Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Dec 1.
Published in final edited form as: Biochim Biophys Acta. 2011 Sep 8;1812(12):1584–1590. doi: 10.1016/j.bbadis.2011.08.013

Calcium channel blocking as a therapeutic strategy for Alzheimer’s disease: the case for isradipine

Thimmappa S Anekonda 1,*, Joseph F Quinn 1
PMCID: PMC3275089  NIHMSID: NIHMS332525  PMID: 21925266

Abstract

Alzheimer’s disease is the most devastating neurodegenerative disorder in the elderly, yet treatment options are severely limited. The drug development effort to modify Alzheimer’s disease pathology by intervention at beta amyloid production sites has been largely ineffective or inconclusive. The greatest challenge has been to identify and define downstream mechanisms reliably predictive of clinical symptoms. Beta amyloid accumulation leads to dysregulation of intracellular calcium by plasma membrane L-type calcium channels located on neuronal somatodendrites and axons in the hippocampus and cortex. Paradoxically, L-type calcium channel subtype Cav1.2 also promotes synaptic plasticity and spatial memory. Increased intracellular calcium modulates amyloid precursor protein processing and affects multiple downstream pathways including increased hyperphosphorylated tau and suppression of autophagyIsradipine is a Federal Drug Administration-approved dihydropyridine calcium channel blocker that binds selectively to Cav1.2 in the hippocampus. Our studies have shown that isradipine in vitro attenuates beta amyloid oligomer toxicity by suppressing calcium influx into cytoplasm and by suppressing Cav1.2 expression. We have previously shown that administration of isradipine to triple transgenic animal model for Alzheimer’s disease was well-tolerated. Our results further suggest that isradipine became bioavailable, lowered tau burden, and improved autophagy function in the brain. A better understanding of brain pharmacokinetics of calcium channel blockers will be critical for designing new experiments with appropriate drug doses in any future clinical trials for Alzheimer’s disease. This review highlights the importance of Cav1.2 channel overexpression, the accumulation of hyperphosphorylated tau and suppression of autophagy in Alzheimer’s disease and modulation of this pathway by isradipine.

Keywords: autophagy, beta amyloid, bioavailability, tau

1. Introduction

Beta amyloid (Aβ) plaque in Alzheimer’s disease (AD) accumulates long before patients become symptomatic; therefore, many therapeutic efforts have now moved to very early intervention. Another strategy to tackle the discordance between Aβ production and AD symptoms is to target mechanisms of neurodegeneration which are “downstream” of Aβ production. The greatest challenge has been to identify critical pathways that directly impact clinical symptoms and then effectively modulate these pathways by pharmacological agents. We and others have observed that a specific set of downstream pathways including dysregulation of intracellular calcium (Ca2+), upregulation of caspase-cleaved tau (tau-C3), hyperphosphorylation of tau (ptau), and loss of cellular housekeeping or autophagy function, may contribute directly to the expression of clinical symptoms. Multiple mechanisms control these seemingly unrelated pathways. A vast amount of current literature substantiates their critical but complex role in AD pathology [1-12].

1.1 Calcium trafficking is a complex process

Ca2+ trafficking is a complex process. A tight functional link exists among channels located on the plasma membrane (Aβ pores; L-type calcium channels, LTCC; N-methyl D-aspartyl receptor, NMDAR), endoplasmic reticulum (ryanodine receptor, RyR; Inositol(1,4,5)P3 receptor, InsP3R; sarco endoplasmic reticulum calcium ATPase, SERCA), and mitochondria [13-16]. Further, presenilin proteins, involved in AD pathogenesis, are located on the ER and can leak Ca2+ into the cytoplasm or interact with RyR, InsP3R, and SERCA to increase their activity [9, 12, 17]. A P86L polymorphism in a novel calcium channel called calcium homeostasis modulator 1 (CALHM1) can influence Aβ production by modulating amyloid precursor protein (APP) processing [11, 12]. Such a complexity involving a bidirectional relationship between Aβ production and calcium homeostasis pathways presents difficult experimental challenges (Figure 1; sections 1 & 2). Recent publications in the triple transgenic mouse model of AD (3xTgAD) implicate roles of RyR, InsP3R and NMDAR in the production of Ca2+ and synaptic plasticity of hippocampal neurons [18-21], but the role of LTCC, especially of Cav1.2, in relation to Aβ, tau, and autophagy is largely unknown. Therefore, in this review, we mainly focus on Cav1.2 relationship to AD pathology.

