Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jun 22.
Published in final edited form as: Prog Mol Biol Transl Sci. 2011;99:265–305. doi: 10.1016/B978-0-12-385504-6.00007-5

Regulation of Caspases in the Nervous System: Implications for Functions in Health and Disease

Carol M Troy *,, Nsikan Akpan *,, Ying Y Jean *,
PMCID: PMC9214546  NIHMSID: NIHMS1816844  PMID: 21238939

Caspases, initially identified as a family of proteases regulating cell death, have been found to have nonapoptotic functions as well. Some family members are critical for mediating programmed cell death in development. After development, caspases are downregulated in the nervous system, but continue to perform important nonapoptotic functions relevant for neurogenesis and synaptic plasticity. In neurodegenerative diseases, where aberrant neuronal death is an outstanding feature, there is an increase in caspase activity. The specific caspase death pathways leading to dysfunction and death have still not been fully clarified, despite the plethora of scientific literature addressing these issues. In this chapter, we will present the current knowledge of caspase activation and activity pathways, the current tools for examining caspases, and functions of caspases in the nervous system in health and in disease. Alzheimer’s Disease, the most common neurodegenerative disorder, and cerebral ischemia, the most common cause of neurologic death, are used to illustrate our current understanding of death signaling in neurodegenerative diseases. A better understanding of how caspases function in health and disease would provide appropriate specific targets for the development of therapeutic interventions for these diseases.

Life and death are exquisitely regulated at the cellular level from development through maturity. During development, neuronal death is the major factor shaping the nervous system. This death is mainly caspase-mediated apoptosis. Once the waves of developmental death have passed (death occurs at different times in different parts of the nervous system), there is downregulation of the death machinery, as the postmitotic neurons should live for the life of the organism. Aberrant neuronal death is a major part of neurodegenerative disorders, but there is still no clear understanding of the processes leading to the phenotypes of the various diseases. Even the type of death that occurs continues to be debated, whether it is apoptotic, necrotic, or autophagic, or some combination of these death mechanisms. Here, we will discuss the role that the caspases play in neuronal function, dysfunction, and death. First, we will discuss the regulation of caspase activation and activity. We will examine the current understanding of caspase function in developmental neuronal death and then illustrate the role of caspases in neuronal death in disease employing two diseases of neuronal loss, Alzheimer’s Disease (AD), which is the most common chronic neurodegenerative disorder, and cerebral ischemia/stroke, the third most common cause of death in Western society, which is an acute neuronal disorder with chronic sequelae.

I. The Caspase Family of Death Proteases

Since the seminal work on death pathways in the model organism Caenorhabditis elegans, studies of apoptosis have increased exponentially. While it is agreed that caspases are critical mediators of apoptosis, many details of the death pathways remain to be explicated. Caspases are a highly conserved family, from worms to humans. One major difference, however, is the number of caspases: in worms, there is one caspase, while in mammals, there are 13 (Fig. 1). This difference raises the question of whether individual mammalian caspases have unique functions. Despite the plethora of studies, this is still an open question. In terms of disease intervention, specific nonredundant functions of individual caspases would provide therapeutic targets. As we increase our understanding of how caspase activation and activity are regulated, we can more critically evaluate current studies and reevaluate former studies.

Fig. 1. Mammalian caspases.

Fig. 1.

Mammalian caspases are schematically represented and grouped by activity. Yellow lines indicate cleavage sites.

In 1992, the interleukin-1β cleaving enzyme (ICE),1,2 now known as caspase-1, was identified and within a few months, ced-3, an enzyme with significant homology to ICE, was identified in C. elegans and found to execute apoptosis.3 Many more mammalian homologs were identified in the ensuing years, and the term “caspase,” for cysteine dependent, aspartate-specific protease, was agreed upon for the mammalian family. The 13 different mammalian caspases can be divided either by structure or by putative actions. From a structural perspective, there are two general groups, caspases with short prodomains (caspases-3, -6, -7, and -14) and with long prodomains (caspases-1, -2, -4, -5, -8, -9, -10, -11, and -12). Caspase-14 is involved in keratinocyte differentiation and does not appear to have a function in the nervous system4 and will not be discussed further. The effectors of apoptosis are the caspases with short prodomains, caspases-3, -6, and -7. The caspases with long prodomains are further subdivided. Caspases-2, -8, -9, and -10 are initiators of apoptosis, although caspase-2 may act as both an initiator and effector. Caspases-1, -4, -5, and -11 process cytokines and contribute to inflammation: caspases-4 and -5 are found only in humans, while caspase-11 is present only in rodents. The inflammatory caspases can also lead to cell death by autocrine mechanisms. Thus, although these caspases may not be direct apoptosis initiators, their activity can lead to death. Caspase-12 is somewhat of an enigma. It may be involved in cytokine processing or in death, particularly in endoplasmic reticulum-associated death. Additionally, while the gene for caspase-12 is expressed in rodents and humans, the protein is expressed in rodents but only in a small number of humans.5 There are studies of caspase-12 that suggest that it acts as a dominant-negative regulator of caspase-1 activity and that the caspase-12 proteolytic activity results only in autoprocessing.6 Work also supports a role for caspase-12 in ER stress as an activator of matrix metalloprotease 3.7

A. Caspase Activation

Caspases are also grouped based on the mechanism of activation (Fig. 2). While the activation of effector caspases is dependent on cleavage at an aspartate residue in the intersubunit linker,8,9 the activation of initiator caspases is more complex and new studies are refining the activation mechanisms. As a generalization, caspases with long prodomains undergo dimerization with a structural change that provides for the formation of an active site.911 The longer intersubunit linker allows flexibility, leading to the formation of an active site. Most of the initiators exist as monomers in the zymogen form; caspase-2 zymogen is a dimer.12 Interaction of the specific adaptor protein with the prodomain leads to dimerization and activation. Once the zymogen is activated, there is limited proteolysis of the interdomain. For caspase-9, proteolysis of the interdomain is not required: uncleavable mutants retain activity.13 However, recent work on caspase-8 shows that both dimerization and cleavage are required for optimal activity.14 This finding suggests that there are subtle differences in activation requirements that have implications for measurements of activation/activity of each caspase.

Fig. 2. Activation mechanisms of effector and initiator caspases.

Fig. 2.

Activation models for initiator and effector caspases are shown. Blue indicates inactive caspase while orange denotes activated caspase. Some of the activated caspases are subject to regulation by IAPs.

The long prodomain caspases have unique activation platforms. The intrinsic pathway of apoptosis centers on the mitochondrial regulation of death (Fig. 3). The apoptosome,15 the caspase-9 activation platform, was the first mammalian activation platform described. Release of cytochrome c from mitochondria leads to an ATP-dependent recruitment of caspase-9 by the adaptor, Apaf-1. Caspase-9 then undergoes limited autocleavage to a p35 fragment, which can be inhibited by XIAP, X-linked inhibitor of apoptosis protein. As noted above, cleavage is not required for activation of caspase-9 and, since the autocleavage allows inhibition of caspase-9 activity, it can actually lead to decreased activity. Caspase-3 can also cleave caspase-9 to a p37 fragment; this cleavage is not an activation step but potentiates activation by preventing XIAP inhibition of caspase-9,16 as described below.

Fig. 3. Caspase regulation: intrinsic pathway.

Fig. 3.

The intrinsic death pathway is activated by permeabilization of the mitochondria, leading to the release of cytochrome c and formation of the apoptosome, the caspase-9 activation platform. Once activated, caspase-9 can cleave and activate caspases-3 and -7. Caspases-3, -7, and -9 are subject to inhibition by XIAP. XIAP can be inhibited by Diablo/SMAC or HtrA2/Omi which are released from permeabilized mitochondria.

The extrinsic death pathway is activated by ligand binding to death receptors (Fig. 4). The caspase-8 activation platform is the death-inducing signaling complex (DISC). Ligand binding to a cell surface death receptor leads to the recruitment of an adaptor protein, which recruits caspase-8. As an example, Fas ligand binds to the fas receptor to recruit FADD and subsequently caspase-8. Binding to FADD dimerizes caspase-8 and then autocleavage provides for complete activation.14,17 Cleavage, without dimerization, by another enzyme, an effector caspase or granzyme B, does not produce activation of caspase-8.14 Recent work suggests that TNF-α induces two different caspase-8 activation pathways, a RIPK1-dependent and a RIPK1-independent pathway.18 There is also phosphorylation-mediated regulation of caspase-8 activity.19

Fig. 4. Caspase regulation: extrinsic pathway.

Fig. 4.

The extrinsic pathway is activated when a ligand binds to a death receptor; binding of FasL to the Fas receptor is shown. This leads to recruitment of an adaptor protein (FADD), which recruits caspase-8, forming the DISC, leading to dimerization and activation of caspase-8. Caspase-8 then cleaves and activates effector caspases. FLIP inhibits the formation of active caspase-8 dimers.

The activation platform for caspase-2 is proposed to be the PIDDosome, a complex of PIDD, RAIDD, and caspase-2.20,21 However, two different lines of PIDD null mice appear to undergo caspase-2-dependent death, although caspase-2 activity was not measured in those studies.22,23 Studies of the PIDD complex in tumor cell lines showed that overexpression of PIDD induced RAIDD-dependent cell death24 and that PIDD function in those cells depends on the cleavage state of PIDD.25 PIDD autocleaves to PIDD-C and PIDD-CC. Partially cleaved PIDD, PIDD-C, complexes with NEMO and leads to NFκB activation and survival signaling. Further cleavage of PIDD produces a fragment, PIDD-CC, that, in the overexpression system, complexes with RAIDD and leads to cell death. There are three isoforms of PIDD26: isoform 1 contains full-length PIDD and can be processed to PIDD-C and PIDD-CC; isoform 2 has a deletion of 146 amino acids at the N-terminus and an 11 amino acid deletion at position aa580; isoform 3 has a 17 amino acid deletion at aa705. Isoform 2 cannot form the PIDD-CC fragment. Expression of these isoforms is tissue- and cell-type specific: isoform 2 is present mainly in transformed cell lines and in normal liver, pancreas, and leukocytes, while isoforms 1 and 3 are found in normal tissue with the exception of the skeletal muscle. Isoform 1 is also expressed in transformed cell lines and isoforms 1 and 2 are found in several tumors. Since isoform 2 cannot form PIDD-CC, it should not interact with RAIDD. It is possible that PIDD function is different in cells with isoform 2 rather than the isoforms that can form PIDD-CC. This hypothesis would have interesting implications for the regulation of caspase-2 function in the different cell types. Caspase-2 activity is also regulated by phosphorylation.2729

Caspase-1 is activated in the inflammasome.3032 Several inflammasomes have been identified, the IPAF (ICE-protease activating factor), the NLRP1 (NACHT-, LRR-, and pyrin-domain containing proteins), and the NLRP2/3 inflammasomes.3134 The basic assembly involves the aforementioned proteins (IPAF or NLRP), which contain pyrin domains, caspase-1, which contains a CARD domain, and an adaptor protein with a CARD domain, such as ASC (apoptosis-associated speck-like protein containing a CARD) or CARDINAL (CARD inhibitor of NFkB-activating ligands). Caspase-1 is incorporated by ASC into the inflammasome through its CARD domain.35,36 The inflammasomes are activated by receptor-mediated signaling through TLRs (toll-like receptors) or in the cytosol by the NLR family in response to stress signals.

B. Endogenous Inhibitors of Caspases, the IAP Family

The Inhibitor of Apoptosis Protein family is classified by the presence of BIR (baculovirus IAP repeat) domains. In mammals, there are eight IAPs (see Ref. 37 for a review). Of these, cIAP1, cIAP2, and XIAP have been shown to bind to caspases, but XIAP is the only one which inactivates caspases.38 Other IAPs do inhibit cell death but not by directly inhibiting caspase activity. XIAP binds to and inactivates active caspases-3 and -7 via the BIR2-linker region and binds to and inactivates caspase-9 via the BIR3 domain. cIAP1 and cIAP2 can also bind to active caspases-3 and -7 but lack the region necessary for inactivating the caspases.39 XIAP-BIR3 binds to the neoepitope of caspase-9 that is generated by autocleavage of caspase-9 at aa315. XIAP cannot bind to the capase-9 neoepitope generated by caspase-3 cleavage of caspase-9 at aa330. Thus, caspase-3 cleavage promotes activity of caspase-9 by preventing the inhibition of caspase-9 by XIAP. In a similar fashion, the p20 fragment of caspase-3 can be inhibited by XIAP, but the p17 fragment cannot be inhibited. The BIR3 domains of cIAP1 and cIAP2 contain only one of the four residues required to inactivate caspase-9.38 Caspase-8 also has an endogenous inhibitor, c-FLIP, which is caspase-8 without the active site.40 FLIP dimerizes with caspase-8, preventing its activation rather than inhibiting active caspase-8.

C. Endogenous Inhibitors of IAPs

There are several endogenous inhibitors of IAPs. Smac/Diablo41,42 and HtrA2/Omi43 are found in the mitochondria of healthy cells, and the N-terminal amino acids of these proteins form the IAP-binding motif (IBM). Mitochondrial permeabilization leads to release of these proteins, which then interfere with the IAP-inhibition of caspases, resulting in potentiation of cell death. Another endogenous inhibitor of IAPs is XAF1.44 In nonneuronal tissues, XAF1 functions as a tumor suppressor. Originally identified as a specific inhibitor of XIAP, it is now reported that XAF1 can also bind to cIAP1 and cIAP2.45 As the function of cIAP1/2 is not quite clear, it is also not clear what the function of XAF1 binding to these molecules would be.

D. Regulation of Mitochondria in Apoptosis

Mitochondrial outer membrane permeabilization (MOMP) allows for selective release of molecules that participate in death. These molecules include cytochrome c, Smac/Diablo, HtrA2/Omi, and AIF. The Bcl2 family of proteins participates in the regulation of MOMP.11 This multimembered family is identified by the presence of BH domains and has both antiapoptotic members, which contain 4 BH domains, and proapoptotic members, which contain either 3 BH domains or only a BH3 domain. Bcl-2 family proteins reside either in the mitochondrial outer membrane or in the cytoplasm of healthy cells.

