Open access

Caspases in Alzheimer’s Disease

Written By

Yan Zhang

Submitted: 20 April 2012 Published: 15 May 2013

DOI: 10.5772/54627

From the Edited Volume

Neurodegenerative Diseases

Edited by Uday Kishore

Chapter metrics overview

2,520 Chapter Downloads

View Full Metrics

1. Introduction

In AD, a significant synaptic loss ranging from 20% to 50% is reported. Biochemistry, electron microscopy and immunocytochemistry have shown a decrease in synaptic density, presynaptic terminals, synaptic vesicle and synaptic protein markers in AD brains compared with the normal aged controls (Terry et al., 1991; Geula, 1998; Larson et al., 1999; Yao et al., 1999; Ashe, 2000; Baloyannis et al., 2000; Terry, 2000; Masliah, 2001; Masliah et al., 2001; Price et al., 2001; Scheff and Price, 2001; Scheff et al., 2001; Stephan et al., 2001; Callahan et al., 2002; Chan et al., 2002; Dodd, 2002). Although synaptic loss is remarkable in AD, it is not specific to AD. Reduction in synaptic density is also found in Pick’s disease, Huntington’s disease, Parkinson’s disease as well as in vascular dementia (Geula, 1998; Larson et al., 1999; Yao et al., 1999; Ashe, 2000; Baloyannis et al., 2000; Terry, 2000; Masliah, 2001; Masliah et al., 2001; Price et al., 2001; Scheff and Price, 2001; Scheff et al., 2001; Stephan et al., 2001; Callahan et al., 2002; Chan et al., 2002; Dodd, 2002).

Since one of the most important physiological functions of synapses is to release and accept neurotransmitters, the changes of activity of these neurotransmitters in neurodegenerative diseases have also been intensively studied (Terry, 2000). In AD, most significant lesions happen in the cholinergic, adrenergic and serotoninergic systems (Davies and Maloney, 1976; Geula, 1998; Larson et al., 1999; Yao et al., 1999; Ashe, 2000; Baloyannis et al., 2000; Terry, 2000; Masliah, 2001; Masliah et al., 2001; Price et al., 2001; Scheff and Price, 2001; Scheff et al., 2001; Stephan et al., 2001; Callahan et al., 2002; Chan et al., 2002; Dodd, 2002). Some other peptidergic neurotransmitters also decrease in AD, such as somatostatin, neuropeptide Y and substance P (Terry, 2000).

Synaptic loss might be one of the first events in AD development (Terry et al., 1991; Terry, 2000; Selkoe, 2002). Decrease in presynaptic terminals, synaptic vesicle and synaptic protein markers occur in very early stage of AD (Ashe, 2000; Terry, 2000; Masliah et al., 2001; Price et al., 2001; Scheff et al., 2001; Callahan et al., 2002; Chan et al., 2002; Dodd, 2002). In the transgenic mice with FAD mutations, synaptophysin, marker for presynaptic protein, decreases before the appearance of Aβ deposits and formation of plaques (Hamos et al., 1989; Masliah et al., 1989; Selkoe, 2002). Most important, the decline of function of synaptic transmission occurs even before synaptic structural changes (Masliah, 2001; Scheff and Price, 2001; Chan et al., 2002; Selkoe, 2002). Long-term potentiation (LTP) is commonly accepted as a measurement for capacity of synaptic plasticity, which is the basis of learning, memory and complex information processing. The incidence and duration of LTP formation are used as an indication for formation and maintenance of working memory. Several lines of FAD mutant transgenic mice show a decline in the formation of LTP and synaptic excitation before the appearance of synaptic loss, plaques and other AD pathology (Geula, 1998; Ashe, 2000; Masliah, 2001; Masliah et al., 2001; Scheff and Price, 2001; Callahan et al., 2002; Chan et al., 2002; Selkoe, 2002). In summary, synaptic loss seems to appear earlier than all other pathological markers and the functional loss of synapses may be responsible for the initiation of cognitive decline in AD patients.

Advertisement

2. Neuronal loss in AD

Synaptic loss and degeneration induce neuronal dysfunction and cell body loss. Neuronal loss in the cerebral cortex and the hippocampus is a hallmark feature of AD. Some of AD patients at late stage of the disease can have a severe decrease in brain volume and weight due to either neuronal loss or atrophy (Smale et al., 1995; Cotman and Su, 1996; Gomez-Isla et al., 1996; Gomez-Isla et al., 1997; Li et al., 1997; Su et al., 1997; Gomez-Isla et al., 1999). Assumption-based and design-based unbiased stereological cell counting show decreased density of neurons in the cerebral cortex, the entorhinal cortex, the association cortex, the basal nucleus of Meynert, the locus coeruleus and the dorsal raphe of AD brains (Bondareff et al., 1982; Lippa et al., 1992; Gomez-Isla et al., 1996; Gomez-Isla et al., 1997; Gomez-Isla et al., 1999; Colle et al., 2000). Profound neuronal loss is especially observed in the entorhinal cortex in the mild AD brains (Gomez-Isla et al., 1996; Gomez-Isla et al., 1997; Gomez-Isla et al., 1999). Besides AD, significant neuronal loss is also observed in the entorhinal cortex in very mild cognitive impairment patient brains (Gomez-Isla et al., 1996; Gomez-Isla et al., 1997; Gomez-Isla et al., 1999). These data suggest that neuronal loss may be one of the early events before formation of SPs and NFTs in AD development.

The loss of cholinergic neurons in AD is widely studied. The hippocampus and cortex receive major cholinergic input from the basal forebrain nuclei (Hohmann et al., 1987). Decrease of choline acetyltransferase activity and acetylcholine synthesis correlate well with the degree of cognitive impairment in AD patients (Mesulam, 1986; Hohmann et al., 1987; Pearson and Powell, 1987). Cholinergic neuronal lesion can be detected in the patients that have showed clinical memory loss symptoms for less than 1 year (Whitehouse et al., 1981; Whitehouse et al., 1982; Francis et al., 1993; Weinstock, 1997). However, markers for dopamine, γ-aminobutyric acid (GABA), or somatostatin are not altered (Whitehouse et al., 1981; Whitehouse et al., 1982; Francis et al., 1993). These results suggest that cholinergic neuronal loss is probably one of the early events in AD. Besides the main pathology discussed above, some other pathologies of AD include granulovacuolar degeneration, cerebral amyloid angiopathy, blood-brain barrier disorder, white matter lesions, neuropil thread and gliosis (Jellinger, 2002a; Jellinger, 2002b, c; Jellinger and Attems, 2003).

As discussed above, stereological cell counting shows that densities of neurons in the AD cerebral cortex, the entorhinal cortex, the association cortex, the basal nucleus of Meynert, the locus coeruleus and the dorsal raphe decrease significantly compared to the age-matched non-AD controls (Bondareff et al., 1982; Lippa et al., 1992; Gomez-Isla et al., 1996; Gomez-Isla et al., 1997; Gomez-Isla et al., 1999; Colle et al., 2000). Neuronal cell loss is one of the first events during AD development. In the mild AD patient brains, remarkable neuronal cell loss of more than 40% is seen in the entorhinal cortex (Gomez-Isla et al., 1996; Gomez-Isla et al., 1997). Even in the mild cognitive impairment patient brains, significant neuronal loss is also observed in the entorhinal cortex (Gomez-Isla et al., 1996). Furthermore, the degree of neuronal loss correlates better with the clinical dementia level in AD than other pathology.

It was thought until recently that neuronal loss is mainly due to passive neurotrophic factor withdrawal. In 1988, Martin et al. (1988) showed that sympathetic neuronal death could be prevented by inhibiting RNA and protein synthesis, indicating that some of the neuronal death might be actively programmed (apoptotic) instead of passive (necrotic).

Advertisement

3. Apoptosis

Apoptosis, or programmed cell death (PCD), is a term proposed by Kerr, Wyllie and Currie in 1972 (Kerr et al., 1972) to describe a common type of cell death characterized by membrane blebbing, cell shrinkage, protein fragmentation, chromatin condensation and DNA degradation followed by rapid engulfment of corpses by surrounding cells (Kerr et al., 1972). It rapidly cleans out dysfunctional cells, limits toxic effects, saves energy and recycles molecules for future de novo synthesis.

Two major apoptotic pathways in mammalian cells are mediated through either death receptors or mitochondria (Figure 1). The so-called extrinsic pathway is initiated by death receptor CD95 (Apo-1/Fas), or other death receptors, such as tumor necrosis factor (TNF) receptor and tumor necrosis factor related apoptosis inducing ligand (TRAIL) receptor. CD95 is linked to pro-caspase-8 by an adapter factor FADD. Pro-caspase-8 has a death effecter domain (DED) which binds to the death domain (DD) on death receptor adapter FADD, and a caspase activation and recruitment domain (CARD), which binds to other downstream caspases and further processes the downstream caspases. Ligand binding signals the activation of pro-caspase-8 to form a tetramer of active caspase-8. Caspase-8 then mediates the cleavage of pro-caspase-3 and initiates the caspase activation cascade, which leads to protein and DNA cleavage and final termination of the cells. In this pathway, caspase-8 activation can be prevented by its natural inhibitors, such as cellular FADD-like interlukin-1-β-converting enzyme (FLICE/caspase-8)-inhibitory proteins (c-FLIPs) (Irmler et al., 1997; Thome et al., 1997) and apoptosis repressor with caspase recruitment domain (ARC) (Koseki et al., 1998) (Figure 1).

