neuronal damage in the rat brainrole of postischemic recirculation in the development of ischemic following severe cerebral ischemia. J Cereb Blood Flow Metabneuronal injury following complete cerebral ischemia. Acta Neuro- 1992;12:2–11.pathol 1981;5:205–20. [34] Abe K, Kogure K, Yamamoto H, Imazawa M, Miyamoto K.

[12] Kumar K, Goosmann M, Krause GS, Nayini NR, Estrada R, Mechanism of arachidonic acid liberation during ischemia in gerbilHoehner TJ et al. Ultrastructural and ionic changes in global cerebral cortex. J Neurochem 1987;48:503–9.ischemic dog brain. Acta Neuropathol (Berlin) 1987;73:393–9. [35] Drenth J, Enzing CM, Kalk KH, Vessies JC. Structure of porcine

[13] Sato M, Hashimoto H, Kosaka F. Histological changes of neuronal pancreatic phospholipase A2. Nature 1976;264:373–7.damage in vegetative dogs induced by 18 minutes of complete

[36] Moskowitz N, Schook W, Puskin S. Regulation of endogenousglobal brain ischemia: two-phase damage of Purkinje cells and

calcium-dependent synaptic membrane phospholipase A2. Brain Reshippocampal CA pyramidal cells. Acta Neuropathol (Berlin)1 1984;290:273–80.1990;80:527–34.

[37] Rehncrona S, Westerberg E, Akesson B, Siesjo B. Brain cortical[14] Ames III A, Wright L, Kowada M, Thurston JM, Majno G. Cerebralfatty acids and phospholipids during and following complete andischemia II: the no-reflow phenomenon. Am J Pathol 1968;52:437–severe incomplete ischemia. J Neurochem 1982;38:84–93.53.

[38] Bazan NG. Effects of ischemia and electroconvulsive shock on free[15] Cerchiari EL, Hoel TM, Safar P, Sclabassi RJ. Protective effects offatty acid pools in the brain. Biochim Biophys Acta 1970;218:1–10.combined superoxide dismutase and deferoxamine on recovery of

[39] Yasuda H, Kishiro K, Izumi N, Nakanishi M. Biphasic liberation ofcerebral blood flow and function after cardiac arrest in dogs. Strokearachidonic and stearic acids during cerebral ischemia. J Neurochem1987;18:869–78.1985;45:168–72.[16] Hall ED, Yonkers PA. Attenuation of post-ischemic cerebral hypo-

[40] Yoshida S, Inoh S, Asano T, Sano K, Kubota M, Shimazaki H et al.perfusion by the 21-aminosteroid U74006F. Stroke 1988;10:340–4.Effect of transient ischemia on free fatty acids and phospholipids in[17] Braughler JM, Pregenzer JF, Chase RL, Duncan LA, Jacobsen EJ,the gerbil brain. J Neurosurg 1980;53:323–31.McCall JM. Novel 21-amino steroids as potent inhibitors of iron-

[41] Katsuki H, Okuda S. Arachidonic acid as a neurotoxic and neuro-dependent lipid peroxidation. J Biol Chem 1987;262:10438–40.trophic substance. Prog Neurobiol 1995;46:607–36.[18] Hossman KA, Kleihues P. Reversibility of ischemic brain damage.

[42] Umemura A. Regional difference in free fatty acids release and theArch Neurol 1973;29:375–84.[19] Meis G, Paschen W, Hossman KA. Cerebral blood flow, glucose action of phospholipase during ischemia in rat brain. No To Shinkei

utilization, regional glucose, and ATP content during the maturation 1990;42:979–86.period of delayed ischemic injury in gerbil brain. J Cereb Blood [43] Demopoulos HB, Flamm ES, Pietronigro DD, Seligman ML. TheFlow Metab 1990;10:638–45. free radical pathology and the microcirculation in the major central

[20] White BC, Hildebrandt JF, Evans AT, Aronson L, Indrieri RJ, nervous system disorders. Acta Physiol Scand 1980;492(Suppl):91–Hoehner TJ et al. Prolonged cardiac arrest and resuscitation in dogs. 119.Brain mitochondrial function with different artificial perfusion [44] Ushijima K, Miyazaki H, Morioka T. Immunohistochemical locali-methods. Ann Emerg Med 1985;14:383–7. zation of glutathione peroxidase in the brain of the rat. Resuscitation

[21] Cooper HK, Zalewska T, Kawakami S, Hossman KA. The effect of 1986;13:97–105.ischemia and recirculation on protein synthesis in the rat brain. J [45] Krause GS, Nayini NR, White BC, Hoenher TJ, Garritano AM,Neurochem 1977;28:929–34. O’Neil BJ et al. Natural course of iron delocalization and lipid

[22] Dienel GA, Pulsinelli WA, Duffy TE. Regional protein synthesis in peroxidation following a 15 minute cardiac arrest in dogs. Annrat brain following acute hemispheric ischemia. J Neurochem Emerg Med 1987;16:1200–5.1980;35:1216–26. [46] Zaleska MM, Floyd RA. Regional lipid peroxidation in rat brain in

¨[23] Siesjo BK. Pathophysiology and treatment of focal cerebral is- vitro: possible role of endogenous iron. Neurochem Reschemia. I. Pathophysiology. J Neurosurg 1992;77:169–84. 1985;10:397–410.

[24] Schoepp DD, Conn PJ. Metabotropic glutamate receptors in brain [47] Halliwell B, Gutteridge J. Oxygen toxicity, oxygen radicals, transi-function and pathology. TIPS 1993;14:13–20. tion metals, and disease. Biochem J 1984;19:1–14.

[25] Siliprandi R, Lipartiti M, Fadda E, Sautter J, Manev H. Activation [48] Freeman BA, Crapo JD. Free radicals and tissue injury. Lab Investof glutamate metabotropic receptor protects retina against N-methyl- 1982;47:412–26.D-aspartate toxicity. Eur J Pharmacol 1992;219:173–4. [49] Aust SD, Morehouse LA, Thomas CE. Role of metals in oxygen

[26] Lipartiti M, Fadda E, Savioni G, Siliprandi R, Sautter J, Arban R et radical reactions. J Free Radic Biol Med 1985;1:3–25.al. In rats, the metaboprobic receptor-triggered hippocampal neuro- [50] Harrison WW, Netsky MG, Brown MD. Trace elements in thenal damage is strain-dependent. Life Sci 1993;52:PL85–90. human brain: copper, iron, and magnesium. Clin Chim Acta

[27] Erecinska M, Silver IA. Relationships between ions and energy 1968;21:55–60.metabolism: cerebral calcium movements during ischaemia and [51] Crichton RR. Interactions between iron metabolism and oxygensubsequent recovery. Can J Physiol Pharmacol 1992;70:S190–3. activation. In: Oxygen free radicals and tissue damage, Ciba

[28] Chan PH, Kerlan R, Fishman RA. Reductions of gamma-amino- foundation symposium, vol. 65, New York: Excerpta Medica, 1979,1 1butyric acid and glutamate and (Na 1K )-ATPase activity in brain pp. 57–66.

slices and synaptosomes by arachidonic acid. J Neurochem [52] Thomas CE, Morehouse LA, Aust SD. Ferritin and superoxide1983;40:309–15. dependent lipid peroxidation. J Biol Chem 1985;260:3275–81.

[29] Braughler JM. Lipid peroxidation-induced inhibition of gamma- [53] Beckman JS. The double-edged role of nitric oxide in brain functionaminobutyric acid uptake in rat brain synaptosomes: protection by and superoxide-mediated injury. J Dev Physiol 1991;15:53–9.glucocorticoids. J Neurochem 1985;44:1282–6. [54] Samdani AF, Dawson TM, Dawson VL. Nitric oxide synthase in

[30] Ames III A. Earliest irreversible changes during ischemia. Am J models of focal ischemia. Stroke 1997;28:1283–8.Emerg Med 1983;2:139–43. [55] Tanaka K, Shirai T, Nagata K, Dembo T, Fukuuchi Y. Immuno-

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 23

histochemical detection of nitrotyrosine in postischemic cerebral [76] Zaleska MM, Wilson DF. Lipid hydroperoxides inhibit reacylationcortex in gerbil. Neurosci Lett 1997;235:85–8. of phospholipids in neuronal membranes. J Neurochem

1989;52:255–60.[56] Kogure K, Watson BD, Busto R, Abe K. Potentiation of lipidperoxides by ischemia in rat brain. Neurochem Res 1982;7:437–54. [77] O’Neil BJ, McKeown TR, DeGracia DJ, Alousi SS, Rafols JA,

White BC. Cell death, calcium mobilization, and immunostaining[57] Watson BD, Busto R, Goldberg WJ, Santiso M, Yoshida S, Ginsbergfor phosphorylated eukaryotic initiation factor 2-alpha (eIF2a) inMD. Lipid peroxidation in vivo induced by reversible globalneuronally-differentiated NB-104 cells: arachidonate and radical-ischemia in rat brain. J Neurochem 1984;42:268–74.mediated injury mechanisms. Resuscitation 1999;41:71–83.[58] Bromont C, Marie C, Bralet J. Increased lipid peroxidation in

[78] Sterz F, Leonov Y, Safar P, Radovsky A, Shearman G. No improvedvulnerable brain regions after transient forebrain ischemia in rats.outcome after prolonged cardiac arrest and treatment with excitatoryStroke 1989;20:918–24.neurotransmitter receptor blocker MK-801 in dogs. Ann Emerg Med[59] Sakamoto A, Ohnishi ST, Ohnishi T, Ogawa R. Relationship1989;18:456.between free radical production and lipid peroxidation during

[79] Pellegrini-Giampietro DE, Cherici G, Alesiani M, Carla V, Moroniischemia-reperfusion injury in rat brain. Brain Res 1991;554:186–F. Excitatory amino acid release and free radical formation may92.cooperate in the genesis of ischemia-induced neuronal damage. J[60] Rosenthal RE, Chanderbhan R, Marshall G, Fiskum G. PreventionNeurosci 1990;10:1035–41.of post-ischemic brain lipid conjugated diene production and

[80] Kumar K, White BC, Krause GS, Indrieri RJ, Evans AT, Hoehner TJneurological injury by hydroxyethyl starch-conjugated deferox-et al. A quantitative morphological assessment of the effect ofamine. Free Radic Biol Med 1992;12:29–33.lidoflazine and deferoxamine therapy for global brain ischemia.[61] Janero DR. Malondialdehyde and thiobarbituric acid reactivity asNeurol Res 1988;10:136–40.diagnostic indices of lipid peroxidation and peroxidative tissue

[81] Rosenthal RE, Chanderbhan R, Marshall G, Fiskum G. Preventioninjury. Free Radic Biol Med 1990;9:515–40.of post-ischemic brain lipid conjugated diene production and[62] Kruman I, Bruce-Keller AJ, Bredesen D, Wang G, Mattson MP.neurological injury by hydroxyethyl starch-conjugated deferox-Evidence that 4-hydroxynonenal mediates oxidative stress-inducedamine. Free Radic Biol Med 1992;12:29–33.neuronal apoptosis. J Neurosci 1999;17:5089–100.

[82] Natale JE, Schott RJ, Hall ED, Braughler JM, D’Alecy LG. Effect[63] Lukacova N, Gottlieb M, Marsala J. Lipid peroxidation and phos-of the aminosteroid U74006F after cardiopulmonary arrest in dogs.pholipid composition in rat brain regions after ischemia and in earlyStroke 1988;19:1371–8.reperfusion periods. Arch Ital Biol 1998;136:167–80.