E. Measurement of Caspase Activation/Activity

Much of the literature regarding caspases should be reevaluated with regard to what the measures of caspases used in a particular study actually showed. Multiple methods have been used to measure caspase activity and activation, including Western blotting, immunocytochemistry, synthetic peptide substrate/inhibitor assays, and fluorescent indicators. Western blotting with specific antibodies can be used to quantify changes in zymogen expression and reveal the appearance of cleavage fragments. The relevance of cleavage to activation state varies among the caspases, as noted above. Proteolytic processing of the zymogen is required for effector caspases (3, 6, and 7) and appearance of the p17/19 and p10/12 fragments indicates that these caspases have been activated. For the initiator caspases, dimerization is required and there is a variable requirement for autocleavage (see above), but appearance of cleaved fragments is not a reliable read-out for activation since it will not distinguish between a cleavage event in the process of activation and a cleavage event that occurred without prior dimerization.

An additional subtle point that is often missed is that caspase activation and activity are not equivalent: IAPs can inhibit the activity of activated caspases. This distinction is critically relevant from a functional perspective. Only an active caspase will enact death. Caspases-3, -7, or -9 can be inhibited by XIAP in situ. Lysis of the cells for Western blotting disrupts IAP-caspase complexes and the Western blot shows cleaved caspases. Thus, activated but inhibited caspases cannot be distinguished from active caspases using Western blotting of cell lysates. For initiator caspases, interpretation of Western blotting data is even more problematic. Both caspase-8 and caspase-9 undergo autoproteolysis when incorporated into their respective activation complexes, although the full-length caspases have activity as well.16,46,47 As stated before, caspase-8 requires further processing for optimal activity, while additional processing of caspase-9 does not substantially increase activity.14 Caspase-2 also undergoes autoproteolysis,12 but it has not yet been clear how much this proteolysis contributes to total caspase-2 activity.

Another frequently used technique is immunocytochemistry, using antibodies which detect either the zymogen and cleaved fragments or are specific for neoepitopes generated during cleavage of zymogens. This technique is excellent for studying changes in the subcellular localization of a caspase, and for appearance of neoepitopes of effector caspases, with the same caveats noted above for Western blotting: appearance of the neoepitope does not necessarily indicate that there is an active caspase. Many studies inaccurately use the appearance of cleaved caspase-3 as a measure of apoptosis but, without another measure, cleaved caspase-3 alone does not mean that the cell is dying, as the caspase can be inhibited by XIAP.

Synthetic peptide pseudosubstrate caspase inhibitors/substrates, such as DEVD or IETD, have been and continue to be used by many groups as a method of distinguishing the role of individual caspases, despite studies published over a decade ago showing that these reagents are not specific for individual caspases.48,49 More recently, a comprehensive study evaluated the specificity of these reagents and concluded that all of the so-called specific caspase inhibitors/substrates are best at detecting caspase-3 activity, and some do not even detect their putative targeted caspase.50 To evaluate data, it is also important to understand how these peptides inhibit caspases: the peptide binds to the active caspase and inhibits downstream events. For in vivo measures of caspase activity, fluorescent versions of these peptides are being utilized as substrates that fluoresce when bound to the active caspase. These peptides bind irreversibly to the active site and cause inhibition of downstream events. Thus, once the fluorescent peptide is added, caspase activity is inhibited. Any cellular events that occur subsequent to the addition of the fluorescent peptide are not the result of caspase activation and could be the result of caspase inhibition. Thus, data acquired using these reagents should be reevaluated.

There are a few methods available to show that a specific caspase is active. One of these utilizes a broad-spectrum caspase substrate/irreversible inhibitor linked to biotin, bVADfmk. This active site affinity ligand binds irreversibly to the active caspase in the cell and inhibits the caspase. Preloading cells with bVADfmk before initiation of death captures initiator caspases and addition of bVADfmk at the time of harvesting cells undergoing death detects active effector caspases.51 The biotinylated affinity ligand–caspase complex is isolated with streptavidin, run on SDS-PAGE, and the specific caspase bound is detected with Western blotting for individual caspases. Studies using this method show that the active initiator caspases isolated by the affinity ligand are uncleaved, supporting the proximity-induced dimerization model of activation (no cleavage necessary initially) for initiator caspases. Adaptation of this method to animal studies could enable the identification of which caspases are active in vivo. Groups are also developing specific fluorescent read-outs of caspase activity. For caspase-2, such a system has been used in cultured cells to show dimerization and activation of caspase-2 after heat shock.52 This system could be adapted for other initiator caspases.

Functional relevance for individual caspases can be obtained using molecular knockdown/knockout of individual caspases. Abrogation of death shows that specific caspases are critical for execution of the death pathway under study. Studies have used antisense and siRNA knockdown of individual caspases. The ability to use siRNA to knockdown caspase-2 has been questioned because of the lack of reproducibility of published work using siRNA.53 However, it appears that it may be the sequence used in the studies in question that is at fault, rather than the ability of siRNA to ablate expression of caspase-2, as siRNA have been successfully used to knockdown other caspases.

Caspase null mice have been valuable tools for studying caspases with a role in development, as discussed below. However, caspase null mice without profound phenotypes suggest either that these caspases do not have a major role in development or that there are compensatory changes in other caspases, which obscure the function of the targeted caspase. Use of conditional knockouts could address these issues.

F. Targets of Caspases

Caspases can cleave many proteins; cleavage is limited (cleavage at one or two sites) as illustrated by the activation of effector caspases by initiator caspases. There have been multiple reports of substrate cleavage changing the activity of the substrate: that is, converting a proapoptotic protein to an antiapoptotic one, or vice versa. Such events serve to amplify the signaling pathways leading to death or survival. Although predictions have been made, careful time courses of substrate cleavage events, combined with specific blockade of these events, will be required to determine the function of such cleavage in individual death pathways.

II. Caspases and Developmental Neuronal Death

During development, programmed cell death, which is mediated by caspases, is the major force that shapes the organism. This process is essential to the formation of a normal nervous system. Two general populations die during neuronal development: neuronal precursors and postmitotic neurons. Caspase-9 and the caspase-9 adapter, Apaf1, are essential for death of the neuronal precursors. Mice lacking either of these proteins have severe malformations of the nervous system.5457 The initial report of the caspase-3 null mice also showed a severe neuronal developmental phenotype, very similar to the caspase-9 null mice, with death of the mice soon after birth.58 Care must be taken when drawing conclusions, because changing the genetic background of the caspase-3 null mice from a mixed background to a C57/Black6 background modified the phenotype drastically59—on this background, neuronal development is grossly normal, and the mice are able to grow to adulthood and to breed. Thus, the dependence of neuronal developmental death on caspase-3 is unclear. It is also not clear that all neuronal developmental death is dependent only on the caspase-9 pathway. The timing of death varies among regions of the brain, from the early embryonic to the early postnatal periods.

Mice lacking many of the other caspases, such as caspase-1, -2, -6, -7, -11, or -12, do not have an obvious neuronal phenotype.5963 These caspases may play a role in regional pruning of neurons or in the plasticity of the nervous system, and be important in the maintenance of the mature nervous system. There is also an issue of compensatory changes in other caspases or caspase regulators, which can confound the simple interpretation of results from mouse knockout studies. This issue is a significant problem with apoptotic genes, where phenotypic selection in developing embryos can severely alter normal expression patterns, as has been shown in caspase-2, -3, and -9 null mice.63,64 Compensations have been shown at the level of expression of other caspases63 or of regulators of caspase activity.64 The relative expression of the caspases and their regulators is a major factor in the choice of execution pathway. It would be wise to keep in mind this potential complexity when evaluating data on caspase involvement in neurodegenerative diseases.

III. Nonapoptotic Caspase Function in the Nervous System

There is increasing evidence that there are nonapoptotic functions of caspases in the nervous system. These data are most compelling for a function for caspases in neurogenesis and synaptic activity.65 Studies of neurogenesis indicate that caspase-3 activity in neuronal progenitors facilitates neurogenesis.66 Inhibition of caspase activity also blocked neurite extension, suggesting a role for caspases in the morphologic changes that occur during neurogenesis. Differentiation of mouse neural stem cells was inhibited by inhibition of caspase activity, and caspase inhibition also blocked p53 phosphorylation and transcriptional activation.67 These data suggest a critical, nonapoptotic function of caspase-3 in neurogenesis.

Neuronal circuits are regulated by selective pruning of synapses, axons, and dendrites. In Drosophila, the initiator caspase Dronc is required for pruning.68 Studies of trophic factor deprivation-mediated death of cultured dorsal root ganglion neurons suggest that there is selective activation of caspase-6 in axons, which leads to axonal pruning but not to death.69 Caspases have also been implicated in neuronal plasticity. In vivo studies in the zebra finch showed that caspase-3 activity is required for the birds to learn a new song.70 The cleaved caspase-3 is in the synapses and is associated with XIAP, suggesting that XIAP modulates the activity of caspase-3, so that synapse remodeling occurs but not death. Studies using DEVDfmk in rats support a role for caspase activity in long-term potentiation71 and in active avoidance learning.72 It has been recently shown that caspase-3 activation is required for long-term depression. LTD was inhibited in caspase-3 null hippocampal slices and XIAP-BIR3, a specific inhibitor of caspase-9, also inhibited LTD.73 These nonapoptotic caspase functions also indicate why it is not appropriate to connote caspase-3 cleavage with apoptosis.

These nonapoptotic functions of caspases need to be better understood for the design of appropriate therapeutic interventions to treat diseases with aberrant caspase activation and neuronal death.

IV. Caspase Function in Neurodegenerative Diseases

The study of diseases requires the use of models of the disease. We will present two diseases that are among the most highly prevalent human neurological disorders of aging. We will consider what the appropriate models are and what the current understanding of the caspase pathways is in each disease.

A. Alzheimer’s Disease

Over the past century, the life span in industrialized countries has increased an average of 25 years. This increased life expectancy has shifted the average age of the population and more people are living beyond 80 years of age. With an increase in the aged population, there is an increase in the prevalence of diseases associated with aging. At age 65, about 2–3% of people have AD, but the incidence of AD doubles for every 5 years of age afterward, and approximately 50% of people over the age of 85 have AD. There are about 5.3 million cases of AD in the United States.74 As a result of increasing life expectancy and expanding population, the number of AD cases should triple over the next 40 years.74 AD is the most common form of dementia among the elderly population and represents the fourth leading cause of death in industrialized countries.75,76

AD was characterized by Alois Alzheimer in 1907 as a progressive impairment of cognitive functions.77 The most frequent symptom is gradual loss of episodic memory, which is memory of recent events. With disease progression, impairment is found in language, cognition, reason, and temporal-spatial orientation, together with other changes in mood and personality. After the initial clinical symptoms, there is continuous and progressive decline leading to mutism, vegetative state, inanition, and finally death. Survival ranges from 5 to 20 years from detection of the first symptoms of the disease to death. The definitive diagnosis of AD is pathologic but biopsies are rarely done and tissue is usually obtained postmortem. Thus, the clinical diagnosis is of possible or probable AD. The two main histopathological hallmarks required for the definitive diagnosis of AD are senile plaques, extracellular congophyllic deposits composed of Aβ peptide, and neurofibrillary tangles (NFTs), intraneuronal filamentous aggregates composed of hyperphosphorylated tau proteins and paired helical filaments (PHF).78

Multiple factors have contributed to the lack of full understanding of the order of events in the development and progression of AD; and these factors include the insidious onset of clinical symptoms, the lack of a definitive clinical diagnosis, and the chronic nature of the disease. The progressive loss of synapses and neurons in limbic and cortical areas, translated functionally as a disconnection between different brain areas, is postulated to lead to the clinical symptoms.79,80 This initial disconnection does not seem to have repercussions on behavior for several decades, since the gradual loss of synapses and neurons seems to start 20–40 years before the manifestation of the first symptoms.81,82 Distribution of NFT correlates better than amyloid plaque burden with the severity of dementia and neuronal death.8387 But, as no studies of progressive pathologic analysis of single patients over time exist, all of the human data should be considered as correlative rather than definitive with regards to molecular causality. The use of imaging of amyloid plaques in AD patients may provide information about the progression of the disease, when correlated with symptoms.

Three genes have been linked to familial AD: amyloid precursor protein (APP),88 located on chromosome 21, and presenilin-1 (PS1)89 and presenilin-2 (PS2),90 located on chromosomes 14 and 1, respectively. About 85% of EOAD cases correspond to mutations in the PS1 gene, and mutations in PS2 or APP are much less frequent.91,92 There have been 157 mutations in PS1 and 10 in PS2 described worldwide. There are families with a clear genetic component that have not been associated with mutations in any of these three genes, which suggests that other genes may be involved.

1. Mouse Models in AD

Knowledge of the genetic mutations in familial AD enabled the development of mouse models expressing these human mutations in APP and PSEN1/2 as tools to study AD. The mouse models do exhibit some of the pathological hallmarks of AD, including amyloid deposits, abnormal tau hyperphosphorylation, and gliosis, and also display impairments of memory and learning.9399 The mice do not have NFTs or neuronal loss. Additional models were generated containing mutant APP and PS1 or both of these and mutant tau, in an attempt to provide a phenocopy for AD.100110 Since some of these models overexpress mutant genes that are not even found in AD cases, the results extracted from these models should be taken with caution.

Although these mice do not exhibit every single hallmark of the disease, they are a valuable tool for understanding the interaction between genetic factors able to modify the in vivo production and deposition of Aβ, identifying molecular targets, and evaluating new therapies designed to stop or slow down the pathological and clinical manifestations of this devastating disease. Some of the mouse models may represent good models of the early, preclinical disease. The models have been exploited to show how plaques develop in real time using 2-photon microscopy on live animals. Surprisingly, these studies revealed that individual plaques develop in several hours and then remain constant over months.111 This technique promises to be useful for examining pathways and structural changes.