The intrinsic pathway is triggered mainly by internal insults such as DNA damage. Damaging insults such as UV irradiation activate tumor suppression gene product p53, a transcriptional factor. There are several hypotheses for p53 activation mechanism. First, stress-activated protein kinases, such as DNA-dependent protein kinase (DNA-PK), phosphorylates p53. This phosphorylation prevents p53 from degradation. For example, DNA-PK can be activated by DNA damage, and then phosphorylate p53 at Ser-15 in the N terminal, which prevents the interaction between p53 and its inhibitory protein MDM-2. In addition to phosphorylation, dephosphorylation, such as the one caused by 14-3-3 at Ser-376, can also enable DNA binding of p53 and activate its function. The second model states that p53 is constitutively active and is regulated by the negative regulator MDM-2. MDM-2 protein can bind to p53 and send it out of the nucleus for degradation. Interestingly, the MDM-2 gene can be activated by p53, therefore, p53 is negatively self-regulated. After activation, p53 acts as a transcriptional factor controlling the expression of certain genes. These genes are involved in cell cycle control (eg. p21, GADD45, 14-3-3, CyclinD1, CyclinG), DNA repair (eg. GADD45, p21), apoptosis (eg. Bax, Bcl-2, FASL, DR5), angiogenesis (eg. TSP-1, BAI1) and cellular stress response (eg. TP53TG1, CSR, PIG3).

During apoptosis, p53 activates transcription of the pro-apoptotic Bax and, at the same time, transcriptionally represses the anti-apoptotic Bcl-2. A family of Bcl-2 proteins is implicated in apoptosis. This family contains three subgroups of proteins, some of them are pro-apoptotic while some are anti-apoptotic. The Group I proteins, such as Bcl-2, have a transmembrane domain and conserved Bcl-2 homology (BH) 1-4 domains. The Group II lacks the BH4 domain, such as Bax, while in the Group III, only the BH3 domain is in common, such as Bid and Bik (Hengartner, 2000). Bax is a pro-apoptotic protein causing the depolarization of mitochondrial membrane and release of cytochrome c from the mitochondria to the cytosol. The detailed mechanism of Bax leading to cytochrome c release is unknown. Bax is located in the cytosol as monomers and upon the apoptotic stimulation, Bax oligomerizes and translocates to the mitochondria. There are several models suggested to explain how Bax oligomers cause intermembrane protein cytochrome c release (Degli Esposti and Dive, 2003). First, Bax oligomers may form a pore structure on the mitochondria outer membrane leading to cytochrome c release. Second, interaction between Bax and other mitochondrial proteins, such as voltage dependent anion selective channel (VDAC) and adenosine nucleotide transporter (ANT) may induce pore formation by VDAC and ANT, through which cytochrome c is released. Third, the pore may form on the membrane by low-selective ion channels and induce cytochrome c release through these channels (Degli Esposti and Dive, 2003). Cytochrome c, together with the adapter molecule Apaf-1 and pro-caspase-9, forms an apoptosome that cleaves pro-caspase-3 into its active form and initiates apoptosis. Bcl-2, the other member of Bcl-2 family in the Group I, is an anti-apoptotic protein preventing Bax-mediated cytochrome c release efficiently.

The two apoptotic pathways cross at Bid, a pro-apoptotic protein from Bcl-2 family Group III. Bid can be cleaved by active caspase-8 and the truncated Bid translocates to the mitochondrial membrane, increases mitochondrial outer membrane permeability, facilitates pore formation and potentiates cytochrome c release on the mitochondrial outer membrane (Figure 1) (Hengartner, 2000).

This apoptosis machinery is self-amplifying. For example, second mitochondria-derived activator of caspase/direct IAP-binding protein with low pI (Smac/DIABLO) and apoptosis-inducing factor (AIF) proteins (Hengartner, 2000; Cregan et al., 2002) are released from the mitochondria with cytochrome c to facilitate apoptosis. In addition, recent studies showed that caspase-3, -6, -7 and –8 can initiate cytochrome c release by activating cytosolic factors (Figure 1). Since cytochrome c acts as an initiator for the caspase activation cascade, this self-amplified loop facilitates cellular apoptosis (Figure 1) (Bossy-Wetzel and Green, 1999).

Figure 1.

Schematic drawing of two major pathways involved in apoptosis. Schematic digram showing the two major pathways mediating apoptosis. The extrinsic pathway is mediated by death receptors, such as FADD, and caspase-8 activation. The intrinsic pathway is induced by DNA damage and mediated by caspase-9. IAP: inhibitor of apoptosis protein, Smac/DIABLO: second mitochondria-derived activator of caspase/direct IAP-binding protein with low pI, AIF: apoptosis inducing factor.

Advertisement

4. Apoptosis involvement in neuronal cell loss of AD

There is evidence for apoptosis involvement in neuronal loss in AD, but the evidence so far is not sufficient to support a significant role for apoptosis in the neuronal cell loss in AD. Evidence of apoptosis in AD is as follows. First, overexpression of FAD-related mutations causes apoptosis in the transfected cell lines, cultured neurons and transgenic mice. For example, overexpression of FAD mutations of APPV642I, APPV642F and APPV642G in COS or F11 cells increases number of apoptotic cells as determined by DNA fragmentation and terminal dUTP nick end labeling (TUNEL) staining assay, which can be inhibited by anti-apoptotic protein Bcl-2 overexpression (Yamatsuji et al., 1996). These results support the role of FAD mutations in inducing apoptosis. Similarly, the data from transgenic mice confirm the above observations. Transgenic mice overexpressing FAD mutant APPV717F develop neuritic dystrophy similar to some pathological features in AD patients (Games et al., 1995; Holcomb et al., 1998). The degenerating neurons in these mice also show typical apoptotic features, such as chromatin segmentation and condensation, and positive TUNEL staining (Nijhawan et al., 2000). However, in these studies, the FAD mutant proteins are normally overexpressed far beyond the physiological levels. However, FAD neurons do not necessarily undergo apoptosis. In the mutant PS1 expressing neurons, increased apoptosis is not reported (Bursztajn et al., 1998). Also, there is no neuronal loss found in mutant PS1 transgenic mice (Takeuchi et al., 2000).

Second, some studies indicate apoptosis in AD patient brains using in situ detection of DNA fragmentation by TUNEL staining (Su et al., 1994; Dragunow et al., 1995; Lassmann et al., 1995; Smale et al., 1995; Anderson et al., 1996; Su et al., 1996; Troncoso et al., 1996; Gervais et al., 1999; Anderson et al., 2000). However, in the AD brain tissues, some TUNEL-positive neurons show typical apoptotic morphology whereas some do not, suggesting degenerating neurons in AD may undergo both apoptosis and passive cell death, necrosis (Su et al., 1994; Troncoso et al., 1996; Lucassen et al., 1997; Yuan and Yankner, 2000). It is now commonly accepted that TUNEL staining sometimes is not specific to apoptosis (Stadelmann et al., 1998). The staining of TUNEL may indicate increased vulnerability of cells to a secondary insult, not necessarily undergoing apoptosis. On the other hand, the number of apoptotic neurons is difficult to measure precisely due to the chronic nature, relatively long progress of the disease and rapid clearance mechanism of dead cells.

Third, there are reports of changes of expression of apoptosis related proteins, such as p53, Bcl-2 and Bcl-xL, in AD brains (Paradis et al., 1996; Kitamura et al., 1997; MacGibbon et al., 1997; Su et al., 1997; Cotman, 1998; Torp et al., 1998; Tortosa et al., 1998). For example, pro-apoptotic protein p53 is increased in AD brains (Kitamura et al., 1997), while anti-apoptotic proteins Bcl-2 and Bcl-xL are decreased in AD brains (Kitamura et al., 1998; Tortosa et al., 1998). Also, another pro-apoptotic protein Bax is increased in AD brains (Paradis et al., 1996; Kitamura et al., 1997; MacGibbon et al., 1997; Su et al., 1997; Cotman, 1998; Torp et al., 1998; Tortosa et al., 1998). However, the regulation of these pro- or anti-apoptotic proteins could be primary or secondary response to insults. On the other hand, the upregulation of either pro- or anti-apoptotic proteins could be explained by either the neurons undergoing apoptosis or neurons responding against apoptotic insults to prevent initiation of apoptosis.