[83] Sterz F, Safar P, Johnson DW, Oku KI, Tisherman SA. Effects of[64] White BC, Krause GS, Nayini NR, Aust SD. Effect of deferoxamineU74006F on multifocal cerebral blood flow and metabolism afteror superoxide dismutase therapy on biochemical markers of neuro-cardiac arrest in dogs. Stroke 1991;22:889–95.nal injury at 8 hours of reperfusion following a 15 minute cardiac

arrest in dogs. Am J Emerg Med 1988;6:569–76. [84] Skjaerlund JM, Krause GS, O’Neil BJ, White BC. The effect of[65] White BC, Rafols JA, DeGracia DJ, Skjaerlund JM, Krause GS. EMHP on post-cardiac arrest survival of rats. Resuscitation

Fluorescent histochemical localization of lipid peroxidation during 1991;22:139–49.brain reperfusion. Acta Neuropathol (Berlin) 1993;86:1–9. [85] Pierucci O, Rickert M. Duplication of E. coli during inhibition of

[66] Hall ED, Oostveen JA, Andrus PK, Anderson DK, Thomas CE. net phospholipid synthesis. J Bacteriol 1985;162:374–82.´ ` ´ `Immunocytochemical method for investigating in vivo neuronal [86] Joseleau-Petit D, Kepes F, Peutat L, D’Ari R, Kepes A. DNA

oxygen radical-induced lipid peroxidation. J Neurosci Methods replication initiation, doubling of rate of phospholipid synthesis, and1997;76:115–22. cell division in E. coli. J Bacteriol 1987;169:3701–6.

[67] Yoshino H, Hattori N, Urabe T, Uchida K, Tanaka M, Mizuno Y. [87] Loewy B, Marczynski GT, Dingwall A, Shapiro L. RegulatoryPostischemic accumulation of lipid peroxidation products in the rat interactions between phospholipid synthesis and DNA replication inbrain: immunohistochemical detection of 4-hydroxy-2-nonenal Caulobacter crescentus. J Bacteriol 1990;172:5523–30.modified proteins. Brain Res 1997;767:81–6. [88] Contreras IR, Bender RA, Mansour J, Henry S, Shapiro L.

[68] Farquhar MG. Traffic of products and membranes through the Golgi Caulobacter cresentus mutant defective in membrane phospholipidcomplex. In: Silverstein KSC, editor, Transport of macromolecules synthesis. J Bacteriol 1979;140:612–9.in cellular systems, Berlin: Dahlem Konferenzen, 1978, pp. 341–62. [89] Cooper RA, Ip SHC, Cassileth PA, Kuo AL. Inhibition of sterol and

[69] Voelker DR. Lipid transport pathways in mammalian cells. Ex- phospholipid synthesis in HL-60 promyelocytic leukemia cells byperientia 1990;46:569–79. inducers of myeloid differentiation. Cancer Res 1981;41:1847–52.

[70] Sun D, Gilboe DD. Ischemia-induced changes in cerebral mito- [90] Siperstein MD. Role of cholesterogenesis and isoprenoid synthesischondrial free fatty acids, phospholipids, and respiration in the rat. J in DNA replication and cell growth. J Lipid Res 1984;25:1462–8.Neurochem 1994;62:1921–8. [91] Benedetti A, Malvaldi G, Fulceri R, Comporti M. Loss of lipid

[71] Barker MO, Brin M. Mechanisms of lipid peroxidation in erythro- peroxidation as a histochemical marker for preneoplastic hepato-cytes of vitamin E deficient rats and in phospholipid model systems. cellular foci of rats. Cancer Res 1984;44:5712–7.Arch Biochem Biophys 1975;166:32–40. [92] Dianzani MU. Lipid peroxidation and cancer: a critical reconsidera-

[72] Sevanian A, Muakkassah-Kelly SF, Montestruque S. The influence tion. Tumori 1989;75:351–7.of phospholipase A2 and glutathione peroxidase on the elimination [93] O’Neil BJ, Alousi SS, White BC, Rafols JA. Ultrastructural conse-of membrane lipid peroxides. Arch Biochem Biophys quences of radical damage before and after differentiation of1983;223:441–52. neuroblastoma-104 cells. Acta Neuropathol (Berlin) 1996;92:75–89.

[73] Au AM, Chan PH, Fishman RA. Stimulation of phospholipase A2 [94] Guroff G. A neutral calcium-activated protease from the solubleactivity by oxygen-derived free radicals in isolated brain capillaries. fraction of rat brain. J Biol Chem 1984;239:149–55.J Cell Biochem 1985;27:449–53. [95] Goll DE, Kleese WC, Okitani A, Kumamoto T, Cong J, Kapprell

21[74] Chan PH, Yurko M, Fishman RA. Phospholipid degradation and H-P. Historical background and current status of the Ca -dependentcellular edema induced by free radicals in brain cortical slices. J proteinase system. In: Mellgren RL, Murachi T, editors, IntracellularNeurochem 1982;38:525–31. calcium-dependent proteolysis, Boca Raton: CRC Press, 1990, pp.

[75] Das DK, Engelman RM, Rousou JA, Breyer RH, Otani H, 3–24.Lemeshow S. Role of membrane phospholipids in myocardial injury [96] Croall DE, DeMartino GN. Calcium-activated neutral proteaseinduced by ischemia and reperfusion. Am J Physiol 1986;251:H71– (Calpain) system: structure, function, and regulation. Physiol Rev9. 1991;3:813–47.

24 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

[97] Goll DE, Thompson VF, Taylor RG, Zalewska T. Is calpain activity [118] Cong J-Y, Goll DE, Peterson AM, Kapprell H-P. The role of21regulated by membranes and autolysis or by calcium and calpastatin. autolysis in activity of the Ca -dependent proteinases (m-calpain

BioEssays 1992;14:549–56. and m-calpain). J Biol Chem 1989;264:10096–103.[98] Siman R, Gall C, Perlmutter L, Christian C, Baudry M, Lynch G. [119] Kuboki M, Ishii H, Kazamz M. Procalpain is activated on the

Distribution of calpain I, an enzyme associated with degenerative plasma membrane and the calpain acts on the membrane. Biochimactivity in rat brain. Brain Res 1985;347:399–403. Biophys Acta 1987;929:164–72.

[99] Hamakubo TR, Kannagi R, Murachi T, Matus A. Distribution of [120] Saido TC, Shibata M, Takenawa T, Murofushi H, Suzuki K.calpains I and II in rat brain. J Neurosci 1986;6:3103–11. Positive regulation of m-calpain action by polyphosphoinositides. J

Biol Chem 1992;267:24585–90.[100] Fukuda T, Adachi E, Kawashamia S, Yoshiya I, Hashimoto PH.Immunohistochemical distribution of calcium-activated neutral [121] Neumar RW, Hagle SM, DeGracia DJ, Krause GS, White BC.proteinases and endogenous CANP inhibitor in the rabbit hip- Brain m-calpain autolysis during global cerebral ischemia. Jpocampus. J Comp Neurol 1990;302:100–9. Neurochem 1996;66:421–4.

[101] Kamakura K, Ishiura S, Imajoh S, Nagata N, Sugita H. Distribution [122] Ruiz-Vela A, Gonzalez de Buitrago G, Martinez AC. Implication ofof calcium-activated neutral protease inhibitor in the central calpain in caspase activation during B cell clonal deletion. EMBO Jnervous system of the rat. J Neurosci Res 1992;31:543–8. 1999;18:4988–98.

[102] Song DK, Malmstrom T, Kater SB, Mykles DL. Calpain inhibitors [123] Kleihues P, Hossmann KA. Protein synthesis in the cat brain afterblock Ca(21)-induced suppression of neurite outgrowth in isolated prolonged cerebral ischemia. Brain Res 1971;35:409–18.hippocampal pyramidal neurons. J Neurosci Res 1994;39:474–81. [124] Cooper HK, Zalewska T, Kawakami S, Hossmann KA. The effect

[103] Seubert P, Lynch G. Plasticity and pathology: brain calpains as of ischemia and recirculation on protein synthesis in the rat brain. Jmodifiers of synaptic structure. In: Mellgren RL, Murachi T, Neurochem 1977;28:929–34.editors, Intracellular calcium-dependent proteolysis, Boca Raton: [125] Nowak Jr. TS, Fried RL, Lust D, Passonneau JV. Changes in brainCRC Press, 1990, pp. 251–64. energy metabolism and protein synthesis following transient bila-

[104] Siman R, Noszek JC, Kegerise C. Calpain I activation is spe- teral ischemia in the gerbil. J Neurochem 1985;44:487–94.cifically related to excitatory amino acid induction of hippocampal [126] Bodsch W, Takahashi K, Barbier A, Grosse Ophoff B, Hossmanndamage. J Neurosci 1989;9:1579–90. KA. Cerebral protein synthesis and ischemia. Prog Brain Res

[105] Schlaepfer WW, Zimmerman U-JP. Mechanisms underlying the 1985;63:197–210.neuronal response to ischemic injury. Calcium-activated [127] Bodsch W, Barbier A, Oehmichen M, Ophoff BG, Hossmann KA.proteolysis of neurofilaments. Prog Brain Res 1985;63:185–96. Recovery of monkey brain after prolonged ischemia II. Protein

[106] Seubert P, Lee K, Lynch G. Ischemia triggers NMDA receptor- synthesis and morphologic alterations. J Cereb Blood Flow Metablinked cytoskeletal proteolysis in hippocampus. Brain Res 1986;6:22–33.1989;492:366–70. [128] Thilmann R, Xie Y, Kleihus P, Kiessling M. Persistent inhibition of

[107] Lee KS, Frank S, Vanderklish P, Arai A, Lynch G. Inhibition of protein synthesis precedes delayed neuronal death in post-ischemicproteolysis protects hippocampal neurons from ischemia. Proc Natl gerbil hippocampus. Acta Neuropathol (Berlin) 1986;71:88–93.Acad Sci USA 1991;88:7233–7. [129] Yoshidomi M, Hayashi T, Abe K, Kogure K. Effects of a new

[108] Rami A, Krieglstein J. Protective effects of calpain inhibitors calcium channel blocker, KB-2796, on protein synthesis of the CA1

against neuronal damage caused by cytotoxic hypoxia in vitro and pyramidal cell and delayed neuronal death following transientischemia in vivo. Brain Res 1993;609:67–70. forebrain ischemia. J Neurochem 1989;53:1589–94.

[109] Roberts-Lewis JM, Siman R. Spectrin proteolysis in the hippocam- [130] Araki T, Kato H, Inoue T, Kogure K. Regional impairment ofpus: a biochemical marker for neuronal injury and neuroprotection. protein synthesis following brief cerebral ischemia in the gerbil.Ann New York Acad Sci 1993;679:78–86. Acta Neuropathol 1990;79:501–5.

14[110] Saido TC, Yokota M, Nagao S, Yamaura I, Tani E, Tsuchiya T et [131] Widmann R, Kuroiwa T, Bonnekoh P, Hossmann KA. [ C]-al. Spatial resolution of fodrin proteolysis in postischemic brain. J Leucine incorporation into brain proteins in gerbils after transientBiol Chem 1993;268:25239–43. ischemia: relationship to selective vulnerability of hippocampus. J

[111] Saito KI, Elce JS, Hamos JE, Nixon RA. Widespread activation of Neurochem 1991;56:789–96.calcium-activated neutral proteinase (calpain) in the brain in [132] Pulsinelli WA, Brierley JB, Plum F. Temporal profile of neuronalAlzheimer disease: a potential molecular basis for neuronal de- damage in a model of transient forebrain ischemia. Ann Neurolgeneration. Proc Natl Acad Sci USA 1993;90:2628–32. 1982;11:491–8.

[112] Squier MKT, Miller ACK, Malkinson AM, Cohen JJ. Calpain [133] Furuta S, Ohta S, Hatakeyama T, Nakamura K, Sakaki S. Recoveryactivation in apoptosis. J Cell Physiol 1994;159:229–37. of protein synthesis in tolerance-induced hippocampal CA1 neu-

[113] Suzuki K. The structure of the calpains and the calpain gene. In: rons after transient forebrain ischemia. Acta Neuropathol (Berlin)Mellgren RL, Murachi T, editors, Intracellular calcium-dependent 1993;86:329–36.proteolysis, Boca Raton: CRC Press, 1990, pp. 25–35. [134] O’Neil BJ, Krause GS, White BC. Thymine glycols and pyrimidine

[114] Maki M, Hatanaka M, Takano E, Murachi T. Structure–function dimers in brain DNA during post-ischemic reperfusion. Resuscita-relationships of calpastatins. In: Mellgren RL, Murachi T, editors, tion 1991;21:41–55.Intracellular calcium-dependent proteolysis, Boca Raton: CRC [135] White BC, DeGracia DJ, Krause GS, Skjaerlund JM, O’Neil BJ,Press, 1990, pp. 37–54. Grossman LI. Brain DNA survives cardiac arrest and reperfusion.