2. In Vitro Models of AD

In vitro models utilizing primary neuronal cultures provide an opportunity to decipher molecular mechanisms of AD. These models have been instrumental in our understanding of how familial AD gene mutations alter Aβ metabolism, secretion, and/or degradation.112 Work using these models has shown that alterations in the γ-secretase complex produce an enhancement of APP processing113,114 and lead to an increase in the intracellular concentration of Aβ species.115 These modifications in the processing of APP alter the Aβ40/Aβ42 ratio, increasing the relative amount of Aβ42.116118 In vitro models have provided critical insights into where in the cell APP is processed and how Aβ is generated and transported within the cell.119

Arguably, the most relevant use of in vitro models is to identify the neurotoxic species triggering the progression of the disease. The lack of correlation between the amyloid plaque burden and the clinical progression of the disease has led to the ongoing debate about the role of amyoid in the etiology of AD. However, studies of soluble oligomeric Aβ species show that early memory impairment and synaptic dysfunction correlate better with the levels of oligomeric Aβ than with the fibrillar species.120 It is important to keep in mind that Aβ assemblies are highly dynamic and are in active equilibrium with the environment. This property makes it very difficult to ascribe specific toxic characteristics to a certain type of aggregation state.

However, several studies have examined the relationship between toxicity and aggregation. In 1998, the characterization of soluble, nonfibrillar oligomeric species (ADDL: Aβ derived diffusible ligands) and demonstration of their neurotoxic properties121 suggested that oligomers, not fibrils, were the toxic species. ADDLs caused neuronal death in primary hippocampal neuron cultures and inhibited long-term potentiation in cultured slices from rat brains.121,122 Another modification of the in vitro systems employs naturally produced, cell-derived Aβ oligomers123,124 that are toxic in cultures and inhibit LTP when injected into rat brain.124 The relevant size of the oligomers is a topic of debate, and oligomers as large as dodecamers have been isolated from human AD brains125 and from transgenic mice.126 These species are also toxic in primary neuronal cultures and impair LTP in hippocampal slices.127

3. Caspases in AD

a. Caspases in AD brains.

Neuronal loss is an outstanding feature of AD. A role for apoptosis in this neuronal loss is suggested by the increased expression of caspases and cleaved caspase substrates in postmortem AD brains.128,129 mRNA expression of caspases-1, -2, -3, -5, -6, -7, -8, and -9 is increased in the brain of AD patients compared to controls.130 Pyramidal neurons from vulnerable regions involved in the disease showed an increase in activated caspase-3 and -6.131,132 Negative correlations have been found between caspase-8 levels and age at disease onset and age at patient death, suggesting a role in disease regulation.133 Postmortem AD brain tissue showed extensive expression of cleaved caspase-8 (p18) in neurons and reactive astrocytes in the hippocampus and the entorhinal cortex134; the immunoreactivity of cleaved caspase-8 is colocalized in tangle-bearing and plaque-associated neurons. As noted above, it is not clear whether this is active caspase-8 or effector-processed (nonactive) caspase-8.

Synaptosomes prepared from AD brain frontal cortices showed an enrichment in caspase-9 compared to nondemented controls.135 While there is a great deal of correlative data showing that caspases are somehow involved in AD, there is not a great deal of specific data for individual caspases. Much of the data either measures changes in expression, which does not necessarily translate into a functional role for proteins that require activation, or utilizes the nonspecific peptide inhibitor/substrates to imply the involvement of specific caspases. The peptide inhibitor/substrate data can be viewed as indications of caspase function, but these data certainly cannot be construed as implicating individual caspases in AD dysfunction pathways.

b. Caspase cleavage of APP.

APP contains caspase cleavage sites and it has been described that caspases-3, -6, -7, and -8 can cleave APP.129,131,136 Cultures of neuronal cells deprived of serum showed activation of caspase-6 and subsequent processing of APP by caspase-6, generating a 6.5-kDa fragment that contains Aβ.131 In this context, caspases may play an active role in Aβ-induced neurotoxicity. Amyloid plaques are enriched in caspase-cleaved APP.129

In vitro systems have been instrumental in defining the cytotoxic properties of APP cleavage fragments, particularly the C-terminal fragments (CTF) derived from γ- and β-secretase cleavage of APP. C99 is toxic to neurons137 and expression of C99 in rodent brains induces Aβ deposition, neurodegeneration, alterations in behavior, and synaptic deficits.138 C99 can be further cleaved by caspases to generate C31 and it may be the C31 that leads to the CTF toxicity.138,139 The specific caspase that cleaves C99 has not been adequately identified, although caspases-8 and -9 have been implicated.140 Expression of C31 alone is cytotoxic, suggesting that the cleavage of CTFs to generate C31 may potentiate susceptibility to apoptosis.135 Mutations in APP to abolish the caspase cleavage site significantly attenuate the cytotoxic effects of C99.139 PDAPP mice carrying the cleavage mutation [PDAPP(D664A) mice] deposit Aβ but do not develop behavioral deficits, supporting a role for C31 in the potentiation of AD deficits.141 However, a recent study using these mice showed that there is no effect of expression of APPD664A on the development of behavioral defects.142 Further work is needed to clarify the functional relevance of C31.

c. Caspase processing of tau.

Caspases do not only process APP; caspase cleavage of tau enhances tau filament polymerization in vitro.143,144 Cleaved caspase-6 in the cortex and hippocampus of human AD brains is colocalized with plaques and NFTs.145,146 There is also an increase in caspase-6 cleaved tau fragments, detected with an antibody to the neoepitope generated when caspase-6 cleaves tau.146 A study of cultured dorsal-root ganglion neurons suggests that NGF-withdrawal leads to APP cleavage, which then induces caspase-6 activation.69 Taken together, these studies suggest that cleaved APP activates caspase-6, which may induce NFT formation. In support of a role for caspases in NFT formation are 2-photon studies. Live imaging from mice expressing mutant tau shows that there is activation of caspases prior to development of tangles. However, this study used a fluorescent caspase substrate/inhibitor (FLICA), which binds irreversibly to active sites, to follow caspase activation, so the causal link between caspase activation and tangle development cannot be inferred, despite the conclusions drawn from this study.147 This work illustrates that caspase activation may be an early event in the death pathway. This is supported by studies discussed below showing that Aβ induces caspase activation in primary neurons within 2 h of addition to the cultures.148

d. Caspases and inflammation in AD.

Inflammation is believed to contribute to the progression of AD.149 AD brains have increased mRNA and protein expression of the inflammatory cytokines IL-1β and IL-18 in both neurons and glia that are colocalized with Aβ and tau.150,151 Microglia isolated from autopsies of AD patients showed stronger IL-1β response to Aβ than did microglia isolated from control brains.152 In the Tg2576 AD mouse model, astrocytes and microglia surround amyloid plaques, and IL-1β expression is upregulated in these astrocytes, suggesting that Aβ induces reactive astrocytes, leading to an increase in the inflammatory cytokine.153

While caspases may not be directly involved in regulating the expression of cytokines, they do regulate their release. Caspase-1 can regulate Aβ-mediated release of cytokines. Aβ treatment of mouse microglia causes increased IL-1β release.154 Caspase-1 activation was assessed in this study by measuring its cleavage profile, which does not indicate whether the cleaved caspase-1 is active (see above), but the increase in IL-1β is strong support for activation of caspase-1.

What is the role of the NLRP caspase-1 inflammasome in the context of AD? No direct study has been done linking AD and caspase-1 activation in the human brain, but NLRP1 protein is found in neurons and oligodendrocytes.155 It has been shown in cell lines and primary mouse macrophages that NLRP3 is activated following intracellular K+ withdrawal.156 In spinal cord neurons treated with valinomycin, a K+ depleting agent, there is activation of the NLRP1 caspase-1 inflammasome.157 Moreover, cerebellar neurons grown in K+ free medium exhibited higher NLRP1 protein levels.158 A link between AD and K+ channel dysfunction has been reported. Aβ has been implicated in inducing a decrease in intracellular K+ levels.159,160 Based on these studies, it can be inferred that increased Aβ in the brain may activate the NLRP1 caspase-1 inflammasome through intracellular K+ depletion.

e. Caspases and Aβ toxicity.

Caspase-2 has been shown to be required for Aβ-mediated death of primary neurons using antisense knockdown; neurons from caspase-2 null mice are resistant to Aβ toxicity.161 Most recently, caspase-2 activity has been measured using the bVAD affinity ligand method in neurons treated with Aβ; measurable activity is induced within 2 h of treatment.148 Other studies of Aβ induction of caspase-2 have employed peptide substrates to measure activity; these measures are not specific for caspase-2 (see above).

Cortical neurons from caspase-12 null mice are resistant to Aβ.161,162 Caspase-12 has been shown to be involved in the signaling pathways associated with ER stress-induced apoptosis in rodent AD models.162,163 However, due to a frame shift mutation in the human caspase-12 gene in most humans, the full-length protein is expressed in only a subset of people and there is no increase of AD in this population.5,164 Hence, caspase-12 is not believed to play any role in human AD. Human caspase-4 shares a high homology with caspase-12, and it is also induced by ER stress.165 Aβ increases caspase-4 cleavage in the human neuroblastoma cell line, SK-N-SH, within 24 h.165

It has also been proposed that Aβ cross-links the cell surface death receptors Fas/TNFR. In neuronal PC12 cells, Aβ induces downregulation of FLIPs, the endogenous caspase-8 inhibitor, leading to cleavage of caspase-8.166,167 Another study utilizing SH-SY5Y neuroblastoma cells revealed that caspase-8 is recruited to the APP complex following Aβ stimulation. Once in the complex, caspase-8 is cleaved to a p10 fragment. It is argued that the cleaved/activated caspase-8 cleaves APP to induce cell death.168 These studies have not shown that removal of caspase-8 blocks Aβ effects.

Calcium and the calmodulin-dependent protein kinase II (CaMKII) could play a partial role in regulating Aβ-induced caspase activity.169 This work points to a potential calcium dysregulation induced by Aβ to activate CaMKII, which is sufficient to elicit caspase activity, measured with peptide substrates. However, CamKII may also phosphorylate caspase-2 to inactivate the caspase.27 The function of CamKII with regard to Aβ and caspase-2 needs to be studied further. The role of calcium in regulating caspase actions is further demonstrated in a study that utilized a human neuronal cell line (hNT), revealing that Aβ induces accumulation of a lipid metabolite, 1-o-hexadecyl-2-acetyl-sn-glycero-3-phosphocholine (C16:0PAF). This lipid product causes elevated intracellular calcium levels as a result of ER stress that leads to caspase activation. Peptide substrates were used to measure the caspase activity.170 Furthermore, m-calpain was activated before detectable caspase activity using the peptide substrates, implicating this enzyme in the regulation of caspase activation.

f. Caspases and Aβ generation.

In addition to responding to Aβ toxicity, caspase-2 or other caspases may play a role in generating Aβ. BACE, the β-secretase that generates Aβ from APP, has been shown to be stabilized by caspase-3 cleavage of GGA3, an adaptor protein involved in BACE trafficking.171 As discussed below, it was shown that in a model of cerebral ischemia, this mechanism leads to an increase in Aβ.

Taken together, caspases are very closely linked to the degeneration process found in AD brains, but the specific caspases involved and the downstream effect of these caspases has not been adequately defined. Figure 5 proposes how caspases may act in AD pathogenesis. Table I summarizes the current evidence of a role for each caspase in AD.

Fig. 5. Proposed caspase actions in Alzheimer’s Disease.

Fig. 5.

The amyloid precursor protein (APP) can be proteolytically processed to release multiple products, including Aβ, C99, and C31. C99 and C31 are toxic to neurons. Aβ can form fibrils, which deposit as amyloid plaques and soluble oligomers and can lead to ER stress, caspase activation, tau hyperphosphorylation, neuronal dysfunction, and neuronal death.

TABLE I.

Caspases Implicated in Alzheimer’s Disease

Caspase Paradigm Measure References
1 AD brain
Aβ treated microglia
mRNA
Casp1 cleavage
In vitro Tau cleavage
128,143,154
2 AD brain
Aβ treated hippocampal neurons
Aβ production from H4-C99 cells
mRNA
Casp2 null neurons siRNA depletion
Casp2 cleavage z-VDVAD-fmk
128,161,235
3 AD brain
Aβ production from H4-C99 cells
Tg4519 mice
mRNA
z-DEVD-fmk
In vitro Tau cleavage
cleaved Casp3 IHC
Casp3 cleavage
128,143,235238
4 Aβ treatment of SK-N-SH cells Casp4 cleavage
siRNA depletion
165,239
5 AD brain mRNA 128
6 AD brain
Tg4510 mice
mRNA
In vitro Tau cleavage
Casp6 cleavage
128,131,143,236
7 AD brain
Tg4510 mice
mRNA
In vitro Tau cleavage
128,143,236
8 AD brain
Aβ production from H4-C99 cells
mRNA
siRNA depletion
z-IETD-fmk
In vitro Tau cleavage
cleaved Casp8 IHC
128,134,143,235,240
9 AD brain mRNA
cleaved Casp9 IHC
128,240
12 Aβ treated cortical neurons
Hippocampal organotypic slices treated with Aβ
Antisense to Casp12
Casp12 null neurons
Casp12 cleavage
162,241

B. Cerebral Ischemia

Stroke, an acute neurodegenerative disorder, is the third most common cause of death and a leading cause of disability in major industrialized countries.172 Approximately 85% of strokes are ischemic, resulting from a thrombus that occludes a major cerebral artery and leads to a focal loss of blood flow. Unless normal blood flow is recovered within a short period of time, under 3 h from onset of occlusion, there is massive cellular death within the ischemic territory. Strokes can also be a consequence of either cardiac arrest or intraparenchymal hemorrhage.