Therefore, in summary, to date, there is no strong evidence showing significant involvement of apoptosis in AD neuronal loss. Since caspases, a family of cysteinyl proteases, play an important role in cell death, especially in apoptosis as discussed in the previous section (Thompson, 1995; Strasser et al., 2000; Yuan and Yankner, 2000), it is of interest to identify which caspase is responsible for human neuronal cell loss, how it is regulated, and whether it can be inhibited. In addition, caspase activation may be easier to use for identification of apoptosis since it is an upstream event of DNA fragmentation. Therefore, the apoptotic cells have not been cleared yet.

Advertisement

5. Caspase involvement in apoptosis and APP metabolism in AD

5.1. Caspases

Caspases (cysteinyl aspartate-specific proteases) belong to a cysteinyl protease family that cleaves specifically after an aspartic acid. To date, 14 caspases (11 of them are found in human) have been identified in mammals. The connection between apoptosis and caspases was first reported by Yuan et al. (1993) that caspase-1 is a homolog to CED-3, a gene regulating cell death in Caenorhaditis elegans (Yuan et al., 1993). The important role of caspases in apoptosis is also supported by the strong correlation between caspase activity and apoptosis in various cell types. Furthermore, caspase inhibition prevents apoptosis both in vitro and in vivo (Yuan et al., 1993; Kuida et al., 1995; Schwartz and Milligan, 1996; Alnemri, 1997; Thornberry, 1997; Li and Yuan, 1999; Yuan and Yankner, 2000).

Inactive caspases contain a pro-domain, a large subunit and a small subunit. According to their pro-domain structure and function, caspases are divided into “inflammatory”, “initiator” and “effecter” caspases (Figure 2A) (Nicholson, 1999; Hengartner, 2000). Caspases are normally present as inactive precursors in cells. After receiving apoptotic signals, caspase pro-enzymes undergo proteolytic processing to remove the N terminal pro-domain and cleave between large and small subunits to produce the active form of a tetrameric enzyme formed by 2 copies of the large subunit and 2 copies of small subunit. To date, three caspase activation pathways are known in mammalian cells: recruitment-activation, trans-activation and autoactivation (Nicholson, 1999). Recruitment-activation is triggered by death receptors of the tumor necrosis factor receptor family. The so-called initiator caspases, namely caspase-2, -8, -9 and –10 (Figure 3A), are thought to be directly activated through the signals from death receptor (Boldin et al., 1996; Muzio et al., 1996). In addition, more recent data show that the initiator caspase-8, -9 and –10 can be activated by homodimerization of their monomeric zymogens, a so called “proximity-induced activation” (Boatright et al., 2003). Trans-activation occurs through downstream or executioner caspases, caspase-3, -6 and –7 that can be activated by direct proteolysis by their upstream initiator caspases. By such trans-activation cascade, apoptosis is well controlled and regulated (Darmon et al., 1995; Martin et al., 1996; Andrade et al., 1998). Caspase activation can also occur by activation of the dormant enzyme molecule by the already active one (autoactivation). The supporting evidence of this mechanism comes from the observation that arginine-glycine-aspartate (RGD) motif-containing peptides can induce caspase-3 autoactivation by triggering conformational changes of pro-caspase-3 (Buckley et al., 1999). A similar mechanism has been suggested for pro-caspase-8 and –2 activation (Hengartner, 2000).

Figure 2.

Caspases are cysteinyl proteases responsible for protein degradation during apoptosis. A. Schematic diagram of caspase category according to function. There are three groups of caspases according to their basic functions: inflammation, initiation and execution. B. Schematic drawing of pro-caspase-6 activation into active caspase-6. Caspase-6 activation by cleavage at the sites between pro-domain and large subunit (p20) as well as large subunit and small subunit (p10). Arrow head: cleavage. C. Schematic drawing of caspase-6-like cleavage sites on APP. There are three caspase-6-like cleavage sites on APP. Arrow: cleavage. TM: transmembrane domain.

After activation, caspases recognize four amino acid substrate sites as their targets and cleave the C terminal to an obligatory aspartic acid (XXXD). According to their specific substrates, caspases are divided into 3 groups: caspase-1, -4, -5 and –13 (substrates: WEHD and YVAD); caspase-2, -3 and -7 (substrate: DEXD) and caspase-6, -8, –9 and -10 (substrates: (I, V, L)EXD) (reviewed by (Thornberry, 1997)). Once activated, caspases cleave downstream substrates in a highly specific and rapid manner. More than 250 substrates are found including downstream caspases or apoptosis-related proteins (e.g. Bid, Bcl-2), structural proteins (e.g. lamins, actin, fodrin, gelsolin), DNA repair proteins (e.g. PARP, p21) and some other proteins involved in neurodegenerative diseases (eg. APP, tau, PSs, Huntingtin) (reviewed by (Bounhar et al., 2002).

Most of the morphological changes in apoptosis described by Kerr et al. (1972) are caused by caspases that are activated specifically in PCD. Caspase cleavage of nuclear lamins results in nuclear shrinking and blebbing (McCarthy et al., 1997; Sakahira et al., 1998). Loss of cell structure may be due to caspase cleavage of cytoskeletal proteins, such as fodrin (Vanags et al., 1996; Janicke et al., 1998) and gelsolin (Kothakota et al., 1997). DNA fragmentation is also caused by caspase-activated DNase (CAD), which is activated by caspases through removing the inhibitory subunit (ICAD) from the inactive CAD enzyme complex (Hengartner, 2000).

5.2. Involvement of caspases in neuronal loss in AD

In general, the evidence supporting the involvement of caspases in neuronal loss in AD is still not conclusive. The involvement of caspases in AD is first suggested by the finding that caspases, acting as proteases, are involved in PSs and APP metabolism and Aβ peptide generation in AD. Caspase-3 directly cleaves PSs during apoptosis (Kim et al., 1997). Caspase-3, -6, -7, -8 and –9 can cleave APP and generate Aβ or Aβ-containing fragments, therefore, giving a possibility of Aβ accumulation in AD (Barnes et al., 1998; Gervais et al., 1999; LeBlanc et al., 1999; Pellegrini et al., 1999; Weidemann et al., 1999). In chick motor neurons, caspase-3 cleaves APP and generates Aβ products (Barnes et al., 1998). In human 293 cells, caspase-9 cleaves APP at the C terminal and produces a “C31” peptide, which is cytotoxic to cells (Lu et al., 2000). Caspase-3 cleaves APP in NT-2 cells and is involved in Aβ formation (Gervais et al., 1999). APP is a caspase substrate in staurosporine-treated NT-2 cells and Fas-treated human Jurkat cells (Pellegrini et al., 1999). In addition, caspase inhibitors can prevent the formation of Aβ (Barnes et al., 1998; Gervais et al., 1999; LeBlanc et al., 1999).

Second, more direct proof of caspase involvement in AD comes from immuno-detection of active caspases in AD brain tissues, although the evidence is not sufficient enough to be conclusive. Some studies have shown the activation of caspase-8 in AD brains (Rohn et al., 2001; Rohn et al., 2002), while others found decreased level of caspase-8 in AD brains (Engidawork et al., 2001a). In yet another study, both inactive and active caspase-8 immunoreactivity are not changed in AD compared to control brains (Engidawork et al., 2001b). The cleavage fragment of caspase-9 is also detected in AD but not in the control brains (Lu et al., 2000; Goyal, 2001; Rohn et al., 2002), but the alteration of two caspase-9 activation co-factors, Apaf-1 and cytochrome c, is not detected (Engidawork et al., 2001b). Furthermore, there is recent evidence showing that pro-caspase-9 is activated through dimerization, but not cleavage (Boatright et al., 2003). Among all caspases, caspase-3 is the most intensively studied since the caspase-3 knockouts develop abnormal brains with significantly excessive numbers of neurons (Cregan et al., 1999; Keramaris et al., 2000; Simpson et al., 2001; Fernando et al., 2002). Some studies report increased caspase-3-like immunoreactivity in AD brains (Masliah et al., 1998; Gervais et al., 1999). However, the protein and mRNA levels of caspase-3 do not appear altered in AD compared to control brains (Desjardins and Ledoux, 1998; LeBlanc et al., 1999). Active caspase-3 is detected in granulovacuolar degeneration, an aging associated pathology that is not necessarily specific to AD (Stadelmann et al., 1999; Roth, 2001). Given that apoptosis results in a rapid clearance of dysfunctional cells, only small amount of caspase activation can be detected at a certain time window. Therefore, the extensive detection of active caspase-3 in some studies may be due to lack of immunospecificity of the caspase-3 antibody. Furthermore, although caspase-3 is critical for apoptosis in many cell types, it does not have significant role in the human neuronal loss in AD (Desjardins and Ledoux, 1998; LeBlanc et al., 1999; Selznick et al., 1999; Stadelmann et al., 1999).

Interestingly, LeBlanc et al. have shown that caspase-6 is activated in serum deprivation induced cell death of human neurons in primary cultured (LeBlanc et al., 1999). Therefore, could caspase-6 be the responsible caspase in human neuronal cell death in AD?