[115] Suzuki K, Saido T, Hirai S. Modulation of cellular signals by Free Radic Biol Med 1991;10:125–35.calpain. Ann New York Acad Sci 1992;674:218–27. [136] White BC, Tribhuwan RC, Vander Lann DJ, DeGracia DJ, Krause

[116] Croall DE, DeMartino GN. Regulation of calcium-dependent GS, Grossman LI. Brain mitochondrial DNA is not damaged byprotease activity in vitro: clues for determining their physiological prolonged cardiac arrest or reperfusion. J Neurochemfunctions. In: Mellgren RL, Murachi T, editors, Intracellular 1992;58:1716–22.

¨calcium-dependent proteolysis, Boca Raton: CRC Press, 1990, pp. [137] Lindvall O, Ernfors P, Bengzon J, Kokaia Z, Smith ML, Siesjo BK103–14. et al. Differential regulation of mRNAs for nerve growth factor,

[117] Hayashi M, Inomata M, Saito Y, Ito H, Kawashima S. Activation brain-derived neurotrophic factor, and neurotrophin-3 in the adultof intracellular calcium-activated neutral proteinase in erythrocytes rat brain following cerebral ischemia and hypoglycemic coma. Procand its inhibition by exogenously added inhibitors. Biochim Natl Acad Sci USA 1992;89:648–52.Biophys Acta 1991;1094:249–56. [138] Xie Y, Herget T, Hallmayer J, Starzinski-Powitz A, Hossmann KA.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 25

Determination of RNA content in postischemic gerbil brain by in factor on the mRNA translocation system and transport of specificsitu hybridization. Metab Brain Dis 1989;4:239–51. poly-A(1) mRNA and poly-A(2) mRNA in isolated nuclei.

Biochemistry 1990;29:2368–78.[139] Nowak Jr. TS, Ikeda J, Nakajima T. 70 kDa heat shock protein andc-fos gene expression after transient ischemia. Stroke [159] Bu X, Hagedorn CH. Platelet-derived growth factor stimulates1990;21(Suppl III):107–11. phosphorylation of the 25 kDa mRNA cap binding protein (eIF-4E)

in human lung fibroblasts. FEBS Lett 1991;283:219–22.[140] Onodera H, Kogure K, Ono Y, Igarashi K, Kiyota Y, Nagaoka A.Proto-oncogene c-fos is transiently induced in the rat cerebral [160] Frederickson RM, Mushynski WE, Sonenberg N. Phosphorylationcortex after forebrain ischemia. Neurosci Lett 1989;98:101–4. of translation initiation factor eIF-4E is induced in a ras-dependent

manner during nerve growth factor-mediated PC12 cell differentia-[141] Kindy MS, Carney JP, Dempsy RJ, Carney JM. Ischemic inductiontion. Mol Cell Biol 1992;12:1239–47.of protooncogene expression in gerbil brain. J Mol Neurosci

1991;2:217–28. [161] Donaldson RW, Hagedorn CH, Cohen S. Epidermal growth factoror okadaic acid stimulates phosphorylation of eukaryotic initiation[142] Matsumoto K, Yamada K, Hayakawa T, Sakaguchi T, Mogami H.factor 4F. J Biol Chem 1991;266:3162–6.RNA synthesis and processing in the gerbil brain after transient

hindbrain ischaemia. Neurol Res 1990;12:45–8. [162] Bu X, Haas DW, Hagedorn CH. Novel phosphorylation sites ofeukaryotic initiation factor-4F and of the p25 and p220 subunits. J[143] Tiffany BR, White BC, Krause GS. Nuclear-envelope nucleosideBiol Chem 1993;268:4975–8.triphosphatase kinetics and mRNA transport following brain

ischemia and reperfusion. Ann Emerg Med 1995;25:809–17. [163] Morley SJ, Dever TE, Etchison DE, Traugh JA. Phosphorylation ofeIF-4F by protein kinase C or multipotential S6 kinase stimulates[144] Krause GS, DeGracia DJ, Skjaerlund JM, O’Neil BJ. Assessmentprotein synthesis at initiation. J Biol Chem 1991;266:4669–72.of free radical-induced damage in brain proteins after ischemia and

reperfusion. Resuscitation 1993;23:59–69. [164] Clemens MJ, Bushell M, Morley SJ. Degradation of eukaryoticpolypeptide chain initiation factor (eIF) 4G in response to induc-[145] Roberts S, Morelos BS. Role of ribonuclease action in phenyl-tion of apoptosis in human lymphoma cell lines. Oncogenealanine-induced disaggregation of rat cerebral polyribosomes. J1998;17:2921–31.Neurochem 1976;26:387–400.

[165] Neumar RW, DeGracia DJ, Konkoly LL, White BC, Krause GS.[146] DeGracia DJ, O’Neil BJ, Frisch C, Krause GS, Skjaerlund JM,Calpain I mediates eukaryotic initiation factor 4G degradationWhite BC et al. Studies of the protein synthesis system in the brainduring global brain ischemia. J Cereb Blood Flow Metabcortex during global ischemia and reperfusion. Resuscitation1998;18:876–81.1993;21:161–70.

[147] Nowak Jr. TS, Carty ER, Lust WD, Passonneau JV. An in vitro [166] DeGracia DJ, Sullivan JM, Neumar RW, Alousi SS, Hikade KR,amino acid incorporation method for assessing the status of in vivo Pittman JE et al. Effect of brain ischemia and reperfusion on theprotein synthesis. Anal Biochem 1984;136:285–92. localization of phosphorylated eukaryotic initiation factor 2a. J

[148] Raley-Susman KM, Lipton P. In vitro ischemia and protein Cereb Blood Flow Metab 1997;17:1291–302.synthesis in the rat hippocampal slice: the role of calcium and [167] Goldstein E, Owen C, White BC, Rafols JA. UltrastructuralNMDA receptor activation. Brain Res 1990;515:27–38. localization of phosphorylated eIF2a [eIF2a(P)] in rat dorsal

[149] Gaitero F, Limas GG, Mendez E, DeHaro C. Purification of a hippocampus during reperfusion. Acta Neuropathol (Berlin)novel heat-stable translational inhibitor from rabbit reticulocyte 1999;98:493–505.lysates. FEBS Lett 1988;236:479–83. [168] McKeown TR, Goldstein EN, Sullivan JM, Rafols JA, White BC,

[150] DeGracia DJ, Neumar RW, White BC, Krause GS. Global brain Krause GS. Nuclear localization and DNA binding properties ofischemia and reperfusion: modifications in eukaryotic initiation phosphorylated eIF2a in vulnerable hippocampal neurons duringfactors are associated with inhibition of translation initiation. J reperfusion. J Cereb Blood Flow Metab 1999;19(Suppl 1):S510.Neurochem 1996;67:2005–12. [169] Sullivan JM, Alousi SS, Hikade KR, Rafols JA, Krause GS, White

[151] Merrick WC. Mechanism and regulation of eukaryotic protein BC. Insulin induces dephosphorylation of eIF2a and restoressynthesis. Microbiol Rev 1992;56:291–315. protein synthesis in vulnerable hippocampal neurons following

[152] Rowlands AG, Panniers R, Henshaw E. The catalytic mechanism transient brain ischemia. J Cereb Blood Flow Metabof guanine nucleotide exchange factor action and competitive 1999;19:1010–9.inhibition by phosphorylated eukaryotic initiation factor 2. J Biol [170] Scorsone KA, Panniers R, Rowlands AG, Henshaw EC. Phos-Chem 1988;263:5526–33. phorylation of eukaryotic initiation factor 2 during physiological

`[153] Alcazar A, Martin ME, Soria E, Rodriguez S, Fando JL, Salinas M. stresses which affect protein synthesis. J Biol ChemPurification and characterization of guinine nucleotide-exchange 1987;262:14538–43.factor, eIF-2B, and p37 calmodulin-binding protein from calf brain. [171] Duncan R, Hershey JWB. Heat shock-induced translational altera-J Neurochem 1995;65:754–61. tions in HeLa cells. J Biol Chem 1984;259:11882–9.

[154] Martin ME, Alcazar A, Fando JL, Garcia AM, Salinas M. [172] Zapata JM, Maroto FG, Sierra JM. Inactivation of mRNA cap-Translation initiation factor eIF-2 subcellular levels and phos- binding protein complex in Drosophila melanogaster embryosphorylation in the developing rat brain. Neurosci Lett under heat shock. J Biol Chem 1991;266:16007–14.1993;156:109–12. [173] Lamphear BJ, Panniers R. Cap binding protein complex that

[155] Ray BM, Hershey JWB, Grifo JA, Merrick WC, Thach RE. Role of restores protein synthesis in heat-shocked Erlich cell lysatesmRNA competition in regulating translation. Further characteriza- contains highly phosphorylated eIF-4E. J Biol Chemtion of mRNA discriminatory initiation factors. Proc Natl Acad Sci 1990;265:5333–6.USA 1983;80:663–7. [174] Lamphear BJ, Panniers R. Heat shock impairs the interaction of

[156] Duncan R, Milburn SC, Hershey JWB. Regulated phosphorylation cap-binding protein complex with 59 mRNA cap. J Biol Chemand low abundance of HeLa cell initiation factor eIF-4F suggest a 1991;266:2789–94.role in translational control. Heat shock effects on eIF-4F. J Biol [175] Joshi-Barve S, DeBenedetti A, Rhoads RE. Preferential translationChem 1987;262:380–8. of heat shock mRNAs in HeLa cells deficient in protein synthesis

[157] Morley SJ, Traugh JA. Differential stimulation of initiation factors initiation factors eIF-4E and eIF-4g. J Biol Chem 1992;267:21038–eIF-4F, eIF-4B, eIF-3 and ribosomal protein S6 by insulin and 43.phorbal esters. J Biol Chem 1990;265:10611–6. [176] Nowak Jr. TS, Bond U, Schlesinger MJ. Heat shock RNA levels in

[158] Schroder HC, Wenger R, Ugarkovic D, Friese K, Bachmann M, brain and other tissues after hyperthermia and transient ischemia. JMuller WE. Differential effect of insulin and epidermal growth Neurochem 1990;54:451–8.

26 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

[177] Nowak Jr. TS. Protein synthesis and the heat shock/stress response [197] Newman GC, Hospod FE, Trowbridge SD, Motwani S, Liu Y.after ischemia. Cerebrovasc Brain Metab Rev 1990;2:345–66. Restoring adenine nucleotides in a brain slice model of cerebral

[178] Dever TE, Yang W, Astrom S, Bystrom AS, Hinnebusch AG. reperfusion. J Cereb Blood Flow Metab 1998;18:675–85.MetModulation of tRNA , eIF-2, and eIF-2B expression shows that [198] Hurst R, Schatz JR, Matts RL. Inhibition of rabbit reticulocytei

GCN4 translation is inversely coupled to the levels of eIF-2-GTP- lysate protein synthesis by heavy metal ions involves the phos-MetMet-tRNA ternary complexes. Mol Cell Biol 1995;15:6351–63. phorylation of the a-subunit of the eukaryotic initiation factor 2. Ji

[179] Gan W, Rhoads RE. Internal initiation of translation directed by the Biol Chem 1987;262:15939–45.59-untranslated region of the mRNA for eIF-4G, a factor involved [199] De Herreros AG, De Haro C, Ochoa S. Mechanism of activation ofin the picornovirus-induced switch from cap-dependent to internal the heme-stabilized translational inhibitor of reticulocyte lysates byinitiation. J Biol Chem 1996;27:623–6. calcium ions and phospholipid. Proc Natl Acad Sci USA

[180] Ohlmann T, Rau M, Pain VM, Morley SJ. The C-terminal domain 1985;82:3119–23.of eukaryotic protein synthesis initiation factor eIF-4G is sufficient [200] Pal JK, Chen JJ, London IM. Tissue distribution of heme-regulatedto support cap-independent translation in the absence of eIF-4E. eIF-2a kinase determined by monoclonal antibodies. BiochemistryEMBO J 1996;15:1371–82. 1991;30:2555–62.