The series of events instigated by ischemia develops over many days. Inflammation and altered microvascular permeability produce tissue edema. Direct effects on cells trigger glutamatergic excitotoxicity, ionic imbalance, and free-radical reactions, among others. There are also dynamic interactions of the neurovascular unit (the complex of neurons, the microvessels that supply them, the supportive cells: astroglia and microglia, and other resident inflammatory cells).173,174

Over the past decade, the mode of cell death induced by ischemia has been shown to be combined necrotic and apoptotic cell death. The center of the lesion, the core, has complete energy depletion, and there is a gradient of energy depletion from the core toward the bordering zone, the penumbra. From a morphologic perspective, the core has been considered mainly necrotic,175,176 while the penumbra has been considered apoptotic.177179 However, it has been suggested that the initial events in the core are apoptotic, and secondary necrosis results from a rapid failure to fully develop the apoptotic program because of the maintained depletion of apoptosis-requiring energy stores in the core.180

Neuronal death in the adult brain does not necessarily exhibit the classic morphological manifestations of apoptosis, as initially described by Kerr et al.,181 while in the neonatal brain, hypoxic-ischemic insults can cause typical morphological changes of apoptosis. In the adult brain, neurons are equipped with antiapoptotic molecules that raise the apoptotic threshold.182 There is a large body of evidence that suggests there is caspase-mediated cell death after ischemia,183 and multiple studies implicate apoptotic pathways as critical mediators of ischemia.184 For more than a decade, it has been suggested that cell death in the CNS following injury can coexist as apoptosis, necrosis, and hybrid forms along an apoptosis–necrosis continuum.185

The damage associated with the sudden return of blood flow, or reperfusion, to the oxygen-starved, energetically compromised tissue is mechanistically and therapeutically important.186 With timely reperfusion, either spontaneous or therapeutic, territory in the penumbra may be salvaged, but a delay in reperfusion will cause cell death. This phenomenon is termed “reperfusion injury.” It is held that much of this injury is produced during transient ischemia by inflammation, excitotoxicity, and apoptotic cell injury,174 and can result in the disruption of the blood–brain barrier (BBB) and formation of brain edema.187 An advantage of modeling stroke is that, unlike other neurodegenerative diseases, in stroke, the primary insult that triggers the pathology is known, and the models provide a highly reproducible, clinically relevant approach for studying this acute insult.

1. Animal Models of Stroke

Animal models are widely used to mimic human stroke. In contrast to other neurodegenerative diseases, there are excellent animal models of stroke that provide evidence for mechanisms, as well as prevention and treatment efficacy in vivo, that allow for control of collateral factors.

While cerebral ischemia can be modeled in large animals (pigs, baboons, etc.), rodent models are the most widely used. Although there are differences in anatomy and functionality between the rodent and the human brain, these models permit a careful dissection of mechanisms of injury and neuroprotection and allow examination of events that occur from onset of the insult to weeks or months after the ischemic event [for a review, see Ref. 188].

There are a variety of models that reflect the assortment of insults in humans. These models can be divided into three subgroups: global ischemia, focal ischemia, and hemorrhagic infarct. Global models, where the major blood vessels to the forebrain are occluded, are models of the consequences of cardiac arrest rather than stroke.189 In focal models, a smaller arterial branch is occluded. Most models occlude the middle cerebral artery (MCA), as occlusion of this vessel is found in the majority of human ischemic strokes.189 Global and focal occlusion models are either permanent or transient, allowing examination of the damage that results from both ischemia and reperfusion. Hemorrhagic stroke models involve the stereotactic injection of collagenase into the cerebral parenchyma.189

An overriding question of much animal model research in the last decade has been the translational value of the models. The lack of efficacy of neuroprotectants against stroke in clinical trials raised the question of the validity of the animal models for preclinical research. A proof of confidence in the value of animal models is that changing several physiological parameters, like reperfusion, hyperglycemia, hyperthermia, or blood pressure, has similar effects on the outcome of stroke in humans and in animal models [for a review, see Ref. 189]. Nevertheless, although animal models mimic the human stroke pathology, small mammalian brains are quite different from human brains in function and morphology, and this may influence the efficacy of the therapies. This phenomenon was recently represented in an in silico model, which considered the differences in the percentage of white matter and glia between rodents and humans.190 If this hypothesis is correct, the efficacy of the therapies should be apparent when animals with a more similar brain composition to humans are used for models.

From a practical, logistical perspective, it is difficult to treat patients at early time points; therefore, clinical investigators should focus on mechanisms that occur later in the ischemic cascade. However, there are also clinical settings where the risk of stroke is increased, such as cardiac surgery, where preventative treatments would be useful. Additionally, it is important to consider how much protection is achieved in preclinical trials, where a reduction of at least 50% of the ischemic damage has been the goal, and also to develop appropriate behavioral tests. There are multiple mechanisms occurring simultaneously and sequentially during stroke. Therefore, it is important to know the chronology and relevance of the different mechanisms that are occurring as a consequence of ischemia and reperfusion. The best treatment strategy might rely on a combination of neuroprotective agents, each geared toward a different death mechanism, administered at the appropriate time to interfere in the multiple ischemia-induced death pathways.

2. In Vitro Models of Ischemia

In vitro models have been utilized for over two decades in the study of cerebral ischemia.191 Much of our understanding of the pathogenesis of stroke derives from studies of cell lines, and primary and organotypic cultures exposed to hypoxia, anoxia, oxygen, and glucose deprivation. These models allow the dissection of pathways in a controlled setting. Of the models under study, the oxygen–glucose deprivation (OGD) model is believed to most resemble the in vivo ischemia setting.192194 Typical insults last for 15–90 min and are immediately followed by reperfusion, that is, addition of glucose to media and return to normoxic conditions.192,193 Acute ischemic insults (10–20 min) are used for electrophysiological analysis in hippocampal neurons195,196 as well as spiny/aspiny striatal neurons.197,198 The discussion below gives a brief overview of work in hippocampal primary cultures or brain slices, unless otherwise stated.

Early evidence with the OGD model suggested that ischemic injury was mainly necrotic and mediated by glutamate excitotoxicity,199201 but subsequent work showed that apoptosis is masked by glutamate receptor activation in OGD in primary cultures.193 Apoptosis in primary cultures is a delayed event (~24–36 h postinsult), while glutamate-mediated necrosis is observed as early as 2 h after the insult.192 As with some other insults, the extent of the insult, in this case OGD, determines how much necrosis occurs. Shorter periods of OGD (~15 min) result in most neurons dying by apoptosis, while longer periods of OGD (~75 min) do not alter apoptotic death, but increase the amount of necrosis.202 The relative timing of apoptosis and necrosis are different in hippocampal brain slices exposed to OGD compared to primary cultures. In slices, apoptosis and glutamate-dependent necrosis both occur within 3 h after the ischemic episode.203205 These discrepancies may signify the importance of the neuron–glia context in studying ischemia in vitro.

The role of caspases has been studied in limited detail in OGD.202,204207 These studies show that caspase-3 is activated by OGD and can be regulated by pan-caspase inhibitors. OGD cells treated with NMDA receptor antagonists undergo apoptotic death that can be inhibited by a pan-caspase inhibitor, z-VAD-fmk.204,206 Another caspase inhibitor (Ac-YVAD-cmk) exhibited similar neuroprotection in OGD brain slices.205 These inhibitors are not specific for individual caspases,50 and therefore, these studies do not clarify which caspases are important in promoting death in this ischemia model. Knockout of caspase-3 is also neuroprotective against OGD.208 Cleavage of caspases-3, -8, and -9 has been observed in microglia exposed to OGD,209 although the causality of the different caspases has not been determined. It has also been shown that cleaved caspase-7 and caspase-3 are found exclusively in microglia and neurons, and not astrocytes in OGD in mixed primary cultures,202 again suggesting that the neuron–glia interactions are important in modeling ischemia.

As will be discussed below, caspase-1 and generation of interleukin-1β are found in animal models of ischemia. Activation of these pathways and subsequent neuronal death are found when SOD-1, the cytoplasmic superoxide dismutase, is downregulated.210,211 This death is mediated by activation of caspase-1, secretion of IL-1β, and generation of peroxynitrite, all molecules that have been shown to be important in the progression of ischemia in vivo.212215 Thus, this model may provide a cellular model for investigation of the mechanism of neuronal death in ischemia.

3. Caspases in Stroke

In 1997, a function for caspases in mammalian neurodegeneration was first shown with the demonstration that a dominant-negative mutant of caspase-1 provided protection against tMCAo.216 During the ensuing years, multiple studies have been published, and it is clear that caspases are key molecules in the death mechanisms induced during ischemia.183,186 We will present data for caspases that are conserved in human and rodents.

a. Caspase-1.

As noted above, the first studies of caspase in neuronal disease showed that ablation of caspase-1 activity provided protection against tMCAo, but those studies used a dominant-negative caspase-1 that could block activity of other caspases in addition to caspase-1.216 This work was followed with studies using caspase-1 null mice which exhibited reduced ischemic damage.217 Indeed, several lines of evidence point to an important role for this caspase. The pro-inflammatory cytokine IL-1β, which is the main target of caspase-1 processing, is rapidly induced by focal/global ischemia.218 Ablation of IL-1β expression provides more than 80% protection from ischemic damage.214 Caspase-1 has been reported to be increased in the core of the stroke180 and in the penumbra.219 A recent review discusses the role of inflammation in acute neurodegeneration.30

b. Caspase-2.

The initial characterization of the caspase-2 null mice showed that the caspase-2 null mice were not protected from tMCAo.61 A more recent study of caspase-2 and PIDD in global ischemia shows a role for PIDD in this model of ischemia.220 However, this study does not directly measure caspase-2 activity and, since the PIDD null mice appear to undergo caspase-2 dependent death22,23 (although neurons have not been studied), it is not clear that caspase-2 is part of the ischemic death mechanism. But it must be considered that, since the caspase-2 null mice have been shown to have an increased expression of caspase-9 and Smac/DIABLO in the brain,64 the compensations in death molecules might mask a role for caspase-2 in ischemia. Thus, the role of caspase-2 and of PIDD in ischemia requires further study.

c. Caspase-3.

Caspase-3 is present in the ischemic penumbra59,180 and its deletion renders mice more resistant to ischemic injury.208 However, although caspase activation is generally described in the penumbra of focal infarcts, immunohistochemical analysis in MCAo has detected neurons containing caspase-3 in the infarct core.221,222 In humans, although there is limited information available, there is a procaspase-3 increase in expression within hours resulting from permanent arterial occlusion,223 and activated caspase-3 and cleaved PARP have been detected in some neurons several days after cardiac arrest with reperfusion.183 Cleaved caspase-3 has also been detected in glial cells 24 h postinfarction.224

d. Caspase-6.

Little data exists with regard to a role for caspase-6 in stroke. Studies of mRNA (see below) showed an increase in caspase-6 expression in rats subjected to pMCAo.

e. Caspase-7.

Much of the data with regard to caspase-7 is overlapping with the data about caspase-3. A clear, distinct role for caspase-7 in stroke has not yet been described. Studies of mRNA (see below) showed an increase in caspase-7 expression in rats subjected to pMCAo.

f. Caspase-8.

Increased expression of caspase-8 has been seen in both the core and in the penumbra,180 as has increased expression of FasL.219

g. Caspase-9.

A limited study of human brains obtained from 4 h to 5 days postinfarction suggests an increase in caspase-9 protein expression peaking at 24 h postinfarct.225 In rodent models, caspase-9 is increased in the penumbra.180

h. mRNA expression.

Harrison et al.226 reported a strict transcriptional regulation of caspase expression following pMCAo in rats. There is a pattern of expression with an increase of caspase-1, -3, -6, -7, -8, and -11 mRNA, and a decrease of caspase-9 was detected at different time points. Caspase-2 mRNA shows no changes. But, in this report, the cellular types expressing these proteases were not studied.

Figure 6 presents an overview of what is presently known about the localization of different caspases during tMCAo. The timing of appearance and cell type location for the different caspases still needs further study. Actual measures of specific caspase activity induced by ischemia are also needed. The knowledge of the pathways and molecules that interconnect these caspases is still elusive, and more research has to be done in this area. Table II summarizes the evidence for a role for each caspase in stroke.

Fig. 6. Caspase activation in cerebral ischemia.

Fig. 6.

Caspases are activated in the ischemic core early in stroke and then in the penumbra as the stroke progresses.

TABLE II.

Caspases Implicated in Ischemia

Caspase Paradigm Measure References
1 tMCAo
pMCAo
Dominant-negative Casp1
Casp1 null mice
ac-YVAD-AFC
mRNA
180,216,217,226,242
2 Global ischemia Caspase-2 cleavage 220
3 tMCAo
pMCAO
Casp3 null mice
WB clC3
ac-DEVD-AFC/WB/IHC
mRNA
180,208,221,226
6 pMCAo mRNA levels 226
7 pMCAo mRNA levels 226
8 pMCAo mRNA levels
ac-IETD-AFC/WB/IHC
180,221,226
9 human stroke tissue
tMCAo rodent
tMCAo canine
IHC C9
Release of C9 from mitochondria
219,225,243

C. Linking Stroke and AD

In this chapter, we have summarized the current knowledge of the roles of caspases in AD and stroke. Caspases can either participate in the development of the disease or in the progression of the disease. Interestingly, a number of studies have revealed a positive correlation between stroke and AD. Cross-epidemiological studies show that patients who have suffered a stroke have an increased likelihood of developing dementia.227229 In a rodent ischemic model, tau hyperphosphorylation is present in the cortices.230 In terms of the relationship between stroke and amyloid, studies point to caspases as key players in modulating between these two conditions. Rodents subjected to tMCAo showed increases in APP and Aβ expression in the ischemic area, corpus callosum, thalamus, striatum, caudate-putamen, and hippocampus within days of reperfusion.231233 The increased expression levels are observed in both neurons and astrocytes in short-term studies ranging from days to weeks. However, over longer survival time, the levels of APP and Aβ gradually decrease.231 This change may be explained by a downregulation of APP and Aβ in astrocytes and/or clearance of these peptides by astrocytes and microglia. Although the expressions of APP and Aβ decrease over time, in the thalamus, the expression profile of Aβ changes from diffused to plaque-like deposits.232 In sum, these rodent studies point to an initial upregulation in APP and Aβ in a diffused pattern in various brain regions following MCAO. Over time, the levels of these proteins are reduced. However, there remains residual damage in the form of plaque-like structures.

The mechanism for elevated APP and Aβ in brain regions following stroke has begun to be addressed by investigators. Administration of zDEVDfmk to rats prior to tMCAo blocked the appearance of cleaved caspase-3 and abrogated the increased expression of BACE1, the enzyme that produces Aβ from APP, and Aβ.233,234 Similar effects were seen in a model of brain trauma that shows increased Aβ levels.233,234 While these studies do not specifically examine caspase-3 function, they do implicate activation of caspases in the production of Aβ, providing a mechanism underlying the clinical observations that stroke or head trauma are risk factors for the development of AD.