5.3. Caspase-6 is activated during human neuronal cell death

5.3.1. Introduction to caspase-6

Caspase-6 (Mch-2), located on human chromosome 4q25, is an “effecter” caspase with a short pro-domain. It recognizes VEID substrates and cleaves after the amino acid D. Its common substrates include APP (LeBlanc et al., 1999), cytoskeleton proteins, such as keratin 18 (Caulin et al., 1997), focal adhesion kinase (Gervais et al., 1998), tau (LeBlanc et al., 1999), β-catenin (Van de Craen et al., 1999), vimentin (Byun et al., 2001) and desmin (Chen et al., 2003), nuclear proteins, such as lamin A (Ruchaud et al., 2002), lamin B (Slee et al., 2001), PARP (Fernandes-Alnemri et al., 1995), DNA topoisomerase I (Samejima et al., 1999) and emerin (Columbaro et al., 2001), several transcriptional factors, such as SATB1, and AP-2α (Galande et al., 2001; Nyormoi et al., 2001). It is highly expressed in the heart, lung, liver, kidney and testis in murine tissues. There is immunodetectable caspase-6 in human brain and neurons (LeBlanc et al., 1999; Zheng et al., 1999; Harrison et al., 2001). Caspase-6 can be activated by caspase-1, -3, -7, -8 and –11 (Fernandes-Alnemri et al., 1995; Chinnaiyan et al., 1996; Orth et al., 1996). Pro-caspase-6 is a ~ 34 kDa protein, that can be cleaved into ~20 kDa (p20) and ~10 kDa (p10) fragments. The p20 and p10 fragments form a tetramer, which is the active form of the enzyme (Figure 3C) (Fernandes-Alnemri et al., 1995). Caspase-6 knockout mice do not show abnormal phenotype during development, which does not rule out the role of caspase-6 in cell death in later stage of life or under stress conditions (Zheng et al., 1999).

5.3.2. Caspase-6 involvement in human neuronal cell death and APP processing

Pro-caspase-6 decreases during apoptosis induced by serum deprivation in human neurons in primary cultures (LeBlanc et al., 1999). Moreover, caspase-6 active 10 kDa fragments is detected only in AD brains, and not in the normal aging control brains (LeBlanc et al., 1999), although thus increase is not dramatic. Given that AD is a long progressive disease, at a certain postmortem time window, there is only limited amount of cell death where caspase activation can be detected. Meanwhile, in contrast to caspase-6, changes in the levels of pro- and active caspase-3 levels are not detectable in AD brains, suggesting that caspase-6, but not caspase-3, may be involved in human neuronal cell death (LeBlanc et al., 1999). Caspase-6 can also alter APP metabolism to generate Aβ-containing fragments. There are several caspase-6-like sites on APP695 (Figure 2C). However, incubating recombinant caspase-6 and APP695-containing neuronal extract does not show APP cleavage. One of these cleavages at 591VKMD594 generates a 6.5 kDa fragment denoted “Capp6.5” containing the Aβ sequence (Figure 2C) (LeBlanc et al., 1999). Although caspase-6 does not directly induce 4 kDa Aβ, pulse-chase experiments showed that this Capp6.5 fragment is able to generate 4 kDa Aβ in human neurons (Figure 2C) (LeBlanc et al., 1999). Caspase-6 can cleave APP close to β-secretase site at D653 to further generate Aβ2-40 or Aβ2-42 (Gervais et al., 1999). In the Swedish mutation of APP, which changes the VKMD653 sequence at the β-secretase site to VNLD653, the caspase-6 cleavage of VNLD-AMC is 6 fold higher than the VKMD-AMC fluorogenic peptide in vitro (Gervais et al., 1999). Also, caspase-6 can cleave APP after the γ-secretase site at VEVD720/A (Gervais et al., 1999). Therefore, caspase-6 may process APP similar to β- and γ-secretase activity. Direct microinjecting caspase-6 into human neurons induces dramatic cell death (Zhang et al., 2000). Taken together, the above evidence suggests that caspase-6 plays important role in human neuronal cell death, Aβ formation, and may be responsible for neuronal loss in AD (LeBlanc et al., 1999).

5.4. Inhibition of active caspases

5.4.1. Natural inhibitors to caspases

The natural inhibitors to caspases include Cowpox virus product cytokine response modifier A (crmA), FLIPs, protease inhibitor 9 (PI-9), p35, ARC and inhibitor of apoptosis proteins (IAPs). CrmA is a member of serpin family, a group of serine protease inhibitors. CrmA inhibits caspases by acting as a pseudosubstrate that binds to active caspases, such as caspase-1, -4, -5, -8 and –9 (Ray et al., 1992; Komiyana et al., 1994). Besides crmA, FLIPs inhibits caspase-8 by acting as the dominant negative form to suppress caspase-8 mRNA expression (Thornberry, 1997). The mammalian homolog of crmA is PI-9. PI-9 mRNA expression can be rapidly induced by estrogen in human liver (Kanamori et al., 2000). PI-9 is a granzyme B inhibitor (GBI). Granzyme B is a 30 to 32 kDa serine protease, which cleaves peptides at aspartyl residue in the killer T thymocytes and natural killer cells. Granzyme B is involved in the perforation of target cells and then initiation of proteolysis that leads to apoptosis. Although PI-9 can inhibit granzyme B and granzyme B-mediated apoptosis, In vitro experiments do not show that PI-9 inhibits active caspases (Bird et al., 1998; Bird, 1999).

p35 is a baculoviral protein that can block the defensive apoptotic response of insect cells to viral infection (Clem et al., 1991; Ekert et al., 1999). p35 inhibits CED-3 in C. elegans and mammalian caspase-1, -3, -6, -7, -8 and -10 (Ekert et al., 1999). P35 is cleaved at its P1 residue by caspases, and the cleaved fragment forms an inhibitory complex to block caspase activation (Zhou et al., 1998). ARC interacts with caspase-2, -8 and CED-3, but not caspase-1, -3, or –9. ARC inhibits caspase-8 enzyme activity in 293 cells, and further attenuates apoptosis induced by FADD through stimulation of death receptors coupled with pro-caspase-8 (Koseki et al., 1998).

IAPs were found in a search for viral genes with a similar function to p35. A group of cellular IAP homologs are found in yeast, C. elegans, Drosophila and vertebrates (Ekert et al., 1999). To date, the IAP family contains about a dozen proteins from viruses, Drosophila, mice and humans. All known IAPs share a baculovirus inhibitory repeat (BIR) domain, which contains a number of conserved residues including a zinc-binding region. Most of the IAPs also contain a RING zinc-binding finger motif at the C terminal. Both BIR motifs and RING finger are important for IAP function (Ekert et al., 1999). The neuronal apoptosis inhibitory protein (NAIP) in human was found by searching for mutations in spinal muscular atrophy (SMA), which is characterized by degeneration of the anterior horn cells in the spinal cord. The NAIP gene is deleted in the SMA patients. During development, excessive neurons are ultimately needed to send out axons, the neurons do not target properly undergo apoptosis. It is suggested that NAIP is involved by preventing apoptosis in the “successful” cells (Miller, 1997).

The most studied IAP is the X-linked IAP (XIAP). XIAP binds to active caspase-9 small subunit through its BIR3 domain and cleaves the small subunit to inactivate caspase-9 (Srinivasula et al., 2001). On the other hand, XIAP binds to active caspase-3 or –7 through BIR2 domain and mask the active site of these caspases (Huang et al., 2001; Riedl et al., 2001; Stennicke et al., 2002). In addition, evidence shows that in insects, some IAPs interact with apoptosis-related proteins, such as Grim, Reaper and Hid (Hay et al., 1995; Miller, 1997; Vucic et al., 1997; Vucic et al., 1998). IAPs can also inhibit caspase-3 directly and cytochrome c-induced caspase-9 (Deveraux et al., 1998). However, up to now, although IAPs may inhibit caspase-6 activation by blocking upstream caspase-9 or –3 activation (Deveraux et al., 1998), there is no evidence showing that mammalian IAPs directly inhibit active caspase-6 enzyme activity.

5.4.2. Synthetic inhibitors to active caspases

Synthetic caspase inhibitors function as pseudosubstrates for active caspases and therefore, they are competitive inhibitors of active caspases (Ekert et al., 1999). The N terminal blocking groups of the pseudosubstrate peptides are usually acetyl- (Ac-) or benzocarbonyl (Z-). The biochemical property of these synthetic inhibitors depends on the chemical group linked to pseudosubstrate peptides. Aldehyde (CHO-) group inhibitors are reversible since there is no covalent bond formed between these inhibitors and caspases. These CHO inhibitors have poor cell membrane permeability, which limits largely the use of these inhibitors on live cells and animals (Schotte et al., 1999; Bounhar et al., 2002). The inhibitors linked to methylketone (chloromethylketone, cmk or fluoromethylketone, fmk) irreversibly inhibit caspases due to the formation of thyomethylketone bonds with the cysteine in the active site of caspases (Bounhar et al., 2002). These inhibitors are membrane permeable, but have less specificity of inhibitory action to caspases (Schotte et al., 1999; Bounhar et al., 2002).