[181] Coldwell MJ, Mitchell SA, Stonley M, MacFarlane M, Willis AE.[201] Haines GK, Ghadge G, Radosevich JA. Expression of p68 protein

Initiation of APAF-1 translation by internal ribosome entry.kinase as recognized by the monoclonal antibody TJ4C4 during

Oncogene 2000;19:899–905.human fetal development. Tumor Biol 1993;14:95–104.

[182] Stein I, Itin A, Einat P, Skaliter R, Grossman Z, Keshet E.[202] Thomas DC, Samuel CE. Mechanism of interferon action: charac-Translation of vascular endothelial growth factor mRNA by

terization of the intermolecular autophosphorylation of PKR, theinternal ribosome entry: implications for translation under hypoxia.interferon-inducible, RNA-dependent protein kinase. J VirolMol Cell Biol 1998;18:3112–9.1995;69:5195–8.[183] Vagner S, Touriol C, Galy B, Audigier S, Gensac MC, Amalric F

[203] Proud CG. PKR: a new name and new roles. TIBS 1995;20:241–6.et al. Translation of CUG- but not AUG-initiated forms of human[204] Petryshyn RA, Li J, Judware R. Activation of the dsRNA-depen-fibroblast growth factor-2 is activated in transformed and stressed

dent kinase. Prog Mol Subcell Biol 1994;14:1–14.cells. J Cell Biol 1996;135:1391–402.[205] Jagus R, Gray MM. Proteins that interact with PKR. Biochimie[184] Martin DP, Schmidt RE, DiStefano PS, Lowry OH, Carter JC,

1994;76:779–91.Johnson EM. Inhibitors of protein synthesis and RNA synthesis[206] Der SD, Yang YL, Weissmann C, Williams BRG. A double-prevent neuronal death caused by nerve growth factor deprivation.

stranded RNA-activated protein kinase-dependent pathway mediat-J Cell Biol 1988;106:829–44.ing stress-induced apoptosis. Proc Natl Acad Sci USA[185] Shigeno T, Yamasaki Y, Kato G, Kusaka K, Mima T, Takakura K1997;94:3279–83.et al. Reduction of delayed neuronal death by inhibition of protein

[207] Takizawa T, Fukuda R, Miyawaki T, Ohashi K, Nakanishi Y.synthesis. Neurosci Lett 1990;120:117–9.[186] Wek RC. eIF-2 kinases: regulators of general and gene-specific Activation of the apoptotic FAS antigen-encoding gene upon

translation initiation. TIBS 1994;19:491–6. influenza virus infection involving spontaneously produced b-[187] Samuel CE. The eIF-2a protein kinases, regulators of translation in interferon. Virology 1995;209:288–96.

eukaryotes from yeasts to humans. J Biol Chem 1993;268:7603–6. [208] Schwemmle M, Clemens MJ, Hilse K, Pfeifer K, Troster H, Muller[188] Harding HP, Zhang Y, Ron D. Protein translation and folding are WE et al. Localization of Epstein–Barr virus-encoded RNAs

coupled by an endoplasmic-reticulum-resident kinase. Nature EBER-1 and EBER-2 in interphase and mitotic Burkitt lymphoma1999;397:271–4. cells. Proc Natl Acad Sci USA 1992;89:10292–6.

[189] Santoyo J, Alcalde J, Mendez R, Pulido D, de Haro C. Cloning and [209] Jeffrey IW, Kadereit S, Meurs EF, Metzger T, Bachman M,characterization of a cDNA encoding a protein synthesis initiation Schwemmle M et al. Nuclear localization of the interferon-induc-factor-2a (eIF-2a) kinase from Drosophila melanogaster. J Biol ible protein kinase PKR in human cells and transfected mouseChem 1997;272:12544–50. cells. Exp Cell Res 1995;218:17–27.

[190] Sood R, Porter AC, Olsen D, Cavener DR, Wek RC. A mammalian [210] Bandyopadhyhay SK, Sen GC. Role of protein phosphorylation inhomologue of GCN2 protein kinase important for translational activation of interferon-stimulated gene factors. J Biol Chemcontrol by phosphorylation of eukaryotic initiation factor-2a. 1992;267:6389–95.Genetics 2000;154:787–801. [211] DeGracia DJ, Adamczyk A, Folbe AJ, Konkkoly LL, Pittman JE,

[191] Donze O, Picard D. HSP90 binds and regulates the ligand-induc- Neumar RW et al. Eukaryotic initiation factor 2a (eIF2a) kinaseible alpha subunit of eukaryotic translation initiation factor kinase and phosphatase activity during post-ischemic brain reperfusion.GCN2. Mol Cell Biol 1999;19:8422–32. Exp Neurol 1998;155:221–7.

[192] Xiao N, Calloway CW, Lipinski CA, Hicks SD, DeFranco DB. [212] Kaufman RJ. Stress signaling from the lumen of the endoplasmicGeldanamycin provides posttreatment protection against glutamate- reticulum: coordination of gene transcriptional and translationalinduced oxidative toxicity in a mouse hippocampal cell line. J controls. Genes Dev 1999;13:1211–33.Neurochem 1999;72:95–101. [213] Brostrom CO, Bocckino SB, Brostrom MA. Identification of a

21[193] Waskiewicz J, Molchanova L, Walajtys-Rode E, Rafalowska U. Ca requirement for protein synthesis in eukaryotic cells. J BiolHypoxia and ischemia modifies histamine metabolism and trans- Chem 1983;258:14390–9.port in brain synaptosomes. Resuscitation 1988;16:287–93. [214] Brostrom CO, Bocckino SB, Brostrom MA, Galuska EM. Regula-

[194] Karkeela JT. Critical evaluation of postmortem changes in human tion of protein synthesis in isolated hepatocytes by calcium-cisternal fluid. Enzymes, electrolytes, acid–base balance, glucose mobilizing hormones. Mol Pharmocol 1985;29:104–11.and glycolysis, free amino acids, and ammonia. Correlation to total [215] Chin KV, Cade C, Brostrom CO, Galuska EM, Brostrom MA.brain ischemia. J Forensic Sci 1993;38:603–16. Calcium-dependent regulation of protein synthesis at translational

[195] Kawamoto T, Ikeda Y, Teramoto A. Protective effect of L-histidine initiation in eukaryotic cells. J Biol Chem 1987;262:16509–14.(singlet oxygen scavenger) on transient forebrain ischemia in the [216] Brostrom CO, Chin KV, Wong WL, Cade C, Brostrom MA.rat. No To Shinkei 1997;49:612–8. Inhibition of translation initiation in eukaryotic cells by calcium

[196] Rolfes RJ, Hinnebusch AG. Translation of the yeast transcriptional ionophore. J Biol Chem 1989;264:1644–9.activator GCN4 is stimulated by purine limitation: implications for [217] Kumar RV, Wolfman A, Panniers R, Henshaw EC. Mechanism ofactivation of the protein kinase GCN2. Mol Cell Biol inhibition of polypeptide chain initiation in calcium-depleted Erlick1993;13:5099–111. ascites tumor cells. J Cell Biol 1989;108:2107–15.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 27

[218] Kimball SR, Jefferson LS. Regulation of protein synthesis by [238] Cohen P, Foulkes JG, Holmes CF, Tonks NK. Protein phosphatasemodulation of intracellular calcium in rat liver. Endocrinol Metab inhibitor-1 and inhibitor-2 from rabbit skeletal muscle. Methods1992;26:E958–64. Enzymol 1988;59:427–37.

[219] Prostko CR, Brostrom MA, Malara EM, Brostrom CO. Phos- [239] Foulkes JG, Jefferson LS, Cohen P. The hormonal control ofphorylation of eukaryotic initiation factor (eIF) 2 alpha and glycogen metabolism: dephosphorylation of protein phosphataseinhibition of eIF-2B in GH3 pituitary cells by perturbation of early inhibitor-1 in vivo in response to insulin. FEBS Lett 1980;112:21–protein processing that induce GRP78. J Biol Chem 4.1992;267:16751–4. [240] Chan CP, McNall SJ, Krebs EG, Fischer EH. Stimulation of protein

[220] Prostko CR, Brostrom MA, Brostrom CO. Reversible phosphoryla- phosphatase activity by insulin and growth factors in 3T3 cells.tion of eukaryotic initiation factor 2 alpha in response to endo- Proc Natl Acad Sci USA 1988;85:6257–61.plasmic reticular signaling. Mol Cell Biochem 1993;127/128:255– [241] Hemmings Jr. HC, Girault JA, Nairn AC, Bertuzzi G, Greengard P.65. Distribution of protein phosphatase inhibitor-1 in brain and

[221] Kimball SR, Antonetti DA, Brawley RM, Jefferson LS. Mechanism peripheral tissues of various species: comparison with DARPP-32.of inhibition of peptide chain initiation by amino acid deprivation J Neurochem 1992;59:1053–61.in perfused rat liver. J Biol Chem 1991;266:1969–76.

[242] Lowenstein PR, Shering AF, MacDougall LK, Cohen P. Immuno-[222] Cohen P. The structure and regulation of protein phosphatases.

localization of protein phosphatase inhibitor-1 in the cerebralAnnu Rev Biochem 1989;58:453–508.

cortex of the rat, cat, and ferret. Brain Res 1995;676:80–92.[223] Ingebritsen TS, Cohen P. The protein phosphatases involved in

[243] Voll CL, Auer RN. Insulin attenuates ischemic brain damagecellular regulation. 1. Classification and substrate specificities. Eurindependent of its hypoglycemic effect. J Cereb Blood Flow MetabJ Biochem 1983;132:255–61.1991;11:1006–14.[224] Redpath NT, Proud CG. Activity of protein phosphatases against

[244] Datta B, Chakrabarti D, Roy AL, Gupta NK. Roles of a 67-kDainitiation factor-2 and elongation factor-2. Biochem Jpolypeptide in reversal of protein synthesis inhibition in heme-1990;272:175–80.deficient reticulocyte lysate. Proc Natl Acad Sci USA[225] Ernst V, Levin DH, Foulkes JG, London IM. Effects of skeletal1988;85:3324–8.muscle protein phosphatase inhibitor-2 on protein synthesis and

[245] Chakraborty A, Saha D, Bose A, Chatterjee M, Gupta NK.protein phosphorylation in rabbit reticulocyte lysates. Proc NatlRegulation of eIF-2 a-subunit phosphorylation in reticulocyteAcad Sci USA 1982;79:7092–6.lysate. Biochemistry 1994;33:6700–6.[226] Foulkes JG, Ernst V, Levin DH. Separation and identification of

[246] Rehemtulla WS, Gupta NK, Kaufman RJ. A eukaryotic translationtype 1 and type 2 protein phosphatases from rabbit reticulocyteinitiation factor 2-associated 67 kDa glycoprotein partially reverseslysates. J Biol Chem 1983;258:1439–43.protein synthesis inhibition by activated double-stranded RNA-[227] Takizawa N, Mizuno Y, Ito Y, Kikuchi K. Tissue distribution ofdependent protein kinase in intact cells. Biochemistryisoforms of type-1 protein phosphatase (PP1) in mouse tissue and

its diabetic alteration. J Biochem 1994;116:411–5. 1996;35:8275–80.[228] Ouimet CC, da Cruz e Silva EF, Greengard P. The alpha and [247] Saha D, Wu S, Bose A, Chatterjee N, Chakraborty A, Chatterjee M

gamma 1 isoforms of protein phosphatase 1 are highly and et al. Viral infection II: hemin induces overexpression of p67 as itspecifically concentrated in dendritic spines. Proc Natl Acad Sci partially prevents appearance of an active p67-deglycosylase inUSA 1995;92:3396–400. baculovirus-infected insect cells. Arch Biochem Biophys

[229] daCruz e Silva EF, Fox CA, Ouimet CC, Gustafson E, Watson SJ, 1997;342:373–82.Greengard P. Differential expression of protein phosphatase 1 [248] Bose A, Saha D, Gupta NK. Viral infection I: regulation of proteinisoforms in mammalian brain. J Neurosci 1995;15:3375–89. synthesis during vaccinia viral infection of animal cells. Arch

[230] Lee EYC, Zhang L, Zhao S, Wei Q, Zhang J, Qi ZQ et al. Biochem Biophys 1997;342:362–72.Phosphorylase phosphatase: new horizons for an old enzyme. Front [249] Owen C, Lipinski C, Page A, White B, Sullivan J, DeGracia D etBiosci 1999;4:270–85. al. Characterization of the eIF2-associated protein p67 during brain

[231] He B, Gross M, Roizman B. The gamma(1)34.5 protein of herpes ischemia and reperfusion. In: Maturation phenomenon in cerebralsimplex virus 1 complexes with protein phosphatase 1alpha to ischemia, vol. IV, Berlin: Springer, 2000, in press.dephosphorylate the alpha subunit of the eukaryotic translation [250] Gerstenfeld L, Beldekas JC, Franzblau C, Sonenshein GE. Cell-initiation factor 2 and preclude the shutoff of protein synthesis by free translation of calf type III collagen: effect of magnesium ondouble-stranded RNA-activated protein kinase. Proc Natl Acad Sci ribosome movement during elongation. J Biol ChemUSA 1997;94:843–8. 1983;258:12058–63.