V. Future Directions

There is an increasing appreciation that caspases have a variety of actions, both death and non-death-related. With this in mind, it is even more important that our understanding of the specific functions of individual caspases in health and disease is increased. In this way, appropriate disease interventions that do not interfere with the non-disease-related actions of caspases can be devised. In order to progress in our understanding of caspase functions, attention must be directed to the tools available to study caspases and the development of better tools for analysis. Multiple studies have now shown that many of the tools used to study caspases are inadequate, yet they continue to be used and to confound the literature. A search of the current literature will show how many studies still use the pseudopeptide inhibitors/substrates as evidence for the function of specific caspases. Many studies still equate caspase cleavage by Western blotting with activity, or caspase-3 cleavage by immunocytochemistry with apoptosis. As of today, the best tools available are molecular manipulation and affinity ligands.

Another issue in the study of human disease is the models available. In the two diseases that we have used to illustrate current knowledge of caspases in neuronal disease, there are different issues. The in vivo models of cerebral ischemia mimic the human disease quite accurately and provide excellent systems for studying death mechanisms. The cellular models of ischemia, while providing the advantages of cell-based models in the study of biochemical and molecular events, do not offer the complexity of the various elements clearly involved in the evolution of the ischemic event. For AD, the in vivo models available at best model preclinical disease. There is no adequate model to mimic the progression of the disease. The current models have little or no neuronal loss, making the study of death mechanisms in these animals difficult. Thus, at present, the cell models and the hippocampal slice systems offer the best models for studying death mechanisms in AD. Better models and more specific tools are in development and are eagerly awaited by the field.