Although synthetic caspase inhibitors are widely used in research, they may not be the ideal candidates for disease treatment since these inhibitors cannot inhibit caspase activation in specific certain cell types. For example, in AD, caspase-6 is the key caspase responsible for neuronal loss. If the synthetic caspase-6 inhibitor is applied and it would inhibit active caspase-6 activity in all types of cells, which has a large potential to develop cancer in cells that proliferate. If a natural inhibitor to active caspase-6 can be activated somehow specifically in human neurons, the risk of oncogenesis in the brain would be greatly reduced.

Since neuronal loss is a striking feature of AD, decreasing or suppressing neuronal cell loss may benefit early AD patients in retaining cognitive capacities and prevent or delay disease progression. Caspases seem to play a significant role in human neuronal cell loss in AD, thus it is intriguing to determine if caspase activity can be inhibited after activation by neuroprotective agents.

References

  1. 1. AlnemriE. S1997Mammalian cell death proteases: a family of highly conserved aspartate specific cysteine proteases. J Cell Biochem 643342
  2. 2. AlorainyI2000Senile scleral plaques: CT. Neuroradiology 42145148
  3. 3. AndersonA. JStoltznerSLaiFSuJNixonR. A2000Morphological and biochemical assessment of DNA damage and apoptosis in Down syndrome and Alzheimer disease, and effect of postmortem issue archival on TUNEL. Neurobiol Aging 21511524
  4. 4. AndersonA. JSuJ. HCotmanC. W1996DNA damage and apoptosis in Alzheimer’s disease: colocalization with c-Jun immunoreactivity, relationship to brain area, and effect of postmortem delay. J Neurosci 1617101719
  5. 5. AndradeFRoySNicholsonDThornberryNRosenACasciola-rosenL1998Granzyme B directly and efficiently cleaves several downstream caspase substrates: implications for CTL-induced apoptosis. Immunity 8451460
  6. 6. ArnoldS. EHymanB. TFloryJDamasioA. RVan HoesenG. W1991The topographical and neuroanatomical distribution of neurofibrillary tangles and neuritic plaques in the cerebral cortex of patients with Alzheimer’s disease. Cereb Cortex 1103116
  7. 7. AsheK. H2000Synaptic structure and function in transgenic APP mice. Ann N Y Acad Sci 9243941
  8. 8. BaloyannisS. JManolidisS. LManolidisL. S2000Synaptic alterations in the vestibulocerebellar system in Alzheimer’s disease--a Golgi and electron microscope study. Acta Otolaryngol 120247250
  9. 9. BarnesN. YLiLYoshikawaKSchwartzL. MOppenheinR. WMilliganC. E1998Increased production of amyloid precursor protein provides a substrate for caspase-3 in dying motoneurons. J Neurosci 1858695880
  10. 10. BirdC. HSuttonV. RSunJ. RHirstC. ENovakAKumarSTrapaniJ. ABirdP. I1998Selective regulation of apoptosis-the cytotoxic lymphocyte serpin protease inhibitor 9 protects against granzyme B-mediated apoptosis without perturbing the Fas cell death pathway. Mol Cell Biol 1863876398
  11. 11. BirdP. I1999Regulation of pro-apoptotic leukocyte granule serine proteinases by intracellular serpins. Immunol Cell Biol 774757
  12. 12. BoatrightK. MRenatusMScottF. LSperandioSShinHPedersenI. MRicciJ. EEdrisW. ASutherlinD. PGreenD. RSalvesenG. S2003A unified model for apical caspase activation. Mol Cell 11529541
  13. 13. BoldinM. PGoncharovT. VGoltsevY. VWallachD1996Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1 and TNF receptor-induced cell death. Cell 85803815
  14. 14. BondareffWMountjoyC. QRothM1982Loss of neurons of origin of the adrenergic projection to cerebral cortex (nucleus locus ceruleus) in senile dementia. Neurology 32164168
  15. 15. Bossy-wetzelEGreenD. R1999Caspases induce cytochrome c release from mitochondria by activating cytosolic factors. J Biol Chem 2741748417490
  16. 16. BounharYTounektiOLeBlanc A (2002Monitoring Caspases in Neuronal Cell Death, 2nd Edition. Totowa: Humana Press Inc.
  17. 17. BraakHBraakE1994Pathology of Alzheimer’s disease. In: Neurodegenerative Disease., 585613Philadelphia: Saunders.
  18. 18. BuckleySDriscollBBarskyLWeinbergKAndersonKWarburtonD1999ERK activation protects against DNA damage and apoptosis in byperoxic rat AEC2. Am J Physiol 277:L159L166.
  19. 19. BursztajnSDe SouzaRMcphieD. L1998Overexpression in neurons of human presenilin-1 familial Alzheimer disease mutant does not enhance apoptosis. J Neurosci 1897909799
  20. 20. ByunYChenFChangRTrivediMGreenK. JCrynsV. L2001Caspase cleavage of vimentin disrupts intermediate filaments and promotes apoptosis. Cell Death Differ 8443450
  21. 21. CallahanL. MVaulesW. AColemanP. D2002Progressive reduction of synaptophysin message in single neurons in Alzheimer disease. J Neuropathol Exp Neurol 61384395
  22. 22. CaulinCSalvesenG. SOshimaR. G1997Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis. J Cell Biol 13813791394
  23. 23. ChanS. LFurukawaKMattsonM. P2002Presenilins and APP in neuritic and synaptic plasticity: implications for the pathogenesis of Alzheimer’s disease. Neuromolecular Med 2167196
  24. 24. ChenFChangRTrivediMCapetanakiYCrynsV. L2003Caspase proteolysis of desmin produces a dominant-negative inhibitor of intermediate filaments and promotes apoptosis. J Biol Chem 27868486853
  25. 25. ChinnaiyanA. MOrthKORourkeKDuanHPoirierG. GDixitVM (1996Molecular ordering of the cell death pathway. Bcl-2 and Bcl-xL function upstream of the CED-3-like apoptotic proteases. J Biol Chem 27145734576
  26. 26. ClemR. JFechheimerMMillerL. K1991Prevention of apoptosis by a baculovirus gene during infection of insect cells. Science 25413881390
  27. 27. ColleM. ADuyckaertsCLaquerriereAPradierLCzechCCheclerFHauwJ. J2000Laminar specific loss of isocortical presenilin 1 immunoreactivity in Alzheimer’s disease. Correlations with the amyloid load and the density of tau-positive neurofibrillary tangles. Neuropathol Appl Neurobiol 26117123
  28. 28. ColumbaroMMattioliELattanziGRutiglianoCOgnibeneAMaraldiN. MSquarzoniS2001Staurosporine treatment and serum starvation promote the cleavage of emerin in cultured mouse myoblasts: involvement of a caspase-dependent mechanism. FEBS Lett 509423429
  29. 29. CotmanC. W1998Apoptosis decision cascades and neuronal degeneration in Alzheimer’s disease. Neurobiol Aging 19:S2932
  30. 30. CotmanC. WSuJ. H1996Mechanism of neuronal death in Alzheimer’s disease. Brain Pathol 6493506
  31. 31. CreganS. PFortinAMacLaurin JG, Callaghan SM, Cecconi F, Yu SW, Dawson TM, Dawson VL, Park DS, Kroemer G, Slack RS (2002Apoptosis-inducing factor is involved in the regulation of caspase-independent neuronal cell death. J Cell Biol 158507517
  32. 32. CreganS. PMacLaurin JG, Craig CG, Robertson GS, Nicholson DW, Park DS, Slack RS (1999Bax-dependent caspase-3 activation is a key determinant in 53apoptosis in neurons. J Neurosci 19:7860-7869.
  33. 33. DarmonA. JNicholsonD. WBleackleyR. C1995Activation of the apoptotic protease CPP32 by cytotoxic T-cell-derived granzyme B. Nature 377446448
  34. 34. DaviesPMaloneyA. J1976Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 2:1403.
  35. 35. DefigueiredoR. JCummingsB. JMundkurP. YCotmanC. W1995Color image analysis in neuroanatomical research: application to senile plaque subtype quantification in Alzheimer’s disease. Neurobiol Aging 16211223
  36. 36. Degli Esposti MDive C (2003Mitochondrial membrane permeabilisation by Bax/Bak. Biochem Biophys Res Commun 304455461
  37. 37. DesjardinsPLedouxS1998Expression of ced-3 and ced-9 homologs in Alzheimer’s disease cerebral cortex. Neurosci Lett 2446972
  38. 38. DeverauxQ. LRoyNStennickH. RVan ArsdaleTZhouQSrinivasulaS. MAlnemriE. SSalvesenG. SReedJ. C1998IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J 1722152223