Figure 1.

Figure 1

A schematic overview of the Aβ-Cav1.2-ptau-autophagy pathway. Three sections of the figure are separated by dashed lines for clarity. Soluble Aβ oligomers, soluble and insoluble Aβ fibrils generated from amyloid precursor protein (APP) processing cause increased expression of Cav1.2 through beta 2 adrenergic receptor (β2AR)/cyclin AMP (cAMP) or other unknown pathways (section 1). Aβ production can be directly modulated by mutations in the calcium homeostasis modulator 1 (CALHM1) receptor located on the endoplasmic reticulum, or in response to treatment with dihydropyridine (DHP) class of calcium channel blockers (CCBs) (section 2). Intracellular or extracellular Aβ can cause over-expression of cav1.2, increased influx of Ca2, disrupted autophagy, and up-regulated ptau expression (section 3). Ca2+-calpain pathway can directly increase ptau levels or dampen autophagy function. Pathological accumulation of autophagosomal vacuoles and ptau will lead to loss of spatial memory and cognition in AD (section 3). Isradipine (ISR) appears to block this pathway.

1.2 L-type calcium channel subtype Cav1.2 is a critical target for calcium channel blockers in AD brains

Aging and Aβ consistently promote Ca2+ influx into neurons by way of L-type calcium channels (LTCCs). Soluble intraneuronal Aβ oligomers, soluble and insoluble Aβ fibrils can increase intracellular Ca2+, impair neuronal function, and adversely affect synaptic functions in AD [13, 22, 23]. Ca2+ increase occurs through over activation of LTCCs [24]. The uncontrolled Ca2+ increase can trigger the overexpression of plasma membrane LTCC subtype Cav1.2 in the hippocampus of AD brains and further exacerbate the pathogenic Ca2+ influx [1, 25, 26]. Cav1.2 is located in cell bodies and dendrites, axonal terminals, and axons of neurons, and glial processes of the hippocampus [2]. Paradoxically, Cav1.2 expression is essential for long-term potentiation (LTP) (independent of NMDAR-dependent LTP), synaptic plasticity, and spatial memory of the hippocampus [1, 2]. Cav1.2KO in mice disrupts remote spatial memory, further confirming the importance of Cav1.2 [1]. Cav1.3, a closely-related LTCC subtype known to mediate the pace-making function of dopaminergic neurons in Parkinson’s disease, has no known role in hippocampal memory function [27, 28].

Our recent study has examined the role of Cav1.2 in response to intracellular oligomeric Aβ because such understanding is an emerging area of relevance to AD [29]. Cav1.2 channel expression is essential for long-term potentiation (LTP) independent of NMDAR-dependent LTP, synaptic plasticity, and spatial memory of the hippocampus [1-3, 5, 18, 28]. Multiple pathways involving generation of intraneuronal or soluble extracellular Aβ can induce protein kinase A (PKA), which in turn binds to LTCC and promotes increased Ca2+ influx [30, 31]. PKA binding to the alpha subunit of Cav1.2 in the cytoplasm causes Cav1.2 overexpression in the membrane [30, 31]. PKA also promotes pathological hyperphosphorylation of tau [32, 33] and suppression of autophagy function in AD [34] (Figure 1; sections 1 & 3).