References

  • 1.Miura M, Zhu H, Rotello R, Hartwieg EA, Yuan J. Induction of apoptosis in fibroblasts by IL-1 beta-converting enzyme, a mammalian homolog of the C. elegans cell death gene ced-3. Cell 1993;75:653–60. [DOI] [PubMed] [Google Scholar]
  • 2.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 1992;356:768–74. [DOI] [PubMed] [Google Scholar]
  • 3.Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell 1993;75:641–52. [DOI] [PubMed] [Google Scholar]
  • 4.Eckhart L, Ban J, Fischer H, Tschachler E. Caspase-14: analysis of gene structure and mRNA expression during keratinocyte differentiation. Biochem Biophys Res Commun 2000;277:655–9. [DOI] [PubMed] [Google Scholar]
  • 5.Fischer H, Koenig U, Eckhart L, Tschachler E. Human caspase 12 has acquired deleterious mutations. Biochem Biophys Res Commun 2002;293:722–6. [DOI] [PubMed] [Google Scholar]
  • 6.Roy S, Sharom JR, Houde C, Loisel TP, Vaillancourt JP, Shao W, et al. Confinement of caspase-12 proteolytic activity to autoprocessing. Proc Natl Acad Sci USA 2008;105:4133–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kim EM, Shin EJ, Choi JH, Son HJ, Park IS, Joh TH, et al. Matrix metalloproteinase-3 is increased and participates in neuronal apoptotic signaling downstream of caspase-12 during endoplasmic reticulum stress. J Biol Chem 2010;285:16444–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Boatright KM, Salvesen GS. Caspase activation. Biochem Soc Symp 2003;70:233–42. [DOI] [PubMed] [Google Scholar]
  • 9.Pop C, Salvesen GS. Human caspases: activation, specificity, and regulation. J Biol Chem 2009;284:21777–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Boatright KM, Renatus M, Scott FL, Sperandio S, Shin H, Pedersen IM, et al. A unified model for apical caspase activation. Mol Cell 2003;11:529–41. [DOI] [PubMed] [Google Scholar]
  • 11.Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 2007;26:1324–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Baliga BC, Read SH, Kumar S. The biochemical mechanism of caspase-2 activation. Cell Death Differ 2004;11:1234–41. [DOI] [PubMed] [Google Scholar]
  • 13.Stennicke HR, Deveraux QL, Humke EW, Reed JC, Dixit VM, Salvesen GS. Caspase-9 can be activated without proteolytic processing. J Biol Chem 1999;274:8359–62. [DOI] [PubMed] [Google Scholar]
  • 14.Oberst A, Pop C, Tremblay AG, Blais V, Denault JB, Salvesen GS, et al. Inducible dimerization and inducible cleavage reveal a requirement for both processes in caspase-8 activation. J Biol Chem 2010;285:16632–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Riedl SJ, Salvesen GS. The apoptosome: signalling platform of cell death. Nat Rev 2007;8:405–13. [DOI] [PubMed] [Google Scholar]
  • 16.Denault JB, Eckelman BP, Shin H, Pop C, Salvesen GS. Caspase 3 attenuates XIAP (X-linked inhibitor of apoptosis protein)-mediated inhibition of caspase 9. Biochem J 2007;405:11–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kang TB, Oh GS, Scandella E, Bolinger B, Ludewig B, Kovalenko A, et al. Mutation of a self-processing site in caspase-8 compromises its apoptotic but not its nonapoptotic functions in bacterial artificial chromosome-transgenic mice. J Immunol 2008;181:2522–32. [DOI] [PubMed] [Google Scholar]
  • 18.Wang L, Du F, Wang X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell 2008;133:693–703. [DOI] [PubMed] [Google Scholar]
  • 19.Jia SH, Parodo J, Kapus A, Rotstein OD, Marshall JC. Dynamic regulation of neutrophil survival through tyrosine phosphorylation or dephosphorylation of caspase-8. J Biol Chem 2008;283:5402–13. [DOI] [PubMed] [Google Scholar]
  • 20.Tinel A, Tschopp J. The PIDDosome, a protein complex implicated in activation of caspase-2 in response to genotoxic stress. Science 2004;304:843–6. [DOI] [PubMed] [Google Scholar]
  • 21.Troy CM, Ribe EM. Caspase-2: vestigial remnant or master regulator? Sci Signal 2008;1:pe42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Manzl C, Krumschnabel G, Bock F, Sohm B, Labi V, Baumgartner F, et al. Caspase-2 activation in the absence of PIDDosome formation. J Cell Biol 2009;185:291–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kim IR, Murakami K, Chen NJ, Saibil SD, Matysiak-Zablocki E, Elford AR, et al. DNA damage- and stress-induced apoptosis occurs independently of PIDD. Apoptosis 2009;14:1039–49. [DOI] [PubMed] [Google Scholar]
  • 24.Berube C, Boucher LM, Ma W, Wakeham A, Salmena L, Hakem R, et al. Apoptosis caused by p53-induced protein with death domain (PIDD) depends on the death adapter protein RAIDD. Proc Natl Acad Sci USA 2005;102:14314–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tinel A, Janssens S, Lippens S, Cuenin S, Logette E, Jaccard B, et al. Autoproteolysis of PIDD marks the bifurcation between pro-death caspase-2 and pro-survival NF-kappaB pathway. EMBO J 2007;26:197–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cuenin S, Tinel A, Janssens S, Tschopp J. p53-induced protein with a death domain (PIDD) isoforms differentially activate nuclear factor-kappaB and caspase-2 in response to genotoxic stress. Oncogene 2008;27:387–96. [DOI] [PubMed] [Google Scholar]
  • 27.Nutt LK, Margolis SS, Jensen M, Herman CE, Dunphy WG, Rathmell JC, et al. Metabolic regulation of oocyte cell death through the CaMKII-mediated phosphorylation of caspase-2. Cell 2005;123:89–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Shin S, Lee Y, Kim W, Ko H, Choi H, Kim K. Caspase-2 primes cancer cells for TRAIL-mediated apoptosis by processing procaspase-8. EMBO J 2005;24:3532–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nutt LK, Buchakjian MR, Gan E, Darbandi R, Yoon SY, Wu JQ, et al. Metabolic control of oocyte apoptosis mediated by 14–3-3zeta-regulated dephosphorylation of caspase-2. Dev Cell 2009;16:856–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Trendelenburg G Acute neurodegeneration and the inflammasome: central processor for danger signals and the inflammatory response? J Cereb Blood Flow Metab 2008;28:867–81. [DOI] [PubMed] [Google Scholar]
  • 31.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002;10:417–26. [DOI] [PubMed] [Google Scholar]
  • 32.Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity 2004;20:319–25. [DOI] [PubMed] [Google Scholar]
  • 33.Petrilli V, Papin S, Tschopp J. The inflammasome. Curr Biol 2005;15:R581. [DOI] [PubMed] [Google Scholar]
  • 34.Tschopp J, Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat Rev Immunol 2010;10:210–5. [DOI] [PubMed] [Google Scholar]
  • 35.Martinon F, Tschopp J. Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ 2007;14:10–22. [DOI] [PubMed] [Google Scholar]
  • 36.Petrilli V, Dostert C, Muruve DA, Tschopp J. The inflammasome: a danger sensing complex triggering innate immunity. Curr Opin Immunol 2007;19:615–22. [DOI] [PubMed] [Google Scholar]
  • 37.Srinivasula SM, Ashwell JD. IAPs: what’s in a name? Mol Cell 2008;30:123–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Eckelman BP, Salvesen GS, Scott FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep 2006;7:988–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Eckelman BP, Salvesen GS. The human anti-apoptotic proteins cIAP1 and cIAP2 bind but do not inhibit caspases. J Biol Chem 2006;281:3254–60. [DOI] [PubMed] [Google Scholar]
  • 40.Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, et al. Inhibition of death receptor signals by cellular FLIP. Nature 1997;388:190–5. [DOI] [PubMed] [Google Scholar]
  • 41.Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000;102:33–42. [DOI] [PubMed] [Google Scholar]
  • 42.Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000;102:43–53. [DOI] [PubMed] [Google Scholar]
  • 43.Hegde R, Srinivasula SM, Zhang Z, Wassell R, Mukattash R, Cilenti L, et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem 2002;277:432–8. [DOI] [PubMed] [Google Scholar]
  • 44.Liston P, Fong WG, Kelly NL, Toji S, Miyazaki T, Conte D, et al. Identification of XAF1 as an antagonist of XIAP anti-Caspase activity. Nat Cell Biol 2001;3:128–33. [DOI] [PubMed] [Google Scholar]
  • 45.Arora V, Cheung HH, Plenchette S, Micali OC, Liston P, Korneluk RG. Degradation of survivin by the X-linked inhibitor of apoptosis (XIAP)-XAF1 complex. J Biol Chem 2007;282:26202–9. [DOI] [PubMed] [Google Scholar]
  • 46.Chang DW, Xing Z, Capacio VL, Peter ME, Yang X. Interdimer processing mechanism of procaspase-8 activation. EMBO J 2003;22:4132–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pop C, Fitzgerald P, Green DR, Salvesen GS. Role of proteolysis in caspase-8 activation and stabilization. Biochemistry 2007;46:4398–407. [DOI] [PubMed] [Google Scholar]
  • 48.Thornberry NA, Rano TA, Peterson EP, Rasper DM, Timkey T, Garcia-Calvo M, et al. A combinatorial approach defines specificities of members of the caspase family and granzyme B Functional relationships established for key mediators of apoptosis. J Biol Chem 1997;272:17907–11. [DOI] [PubMed] [Google Scholar]
  • 49.Talanian RV, Quinlan C, Trautz S, Hackett MC, Mankovich JA, Banach D, et al. Substrate specificities of caspase family proteases. J Biol Chem 1997;272:9677–82. [DOI] [PubMed] [Google Scholar]
  • 50.McStay GP, Salvesen GS, Green DR. Overlapping cleavage motif selectivity of caspases: implications for analysis of apoptotic pathways. Cell Death Differ 2008;15:322–31. [DOI] [PubMed] [Google Scholar]
  • 51.Tu S, McStay GP, Boucher LM, Mak T, Beere HM, Green DR. In situ trapping of activated initiator caspases reveals a role for caspase-2 in heat shock-induced apoptosis. Nat Cell Biol 2006;8:72–7. [DOI] [PubMed] [Google Scholar]
  • 52.Bouchier-Hayes L, Oberst A, McStay GP, Connell S, Tait SW, Dillon CP, et al. Characterization of cytoplasmic caspase-2 activation by induced proximity. Mol Cell 2009;35:830–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kitevska T, Spencer DM, Hawkins CJ. Caspase-2: controversial killer or checkpoint controller? Apoptosis 2009;14:829–48. [DOI] [PubMed] [Google Scholar]
  • 54.Kuida K, Haydar TF, Kuan CY, Gu Y, Taya C, Karasuyama H, et al. Reduced apoptosis and cytochrome c-mediated caspase activation in mice lacking caspase 9. Cell 1998;94:325–37. [DOI] [PubMed] [Google Scholar]
  • 55.Cecconi F, Alvarez-Bolado G, Meyer BI, Roth KA, Gruss P. Apaf1 (CED-4 homolog) regulates programmed cell death in mammalian development. Cell 1998;94:727–37. [DOI] [PubMed] [Google Scholar]
  • 56.Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Soengas MS, et al. Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell 1998;94:339–52. [DOI] [PubMed] [Google Scholar]
  • 57.Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, Hakem R, et al. Apaf1 is required for mitochondrial pathways of apoptosis and brain development. Cell 1998;94:739–50. [DOI] [PubMed] [Google Scholar]
  • 58.Kuida K, Zheng TS, Na S, Kuan C, Yang D, Karasuyama H, et al. Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice. Nature 1996;384:368–72. [DOI] [PubMed] [Google Scholar]
  • 59.Namura S, Zhu J, Fink K, Endres M, Srinivasan A, Tomaselli KJ, et al. Activation and cleavage of caspase-3 in apoptosis induced by experimental cerebral ischemia. J Neurosci 1998;18:3659–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kuida K, Lippke JA, Ku G, Harding MW, Livingston DJ, Su MS, et al. Altered cytokine export and apoptosis in mice deficient in interleukin-1 beta converting enzyme. Science 1995;267:2000–3. [DOI] [PubMed] [Google Scholar]
  • 61.Bergeron L, Perez GI, Macdonald G, Shi L, Sun Y, Jurisicova A, et al. Defects in regulation of apoptosis in caspase-2-deficient mice. Genes Dev 1998;12:1304–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang S, Miura M, Jung YK, Zhu H, Li E, Yuan J. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell 1998;92:501–9. [DOI] [PubMed] [Google Scholar]
  • 63.Zheng TS, Hunot S, Kuida K, Momoi T, Srinivasan A, Nicholson DW, et al. Deficiency in caspase-9 or caspase-3 induces compensatory caspase activation. Nat Med 2000;6:1241–7. [DOI] [PubMed] [Google Scholar]
  • 64.Troy CM, Rabacchi SA, Hohl JB, Angelastro JM, Greene LA, Shelanski ML. Death in the balance: alternative participation of the caspase-2 and -9 pathways in neuronal death induced by nerve growth factor deprivation. J Neurosci 2001;21:5007–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.D’Amelio M, Cavallucci V, Cecconi F. Neuronal caspase-3 signaling: not only cell death. Cell Death Differ 2010;17:1104–14. [DOI] [PubMed] [Google Scholar]
  • 66.Fernando P, Brunette S, Megeney LA. Neural stem cell differentiation is dependent upon endogenous caspase 3 activity. FASEB J 2005;19:1671–3. [DOI] [PubMed] [Google Scholar]
  • 67.Aranha MM, Sola S, Low WC, Steer CJ, Rodrigues CM. Caspases and p53 modulate FOXO3A/Id1 signaling during mouse neural stem cell differentiation. J Cell Biochem 2009;107:748–58. [DOI] [PubMed] [Google Scholar]
  • 68.Williams DW, Kondo S, Krzyzanowska A, Hiromi Y, Truman JW. Local caspase activity directs engulfment of dendrites during pruning. Nat Neurosci 2006;9:1234–6. [DOI] [PubMed] [Google Scholar]
  • 69.Nikolaev A, McLaughlin T, O’Leary DD, Tessier-Lavigne M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 2009;457:981–9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 70.Huesmann GR, Clayton DF. Dynamic role of postsynaptic caspase-3 and BIRC4 in zebra finch song-response habituation. Neuron 2006;52:1061–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Gulyaeva NV, Kudryashov IE, Kudryashova IV. Caspase activity is essential for long-term potentiation. J Neurosci Res 2003;73:853–64. [DOI] [PubMed] [Google Scholar]
  • 72.Stepanichev MY, Kudryashova IV, Yakovlev AA, Onufriev MV, Khaspekov LG, Lyzhin AA, et al. Central administration of a caspase inhibitor impairs shuttle-box performance in rats. Neuroscience 2005;136:579–91. [DOI] [PubMed] [Google Scholar]
  • 73.Li Z, Jo J, Jia JM, Lo SC, Whitcomb DJ, Jiao S, et al. Caspase-3 activation via mitochondria is required for long-term depression and AMPA receptor internalization. Cell 2010;141:859–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gu Y, Nieves JW, Stern Y, Luchsinger JA, Scarmeas N. Food combination and Alzheimer disease risk: a protective diet. Arch Neurol 2010;67:699–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pappolla MA, Chyan YJ, Poeggeler B, Frangione B, Wilson G, Ghiso J, et al. An assessment of the antioxidant and the antiamyloidogenic properties of melatonin: implications for Alzheimer’s disease. J Neural Transm 2000;107:203–31. [DOI] [PubMed] [Google Scholar]
  • 76.Blennow K, de Leon MJ, Zetterberg H. Alzheimer’s disease. Lancet 2006;368:387–403. [DOI] [PubMed] [Google Scholar]
  • 77.Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde” Clin Anat 1995;8:429–31. [DOI] [PubMed] [Google Scholar]
  • 78.Small GW, Rabins PV, Barry PP, Buckholtz NS, DeKosky ST, Ferris SH, et al. Diagnosis and treatment of Alzheimer disease and related disorders consensus statement of the American Association for Geriatric Psychiatry, the Alzheimer’s Association, and the American Geriatrics Society. JAMA 1997;278:1363–71. [PubMed] [Google Scholar]
  • 79.Terry RD, Peck A, DeTeresa R, Schechter R, Horoupian DS. Some morphometric aspects of the brain in senile dementia of the Alzheimer type. Ann Neurol 1981;10:184–92. [DOI] [PubMed] [Google Scholar]
  • 80.Scott SA, DeKosky ST, Sparks DL, Knox CA, Scheff SW. Amygdala cell loss and atrophy in Alzheimer’s disease. Ann Neurol 1992;32:555–63. [DOI] [PubMed] [Google Scholar]
  • 81.Khachaturian ZS. Toward a comprehensive theory of Alzheimer’s disease—challenges, caveats, and parameters. Ann NY Acad Sci 2000;924:184–93. [DOI] [PubMed] [Google Scholar]
  • 82.Khachaturian ZS. Neurobiology of aging: Alzheimer’s clinical review series. Bridging bench to bedside: clinical problems in search of basic solution. Neurobiol Aging 2000;21:843. [DOI] [PubMed] [Google Scholar]
  • 83.Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW. The topographical and neuroanatomical distribution of neurofibrillary tangles and neuritic plaques in the cerebral cortex of patients with Alzheimer’s disease. Cereb Cortex 1991;1:103–16. [DOI] [PubMed] [Google Scholar]
  • 84.Braak H, Braak E. Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections. Brain Pathol 1991;1:213–6. [DOI] [PubMed] [Google Scholar]
  • 85.Braak H, Braak E. Morphological changes in the human cerebral cortex in dementia. J Hirnforsch 1991;32:277–82. [PubMed] [Google Scholar]
  • 86.Arriagada PV, Marzloff K, Hyman BT. Distribution of Alzheimer-type pathologic changes in nondemented elderly individuals matches the pattern in Alzheimer’s disease. Neurology 1992;42:1681–8. [DOI] [PubMed] [Google Scholar]
  • 87.Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 1992;42:631–9. [DOI] [PubMed] [Google Scholar]
  • 88.Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F, Fidani L, et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 1991;349:704–6. [DOI] [PubMed] [Google Scholar]