  39. 39. DoddP. R2002Excited to death: different ways to lose your neurones. Biogerontology 35156
  40. 40. DragunowMFaullR. LLawlorPBeilharzE. JSingletonKWalkerE. BMeeE1995In situ evidence for DNA fragmentation in Huntington’s disease striatum and Alzheimer’s disease temporal lobes. Neuroreport 610531057
  41. 41. EkertP. GSilkeJVauxD. L1999Caspase inhibitors. Cell Death Differe 610811086
  42. 42. EngidaworkEGulesserianTYooB. CCairnsNLubecG2001aAlteration of caspases and apoptosis-related proteins in brains of patients with Alzheimer’s disease. Biochem Biophys Res Commun 2818493
  43. 43. EngidaworkEGulesserianTYooB. CCairnsNLubecG2001bAlternation of caspase- and apoptosis-related proteins in brains of patients with Alzheimer’s disease. Biochem Biophys Res Comm 2818493
  44. 44. Fernandes-alnemriTLitwackGAlnemriE. S1995Mch2, a new member of the apoptotic Ced-3/Ice cysteine protease gene family. Cancer Res 5527372742
  45. 45. FernandoPKellyJ. FBalazsiKSlackR. SMegeneyL. A2002Caspase 3 activity is required for skeletal muscle differentiation. Proc Natl Acad Sci U S A 991102511030
  46. 46. FrancisP. TWebsterM. TChessellI. PHolmesCStratmannG. CProcterA. WCrossA. JGreenA. RBowenD. M1993Neurotransmitters and second messengers in aging and Alzheimer’s disease. Ann N Y Acad Sci 6951926
  47. 47. GalandeSDickinsonL. AMianI. SSikorskaMKohwi-shigematsuT2001SATB1 cleavage by caspase 6 disrupts PDZ domain-mediated dimerization, causing detachment from chromatin early in T-cell apoptosis. Mol Cell Biol 2155915604
  48. 48. GamesDAdamsDAlesandriniRBarbourRBertheletteP1995Alzheimer-type neuropathology in transgenic mice overexpression 717Fbeta-amyloid precursor protein. Nature 373:523-527.
  49. 49. GarverT. DHarrisK. ALehmanR. ALeeV. MTrojanowskiJ. QBillingsleyM. L1994Tau phosphorylation in human, primate, and rat brain: evidence that a pool of tau is highly phosphorylated in vivo and is rapidly dephosphorylated in vitro. J Neurochem 6322792287
  50. 50. GervaisF. GThornberryN. ARuffoloS. CNicholsonD. WRoyS1998Caspases cleave focal adhesion kinase during apoptosis to generate a FRNK-like polypeptide. J Biol Chem 2731710217108
  51. 51. Gervais FG, Xu D, Robertson GS, Vaillancourt JP, Zhu Y, J. H, LeBlanc A, Smith D, Rigby M, Shearman MS, Clarke EE, Zheng H, van Der Ploeg LHT, Ruffolo SC, Thornberry NS, Xanthoudakis S, Zamboni RJ, Roy S, Nicholson DW (1999) Involvement of caspases in proteolytic cleavage of Alzheimer’s amyloid-b precursor protein and amyloidogenic Ab peptide formation. Cell 97:395-406.
  52. 52. GeulaC1998Abnormalities of neural circuitry in Alzheimer’s disease: hippocampus and cortical cholinergic innervation. Neurology 51:S1829discussion S65-17.
  53. 53. GlennerG. GWongC. W1984Alzheimer’s disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. biochem Biophys Res Comm 120885890
  54. 54. GoldM2002Tau therapeutics for Alzheimer’s disease: the promise and the challenges. J Mol Neurosci 19331334
  55. 55. Gomez-islaTGrowdonW. BMcnamaraMNewellKGomez-tortosaEHedley-whyteE. THymanB. T1999Clinicopathologic correlates in temporal cortex in dementia with Lewy bodies. Neurology 5320032009
  56. 56. Gomez-islaTHollisterRWestHMuiSGrowdonJ. HPetersenR. CParisiJ. EHymanB. T1997Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann Neurol 411724
  57. 57. Gomez-islaTPriceJ. LMckeelD. WJr., Morris JC, Growdon JH, Hyman BT (1996Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer’s disease. J Neurosci 1644914500
  58. 58. GoyalL2001Cell death inhibition: Keeping caspases in check. Cell 104:805.
  59. 59. HamosJ. EDegennaroL. JDrachmanD. A1989Synaptic loss in Alzheimer’s disease and other dementias. Neurology 39355361
  60. 60. HardyJ2003The Relationship between Amyloid and Tau. J Mol Neurosci 20203206
  61. 61. HarrisonD. CDavisR. PBondB. CCampbellC. AJamesM. FParsonsA. APhilpottK. L2001Caspase mRNA expression in a rat model of focal cerebral ischemia. Brain Res Mol Brain Res 89133146
  62. 62. HartigWKleinCBrauerKSchuppelK. FArendtTBrucknerGBiglV2000Abnormally phosphorylated protein tau in the cortex of aged individuals of various mammalian orders. Acta Neuropathol (Berl) 100305312
  63. 63. HayB. AWassamanD. ARubinG. M1995Drosophila homologs of baculoirus inhibitor of apoptosis proteins function to block cell death. Cell 8312531262
  64. 64. HengartnerM. O2000The biochemistry of apoptosis. Nature 407770776
  65. 65. HohmannG. FWenkG. LLowensteinPBrownM. ECoyleJ. T1987Age-related recurrence of basal forebrain lesion-induced cholinergic deficits. Neurosci Lett 82253259
  66. 66. HolcombLGordonM. NMcgowanEYuXBenkovicS1998Accelerate Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat Med 497100
  67. 67. HuangYParkY. CRichR. LSegalDMyszkaD. GWuH2001Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell 104781790
  68. 68. IrmlerMThomeMHahneMSchneiderPHofmannKSteinerVBodmerJ. LSchroterMBurnsKMattmannCRimoldiDFrenchL. ETschoppJ1997Inhibition of death receptor signals by cellular FLIP. Nature 388190195
  69. 69. JanickeR. UNgPSprengartM. LPorterA. G1998Caspase-3 is required for alpha-fodrin cleavage but dispensable for cleavage of other death substrates in apoptosis. J Biol Chem 2731554015545
  70. 70. JellingerK2002aPrevalence of Alzheimer’s disease in very elderly people: a prospective neuropathological study. Neurology 58671672author reply 671-672.
  71. 71. JellingerK. A2002bVascular-ischemicdementia: an update. J Neural Transm Suppl:123
  72. 72. JellingerK. A2002cAlzheimer disease and cerebrovascular pathology: an update. J Neural Transm 109813836
  73. 73. JellingerK. AAttemsJ2003Incidence of cerebrovascular lesions in Alzheimer’s disease: a postmortem study. Acta Neuropathol (Berl) 1051417
  74. 74. KanamoriHKriegSMaoCDi Pippo VA, Wang S, Zajchowski DA, Shapiro DJ (2000Proteinase inhibitor 9, an inhibitor of granzyme B-mediated apoptosis, is a primary estrogen-inducible gene in human liver cells. J Biol Chem 27558675873
  75. 75. KeramarisEStefanisLMacLaurin J, Harada N, Takaku K, Ishikawa T, Taketo MM, Robertson GS, Nicholson DW, Slack RS, Park DS (2000Involvement of caspase 3 in apoptotic death of cortical neurons evoked by DNA damage. Mol Cell Neurosci 15368379
  76. 76. KerrJ. EWyllieA. HCurrieA. R1972Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26239257
  77. 77. KimT. WPettingellW. HJungY. KKovacsD. MTanziR. E1997Alternative cleavage of Alzheimer-associated presenilins during apoptosis by a caspase-3 family protease. Science 277373376
  78. 78. KitamuraYShimohamaSKamoshimaWMatsuokaYNomuraYTaniguchiT1997Changes of 53in the brains of patients with Alzheimer’s disease. Biochem Biophys Res Commun 232:418-421.
  79. 79. KitamuraYShimohamaSKamoshimaWOtaTMatsuokaYNomuraYSmithM. APerryGWhitehouseP. JTaniguchiT1998Alteration of proteins regulating apoptosis, Bcl-2, Bcl-x, Bax, Bak, Bad, ICH-1 and CPP32, in Alzheimer’s disease. Brain Res 780260269
  80. 80. KomiyanaTRayC. APickupD. JHowardA. DThornberryN. APetersonE. PSalvesenG1994Inhibitorion of interleukin-1-Beta converting enzyme by the cowpox virus serpin CrmA-an example of cross-class inhibition. J Biol Chem 2691933119337
  81. 81. KosekiTInoharaHChenSNEzG1998Arean inhibitor of apoptosis expressed in skeletal muscle and heart that interacts selectively with caspases. Proc Natl Acad Sci USA 9551565160
  82. 82. KothakotaSAzumaTReinhardCKlippelATangJChuKMcgarryT. JKirschnerM. WKothsKKwiatkowskiD. JWilliamsL. T1997Caspase-3-generated fragment of gelsolin: effector of morphological change in apoptosis. Science 278294298