[232] Szyszka R, Kudlicki W, Kramer G, Hardesty B, Galabru J, [251] Peterrson I, Kurland CG. Ribosomal protein L7/L12 is required forHovanessian A. A type 1 phosphoprotein phosphatase active with optimal translation. Proc Natl Acad Sci USA 1980;77:4007–10.phosphorylated M 568,000 initiation factor 2 kinase. J Biol Chem [252] VanDuffel L, Peters B, Rombauts W. Isolation and characteriza-r

1989;264:3827–31. tions of active ribosomal subunits from human placenta. J Biochem[233] Hubbard MJ, Cohen P. On target with a new mechanism for the 1975;57:481–91.

regulation of protein phosphorylation. Trends Biochem Sci [253] Hata R, Macada K, Hermann D, Miles G, Hossmann KA.1993;18:172–7. Dynamics of regional brain metabolism and gene expression after

[234] Srinivasan M, Begum N. Regulation of protein phosphatase 1 and middle cerebral occlusion in mice. J Cereb Blood Flow Metab2A activities by insulin during myogenesis in rat skeletal muscle 2000;20:306–15.cells in culture. J Biol Chem 1994;269:12514–20. [254] Bredesen DE. Keeping neurons alive: the molecular control of

[235] Chen J, Martin BL, Brautigan DI. Regulation of protein serine- apoptosis. Neuroscientist 1996;2:211–6.threonine phosphatase type-2A by tyrosine phosphorylation. Sci- [255] Hale AJ, Smith CA, Sutherland LC, Stoneman VEA, Longthorneence 1992;257:1261–4. VL, Culhane AC et al. Apoptosis: molecular regulation of cell

[236] Katayose Y, Li M, Al-Murrani SWK, Shenolikar S, Damuni A. death. Eur J Biochem 1995;236:1–26.PP2A PP2AProtein phosphatase 2A inhibitors, I and I , associate with [256] Arends MJ, Morris RG, Wyllie AH. Apoptosis. The role of the1 2

and modify the substrate specificity of protein phosphatase 1. J endonuclease. Am J Pathol 1990;136:593–608.Biol Chem 2000;275:9209–14. [257] Jacobson MD, Burne JF, Raff MC. Programmed cell death and

[237] Oliver CJ, Shenolikar S. Physiologic importance of protein phos- Bcl-2 protection in the absence of a nucleus. EMBO Jphatase inhibitors. Front Biosci 1998;3:961–72. 1994;13:1899–910.

28 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

[258] Ellis RE, Jacobson DM, Horvitz HR. Genes required for the novel zinc finger protein that inhibits apoptosis. J Biol Chemengulfment of cell corpses during programmed cell death in C. 1995;270:12343–6.elegans. Genetics 1991;129:79–94. [278] DeValck D, Jin DY, Heyninck K, Van De Craen M, Contreras R,

[259] Nagata S, Golstein P. The Fas death factor. Science Fiers W et al. The zinc finger protein A20 interacts with a novel1995;267:1449–56. anti-apoptotic protein which is cleaved by specific caspases.

Oncogene 1999;18:4182–90.[260] Hu WH, Johnson H, Shu HB. Tumor necrosis factor-relatedapoptosis-inducing ligand receptors signal NF-kB and JNK activa- [279] Darville MI, Ho YS, Eizirik DL. NF-kB is required for cytokine-tion and apoptosis through distinct pathways. J Biol Chem induced manganese superoxide dismutase expression in insulin-1999;274:30603–10. producing cells. Endocrinology 2000;141:153–62.

[261] Ahmad M, Srinivasula SM, Wang L, Talanian RV, Litwack G, [280] Xu D, Bureau Y, McIntyre DC, Nicholson DW, Liston P, Zhu Y etFernandes-Alnemri T et al. CRADD, a novel human apoptotic al. Attenuation of ischemia-induced cellular and behavioral deficitsadaptor molecule for caspase-2 and FasL/ tumor necrosis factor by X chromosome-linked inhibitor of apoptosis protein overexpres-receptor-interacting protein RIP. Cancer Res 1997;57:615–9. sion in the rat hippocampus. J Neurosci 1999;19:5026–33.

[262] Muzio M, Salvesen GS, Dixit VM. FLICE-induced apoptosis in a [281] Li C, Ha T, Liu L, Browder W, Kao RL. Adenosine preventscell-free system. Cleavage of caspase zymogens. J Biol Chem activation of transcription factor NF-kB and enhances activator1997;272:2952–6. protein-1 binding activity in ischemic rat heart. Surgery

2000;127:161–9.[263] Zhou Q, Snipas S, Orth K, Muzio M, Dixit VM, Salvesen GS.Target protease specificity of the viral serpin CrmA. Analysis of [282] Clemens JA, Stephenson DT, Dixon EP, Smalstig EB, Mincy RE,five caspases. J Biol Chem 1997;272:7797–800. Rash KS et al. Global cerebral ischemia activates nuclear factor-

kappaB prior to evidence of DNA fragmentation. Brain Res Mol[264] Bertin J, Armstrong RC, Ottilie S, Martin DA, Wang Y, Banks S etBrain Res 1997;48:187–96.al. Death effector domain-containing herpesvirus and poxvirus

proteins inhibit both FAS- and TNFR1-induced apoptosis. Proc [283] Clemens JA, Stephenson DT, Yin T, Smalstig EB, Panetta JA,Natl Acad Sci USA 1997;94:1172–6. Little SP. Drug-induced neuroprotection from global ischemia is

associated with prevention of persistent but not transient activation[265] Kajstura J, Cheng W, Reiss K, Clark WA, Sonnenblick EH,of nuclear factor-kappaB. Stroke 1998;29:677–82.Krajewski S et al. Apoptotic and necrotic myocyte cell deaths are

independent contributing variables in infarct size in rats. Lab Invest [284] Botchkina GI, Geimonen E, Bilof ML, Villarreal O, Tracey KJ.1996;74:86–107. Loss of NF-kB activity during cerebral ischemia and TNF cytotox-

¨ icity. Mol Med 1999;5:372–81.[266] Bromme HJ, Holtz J. Apoptosis in the heart: when and why? MolCell Biochem 1996;163/164:261–75. [285] Zaifang Y, Zhou D, Bruce-Keller AJ, Kindy MS, Mattson MP.

[267] Matsuyama T, Hata R, Tagaya M, Yamamoto Y, Nakajima T, Lack of the p50 subunit of nuclear factor-kB increases theFuruyama J et al. Fas antigen mRNA induction in postischemic vulnerability of hippocampal neurons to excitotoxic injury. Jmurine brain. Brain Res 1994;657:342–6. Neurosci 1999;19:8856–65.

[268] Martin-Villalba A, Herr I, Jeremias I, Hahne M, Brandt R, Vogel J [286] Cain K, Bratton SB, Langlais C, Walker G, Brown DG, Sun XM etet al. CD95 ligand (Fas-L/APO-1L) and tumor necrosis factor- al. Apaf-1 oligomerizes into biologically active |700-kDa andrelated apoptosis-inducing ligand mediate ischemia-induced apop- inactive |1.4 MDa apoptosome complexes. J Biol Chemtosis in neurons. J Neurosci 1999;19:3809–17. 2000;275:6067–70.

[269] Herr I, Martin-Villalba A, Kurz E, Roncaioli P, Schenkel J, Cifone [287] Irmler M, Hofmann K, Vaux D, Tschopp J. Direct physicalMG et al. FK506 prevents stroke-induced generation of ceramide interaction between the C. elegans ‘death proteins’ CED-3 andand apoptosis signaling. Brain Res 1999;826:210–9. CED-4. FEBS Lett 1997;406:189–90.

[270] Roth W, Isenmann S, Naumann U, Kugler S, Bahr M, Dichgans J [288] James C, Gschmeissner S, Fraser A, Evan GI. CED-4 induceset al. Locoregional Apo2L/TRAIL eradicates intracranial human chromatin condensation in S. pombe and is inhibited by directmalignant glioma xenografts in athymic mice in the absence of physical interaction with CED-9. Curr Biol 1997;7:246–52.neurotoxicity. Biochem Biophys Res Commun 1999;265:479–83. [289] Spector MS, Desnoyers S, Hoeppner DJ, Hengartner MO. Inter-

[271] Zhang ZG, Chopp M, Goussev A, Power C. Tumor necrosis factor action between the C. elegans cell-death regulators CED-9 andreceptor expression following focal embolic cerebral ischemia in CED-4. Nature 1997;385:653–6.mice. J Cereb Blood Flow Metab 1997;17(Suppl 1):S531. [290] Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human

[272] Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter protein homologous to C. elegans CED-4, participates in cyto-MK et al. Altered neuronal and microglial responses to excitotoxic chrome-c-dependent activation of caspase 3. Cell 1997;90:405–13.and ischemic brain injury in mice lacking TNF receptors. Nat Med [291] Krajewski S, Krajewski M, Ellerby LM, Welsh K, Xie Z, Deveraux1996;2:788–94. QL et al. Release of caspase-9 from mitochondria during neuronal

[273] Reuther JY, Baldwin AS. Apoptosis promotes a caspase-induced apoptosis and cerebral ischemia. Proc Natl Acad Sci USAamino-terminal truncation of IkBa that functions as a stable 1999;96:5752–7.inhibitor of NF-kB. J Biol Chem 1999;274:20664–70. [292] Fujimura M, Morita-Furimura Y, Murakami K, Kawase M, Chan

[274] Natoli G, Costanzo A, Guido F, Moretti F, Bernardo A, Burgio VL PH. Cytosolic redistribution of cytochrome-c after transient focalet al. Nuclear factor kB-independent cytoprotective pathways cerebral ischemia in rats. J Cereb Blood Flow Metaboriginating at tumor necrosis factor receptor-associated factor 2. J 1998;18:1239–47.Biol Chem 1998;273:31262–72. [293] Nakatsuka H, Ohta S, Tanaka J, Toku K, Kumon Y, Maeda N et al.