  • 89.Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 1995;375:754–60. [DOI] [PubMed] [Google Scholar]
  • 90.Levy-Lahad E, Wijsman EM, Nemens E, Anderson L, Goddard KA, Weber JL, et al. A familial Alzheimer’s disease locus on chromosome 1. Science 1995;269:970–3. [DOI] [PubMed] [Google Scholar]
  • 91.Kowalska A, Pruchnik-Wolinska D, Florczak J, Modestowicz R, Szczech J, Kozubski W, et al. Genetic study of familial cases of Alzheimer’s disease. Acta Biochim Pol 2004;51:245–52. [PubMed] [Google Scholar]
  • 92.Kowalska A, Pruchnik-Wolinska D, Florczak J, Szczech J, Kozubski W, Rossa G, et al. Presenilin 1 mutations in Polish families with early-onset Alzheimer’s disease. Folia Neuropathol 2004;42:9–14. [PubMed] [Google Scholar]
  • 93.Games D, Adams D, Alessandrini R, Barbour R, Berthelette P, Blackwell C, et al. Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 1995;373:523–7. [DOI] [PubMed] [Google Scholar]
  • 94.Hsiao KK, Borchelt DR, Olson K, Johannsdottir R, Kitt C, Yunis W, et al. Age-related CNS disorder and early death in transgenic FVB/N mice overexpressing Alzheimer amyloid precursor proteins. Neuron 1995;15:1203–18. [DOI] [PubMed] [Google Scholar]
  • 95.Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, et al. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996;274:99–102. [DOI] [PubMed] [Google Scholar]
  • 96.Johnson-Wood K, Lee M, Motter R, Hu K, Gordon G, Barbour R, et al. Amyloid precursor protein processing and A beta42 deposition in a transgenic mouse model of Alzheimer disease. Proc Natl Acad Sci USA 1997;94:1550–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sturchler-Pierrat C, Abramowski D, Duke M, Wiederhold KH, Mistl C, Rothacher S, et al. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc Natl Acad Sci USA 1997;94:13287–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Terai K, Iwai A, Kawabata S, Tasaki Y, Watanabe T, Miyata K, et al. beta-amyloid deposits in transgenic mice expressing human beta-amyloid precursor protein have the same characteristics as those in Alzheimer’s disease. Neuroscience 2001;104:299–310. [DOI] [PubMed] [Google Scholar]
  • 99.Callahan MJ, Lipinski WJ, Bian F, Durham RA, Pack A, Walker LC. Augmented senile plaque load in aged female beta-amyloid precursor protein-transgenic mice. Am J Pathol 2001;158:1173–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Gotz J, Probst A, Spillantini MG, Schafer T, Jakes R, Burki K, et al. Somatodendritic localization and hyperphosphorylation of tau protein in transgenic mice expressing the longest human brain tau isoform. EMBO J 1995;14:1304–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gotz J, Chen F, Barmettler R, Nitsch RM. Tau filament formation in transgenic mice expressing P301L tau. J Biol Chem 2001;276:529–34. [DOI] [PubMed] [Google Scholar]
  • 102.Brion JP, Tremp G, Octave JN. Transgenic expression of the shortest human tau affects its compartmentalization and its phosphorylation as in the pretangle stage of Alzheimer’s disease. Am J Pathol 1999;154:255–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Spittaels K, Van den Haute C, Van Dorpe J, Bruynseels K, Vandezande K, Laenen I, et al. Prominent axonopathy in the brain and spinal cord of transgenic mice overexpressing four-repeat human tau protein. Am J Pathol 1999;155:2153–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Probst A, Gotz J, Wiederhold KH, Tolnay M, Mistl C, Jaton AL, et al. Axonopathy and amyotrophy in mice transgenic for human four-repeat tau protein. Acta Neuropathol 2000;99:469–81. [DOI] [PubMed] [Google Scholar]
  • 105.Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 2000;25:402–5. [DOI] [PubMed] [Google Scholar]
  • 106.Tanemura K, Akagi T, Murayama M, Kikuchi N, Murayama O, Hashikawa T, et al. Formation of filamentous tau aggregations in transgenic mice expressing V337M human tau. Neurobiol Dis 2001;8:1036–45. [DOI] [PubMed] [Google Scholar]
  • 107.Tanemura K, Murayama M, Akagi T, Hashikawa T, Tominaga T, Ichikawa M, et al. Neurodegeneration with tau accumulation in a transgenic mouse expressing V337M human tau. J Neurosci 2002;22:133–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lim F, Hernandez F, Lucas JJ, Gomez-Ramos P, Moran MA, Avila J. FTDP-17 mutations in tau transgenic mice provoke lysosomal abnormalities and Tau filaments in forebrain. Mol Cell Neurosci 2001;18:702–14. [DOI] [PubMed] [Google Scholar]
  • 109.Tatebayashi Y, Miyasaka T, Chui DH, Akagi T, Mishima K, Iwasaki K, et al. Tau filament formation and associative memory deficit in aged mice expressing mutant (R406W) human tau. Proc Natl Acad Sci USA 2002;99:13896–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gotz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 2001;293:1491–5. [DOI] [PubMed] [Google Scholar]
  • 111.Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia-Alloza M, de Calignon A, Rozkalne A, et al. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer’s disease. Nature 2008;451:720–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.De Jonghe C, Esselens C, Kumar-Singh S, Craessaerts K, Serneels S, Checler F, et al. Pathogenic APP mutations near the gamma-secretase cleavage site differentially affect Abeta secretion and APP C-terminal fragment stability. Hum Mol Genet 2001;10:1665–71. [DOI] [PubMed] [Google Scholar]
  • 113.Golde TE, Janus C. Homing in on intracellular Abeta? Neuron 2005;45:639–42. [DOI] [PubMed] [Google Scholar]
  • 114.Golde TE. The Abeta hypothesis: leading us to rationally-designed therapeutic strategies for the treatment or prevention of Alzheimer disease. Brain Pathol 2005;15:84–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kowalska A Genetic aspects of amyloid beta-protein fibrillogenesis in Alzheimer’s disease. Folia Neuropathol 2004;42:235–7. [PubMed] [Google Scholar]
  • 116.Theuns J, Marjaux E, Vandenbulcke M, Van Laere K, Kumar-Singh S, Bormans G, et al. Alzheimer dementia caused by a novel mutation located in the APP C-terminal intracytosolic fragment. Hum Mutat 2006;27:888–96. [DOI] [PubMed] [Google Scholar]
  • 117.Theuns J, Brouwers N, Engelborghs S, Sleegers K, Bogaerts V, Corsmit E, et al. Promoter mutations that increase amyloid precursor-protein expression are associated with Alzheimer disease. Am J Hum Genet 2006;78:936–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Walker ES, Martinez M, Brunkan AL, Goate A. Presenilin 2 familial Alzheimer’s disease mutations result in partial loss of function and dramatic changes in Abeta 42/40 ratios. J Neurochem 2005;92:294–301. [DOI] [PubMed] [Google Scholar]
  • 119.Duyckaerts C, Potier MC, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta Neuropathol 2008;115:5–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Koffie RM, Meyer-Luehmann M, Hashimoto T, Adams KW, Mielke ML, Garcia-Alloza M, et al. Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci USA 2009;106:4012–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA 1998;95:6448–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wang HW, Pasternak JF, Kuo H, Ristic H, Lambert MP, Chromy B, et al. Soluble oligomers of beta amyloid (1–42) inhibit long-term potentiation but not long-term depression in rat dentate gyrus. Brain Res 2002;924:133–40. [DOI] [PubMed] [Google Scholar]
  • 123.Walsh DM, Tseng BP, Rydel RE, Podlisny MB, Selkoe DJ. The oligomerization of amyloid beta-protein begins intracellularly in cells derived from human brain. Biochemistry 2000;39:10831–9. [DOI] [PubMed] [Google Scholar]
  • 124.Walsh DM, Klyubin I, Fadeeva JV, Rowan MJ, Selkoe DJ. Amyloid-beta oligomers: their production, toxicity and therapeutic inhibition. Biochem Soc Trans 2002;30:552–7. [DOI] [PubMed] [Google Scholar]
  • 125.Barghorn S, Nimmrich V, Striebinger A, Krantz C, Keller P, Janson B, et al. Globular amyloid beta-peptide oligomer—a homogenous and stable neuropathological protein in Alzheimer’s disease. J Neurochem 2005;95:834–47. [DOI] [PubMed] [Google Scholar]
  • 126.Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature 2006;440:352–7. [DOI] [PubMed] [Google Scholar]
  • 127.Nimmrich V, Grimm C, Draguhn A, Barghorn S, Lehmann A, Schoemaker H, et al. Amyloid beta oligomers (A beta(1–42) globulomer) suppress spontaneous synaptic activity by inhibition of P/Q-type calcium currents. J Neurosci 2008;28:788–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cotman CW, Anderson AJ. A potential role for apoptosis in neurodegeneration and Alzheimer’s disease. Mol Neurobiol 1995;10:19–45. [DOI] [PubMed] [Google Scholar]
  • 129.Gervais FG, Xu D, Robertson GS, Vaillancourt JP, Zhu Y, Huang J, et al. Involvement of caspases in proteolytic cleavage of Alzheimer’s amyloid-beta precursor protein and amyloidogenic A beta peptide formation. Cell 1999;97:395–406. [DOI] [PubMed] [Google Scholar]
  • 130.Pompl PN, Yemul S, Xiang Z, Ho L, Haroutunian V, Purohit D, et al. Caspase gene expression in the brain as a function of the clinical progression of Alzheimer disease. Arch Neurol 2003;60:369–76. [DOI] [PubMed] [Google Scholar]
  • 131.LeBlanc A, Liu H, Goodyer C, Bergeron C, Hammond J. Caspase-6 role in apoptosis of human neurons, amyloidogenesis, and Alzheimer’s disease. J Biol Chem 1999;274:23426–36. [DOI] [PubMed] [Google Scholar]
  • 132.Stadelmann C, Deckwerth TL, Srinivasan A, Bancher C, Bruck W, Jellinger K, et al. Activation of caspase-3 in single neurons and autophagic granules of granulovacuolar degeneration in Alzheimer’s disease. Evidence for apoptotic cell death. Am J Pathol 1999;155:1459–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Matsui T, Ramasamy K, Ingelsson M, Fukumoto H, Conrad C, Frosch MP, et al. Coordinated expression of caspase 8, 3 and 7 mRNA in temporal cortex of Alzheimer disease: relationship to formic acid extractable abeta42 levels. J Neuropathol Exp Neurol 2006;65:508–15. [DOI] [PubMed] [Google Scholar]
  • 134.Rohn TT, Head E, Nesse WH, Cotman CW, Cribbs DH. Activation of caspase-8 in the Alzheimer’s disease brain. Neurobiol Dis 2001;8:1006–16. [DOI] [PubMed] [Google Scholar]
  • 135.Lu DC, Rabizadeh S, Chandra S, Shayya RF, Ellerby LM, Ye X, et al. A second cytotoxic proteolytic peptide derived from amyloid beta-protein precursor. Nat Med 2000;6:397–404. [DOI] [PubMed] [Google Scholar]
  • 136.Pellegrini L, Passer BJ, Tabaton M, Ganjei JK, D’Adamio L. Alternative, non-secretase processing of Alzheimer’s beta-amyloid precursor protein during apoptosis by caspase-6 and -8. J Biol Chem 1999;274:21011–6. [DOI] [PubMed] [Google Scholar]
  • 137.McPhie DL, Golde T, Eckman CB, Yager D, Brant JB, Neve RL. beta-Secretase cleavage of the amyloid precursor protein mediates neuronal apoptosis caused by familial Alzheimer’s disease mutations. Brain Res Mol Brain Res 2001;97:103–13. [DOI] [PubMed] [Google Scholar]
  • 138.Koo EH. The beta-amyloid precursor protein (APP) and Alzheimer’s disease: does the tail wag the dog? Traffic 2002;3:763–70. [DOI] [PubMed] [Google Scholar]
  • 139.Zheng H, Koo EH. The amyloid precursor protein: beyond amyloid. Mol Neurodegener 2006;1:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Lu DC, Soriano S, Bredesen DE, Koo EH. Caspase cleavage of the amyloid precursor protein modulates amyloid beta-protein toxicity. J Neurochem 2003;87:733–41. [DOI] [PubMed] [Google Scholar]
  • 141.Galvan V, Zhang J, Gorostiza OF, Banwait S, Huang W, Ataie M, et al. Long-term prevention of Alzheimer’s disease-like behavioral deficits in PDAPP mice carrying a mutation in Asp664. Behav Brain Res 2008;191:246–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Harris JA, Devidze N, Halabisky B, Lo I, Thwin MT, Yu GQ, et al. Many neuronal and behavioral impairments in transgenic mouse models of Alzheimer’s disease are independent of caspase cleavage of the amyloid precursor protein. J Neurosci 2010;30:372–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Gamblin TC, Chen F, Zambrano A, Abraha A, Lagalwar S, Guillozet AL, et al. Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer’s disease. Proc Natl Acad Sci USA 2003;100:10032–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Rissman RA, Poon WW, Blurton-Jones M, Oddo S, Torp R, Vitek MP, et al. Caspase-cleavage of tau is an early event in Alzheimer disease tangle pathology. J Clin Invest 2004;114:121–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Guo H, Albrecht S, Bourdeau M, Petzke T, Bergeron C, LeBlanc AC. Active caspase-6 and caspase-6-cleaved tau in neuropil threads, neuritic plaques, and neurofibrillary tangles of Alzheimer’s disease. Am J Pathol 2004;165:523–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Albrecht S, Bogdanovic N, Ghetti B, Winblad B, LeBlanc AC. Caspase-6 activation in familial alzheimer disease brains carrying amyloid precursor protein or presenilin i or presenilin II mutations. J Neuropathol Exp Neurol 2009;68:1282–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, et al. Caspase activation precedes and leads to tangles. Nature 2010;464:1201–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Tizon B, Ribe EM, Mi W, Troy CM, Levy E. Cystatin C protects neuronal cells from amyloid-beta-induced toxicity. J Alzheimers Dis 2010;19:885–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Heneka MT, O’Banion MK. Inflammatory processes in Alzheimer’s disease. J Neuroimmunol 2007;184:69–91. [DOI] [PubMed] [Google Scholar]
  • 150.Ojala J, Alafuzoff I, Herukka SK, van Groen T, Tanila H, Pirttila T. Expression of interleukin-18 is increased in the brains of Alzheimer’s disease patients. Neurobiol Aging 2009;30:198–209. [DOI] [PubMed] [Google Scholar]
  • 151.Zuliani G, Ranzini M, Guerra G, Rossi L, Munari MR, Zurlo A, et al. Plasma cytokines profile in older subjects with late onset Alzheimer’s disease or vascular dementia. J Psychiatr Res 2007;41:686–93. [DOI] [PubMed] [Google Scholar]
  • 152.Lue LF, Rydel R, Brigham EF, Yang LB, Hampel H, Murphy GM Jr., et al. Inflammatory repertoire of Alzheimer’s disease and nondemented elderly microglia in vitro. Glia 2001;35:72–9. [DOI] [PubMed] [Google Scholar]
  • 153.Apelt J, Schliebs R. Beta-amyloid-induced glial expression of both pro- and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res 2001;894:21–30. [DOI] [PubMed] [Google Scholar]
  • 154.Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol 2008;9:857–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kummer JA, Broekhuizen R, Everett H, Agostini L, Kuijk L, Martinon F, et al. Inflammasome components NALP 1 and 3 show distinct but separate expression profiles in human tissues suggesting a site-specific role in the inflammatory response. J Histochem Cytochem 2007;55:443–52. [DOI] [PubMed] [Google Scholar]
  • 156.Petrilli V, Papin S, Dostert C, Mayor A, Martinon F, Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ 2007;14:1583–9. [DOI] [PubMed] [Google Scholar]
  • 157.de Rivero Vaccari JP, Lotocki G, Marcillo AE, Dietrich WD, Keane RW. A molecular platform in neurons regulates inflammation after spinal cord injury. J Neurosci 2008;28:3404–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Frederick Lo C, Ning X, Gonzales C, Ozenberger BA. Induced expression of death domain genes NALP1 and NALP5 following neuronal injury. Biochem Biophys Res Commun 2008;366:664–9. [DOI] [PubMed] [Google Scholar]
  • 159.Colom LV, Diaz ME, Beers DR, Neely A, Xie WJ, Appel SH. Role of potassium channels in amyloid-induced cell death. J Neurochem 1998;70:1925–34. [DOI] [PubMed] [Google Scholar]
  • 160.Yu SP, Farhangrazi ZS, Ying HS, Yeh CH, Choi DW. Enhancement of outward potassium current may participate in beta-amyloid peptide-induced cortical neuronal death. Neurobiol Dis 1998;5:81–8. [DOI] [PubMed] [Google Scholar]
  • 161.Troy CM, Rabacchi SA, Friedman WJ, Frappier TF, Brown K, Shelanski ML. Caspase-2 mediates neuronal cell death induced by beta-amyloid. J Neurosci 2000;20:1386–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, et al. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 2000;403:98–103. [DOI] [PubMed] [Google Scholar]
  • 163.Song S, Lee H, Kam TI, Tai ML, Lee JY, Noh JY, et al. E2–25K/Hip-2 regulates caspase-12 in ER stress-mediated Abeta neurotoxicity. J Cell Biol 2008;182:675–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Saleh M, Vaillancourt JP, Graham RK, Huyck M, Srinivasula SM, Alnemri ES, et al. Differential modulation of endotoxin responsiveness by human caspase-12 polymorphisms. Nature 2004;429:75–9. [DOI] [PubMed] [Google Scholar]
  • 165.Hitomi J, Katayama T, Eguchi Y, Kudo T, Taniguchi M, Koyama Y, et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death. J Cell Biol 2004;165:347–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Vaisid T, Barnoy S, Kosower NS. Calpain activates caspase-8 in neuron-like differentiated PC12 cells via the amyloid-beta-peptide and CD95 pathways. Int J Biochem Cell Biol 2009;41:2450–8. [DOI] [PubMed] [Google Scholar]