  83. 83. KuidaKLippkeJ. AKuGHardingM. WLivingstonD. JSuM. SFlavellR. A1995Altered cytokine export and apoptosis in mice deficient in interleukin-1 beta converting enzyme. Science 26720002003
  84. 84. LarsonJLynchGGamesDSeubertP1999Alterations in synaptic transmission and long-term potentiation in hippocampal slices from young and aged PDAPP mice. Brain Res 8402335
  85. 85. LassmannHBancherCBreitschopfHWegielJBobinskiM1995Cell death in Alzheimer’s disease evaluated by DNA fragmentation in situ. Acta Neuropathol 893541
  86. 86. LeBlanc ALiu H, Goodyer C, Bergeron C, Hammond J (1999Caspase-6 role in apoptosis of human neurons, amyloidogenesis, and Alzheimer’s disease. J Biol Chem 2742342623436
  87. 87. Lee VMYBalin BJ, Otvos LJ, Trojanowski JQ (1991A68: a major subunit of paired helical filaments and derivatized forms of normal tau. Science 251675678
  88. 88. LiHYuanJ1999Deciphering the pathways of life and death. Curr Opin Cell Biol 11261266
  89. 89. LiW. PChanW. YLaiH. WYewD. T1997Terminal dUTP nick end labeling (TUNEL) positive cells in the different regions of the brain in normal aging and Alzheimer patients. J Mol Neurosci 87582
  90. 90. LippaC. FHamosJ. EPulaski-saloDDegennaroL. JDrachmanD. A1992Alzheimer’s disease and aging: effects on perforant pathway perikarya and synapses. Neurobiol Aging 13405411
  91. 91. LuD. CRabizadehSChandraSShayyaR. FEllerbyL. MYeXSalvesenG. SKooE. HBredesenD. E2000A second cytotoxic proteolytic peptide derived from amyloid b-protein precursor. Nature Med 6397404
  92. 92. LucassenP. JChungW. CKamphorstWSwaabD. F1997DNA damage distribution in the human brain as shown by in situ end labeling; area-specific differences in aging and Alzheimer disease in the absence of apoptotic morphology. J Neurophathol Exp Neuorl 56887900
  93. 93. MacGibbon GALawlor PA, Sirimanne ES, Walton MR, Connor B, Young D, Williams C, Gluckman P, Faull RL, Hughes P, Dragunow M (1997Bax expression in mammalian neurons undergoing apoptosis, and in Alzheimer’s disease hippocampus. Brain Res 750223234
  94. 94. MartinS. JFinucaneD. MAmarante-mendesG. POBrienG. AGreenDR (1996Phosphatidylserine externalization during CD95-induced apoptosis of cells and cytoplasts requires ICE/CED-3 protease activity. J Biol Chem 2712875328756
  95. 95. MasliahE2001Recent advances in the understanding of the role of synaptic proteins in Alzheimer’s Disease and other neurodegenerative disorders. J Alzheimers Dis 3121129
  96. 96. MasliahEMalloryMAlfordMDeteresaRHansenL. AMckeelD. WJr., Morris JC (2001Altered expression of synaptic proteins occurs early during progression of Alzheimer’s disease. Neurology 56127129
  97. 97. MasliahEMalloryMAlfordMTanakaSHansenL. A1998Caspase dependent DNA fragmentation might be associated with excitotoxicity in Alzheimer disease. J Neuropathol Exp Neurol 5710411052
  98. 98. MasliahETerryR. DDeteresaR. MHansenL. A1989Immunohistochemical quantification of the synapse-related protein synaptophysin in Alzheimer disease. Neurosci Lett 103234239
  99. 99. MastersC. LSimmsGWeinmanN. AMulthaupGMcdonaldB. LBeyreutherK1985Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA 8242454249
  100. 100. MccarthyN. JWhyteM. KGilbertC. SEvanG. I1997Inhibition of Ced-3/ICE-related proteases does not prevent cell death induced by oncogenes, DNA damage, or the Bcl-2 homologue Bak. J Cell Biol 136215227
  101. 101. MckhannGDrachmanDFolsteinMKatzmanRPriceD. LStadlanE. M1984Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s disease. Neurology 34939944
  102. 102. MesulamM. M1986Alzheimer plaques and cortical cholinergic innervation. Neuroscience 17275276
  103. 103. MichaelisM. LDobrowskyR. TLiG2002Tau neurofibrillary pathology and microtubule stability. J Mol Neurosci 19289293
  104. 104. MillerL. K1997Baculovirus interaction with host apoptotic pathways. J Cell Physiol 173178182
  105. 105. MoriHTakiokOgawaraMSelkieD1992Mass spectrometry of purified amyloid b protein in Alzheimer’s disease. J Biol Chem 2671708217086
  106. 106. MorrisJ. CMcKeel DWJ, Storandt M, Rubin EH, Price JL, Grant EA, Ball MJ, Berg L (1991Very mild Alzheimer’s disease: informant-based clinical, psychometric, and pathologic distinction from normal aging. Neurology 41469478
  107. 107. MuzioMChinnaiyanA. MKischkelF. CORourkeKShevchenkoANiJScaffidiCBretzJ. DZhangMGentzRMannMKrammerP. HPeterM. EDixitV. M1996Flicea novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex. Cell 85817827
  108. 108. NicholsonD. W1999Caspase structure, proteilytic substrates, and function during apoptotic cell death. Cell Death Differe 610281042
  109. 109. NijhawanDHonarpourNWangX2000Apoptosis in neural development and disease. Annu Rev Neurosci 237387
  110. 110. NyormoiOWangZDoanDRuizMMcconkeyDBar-eliM2001Transcription factor AP-2alpha is preferentially cleaved by caspase 6 and degraded by proteasome during tumor necrosis factor alpha-induced apoptosis in breast cancer cells. Mol Cell Biol 2148564867
  111. 111. OrthKORourkeKSalvesenG. SDixitVM (1996Molecular ordering of apoptotic mammalian CED-3/ICE-like proteases. J Biol Chem 2712097720980
  112. 112. ParadisEDouillardHKoutroumanisMGoodyerCLeBlanc A (1996Amyloid beta peptide of Alzheimer’s disease downregulates Bcl-2 and upregulates bax expression in human neurons. J Neurosci 1675337539
  113. 113. PearsonR. CPowellT. P1987Anterograde vs. retrograde degeneration of the nucleus basalis medialis in Alzheimer’s disease. J Neural Transm Suppl 24139146
  114. 114. PellegriniLPasserB. JTabatonMGanjeiJ. KDAdamioL1999Alternativenon-secretase processing of Alzheimer’s b-amyloid precursor protein during apoptosis by caspase-6 and-8. J Biol Chem 2742101121016
  115. 115. PriceD. LSisodiaS. S1998Mutant genes in familial Alzheimer’s disease and transgenic models. Annu Reve Neurosci 21479505
  116. 116. PriceJ. LMckeelD. WJr., Morris JC (2001Synaptic loss and pathological change in older adults--aging versus disease? Neurobiol Aging 22351352
  117. 117. RayC. ABlackR. AKronheimS. RGreensheetT. ASleathP. RSalvesenG. SPickupD. J1992Viral inhibition of inflammation: cowpox virus encodes an inhibitor of the interleukin-1 beta converting enzyme. Cell 69597604
  118. 118. RiedlS. JRenatusMSchwarzenbacherRZhouQSunCFesikS. WLiddingtonR. CSalvesenG. S2001Structural basis for the inhibition of caspase-3 by XIAP. Cell 104791800
  119. 119. RoderH2003Prospect of therapeutic approaches to tauopathies. J Mol Neurosci 20195202
  120. 120. RoherALowensonJClarkeSWoodsSCotterRGowingEBallM. J1993beta-Amyloid-(1-42) is a major component of cerebrovascular amyloid deposits: implications for the pathology of Alzheimer’s disease. Proc Natl Acad Sci USA 901083610840
  121. 121. RohnT. THeadENesseW. HCotmanC. WCribbsD. H2001Activation of caspase-8 in the Alzheimer’s disease brain. Neurobiol Dis 810061016
  122. 122. RohnT. TRissmanR. ADavisM. CKimY. ECotmanC. WHeadE2002Caspase-9 activation and caspase cleavage of tau in the Alzheimer’s disease brain. Neurobiol Dis 11341354
  123. 123. RothK. A2001Caspases, apoptosis, and Alzheimer disease: Causation, correlation, and confusion. J Neuropathol Exp Neurol 60829838
  124. 124. RuchaudSKorfaliNVillaPKottkeT. JDingwallCKaufmannS. HEarnshawW. C2002Caspase-6 gene disruption reveals a requirement for lamin A cleavage in apoptotic chromatin condensation. Embo J 2119671977
  125. 125. SakahiraHEnariMNagataS1998Cleavage of CAD inhibitor in CAD activation and DNA degradation during apoptosis. Nature 3919699
  126. 126. SamejimaKSvingenP. ABasiG. SKottkeTMesnerP. WJr., Stewart L, Durrieu F, Poirier GG, Alnemri ES, Champoux JJ, Kaufmann SH, Earnshaw WC (1999Caspase-mediated cleavage of DNA topoisomerase I at unconventional sites during apoptosis. J Biol Chem 27443354340