It is important to recognize that Cav1.2-independent pathways may also directly influence Aβ production, ptau or autophagy functions. Aβ-toxicity that is independent of Cav1.2 may occur when intraneuronal Aβ directly enters mitochondria and disrupts cellular energy balance or promotes free radical production [35]. Alternatively, free radicals can be generated from APP in autophagosomal compartments and disrupt autophagy function without affecting Cav1.2 [36].

1.3 Autophagy dysfunction in AD

The emerging literature suggests that autophagy, a self-cleaning cellular housekeeping mechanism, plays an important role in AD pathology [36]. Previously, it was assumed that autophagy was inducible only in response to stress but not essential for cell function. The most recent literature, however, provides unequivocal evidence for the constitutive role of autophagy in cellular homeostasis, thus making it essential for neuronal survival [37, 38]. Autophagosomal vacuoles accumulate in AD [7, 36, 39, 40], which can lead to an accelerated accumulation of ptau and tau-C3 [41-43]. Accumulated ptau, in turn, can rupture lysosomes, causing them to prematurely release proteolytic enzymes into the cytoplasm and disrupt normal autophagy function leading to neuronal death by apoptosis in AD [44-46]. A heterozygous deletion of the autophagy marker beclin-1 in Tg2576 mice increases intraneuronal Aβ accumulation, extracellular Aβ deposits, and neurodegeneration [47], suggesting that autophagy plays a key protective role against AD. Autophagy function is suppressed in several mouse models including 3xTg-AD [48-50]. Indeed one of the main functions of autophagy is to regulate mitochondrial function by enzymatic degradation of dysfunctional mitochondria and by clearing misfolded proteins in the cell [41, 51]. Our studies in Tg2576 mice provide strong evidence for mitochondrial deposition of Aβ leading to generation of free radicals and a protective role of autophagy against AD pathology [35].

1.4 Caspase-cleaved and hyperphosphorylated tau

A considerable effort has been made in the past few decades to understand tau-associated pathology in AD [52-55]. There seems to be a general consensus among experts that: (1) hyperphosphorylated tau appears early in the disease progression and is more prone to aggregation and tangle formation than the normal tau [52, 56, 57]. Increased Ca2+ can promote ptau via calpain-dependent pathway [58, 59]; (2) the presence of more 3-repeat tau conformations relative to 4-repeats can be pathogenic in AD [55, 60]; and (3) caspase-cleaved tau-C3 is fibrillogenic with much greater propensity for aggregation and formation of ptau and neurofibrillary tangles [53, 61, 62]. The most exciting and numerous roles of tau-C3 in AD pathology are still emerging in the literature. When wild type tau is cleaved mainly by caspase-3 at Asp421 site, a tau-C3 is formed [56, 62]. A recent study suggests that caspase activation may precede tangle formation in tau transgenic Tg4510 mice [63]. Some suggest that tau-C3 is present in tauopathies in the absence of Aβ pathology [43]. Lysosomal dysfunction may promote the increased formation of tau-C3 [44]. Thus, these emerging studies strongly suggest that the clearance pathway of tau-C3 is tightly linked to autophagy-lysosomal pathways in AD. Furthermore, tau-C3 is preferentially degraded by the macroautophagy pathway [41]. Ca2+ toxicity or overexpression of Cav1.2 can also lead to mislocalization of tau into somatodendritic regions instead of normal axonal localization [64]. In somatodendritic regions, tau can damage microtubules and spines and become ptau [64].

These studies together provide a strong evidence for complex roles of Aβ, Cav1.2, Cai2+, tau-C3, ptau, autophagy-lysosomal dysfunction in orchestrating AD pathology. We call this nexus of toxicity in short: Aβ-Cav1.2-ptau-autophagy pathway.