[275] Chu ZL, McKinsey TA, Liu L, Gentry JJ, Malim MH, Ballard DW. Release of cytochrome-c from mitochondria to cytosol in gerbilSuppression of tumor necrosis factor-induced cell death by inhib- hippocampal CA1 neurons after transient forebrain ischemia. Brainitor of apoptosis c-IAP2 is under NF-kB control. Proc Natl Acad Res 1999;849:216–9.Sci USA 1997;94:10057–62. [294] Andreyev AY, Fiskum G. Cytochrome-c release from brain mito-

[276] Wang CY, Mayo MW, Korneluk RG, Goddel DV, Baldwin AS. chondria is independent of the mitochondrial permeability transi-NF-kB anti-apoptosis: induction of TRAF1 And TRAF2 and c- tion. FEBS Lett 1998;439:373–6.IAP1 and c-IAP2 to suppress caspase-8 activation. Science [295] Blomgren K, Hallin U, Puka-Sundvall M, Hagberg H. Caspase–1998;281:1680–3. calpain interaction and calpastatin as a stress protein in neonatal rat

[277] Sarma V, Lin Z, Clark L, Rust BM, Tewari M, Noelle RJ et al. hypoxia-ischemia. J Cereb Blood Flow Metab 1999;19(SupplActivation of the B-cell surface receptor CD40 induces A20, a 1):S49.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 29

[296] Datta SR, Brunet A, Greenberg ME. Cellular survival: a play in DE et al. Bcl-2 expression in neural cells blocks activation ofthree Akts. Genes Dev 1999;13:2905–27. ICE/CED-3 family proteases during apoptosis. J Neurosci

1996;16:5654–60.[297] Gillardon F, Botiger BW, Schmitz B, Hossmann KA. Activation ofCPP-32, an ICE-related protease, in hippocampal neurons follow- [315] Henderson S, Huen D, Rowe M, Dawson C, Johnson G, Rickinsoning ischemia and epilepsy. J Cereb Blood Flow Metab A. Epstein–Barr virus-encoded BHRF1 protein, a viral homolgue1997;17(Suppl 1):S445. of Bcl-2, protects human B cells from programmed cell death. Proc

Natl Acad Sci USA 1993;90:8479–83.[298] Martins LM, Kottke T, Mesner PW, Basi GS, Sinha S, Frigon Jr. N˜et al. Activation of multiple interleukin-1b converting enzyme [316] Lam M, Dubyak G, Chen L, Nunez G, Miesfeld RL, Distelhorst

homologues in cytosol and nuclei of HL-60 cells during etoposide- CW. Evidence that Bcl-2 represses apoptosis by regulating endo-21induced apoptosis. J Biol Chem 1997;272:7421–30. plasmic reticulum-associated Ca fluxes. Proc Natl Acad Sci USA

1994;91:6569–73.[299] Margolin N, Raybuck SA, Wilson KP, Chen W, Fox T, Gu Y et al.Substrate and inhibitor specificity of interleukin-1 beta-converting [317] Antonsson B, Conti F, Ciavatta AM, Montessuit S, Lewis S,enzyme and related caspases. J Biol Chem 1997;272:7223–8. Martinou I et al. Inhibition of Bax channel-forming activity by

Bcl-2. Science 1997;277:370–2.[300] Van de Craen M, Vandenabeele P, Declercq W, Van den Brande I,Van Loo G, Molemans F et al. Characterization of seven murine [318] Antonsson B, Montessuit S, Lauper S, Eskes R, Martinou JC. Baxcaspase family members. FEBS Lett 1997;403:61–9. oligomerization is required for channel-forming activity in lipo-

somes and to trigger cytochrome-c release from mitochondria.[301] Le Romancer M, Cosulich SC, Jackson SP, Clarke PR. CleavageBiochem J 2000;345(Pt 2):271–8.and inactivation of DNA-dependent protein kinase catalytic subunit

during apoptosis in Xenopus egg extracts. J Cell Sci 1996;109(Pt [319] Snider BJ, Gottron FJ, Choi DW. Apoptosis and necrosis in13):3121–7. cerebrovascular disease. Ann New York Acad Sci 1999;893:243–

53.[302] Rosenthal DS, Ding R, Simbulan-Rosenthal CMG, Cherney B,Vanek P, Samulson M. Detection of DNA breaks in apoptotic cells [320] Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serineutilizing the DNA binding domain of poly(ADP-ribose) polymerase phosphorylation of death agonist BAD in response to survivalwith fluorescence microscopy. Nucleic Acids Res 1997;15:1437– factor results in binding 14-3-3 not Bcl-Xl. Cell 1996;87:619–28.41. [321] Datta SR, Dudek H, Tao X, Masters S, Fu HS, Gotoh Y et al. Akt

[303] Whitacre CM, Hashimoto H, Tsai ML, Chatterjee S, Berger SJ, phosphorylation of Bad couples survival signals to the cell intrinsicBerger NA. Involvement of NAD-poly(ADP-ribose) metabolism in death machinery. Cell 1997;91:231–41.p53-regulation and its consequences. Cancer Res 1995;55:3697– [322] Kennedy SG, Kandel ES, Cross TK, Hay N. Akt /protein kinase B701. inhibits cell death by preventing the release of cytochrome c from

[304] Shah GM, Poirer D, Desnoyers S, Saint-Martin S, Hoflack JC, mitochondria. Mol Cell Biol 1999;19:5800–10.Rong P et al. Complete inhibition of poly(ADP-ribose) polymerase [323] Wang HG, Pathan N, Ethell IM, Krajewski S, Yamaguchi Y,

21activity prevents the recovery of C3H10T1/2 cells from oxidative Shibasaki F et al. Ca -induced apoptosis through calcineurinstress. Biochim Biophys Acta 1996;1312:1–7. dephosphorylation of Bad. Science 1999;284:339–43.

[305] Gaken JA, Tavassoli M, Gan SU,Vallian S, Giddings I, Darling DC [324] Srivastava SP, Kumar KU, Kaufman RJ. Phosphorylation ofet al. Efficient retroviral infection of mammalian cells is blocked eukaryotic translation initiation factor 2 mediates apoptosis inby inhibition of poly(ADP-ribose) polymerase activity. J Virol response to activation of the double-stranded RNA-dependent1996;70:3992–4000. protein kinase. J Biol Chem 1998;273:2416–23.

[306] Mashima T, Naito M, Noguchi K, Miller DK, Nicholson DW, [325] Gil J, Alcami J, Esteban M. Induction of apoptosis by double-Tsuruo T. Actin cleavage by CPP-32/apopain during the develop- stranded-RNA-dependent protein kinase (PKR) involves the alphament of apoptosis. Oncogene 1997;14:1007–12. subunit of eukaryotic translation initiation factor 2 and NF-kB.

[307] Kuida K, Zheng TS, Na S, Kuan C, Yang D, Karasuyama H et al. Mol Cell Biol 1999;19:4653–63.Decreased apoptosis in the brain and premature lethality in CPP32- [326] Kaufman RJ, Kumar KU, Wu S, Srivastava SP. Regulation of thedeficient mice. Nature 1996;384:368–72. double-stranded RNA-dependent protein kinase (PKR) and its role

[308] Ni B, Wu X, Du Y, Su Y, Hamilton-Byrd E, Rockey PK et al. in apoptosis. Translational control. Cold Spring Harbor Laborator-Cloning and expression of a rat brain interleukin-1b-converting ies, 1998:330.enzyme (ICE)-related protease (IRP) and its possible role in [327] Kohno K, Higuchi T, Ohta S, Kohno K, Kumon Y, Sakaki S.apoptosis of cultured cerebellar granule neurons. J Neurosci Neuroprotective nitric oxide synthase inhibitor reduces intracellular1997;17:1561–9. calcium accumulation following transient global ischemia in gerbil.

[309] Namura S, Zhu J, Fink K, Endres M, Srinivasan A, Tomaselli KJ et Neurosci Lett 1997;224:17–20.al. Activation and cleavage of caspase-3 in apoptosis induced by [328] Sharp AH, McPherson PS, Dawson TM, Aoki C, Campbell KP,experimental cerebral ischemia. J Neurosci 1998;18:3659–68. Snyder SH. Differential immunohistochemical localization of

21[310] Hara H, Friedlander RM, Gagliardini V, Ayata C, Fink K, Huang Z inositol 1,4,5-trisphosphate- and ryanodine-sensitive Ca releaseet al. Inhibition of interleukin 1b converting enzyme family channels in rat brain. J Neurosci 1993;13:3051–63.proteases reduces ischemic and excitotoxic neuronal damage. Proc [329] Uehara A, Yasukochi M, Imanaga I. Modulation of ryanodine

21Natl Acad Sci USA 1997;94:2007–12. binding to the cardiac Ca release channel by arachidonic acid. J[311] Troy CM, Stefanis L, Greene LA, Shelanski ML. Nedd2 is Mol Cell Cardiol 1996;18:43–51.

required for apoptosis after trophic factor withdrawal, but not [330] Marks AM. Intracellular calcium-release channels: regulators ofsuperoxide dismutase (SOD1) downregulation, in sympathetic cell life and death. Am J Physiol 1997;272:H597–605.neurons and PC12 cells. J Neurosci 1997;17:1911–8. [331] Marks AR. Cellular functions of immunophilins. Physiol Rev

[312] Allet B, Hochmann A, Martinou I, Berger A, Missotten M, 1996;76:631–49.Antonsson B et al. Dissecting processing and apoptotic activity of a [332] Cameron AM, Steiner JP, Sabatini DM, Kaplin AL, Walensky LD,cysteine protease by mutant analysis. J Cell Biol 1996;135:479–86. Snyder SH. Immunophilin FK506 binding protein associated with

[313] Gu Y, Sarnecki C, Aldape RA, Livingston DJ, Su MS. Cleavage of inositol 1,4,5-trisphosphate receptor modulates calcium flux. Procpoly(ADP-ribose) polymerase by interleukin-1 beta converting Natl Acad Sci USA 1995;92:1784–8.enzyme and its homologs TX and Nedd-2. J Biol Chem [333] Timermann AP, Wiederrecht G, Marcy A, Fleischer S. Characteri-1995;270:18715–8. zation of an exchange reaction between soluble FKBP-12 and the

[314] Srinivasan A, Foster LM, Testa MP, Ord T, Keane RW, Bredesen FKBP–ryanodine receptor complex. Modulation by FKBP mutants

30 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

deficient in peptidyl-prolyl isomerase activity. J Biol Chem of nitric oxide synthase and protects against glutamate neuro-1995;270:2451–9. toxicity. Proc Natl Acad Sci USA 1993;90:9808–12.

[334] Sharkey J, Butcher SP. Immunophilins mediate the neuroprotective [353] Wiederrecht G, Hong S, Chan HK, March A, Martin M, Calaycay Jeffects of FK506 in focal cerebral ischemia. Nature 1994;371:336– et al. Characterization of high molecular weight FK-506 binding9. activities reveals a novel FK-506 binding protein as well as a

protein complex. J Biol Chem 1992;267:21753–60.[335] Katayama Y, Kamlya T, Muramatsu H, Abe H, Terashi A.Immunosuppressant FK506 enhances immediate-early gene expres- [354] Steiner JP, Connolly MA, Valentine HL, Hamilton GS, Dawsonsion and prevents delayed neuronal death in the gerbil hippocam- TM, Hester L et al. Neurotrophic actions of nonimmunosuppres-pus. J Cereb Blood Flow Metab 1997;17(Suppl 1):S492. sive analogues of immunosuppressive drugs FK506, rapamycin and

cyclosporin A. Nat Med 1997;3:421–8.[336] Kamlya T, Katayama Y, Aoyama S, Muramatsu H, Abe H, TerashiA. Neuroprotective effects of immunosuppressant FK506 in focal [355] Toung TJ, Bhardwaj A, Dawson VL, Dawson TM, Traystman RJ,cerebral ischemia in the rat: the effect on cerebral infarction, brain Hurh PD. Neuroprotective FK506 does not alter in vivo nitricedema, immediate-early genes, and heat shock protein hsp72. J oxide production during ischemia and early reperfusion in rats.Cereb Blood Flow Metab 1997;17(Suppl 1):S372. Stroke 1999;30:1279–85.

[337] Thomson AW, Bonham CA, Zeevi A. Mode of action of tacrolimus [356] Schuler M, Bossy-Wetzel E, Goldstein JC, Fitzgerald P, Green DR.(FK506): molecular and cellular mechanisms. Ther Drug Monit p53 induces apoptosis by caspase activation through mitochondrial1995;17:584–91. cytochrome-c release. J Biol Chem 2000;275:7337–42.

[338] Bierer BE, Schreiber SL, Burakoff SJ. The effect of the immuno- [357] Larsson LG, Gray HE, Totterman T, Pettersson U, Nilsson K.suppressant FK-506 on alternate pathways of T cell activation. Eur Drastically increased expression of myc and fos proto-oncogenesJ Immunol 1991;21:439–45. during in vitro differentiation of chronic lymphocytic cells. Proc

Natl Acad Sci USA 1987;84:223–7.[339] Odom A, Del Poeta M, Perfect J, Jeitman J. The immunosuppres-sant FK506 and its nonimmunosuppressive analog L-685,818 are [358] Sheng M, Greenberg ME. The regulation and function of c-fos andtoxic to Cryptococcus neoformans by inhibition of a common other immediate early genes in the nervous system. Neurontarget protein. Antimicrob Agents Chemother 1997;41:156–61. 1990;4:477–85.