  • 167.Vaisid T, Kosower NS, Elkind E, Barnoy S. Amyloid beta peptide toxicity in differentiated PC12 cells: calpain-calpastatin, caspase, and membrane damage. J Neurosci Res 2008;86:2314–25. [DOI] [PubMed] [Google Scholar]
  • 168.Lu DC, Shaked GM, Masliah E, Bredesen DE, Koo EH. Amyloid beta protein toxicity mediated by the formation of amyloid-beta protein precursor complexes. Ann Neurol 2003;54:781–9. [DOI] [PubMed] [Google Scholar]
  • 169.Lin KF, Chang RC, Suen KC, So KF, Hugon J. Modulation of calcium/calmodulin kinase-II provides partial neuroprotection against beta-amyloid peptide toxicity. Eur J Neurosci 2004;19:2047–55. [DOI] [PubMed] [Google Scholar]
  • 170.Ryan SD, Whitehead SN, Swayne LA, Moffat TC, Hou W, Ethier M, et al. Amyloid-{beta}42 signals tau hyperphosphorylation and compromises neuronal viability by disrupting alkylacyl-glycerophosphocholine metabolism. Proc Natl Acad Sci USA 2008;97:60–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Tesco G, Koh YH, Kang EL, Cameron AN, Das S, Sena-Esteves M, et al. Depletion of GGA3 stabilizes BACE and enhances beta-secretase activity. Neuron 2007;54:721–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Warlow CP. Epidemiology of stroke. Lancet 1998;352(Suppl. 3):1–4. [DOI] [PubMed] [Google Scholar]
  • 173.del Zoppo GJ. Stroke and neurovascular protection. N Engl J Med 2006;354:553–5. [DOI] [PubMed] [Google Scholar]
  • 174.Ribe EM, Serrano-Saiz E, Akpan N, Troy CM. Mechanisms of neuronal death in disease: defining the models and the players. Biochem J 2008;415:165–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Garcia JH, Liu KF, Ho KL. Neuronal necrosis after middle cerebral artery occlusion in Wistar rats progresses at different time intervals in the caudoputamen and the cortex. Stroke 1995;26:636–42 [Discussion: 643]. [DOI] [PubMed] [Google Scholar]
  • 176.Lipton P Ischemic cell death in brain neurons. Physiol Rev 1999;79:1431–568. [DOI] [PubMed] [Google Scholar]
  • 177.Linnik MD, Zobrist RH, Hatfield MD. Evidence supporting a role for programmed cell death in focal cerebral ischemia in rats. Stroke 1993;24:2002–8 [Discussion: 2008–2009]. [DOI] [PubMed] [Google Scholar]
  • 178.Charriaut-Marlangue C, Margaill I, Represa A, Popovici T, Plotkine M, Ben-Ari Y. Apoptosis and necrosis after reversible focal ischemia: an in situ DNA fragmentation analysis. J Cereb Blood Flow Metab 1996;16:186–94. [DOI] [PubMed] [Google Scholar]
  • 179.Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, et al. Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell 1995;80:401–11. [DOI] [PubMed] [Google Scholar]
  • 180.Benchoua A, Guegan C, Couriaud C, Hosseini H, Sampaio N, Morin D, et al. Specific caspase pathways are activated in the two stages of cerebral infarction. J Neurosci 2001;21:7127–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972;26:239–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Mattson MP. Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol 2000;1:120–9. [DOI] [PubMed] [Google Scholar]
  • 183.Love S. Apoptosis and brain ischaemia. Prog Neuropsychopharmacol Biol Psychiatry 2003;27:267–82. [DOI] [PubMed] [Google Scholar]
  • 184.Ferrer I Apoptosis: future targets for neuroprotective strategies. Cerebrovasc Dis 2006;21 (Suppl 2):9–20. [DOI] [PubMed] [Google Scholar]
  • 185.Martin LJ, Al-Abdulla NA, Brambrink AM, Kirsch JR, Sieber FE, Portera-Cailliau C. Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation: a perspective on the contributions of apoptosis and necrosis. Brain Res Bull 1998;46:281–309. [DOI] [PubMed] [Google Scholar]
  • 186.Pan J, Konstas AA, Bateman B, Ortolano GA, Pile-Spellman J. Reperfusion injury following cerebral ischemia: pathophysiology, MR imaging, and potential therapies. Neuroradiology 2007;49:93–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Kahles T, Luedike P, Endres M, Galla HJ, Steinmetz H, Busse R, et al. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 2007;38:3000–6. [DOI] [PubMed] [Google Scholar]
  • 188.Traystman RJ. Animal models of focal and global cerebral ischemia. ILAR J 2003;44:85–95. [DOI] [PubMed] [Google Scholar]
  • 189.Richard Green A, Odergren T, Ashwood T. Animal models of stroke: do they have value for discovering neuroprotective agents? Trends Pharmacol Sci 2003;24:402–8. [DOI] [PubMed] [Google Scholar]
  • 190.Dronne MA, Grenier E, Chapuisat G, Hommel M, Boissel JP. A modelling approach to explore some hypotheses of the failure of neuroprotective trials in ischemic stroke patients. Prog Biophys Mol Biol 2008;97:60–78. [DOI] [PubMed] [Google Scholar]
  • 191.Rothman SM. Synaptic activity mediates death of hypoxic neurons. Science 1983;220:536–7. [DOI] [PubMed] [Google Scholar]
  • 192.Goldberg MP, Choi DW. Combined oxygen and glucose deprivation in cortical cell culture: calcium-dependent and calcium-independent mechanisms of neuronal injury. J Neurosci 1993;13:3510–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Gwag BJ, Lobner D, Koh JY, Wie MB, Choi DW. Blockade of glutamate receptors unmasks neuronal apoptosis after oxygen-glucose deprivation in vitro. Neuroscience 1995;68:615–9. [DOI] [PubMed] [Google Scholar]
  • 194.Laake JH, Haug FM, Wieloch T, Ottersen OP. A simple in vitro model of ischemia based on hippocampal slice cultures and propidium iodide fluorescence. Brain Res Brain Res Protoc 1999;4:173–84. [DOI] [PubMed] [Google Scholar]
  • 195.Schurr A, Payne RS, Miller JJ, Rigor BM. Study of cerebral energy metabolism using the rat hippocampal slice preparation. Methods 1999;18:117–26. [DOI] [PubMed] [Google Scholar]
  • 196.Schurr A, Tseng MT, West CA, Rigor BM. Taurine improves the recovery of neuronal function following cerebral hypoxia: an in vitro study. Life Sci 1987;40:2059–66. [DOI] [PubMed] [Google Scholar]
  • 197.Calabresi P, Centonze D, Bernardi G. Cellular factors controlling neuronal vulnerability in the brain: a lesson from the striatum. Neurology 2000;55:1249–55. [DOI] [PubMed] [Google Scholar]
  • 198.Pedersen JZ, Bernardi G, Centonze D, Pisani A, Rossi L, Rotilio G, et al. Hypoglycemia, hypoxia, and ischemia in a corticostriatal slice preparation: electrophysiologic changes and ascorbyl radical formation. J Cereb Blood Flow Metab 1998;18:868–75. [DOI] [PubMed] [Google Scholar]
  • 199.Benveniste H, Drejer J, Schousboe A, Diemer NH. Elevation of the extracellular concentrations of glutamate and aspartate in rat hippocampus during transient cerebral ischemia monitored by intracerebral microdialysis. J Neurochem 1984;43:1369–74. [DOI] [PubMed] [Google Scholar]
  • 200.Choi DW, Rothman SM. The role of glutamate neurotoxicity in hypoxic-ischemic neuronal death. Annu Rev Neurosci 1990;13:171–82. [DOI] [PubMed] [Google Scholar]
  • 201.Simon RP, Swan JH, Griffiths T, Meldrum BS. Blockade of N-methyl-d-aspartate receptors may protect against ischemic damage in the brain. Science 1984;226:850–2. [DOI] [PubMed] [Google Scholar]
  • 202.Malagelada C, Xifro X, Minano A, Sabria J, Rodriguez-Alvarez J. Contribution of caspase-mediated apoptosis to the cell death caused by oxygen–glucose deprivation in cortical cell cultures. Neurobiol Dis 2005;20:27–37. [DOI] [PubMed] [Google Scholar]
  • 203.Meli E, Pangallo M, Baronti R, Chiarugi A, Cozzi A, Pellegrini-Giampietro DE, et al. Poly (ADP-ribose) polymerase as a key player in excitotoxicity and post-ischemic brain damage. Toxicol Lett 2003;139:153–62. [DOI] [PubMed] [Google Scholar]
  • 204.Moroni F, Meli E, Peruginelli F, Chiarugi A, Cozzi A, Picca R, et al. Poly(ADP-ribose) polymerase inhibitors attenuate necrotic but not apoptotic neuronal death in experimental models of cerebral ischemia. Cell Death Differ 2001;8:921–32. [DOI] [PubMed] [Google Scholar]
  • 205.Ray AM, Owen DE, Evans ML, Davis JB, Benham CD. Caspase inhibitors are functionally neuroprotective against oxygen glucose deprivation induced CA1 death in rat organotypic hippocampal slices. Brain Res 2000;867:62–9. [DOI] [PubMed] [Google Scholar]
  • 206.Gottron FJ, Ying HS, Choi DW. Caspase inhibition selectively reduces the apoptotic component of oxygen-glucose deprivation-induced cortical neuronal cell death. Mol Cell Neurosci 1997;9:159–69. [DOI] [PubMed] [Google Scholar]
  • 207.Taranukhin AG, Taranukhina EY, Saransaari P, Djatchkova IM, Pelto-Huikko M, Oja SS. Taurine reduces caspase-8 and caspase-9 expression induced by ischemia in the mouse hypothalamic nuclei. Amino Acids 2008;34:169–74. [DOI] [PubMed] [Google Scholar]
  • 208.Le DA, Wu Y, Huang Z, Matsushita K, Plesnila N, Augustinack JC, et al. Caspase activation and neuroprotection in caspase-3-deficient mice after in vivo cerebral ischemia and in vitro oxygen glucose deprivation. Proc Natl Acad Sci USA 2002;99:15188–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Shang YC, Chong ZZ, Hou J, Maiese K. FoxO3a governs early microglial proliferation and employs mitochondrial depolarization with caspase 3, 8, and 9 cleavage during oxidant induced apoptosis. Curr Neurovasc Res 2009;6:223–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Troy CM, Derossi D, Prochiantz A, Greene LA, Shelanski ML. Downregulation of Cu/Zn superoxide dismutase leads to cell death via the nitric oxide-peroxynitrite pathway. J Neurosci 1996;16:253–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Davidson TJ, Harel S, Arboleda VA, Prunell GF, Shelanski ML, Greene LA, et al. Highly efficient small interfering RNA delivery to primary mammalian neurons induces MicroRNA-like effects before mRNA degradation. J Neurosci 2004;24:10040–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Fujimura M, Morita-Fujimura Y, Noshita N, Sugawara T, Kawase M, Chan PH. The cytosolic antioxidant copper/zinc-superoxide dismutase prevents the early release of mitochondrial cytochrome c in ischemic brain after transient focal cerebral ischemia in mice. J Neurosci 2000;20:2817–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Kondo T, Reaume AG, Huang TT, Carlson E, Murakami K, Chen SF, et al. Reduction of CuZn-superoxide dismutase activity exacerbates neuronal cell injury and edema formation after transient focal cerebral ischemia. J Neurosci 1997;17:4180–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Boutin H, LeFeuvre RA, Horai R, Asano M, Iwakura Y, Rothwell NJ. Role of IL-1alpha and IL-1beta in ischemic brain damage. J Neurosci 2001;21:5528–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Rothwell N Interleukin-1 and neuronal injury: mechanisms, modification, and therapeutic potential. Brain Behav Immun 2003;17:152–7. [DOI] [PubMed] [Google Scholar]
  • 216.Friedlander RM, Gagliardini V, Hara H, Fink KB, Li W, MacDonald G, et al. Expression of a dominant negative mutant of interleukin-1 beta converting enzyme in transgenic mice prevents neuronal cell death induced by trophic factor withdrawal and ischemic brain injury. J Exp Med 1997;185:933–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Schielke GP, Yang GY, Shivers BD, Betz AL. Reduced ischemic brain injury in interleukin-1 beta converting enzyme-deficient mice. J Cereb Blood Flow Metab 1998;18:180–5. [DOI] [PubMed] [Google Scholar]
  • 218.Allan SM, Tyrrell PJ, Rothwell NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol 2005;5:629–40. [DOI] [PubMed] [Google Scholar]
  • 219.Ferrer I, Planas AM. Signaling of cell death and cell survival following focal cerebral ischemia: life and death struggle in the penumbra. J Neuropathol Exp Neurol 2003;62:329–39. [DOI] [PubMed] [Google Scholar]
  • 220.Niizuma K, Endo H, Nito C, Myer DJ, Kim GS, Chan PH. The PIDDosome mediates delayed death of hippocampal CA1 neurons after transient global cerebral ischemia in rats. Proc Natl Acad Sci USA 2008;105:16368–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Velier JJ, Ellison JA, Kikly KK, Spera PA, Barone FC, Feuerstein GZ. Caspase-8 and caspase-3 are expressed by different populations of cortical neurons undergoing delayed cell death after focal stroke in the rat. J Neurosci 1999;19:5932–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Guegan C, Sola B. Early and sequential recruitment of apoptotic effectors after focal permanent ischemia in mice. Brain Res 2000;856:93–100. [DOI] [PubMed] [Google Scholar]
  • 223.Love S, Barber R, Srinivasan A, Wilcock GK. Activation of caspase-3 in permanent and transient brain ischaemia in man. NeuroReport 2000;11:2495–9. [DOI] [PubMed] [Google Scholar]
  • 224.Giffard RG, Swanson RA. Ischemia-induced programmed cell death in astrocytes. Glia 2005;50:299–306. [DOI] [PubMed] [Google Scholar]
  • 225.Duan SR, Wang JX, Wang J, Xu R, Zhao JK, Wang DS. Ischemia induces endoplasmic reticulum stress and cell apoptosis in human brain. Neurosci Lett 2010;475:132–5. [DOI] [PubMed] [Google Scholar]
  • 226.Harrison DC, Davis RP, Bond BC, Campbell CA, James MF, Parsons AA, et al. Caspase mRNA expression in a rat model of focal cerebral ischemia. Brain Res Mol Brain Res 2001;89:133–46. [DOI] [PubMed] [Google Scholar]
  • 227.Desmond DW, Moroney JT, Sano M, Stern Y. Incidence of dementia after ischemic stroke: results of a longitudinal study. Stroke 2002;33:2254–60. [DOI] [PubMed] [Google Scholar]
  • 228.Tatemichi TK, Desmond DW, Stern Y, Paik M, Sano M, Bagiella E. Cognitive impairment after stroke: frequency, patterns, and relationship to functional abilities. J Neurol Neurosurg Psychiatry 1994;57:202–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Tatemichi TK, Paik M, Bagiella E, Desmond DW, Stern Y, Sano M, et al. Risk of dementia after stroke in a hospitalized cohort: results of a longitudinal study. Neurology 1994;44:1885–91. [DOI] [PubMed] [Google Scholar]
  • 230.Wen Y, Yang S, Liu R, Brun-Zinkernagel AM, Koulen P, Simpkins JW. Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and Alzheimer’s disease-like tauopathy in female rats. J Biol Chem 2004;279:22684–92. [DOI] [PubMed] [Google Scholar]
  • 231.Nihashi T, Inao S, Kajita Y, Kawai T, Sugimoto T, Niwa M, et al. Expression and distribution of beta amyloid precursor protein and beta amyloid peptide in reactive astrocytes after transient middle cerebral artery occlusion. Acta Neurochir (Wien) 2001;143:287–95. [DOI] [PubMed] [Google Scholar]
  • 232.van Groen T, Puurunen K, Maki HM, Sivenius J, Jolkkonen J. Transformation of diffuse beta-amyloid precursor protein and beta-amyloid deposits to plaques in the thalamus after transient occlusion of the middle cerebral artery in rats. Stroke 2005;36:1551–6. [DOI] [PubMed] [Google Scholar]
  • 233.Xiong M, Zhang T, Zhang LM, Lu SD, Huang YL, Sun FY. Caspase inhibition attenuates accumulation of beta-amyloid by reducing beta-secretase production and activity in rat brains after stroke. Neurobiol Dis 2008;32:433–41. [DOI] [PubMed] [Google Scholar]
  • 234.Abrahamson EE, Ikonomovic MD, Ciallella JR, Hope CE, Paljug WR, Isanski BA, et al. Caspase inhibition therapy abolishes brain trauma-induced increases in Abeta peptide: implications for clinical outcome. Exp Neurol 2006;197:437–50. [DOI] [PubMed] [Google Scholar]
  • 235.Chae SS, Yoo CB, Jo C, Yun SM, Jo SA, Koh YH. Caspases-2 and -8 are involved in the presenilin1/gamma-secretase-dependent cleavage of amyloid precursor protein after the induction of apoptosis, J Neurosci Res 2010;88:1926–33. [DOI] [PubMed] [Google Scholar]
  • 236.de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, et al. Caspase activation precedes and leads to tangles, Nature 2010;464:1201–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Su JH, Zhao M, Anderson AJ, Srinivasan A, Cotman CW. Activated caspase-3 expression in Alzheimer’s and aged control brain: correlation with Alzheimer pathology. Brain Res 2001;898:350–7. [DOI] [PubMed] [Google Scholar]
  • 238.Louneva N, Cohen JW, Han LY, Talbot K, Wilson RS, Bennett DA, et al. Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer’s disease. Am J Pathol 2008;173:1488–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Matsuzaki S, Hiratsuka T, Kuwahara R, Katayama T, Tohyama M. Caspase-4 is partially cleaved by calpain via the impairment of Ca2+ homeostasis under the ER stress, Neurochem Int 2010;56:352–6. [DOI] [PubMed] [Google Scholar]
  • 240.Rohn TT, Rissman RA, Davis MC, Kim YE, Cotman CW, Head E. Caspase-9 activation and caspase cleavage of tau in the Alzheimer’s disease brain. Neurobiol Dis 2002;11:341–54. [DOI] [PubMed] [Google Scholar]
  • 241.Ishige K, Takagi N, Imai T, Rausch WD, Kosuge Y, Kihara T, et al. Role of caspase-12 in amyloid beta-peptide-induced toxicity in organotypic hippocampal slices cultured for long periods. J Pharmacol Sci 2007;104:46–55. [DOI] [PubMed] [Google Scholar]
  • 242.Friedlander RM, Brown RH, Gagliardini V, Wang J, Yuan J. Inhibition of ICE slows ALS in mice [letter] [published erratum appears in Nature 1998 Apr 9;392(6676):560]. Nature 1997;388:31. [DOI] [PubMed] [Google Scholar]
  • 243.Krajewska M, Rosenthal RE, Mikolajczyk J, Stennicke HR, Wiesenthal T, Mai J, et al. Early processing of Bid and caspase-6, -8, -10, -14 in the canine brain during cardiac arrest and resuscitation. Exp Neurol 2004;189:261–79. [DOI] [PubMed] [Google Scholar]

RESOURCES