2. Calcium channel blockers to break the nexus of toxicity

Usefulness of LTCC blockers (CCBs) against AD pathology is controversial. Epidemiological studies suggest that CCBs prevent [65] or slow the rate of progression of AD [66, 67]. A large clinical trial with nimodipine did not show significant benefits from the primary outcome measures but has shown moderate benefits for treatment of AD in the secondary outcome measures [68, 69]. In a recent study, however, nimodipine selectively stimulated the secretion of Aβ1-42 in vitro and in the plasma of Tg2576 mouse model of AD, questioning the usefulness of this CCB for AD [70]. Several clinical studies have suggested that CCBs used as antihypertensive drugs may prevent cognitive decline in hypertensive subjects [71-74], but none have demonstrated a role for CCBs in AD per se. Two dihydropyridine CCBs were among the eight FDA-approved drugs selected from 485 small biomolecules screened for their ability to induce autophagy without causing toxic effects in human neuroglioma H4 cells [75]. Further, dihydropyridine-based derivatives act as potent activators of anti-aging neuroprotective protein sirtuin 1 [76], which appears to regulate autophagy function [77].

Two recent in vitro studies have shown usefulness of DHP CCBs on Aβ production and Aβ1-42 transcytosis across an in vitro blood-brain barrier (BBB) created by an apical “blood” and basolateral “brain” layers of human brain microvascular endothelial cells [78, 79]. Nivladipine, nitrendipine, and amlodipine reduced Aβ production in 7W Chinese hamster ovary (CHO) cells, which have been stably transfected with human APP751 [79]. This study further shows an improved Aβ clearance by nilvadipine and nitrendipine, and an improved explorative activity for animals treated with nivaldipine in a transgenic animal model of AD (Tg PS1/APPsw) [79]. The BBB transcytosis of Aβ1-42 also increased for several of DHP CCBs including nitrendipine, nicardipine, nimodipine, and nilvadipine [78]. In their study, isradipine had no effect on Aβ1-42 transcytosis. Furthermore, CCBs (nitrendipine, clinidipine, nivaldipine) promoted Aβ1-42 clearance across the BBB in wild type mice; and in animals treated with human Aβ1-42, nilvadipine improved the cognitive functions of the animals in Morris water maze test [78]. These studies clearly suggest that DHP CCBs possess non-channeling functions that are independent of their calcium channel blocking ability, suggesting a need for thorough validation of CCBs for AD.

2.1 Why isradipine a suitable CCB for treatment of AD?

Emerging studies in models of Parkinson’s disease (PD) show neuroprotection by isradipine, an FDA-approved dihydropyridine class of CCB for hypertension [27, 80-82]. In these studies, isradipine blocks LTCC subtype cav1.3 function in dopaminergic neuron of the substantia nigra and modulates autonomous pacemaking function [27, 82, 83]. Isradipine provides protection against stroke and brain ischemia in rat models for hypertension [84, 85]. Early studies focusing on binding properties of [3H]isradipine in AD and control brains suggest that the hippocampal CA1 region experiences greater cell loss in response to increased expression of LTCCs in AD brains relative to control brains [86, 87], suggesting that isradipine treatment is likely to modulate the over-expressing LTCCs in the CA1 region and preserve the hippocampal function. Isradipine can also attenuate over active LTCC function as well as oxidative stress-induced apoptotic cell death in hypobaric hypoxia model of rats and preserve their memory function [88]. Our preliminary studies have clearly shown the superior effects of isradipine over nimodipine in vitro [29]. We also predict that the brain bioavailability of therapeutic doses of isradipine is superior to that of nimodipine, as bioavailability studies in animals generally support this assertion [89, 90].