[340] Fischer S, Michnick S, Karplus M. A mechanism for rotamase [359] Angel P, Karin M. The role of Jun, Fos and the AP-1 complex incatalysis by the FK506 binding protein (FKBP). Biochemistry cell-proliferation and transformation. Biochim Biophys Acta1993;32:13830–7. 1991;1072:129–57.

[341] Galat A. Peptidylproline cis–trans-isomerases: immunophilins. Eur [360] Ransone LJ, Kerr LD, Schmitt MJ, Wamsley P, Verma IM. TheJ Biochem 1993;216:689–707. bZIP domains of Fos and Jun mediate a physical association with

[342] Montague JW, Hughes FM, Cidlowski JA. Native recombinant the TATA box-binding protein. Gene Expr 1993;3:37–48.cyclophilins A, B, and C degrade DNA independently of pep- [361] Uemura Y, Kowall NW, Beal MF. Global ischemia induces NMDAtidylprolyl cis–trans-isomerase activity. Potential roles of receptor-mediated c-fos expression in neurons resistant to injury incyclophilins in apoptosis. J Biol Chem 1997;272:6677–84. gerbil hippocampus. Brain Res 1991;542:343–7.

[343] Montague JW, Gaido ML, Frye C, Cidlowski JA. The calcium- [362] Wessel TC, Joh TH, Volpe BT. In situ hybridization analysis ofdependent nuclease from apoptotic rat thymocytes is homologous c-fos and c-jun expression in the rat brain following transientwith cyclophilin. Recombinant cyclophilins A, B, and C have forebrain ischemia. Brain Res 1991;567:231–40.nuclease activity. J Biol Chem 1994;169:18877–80. [363] Jorgensen MB, Deckert J, Wright DC, Gehlert DR. Delayed c-fos

[344] Wine RN, Ku WW, Li LH, Chapin RE. Cyclophilin A is present in proto-oncogene expression in the rat hippocampus induced byrat germ cells and is associated with spermatocyte apoptosis. Biol transient global ischemia: an in situ hybridization study. Brain ResReprod 1997;56:439–46. 1989;484:393–8.

[345] Fraser MJ, Tynan SJ, Papaioannou A, Ireland CM, Pittman SM. [364] Uemura Y, Kowall NW, Moskowitz MA. Focal ischemia in ratsEndo-exonuclease of human leukaemic cells: evidence for a role in causes time-dependent expression of c-Fos immunoreactivity inapoptosis. J Cell Sci 1996;109:2343–60. widespread regions of ipsilateral cortex. Brain Res 1991;552:99–

[346] Hughes FM, Cidlowski JA. Potassium is a critical regulator of 105.apoptotic enzymes in vitro and in vivo. Adv Enzyme Regul [365] Jorgensen MB, Johansen FF, Diemer NH. Post-ischemic and kanic1999;39:157–71. acid-induced c-Fos protein expression in the rat hippocampus. Acta

[347] Sakahira H, Enari M, Nagato S. Functional differences of two Neurol Scand 1991;84:352–6.forms of the inhibitor of caspase-activated DNAse, ICAD-L, and [366] Zhang Y, Widmayer MA, Zhang B, Cui JK, Baskin DS. Suppres-ICAD-S. J Biol Chem 1999;274:15740–4. sion of post-ischemic-induced FOS protein expression by an

[348] Tanveer A, Virji S, Andreeva L, Totty NF, Hsuan JJ, Ward JM et antisense oligonucleotide to c-fos mRNA leads to increased tissueal. Involvement of cyclophilin D in the activation of a mito- damage. Brain Res 1999;832:112–7.

21chondrial pore by Ca and oxidant stress. Eur J Biochem [367] Schiaffonati L, Rappocciolo E, Tacchini L, Cairo G, Bernelli-1996;238:166–72. Zazzera A. Reprogramming of gene expression in post-ischemic rat

[349] Andreeva L, Tanveer A, Crompton M. Evidence for involvement liver: induction of proto-oncogenes and hsp-70 gene family. J Cellof a membrane-associated cyclosporin-A binding protein in the Physiol 1990;143:79–87.

21Ca -activated inner membrane pore of heart mitochondria. Eur J [368] Pelham HRB. Speculations on the functions of the major heatBiochem 1995;230:1125–32. shock proteins and glucose regulated proteins. Cell 1986;46:959–

[350] Schweizer M, Schlegel J, Baumgartner D, Richter C. Sensitivity of 69.mitochondrial peptidyl-prolyl cis–trans isomerase, pyridine [369] Beckmann RP, Mizzen LA, Welch WJ. Interaction of HSP-70 with

21nucleotide hydrolysis, and Ca release to cyclosporin A and newly synthesized proteins: implications for protein folding andrelated compounds. Biochem Pharmacol 1993;45:641–6. assembly. Science 1990;248:850–4.

[351] Liu J, Albers MW, Wandless TJ, Luan S, Alberg DG, Belshaw PJ [370] Hightower LE. Heat shock, stress proteins, chaperones, andet al. Inhibition of T cell signaling by immunophilin–ligand proteotoxicity. Cell 1991;66:191–7.complexes correlates with loss of calcineurin phosphatase activity. [371] Schlesinger MJ. Heat shock proteins: the search for functions. JBiochemistry 1992;31:3896–901. Cell Biol 1986;103:321–5.

[352] Dawson TM, Steiner JP, Dawson VL, Dinerman JL, Uhl GR, [372] Deshaies RJ, Koch BD, Schekman R. The role of stress proteins inSnyder SH. Immunosuppressant FK506 enhances phosphorylation membrane biogenesis. TIBS 1988;13:384–8.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 31

[373] Burdon RH. Heat shock and heat shock proteins. J Biochem [395] Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen1986;240:313–24. AM et al. Localization and characterization of insulin receptors in

[374] Rordorf G, Koroshetz WJ, Bonventre JV. Heat shock protects rat brain and pituitary gland using in vitro autoradiography andcultured neurons from glutamate toxicity. Neuron 1991;7:1043–51. computerized densitometry. Endocrinology 1987;121:1562–70.

[375] Ting LP, Tu CL, Chou CK. Insulin-induced expression of human [396] Hill JM, Lesniack MA, Pert CB, Roth J. Autoradiographic localiza-heat shock protein gene hsp-70. J Biol Chem 1989;264:3404–8. tion of insulin receptors in rat brain: prominence in olfactory and

[376] Nowak Jr. TS. Localization of 70 kDa stress protein mRNA limbic areas. Neuroscience 1986;17:1127–38.induction in gerbil brain after ischemia. J Cereb Blood Flow Metab [397] Wanaka A, Johnson Jr. EM, Milbrandt J. Localization of FGF1991;11:432–9. receptor mRNA in the adult rat central nervous system by in situ

[377] Nowak Jr. TS. Synthesis of a stress protein following transient hybridization. Neuron 1990;5:267–81.ischemia in the gerbil. J Neurochem 1985;45:1635–41. [398] Cheng B, Mattson MP. NGF and b-FGF protect rat hippocampal

[378] Gonzalez MF, Lowenstein FD, Fernyak S, Hisanaga K, Simon R, and human cortical neurons against hypoglycemic damage bySharp FR. Induction of heat shock protein 72-like immunoreactivi- stabilizing calcium homeostasis. Neuron 1991;7:1031–41.ty in the hippocampal formation following transient global is- [399] Taniuchi M, Schweitzer JB, Johnson EM. Nerve growth factorchemia. Brain Res Bull 1991;26:241–50. receptor molecules in rat brain. Proc Natl Acad Sci USA

[379] Vass K, Welch WJ, Nowak Jr. TS. Localization of 70 kDa stress 1986;83:1950–4.protein induction in gerbil brain after ischemia. Acta Neuropathol [400] Koh S, Oyler GA, Higgins GA. Localization of nerve growth factor(Berlin) 1988;77:128–35. receptor messenger RNA and protein in the adult rat brain. Exp

[380] Higo H, Lee JY, Satow Y, Higo K. Elevated expression of proto- Neurol 1989;106:209–21.oncogenes accompany enhanced induction of heat-shock genes [401] Unsicker K, Flanders KC, Cissel DS, Lafyatis R, Sporn MB.after exposure of rat embryos in utero to ionizing radiation. Teratog Transforming growth factor beta isoforms in the adult rat centralCarcinog Mutag 1989;9:191–8. and peripheral nervous system. Neuroscience 1991;44:613–25.

[381] Privalle CT, Fridovich I. Induction of superoxide dismutase in E. [402] Marks JL, Porte Jr. D, Baskin DG. Localization of type I insulin-coli by heat shock. Proc Natl Acad Sci USA 1987;84:2723–6. like growth factor receptor messenger RNA in the adult rat brain

[382] Drummond IA, McClure SA, Poenie M, Tsien RY, Steinhardt RA. by in situ hybridization. Mol Endocrinol 1991;5:1158–68.Large changes in intracellular pH and calcium observed during [403] Knusel B, Hefti F. Trophic actions of IGF-I, IGF-II and insulin onheat shock are not responsible for the induction of heat shock cholinergic and dopaminergic brain neurons. Adv Exp Med Biolproteins in Drosophila melanogaster. Mol Cell Biol 1986;6:1767– 1991;293:351–60.75. [404] Lauterio TJ. Regulation and physiological function of insulin-like

[383] Urano F, Wang XZ, Bertolotti A, Zhang Y, Chung P, Harding HP et growth factors in the central nervous system. Adv Exp Med Biolal. Coupling of stress in the ER to activation of JNK protein 1991;293:419–30.kinases by transmembrane protein kinase IRE1. Science [405] Breese CR, D’Costa A, Booze RM, Sonntag WE. Distribution of2000;287:664–6. insulin-like growth factor 1 (IGF-1) and 2 (IGF-2) receptors in the

[384] Behrens A, Sibilia M, Wagner EF. Amino-terminal phosphorylation hippocampal formation of rats and mice. Adv Exp Med Biolof c-Jun regulates stress-induced apoptosis and cellular prolifer- 1991;293:449–58.ation. Nat Genet 1999;21:326–9. [406] Marks JL, King MG, Baskin DG. Localization of insulin and type

[385] Herdegen T, Claret FX, Kallunki T, Martin-Villalba A, Winter C, 1 IGF receptors in rat brain by in vitro autoradiography and in situHunter T et al. Lasting N-terminal phosphorylation of c-Jun and hybridization. Adv Exp Med Biol 1991;293:459–70.activation of c-Jun N-terminal kinases after neuronal injury. J [407] Plata-Salaman CR. Epidermal growth factor and the nervousNeurochem 1998;18:5124–35. system. Peptides 1991;12:653–63.

[386] Xanthoudakis S, Miao G, Wang F, Pan YCE, Curran T. Redox [408] Kiyota Y, Takami K, Iwane M, Shino A, Miyamoto M, Tsukuda Ractivation of Fos–Jun DNA binding is mediated by a DNA repair et al. Increase in basic fibroblast growth factor-like immuno-enzyme. EMBO J 1992;11:3323–35. reactivity in rat brain after forebrain ischemia. Brain Res

[387] Xanthoudakis S, Miao GG, Curran T. The redox and DNA-repair 1991;545:322–8.activities of Ref-1 are encoded by non-overlapping domains. Proc [409] Gluckman P, Klempt N, Guan J, Mallard C, Sirimanne E,Natl Acad Sci USA 1994;91:23–7. Dragunow M et al. A role for IGF-1 in the rescue of CNS neurons

[388] Zinszner H, Kuroda M, Wang XZ, Batchvarova N, Lightfoot RT, following hypoxic-ischemic injury. Biochem Biophys Res Com-Remotti H et al. CHOP is implicated in programmed cell death in mun 1992;182:593–9.response to impaired function of the endoplasmic reticulum. Genes [410] Varon S, Hagg T, Manthorpe M. Nerve growth factor in CNSDev 1998;12:982–95. repair and regeneration. Adv Exp Med Biol 1991;296:267–76.