  127. 127. ScheffS. WPriceD. A2001Alzheimer’s disease-related synapse loss in the cingulate cortex. J Alzheimers Dis 3495505
  128. 128. ScheffS. WPriceD. ASparksD. L2001Quantitative assessment of possible age-related change in synaptic numbers in the human frontal cortex. Neurobiol Aging 22355365
  129. 129. SchottePDeclercqWVan HuffelSVandenabeelePBeyaertR1999Non-specific effects of methyl ketone peptide inhibitors of caspases. FEBS Lett 442117121
  130. 130. SchwartzL. MMilliganC. E1996Cold thoughts of death : the role of ICE proteases in neuronal cell death. Trends Neurosci 19555562
  131. 131. SelkoeD. J2002Alzheimer’s disease is a synaptic failure. Science 298789791
  132. 132. SelznickL. AHoltzmanD. MHanB. HGokdenMSrinivassanA. NJohnson EMJ, Roth KA (1999In situ immunodetection of neuronal caspase-3 activation in Alzheimer disease. J Neurophathol Exp Neurol 5810201026
  133. 133. SimpsonM. TMacLaurin JG, Xu D, Ferguson KL, Vanderluit JL, Davoli MA, Roy S, Nicholson DW, Robertson GS, Park DS, Slack RS (2001Caspase 3 deficiency rescues peripheral nervous system defect in retinoblastoma nullizygous mice. J Neurosci 2170897098
  134. 134. SleeE. AAdrainCMartinS. J2001Executioner caspase-3,-6, and-7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem 27673207326
  135. 135. SmaleGNicholsN. RBradyD. RFinchC. EHorton WEJ (1995Evidence for apoptotic cell death in Alzheimer’s disease. Exp Neurol 133225230
  136. 136. SrinivasulaS. MHegdeRSalehADattaPShiozakiEChaiJLeeR. ARobbinsP. DFernandes-alnemriTShiYAlnemriE. S2001A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 410112116
  137. 137. StadelmannCBrk WBancherCJellingerKLassmannH1998Alzheimer disease: DNA fragmentation indicates increased neuronal vulnerability, but not apoptosis. J Neuropathol Exp Neurol 57456464
  138. 138. StadelmannCDeckwerthTSrinivasanABancherCBrk WJellingerKLassmannH1999Activation of caspase-3 in single neurons and autophagic granules of granulovacuolar degeneration in Alzheimer’s disease. Am J Path 15514591466
  139. 139. StennickeH. RRyanC. ASalvesenG. S2002Reprieval from execution: the molecular basis of caspase inhibition. Trends Biochem Sci 2794101
  140. 140. StephanALarocheSDavisS2001Generation of aggregated beta-amyloid in the rat hippocampus impairs synaptic transmission and plasticity and causes memory deficits. J Neurosci 2157035714
  141. 141. StrasserAOConnorLDixit2000Apoptosis signaling. Annu Rev Biochem 69:217-245.
  142. 142. SuJ. HAndersonA. JCummingsB. JCotmanC. W1994Immunohistochemical evidence for apoptosis in Alzheimer’s disease. Neuroreport 525292533
  143. 143. SuJ. HDengGCotmanC. W1997Bax protein expression is increased in Alzheimer’s brain: correlations with DNA damage, Bcl-2 expression, and brain pathology. J NeuropatholExp Neurol 568693
  144. 144. SuJ. HSatouTAndersonA. JCotmanC. W1996Up-regulation of Bcl-2 is associated with neuronal DNA damage in Alzheimer’s disease. Neuroreport 7437440
  145. 145. TakeuchiAIrizarryM. CDuffK2000Age-related amyloid beta deposition in transgenic mice overexpressing both Alzheimer mutant presenilin 1 and amyloid beta precursor protein Swedish mutant is not associated with global neuronal loss. Am J Path 157331339
  146. 146. TerryR. D2000Cell death or synaptic loss in Alzheimer disease. J Neuropathol Exp Neurol 5911181119
  147. 147. TerryR. DMasliahESalmonD. P1991Physical basis of cognitive alterations in Alzheimer’s disease: Synaptic loss is a major correlate of cognitive impairment. Ann Neurol 30572580
  148. 148. ThomeMSchneiderPHofmannKFickenscherHMeinlENeipelFMattmannCBurnsKBodmerJ. LSchroterMScaffidiCKarammerP. HPeterM. ETachoppJ1997Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis induced by death receptors. Nature 386517521
  149. 149. ThompsonC. S1995Apoptosis in the pathogenesis and treatment of disease. Science 266714561462
  150. 150. ThornberryN. A1997The caspase family of cysteine proteases. Br Med Bull 53478490
  151. 151. TorpRSuJ. HDengGCotmanC. W1998GADD45 is induced in Alzheimer’s disease, and protects against apoptosis in vitro. Neurobiol Dis 5245252
  152. 152. TortosaALopezEFerrerI1998Bcl-2 and Bax protein expression in Alzheimer’s disease. Acta Neuropathol (Berl) 95407412
  153. 153. TroncosoJ. CSukhovR. RKawasC. HKoliatsosV. E1996In situ labeling of dying cortical neurons in normal aging and in Alzheimer’s disease: correlations with senile plaques and disease progression. J Nuropathol Exp Neurol 5511341142
  154. 154. TsengB. PEslerW. PClishC. BStimsonE. RGhilardiJ. RVintersH. VMantyhP. WLeeJ. PMaggioJ. E1999Deposition of monomeric, not oligomeric, Abeta mediates growth of Alzheimer’s disease amyloid plaques in human brain preparations. Biochemistry 381042410431
  155. 155. UrbancBCruzLBuldyrevS. VHavlinSIrizarryM. CStanleyH. EHymanB. T1999Dynamics of plaque formation in Alzheimer’s disease. Biophys J 7613301334
  156. 156. Van de Craen MBerx G, Van den Brande I, Fiers W, Declercq W, Vandenabeele 1999Proteolytic cleavage of beta-catenin by caspases: an in vitro analysis. FEBS Lett 458:167-170.
  157. 157. VanagsD. MPorn-aresM. ICoppolaSBurgessD. HOrreniusS1996Protease involvement in fodrin cleavage and phosphatidylserine exposure in apoptosis. J Biol Chem 2713107531085
  158. 158. VucicDKaiserW. JHarveyA. JMillerL. K1997Inhibition of reaper-induced apoptosis by interaction with inhibitor of apoptosis proteins (IAPs). Proc Natl Acd Sci USA 941018310188
  159. 159. VucicDKaiserW. JMillerL. K1998Inhibitor of apoptosis proteins physically interact with and block apoptosis induced by Drosophila proteins hid and grim. Mol Cell Biol 1833003309
  160. 160. WeidemannAPaligaKDrwangUReinhardF. BSchuckertOEvinGMastersC. L1999Proteolytic processing of the Alzheimer/s disease amyloid precursor protein within its cytoplasmic domain by caspase-like proteases. J Biol Chem 27458235829
  161. 161. WeinstockM1997Possible role of the cholinergic system and disease models. J Neural Transm Suppl 4993102
  162. 162. WhitehouseP. JPriceD. LClarkA. WCoyleJ. TDelongM. R1981Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann Neurol 10122126
  163. 163. WhitehouseP. JPriceD. LStrubleR. GClarkA. WCoyleJ. TDelonM. R1982Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain. Science 21512371239
  164. 164. YamatsujiTOkamotoTTakedaSMurayamaYTanakaNNishinotoI1996Expression of 642APP mutant causes cellular apoptosis as Alzheimer trait-linked phenotype. EMBO J 15:498-509.
  165. 165. YaoP. JMorschRCallahanL. MColemanP. D1999Changes in synaptic expression of clathrin assembly protein AP180 in Alzheimer’s disease analysed by immunohistochemistry. Neuroscience 94389394
  166. 166. YuanJShahamSLedouxSEllisH. MHorvitzH. R1993TheCElegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1beta-converting enzyme. Cell 75641652
  167. 167. YuanJYanknerB. A2000Apoptosis in the nervous system. Nature 407802809
  168. 168. ZhangYGoodyerCLeBlanc A (2000Selective and protracted apoptosis in human primary neurons microinjected with active caspase-3,-6,-7, and-8. J Neurosci 2083848389
  169. 169. ZhengT. SHunotSKuidaKFlavellR. A1999Caspase knockouts: matters of life and death. Cell Death Differe 610431053
  170. 170. ZhouQKrebsJ. FSnipasS. JPriceAAlnemriE. STomaselliK. JSalvesenG. S1998Interaction of the baculovirus anti-apoptotic protein 35with caspase-specificity, kinetics, and characterization of the caspase/p35 complex. Biochemistry 37:10757-10765.

Written By

Yan Zhang

Submitted: 20 April 2012 Published: 15 May 2013