[389] Ubeda M, Vallejo M, Habener JF. CHOP enhancement of gene [411] Mattson MP, Murrain M, Guthrie PB, Kater SB. Fibroblast growthtranscription by interactions with Jun/Fos AP-1 complex proteins. factor and glutamate: opposing roles in the generation and degene-Mol Cell Biol 1999;19:7589–99. ration of hippocampal neuroarchitecture. J Neurosci 1989;9:3728–

[390] White BC, Grossman LI, O’Neil BJ, DeGracia DJ, Neumar RW, 40.Rafols JA et al. Global brain ischemia and reperfusion. Ann Emerg [412] Shigeno T, Mima T, Takakura K, Graham DI, Kato G, HashimotoMed 1996;27:588–94. Y et al. Amelioration of delayed neuronal death in the hippocam-

[391] The control of cell proliferation. In: Watson JD, Hopkins NH, pus by nerve growth factor. J Neurosci 1991;11:2914–9.Roberts JW, Stietz JA, Weiner AM, editors. Molecular biology of [413] Voll CL, Auer RN. Post-ischemic seizures and necrotizing is-the gene. 4th ed. Menlo Park, CA: Benjamin/Cummings, chemic brain damage: neuroprotective effect of post-ischemic1987:962–92. diazepam and insulin. Neurology 1991;41:423–8.

[392] McKay R, Valtz N, Cunningham M, Hayes T. Mechanisms [414] LeMay DR, Gehua L, Zelenock GB, D’Alecy G. Insulin adminis-regulating cell number and type in the mammalian central nervous tration protects neurological function in cerebral ischemia in rats.system. Cold Spring Harbor Symp Quant Biol 1990;55:291–301. Stroke 1988;19:1411–9.

[393] Cattaneo E, McKay R. Proliferation and differentiation of neuronal [415] Standaert ML, Pollet RJ. Insulin-glycerolipid mediators and genestem cells regulated by nerve growth factor. Nature 1990;347:762– expression. FASEB J 1988;2:2453–61.5. [416] Farese RV, Konda TS, Davis JS, Standaert ML, Pellet RJ, Cooper

[394] Havarankova J, Roth J. Insulin receptors are widely distributed in DR. Insulin rapidly increases diacylglycerol by activating de novothe central nervous system of the rat. Nature 1978;272:827–9. phosphatidic acid synthesis. Science 1987;236:586–9.

32 B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33

[417] Yarden Y. Growth factor receptor tyrosine kinases. Annu Rev Inhibition of DNA synthesis by a farnesyltransferase inhibitors6kinvolves inhibition of the p70 pathway. J Biol ChemBiochem 1988;57:443–78.

1999;274:4743–8.[418] Pillion DJ, Kim SJ, Kim H, Meezon E. Insulin signal transduction:[436] DeLuca JP, Garnache AK, Rulfs J, Miller TB. Wortmannin inhibitsthe role of protein phosphorylation. Am J Med 1992;303:40–52.

insulin-stimulated activation of protein phosphatase 1 in rat[419] Sacks DB, Fujita-Yamaguchi Y, Gale RD, McDonald JM.cardiomyocytes. Am J Physiol 1999;276:H1520–6.Tyrosine-specific phosphorylation of calmodulin by the insulin

[437] Jiang K, Coppola D, Crespo NC, Nicosia SV, Hamilton AD, Sebtireceptor kinase purified from human placenta. Biochem JSM et al. The phosphoinositide 3-OH kinase /AKT2 pathway is a1989;263:803–12.critical target for farnesyltransferase inhibitor-induced apoptosis.[420] Laurino JP, Colca JR, Pearson JD, DeWald DB, McDonald JM.Mol Cell Biol 2000;20:139–48.The in vitro phosphorylation of calmodulin by the insulin receptor

[438] Heck S, Lezouale’h G, Engert S, Behl C. Insulin-like growthtyrosine kinase. Arch Biochem Biophys 1988;265:8–21.factor-1-mediated neuroprotection against oxidative stress is asso-[421] Moss AM, Unger JW, Moxley RT, Livingston JN. Location ofciated with activation of nuclear factor kB. J Biol Chemphosphotyrosine-containing proteins by immunocytochemistry in1999;274:9828–35.the rat forebrain corresponds to the distribution of the insulin

[439] Kaliman P, Canicio J, Testar X, Palacin M, Zorzano A. Insulin-likereceptor. Proc Natl Acad Sci USA 1990;87:4453–7.

growth factor-II, phosphatidylinositol 3-kinase, nuclear factor-kap-[422] Lee-Kwon W, Park D, Baskar PV, Kole S, Bernier M. Antiapop-

paB and inducible nitric-oxide synthase define a commontotic signaling by the insulin receptor in chinese hamster ovary

myogenic signaling pathway. J Biol Chem 1999;274:17437–44.cells. Biochemistry 1998;37:15747–57. [440] Philpott KL, McCarthy MJ, Klippel A, Rubia LL. Activated

[423] Bertrand F, Afti A, Cadoret A, L’Allemain G, Robin H, Lascols O phosphatidylinositol 3-kinase and Akt promote survival of superioret al. A role for nuclear factor kB in the antiapoptotic function of cervical neurons. J Cell Biol 1997;139:809–15.insulin. J Biol Chem 1998;273:2931–8. [441] Xue L, Murray JH, Tolkovsky AM. The Ras /phosphatidylinositol

[424] Kulik G, Klippel A, Weber MJ. Antiapoptotic signalling by the 3-kinase and Ras /ERK pathways function as independent survivalinsulin-like growth factor 1 receptor, phosphatidylinositol 3-kinase, modules each of which inhibits a distinct apoptotic signalingand Akt. Mol Cell Biol 1997;17:1595–606. pathway in sympathetic neurons. J Biol Chem 2000;275:8817–24.

[425] Parrizas M, Saltiel AR, LeRoith D. Insulin-like growth factor 1 [442] Dufner A, Andjelkovic M, Burgering BM, Hemmings BA, Thomasinhibits apoptosis using the phosphatidylinositol 39-kinase and G. Protein kinase B localization and activation differentially affectmitogen-activated protein kinase pathways. J Biol Chem S6 kinase 1 activity and eukaryotic translation initiation factor1997;272:154–61. 4E-binding protein 1 phosphorylation. Mol Cell Biol

[426] Bertrand F, Desbois-Mouthon C, Cadoret A, Prunier C, Robin H, 1999;19:4525–34.Capeau J et al. Insulin antiapoptotic signaling involves insulin [443] Staal SP. Molecular cloning of the akt oncogene and its humanactivation of the nuclear factor kB-dependent survival genes homologs Akt1 and Akt2 : amplification of Akt1 in a primaryencoding tumor necrosis factor receptor-associated factor 2 and human gastric adenocarcinoma. Proc Natl Acad Sci USAmanganese-superoxide dismutase. J Biol Chem 1999;274:30596– 1987;84:5034–7.602. [444] Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franks TF,

[427] Gonzalez-Zulueta M, Feldman AB, Klesse LJ, Kalb RG, Dillman Stanbridge E et al. Regulation of cell death protease caspase-9 byJF, Parada LF et al. Requirement for nitric oxide activation of phosphorylation. Science 1998;282:1318–21.

rasp21 /extracellular regulated kinase in neuronal ischemic pre- [445] Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS et al. Aktconditioning. Proc Natl Acad Sci 2000;97:436–41. promotes cell survival by phosphorylating and inhibiting a For-

[428] Teng KK, Esposito DK, Schwartz GD, Lander HM, Hempstead khead transcription factor. Cell 1999;96:857–68.BL. Activation of c-Ha-Ras by nitric oxide modulates survival [446] Biggs WH, Meisenhelder J, Hunter T, Cavener WK, Arden KC.responsiveness in neuronal PC12 cells. J Biol Chem Protein kinase B/Akt-mediated phosphorylation promotes nuclear1999;274:37315–20. exclusion of the winged helix transcription factor FKHR1. Proc

[429] Kim E, Ambroziak P, Otto JC, Taylor B, Ashby M, Shannon K et Natl Acad Sci USA 1999;96:7421–6.al. Disruption of the mouse Rce-1 gene results in defective ras [447] Ozes ON, Mayo ID, Gustin JA, Pfeiffer SR, Preiffer LM, Donnerprocessing and mislocalization of Ras within cells. J Biol Chem DB. NF-kB activation by tumor necrosis factor requires the Akt1999;274:8383–90. serine-threonine kinase. Nature 1999;401:82–5.

[430] Goalstone ML, Leitner JW, Wall K, Dolgonos L, Rother KL, Accili [448] Madrid LV, Wang CY, Guttridge DC, Schottelius AJ, Baldwin AS,D et al. Effect of insulin on farnesyltransferase. Specificity of Mayo MW. Akt suppresses apoptosis by stimulating the transacti-insulin action and potentiation of nuclear effects of insulin-like vation potential of the RelA/p65 subunit of NF-kB. Mol Cell Biolgrowth factor-1, epidermal growth factor, and platelet-derived 2000;20:1626–38.growth factor. J Biol Chem 1998;273:23892–6. [449] Mendez R, Myers MG, White MF, Rhoads RE. Stimulation of

[431] Nobutaka S, Urano J, Tamanoi F. Farnesyltransferase inhibitors protein synthesis, eukaryotic translation initiation factor 4E phos-induce cytochrome-c release and caspase 3 activation preferentially phorylation, and PHAS-1 phosphorylation by insulin requiresin transformed cells. Proc Natl Acad Sci USA 1998;95:15356–61. insulin receptor substrate 1 and phosphatidylinositol 3-kinase. Mol

[432] Strong JE, Coffey MC, Tang D, Sabina P, Lee PW. The molecular Cell Biol 1996;16:2857–64.basis of viral oncolysis: usurpation of the Ras signaling pathway by [450] Van Weeren PC, de Bruyn KM, de Vries-Smits AM, van Lint J,reovirus. EMBO J 1998;17:3351–62. Burgering BM. Essential role for protein kinase B (PKB) in

[433] Kawsome H, Papst P, Webb S, Keller GM, Johnson GL, Gelfand insulin-induced glycogen synthase kinase 3 inactivation. Charac-s6kEW et al. Targeted disruption of p70 defines its role in protein terization of dominant-negative PKB. Biol Chem 1998;273:13150–

synthesis and rapamycin sensitivity. Proc Natl Acad Sci USA 6.´1998;95:5033–8. [451] Quevedo C, Alcazar A, Salinas M. Two different signal transduc-

[434] Welsh GI, Miller CM, Loughlin AJ, Price NT, Proud CG. tion pathways are implicated in the regulation of initiation factorRegulation of eukaryotic initiation factor eIF2B: glycogen synthase 2B activity in insulin-like growth factor-1-stimulated neuronalkinase-3 phosphorylates a conserved serine which undergoes cells. J Biol Chem 2000;275:19192–7.dephosphorylation in response to insulin. FEBS Lett [452] Widmann C, Gibson S, Johnson GL. Caspase-dependent cleavage1998;421:125–30. of signaling proteins during apoptosis: a turn-off mechanism for

[435] Law BK, Norgaard P, Gnudi L, Kahn BB, Poulson HS, Moses HL. anti-apoptotic signals. J Biol Chem 1998;273:7141–7.

B.C. White et al. / Journal of the Neurological Sciences 179 (2000) 1 –33 33

[453] Drews G, Debuyser A, Nenquin M, Henquin JC. Galanin and [456] Iwahashi H, Morishita H, Ishii T, Sugata R, Kido R. Enhancementepinephrine act on distinct receptors to inhibit insulin release by by catechols of hydroxyl-radical formation in the presence of ferric

1the same mechanisms, including an increase in K permeability of ions and hydrogen peroxide. J Biochem 1989;105:429–34.the b-cell membrane. Endocrinology 1990;126:1646–53. [457] Tanaka M, Sotomatsu A, Kanai H, Hirai S. Dopa and dopamine

[454] Yu KT, Pessin JE, Czech MP. Regulation of insulin receptor kinase cause cultured neuronal death in the presence of iron. J Neurol Sciby multisite phosphorylation. Biochimie 1985;67:1081–90. 1991;101:198–203.

[455] Sotomatsu A, Nakano M, Hirai S. Phospholipid peroxidation [458] Tirosh A, Potashnik R, Bashan N, Rudich A. Oxidative stress31 21induced by the catechol–Fe (Cu ) complex: a possible mecha- disrupts insulin-induced cellular redistribution of insulin receptor

nism of nigrostriatal cell damage. Arch Biochem Biophys substrate-1 and phosphatidylinositol 3-kinase in 3T3-L11990;283:334–41. adipocytes. J Biol Chem 1999;274:10595–602.