A recent isradipine safety and tolerability study in PD provides valuable guidelines for how to evaluate the neuroprotective effects of isradipine in clinical trials [80]. In this study, subjects (average age = ~59 years) with early PD were treated with increasing doses of controlled release isradipine (5-20 mg daily doses) over a period of eight weeks. There was a dose-dependent tolerability for isradipine (94% for 5 mg; 87% for 10 mg; 68% for 15 mg; 52% for 20 mg). Isradipine did not show any significant impact on blood pressure or motor disability, but leg edema and dizziness were the two frequent adverse symptoms commonly observed in this study, leading to a conclusion that 10 mg daily dose of isradipine was a safe treatment [80]. In a Danish case-control study (average age=72 years), the effect of DHP class of CCBs was evaluated retrospectively for their neuroprotective effects on PD [27]. The commonly prescribed centrally-acting DHP CCBs (nimodipine, isradipine, nitrendipine, nifedipine), as opposed to non-DHP class of CCBs, provided up to 27% risk reduction for PD, irrespective of treatment length and duration [27]. These studies appear to suggest that careful clinical trials using isradipine in older patients are highly feasible, safe, and unlikely to cause adverse effects on the memory-related function of Cav1.2.

2.1.1 In vitro studies

Our recent publication is the first such report to show neuroprotection of isradipine against Aβ-induced Ca2+ toxicity in human neuroblastoma/MC65 cells [29]. MC65 cells that are stably transfected with the APP-C99 gene conditionally express a fusion protein fragment of the amino-17 and carboxyl-99 residues [91, 92]. APP-C99 gene expression in these cells is controlled by a tetracycline-responsive promoter whose activity is repressed in the presence of tetracycline or induced by withdrawing tetracycline from the growth medium. Removal of tetracycline leads to expression of the C-terminal APP fragment and subsequent processing of this fragment into Aβ monomers followed by accumulation of intracellular Aβ oligomers, and precipitous cell death by about 72 hours. In this cell culture model we tested the role of cav1.2 expression on Cai2+ influx and cell survival as well as protective effects of four CCBs (diltiazem, verapamil, nimodipine, isradipine) against Cai2+-induced toxicity. Isradipine was the most potent of the four CCBs tested [29]. This study suggests that intracellular Aβ oligomers trigger increased intracellular Ca2+ influx primarily through Cav1.2 channels. None of these CCBs prevented the formation of intracellular oligomers. Isradipine provided protection against Ca2+ toxicity by both blocking calcium influx and suppressing cav1.2 expression downstream of Aβ formation [29]. Our study results also indicate that Ca2+ levels are tightly controlled and cells respond to small variations in Ca2+ and this response is sensitive to small concentrations of isradipine [29].

2.1.2 In vivo studies

We also tested isradipine for its neuroprotective functions in four evolutionarily divergent species including models for AD: human neuroblastoma/MC65 cells, transgenic Drosophila, Monduca, and 3xTg-AD mice [93]. In transgenic Drosophila model, 250 μM isradipine increased the survival of flies from 6.5% to 12% (p<0.5) against APP695 toxicity [93]. In moth/Monduca sexta experiments, embryonic culture preparations were exposed to exogenous Aβ1-42 peptide to determine the toxic effects of this peptide on neuronal development and growth in the presence or absence of 10 μM isradipine [93]. Treatment with Aβ1-42 induces concentration-dependent perturbations in the extent of migration and outgrowth of enteric nervous system. A simultaneous treatment with isradipine prevents the deleterious effects of Aβ [93]. Our studies further show that subcutaneous administration of isradipine (3 μg/g/day; 60-day release) to 3xTgAD mice was well-tolerated and isradipine became available to the brain [29, 93]. The 3xTgAD mouse model, which harbors PS1(M146V), APPswe and tau(P301L) transgenes, develops age-dependent and region-specific Aβ and tau aggregations in the cortex and hippocampus that closely mimics the pathology found in human AD [94, 95]. A small cohort of 17-month-old female 3xTgAD mice (an age well after the appearance of AD pathology) were implanted with carrier-bound isradipine pellets (n=3, 3 mg/Kg/day, 60-day release, Innovative Research of America) or placebo control pellets (n=4). Isradipine was well-tolerated as evidenced by average weekly body weights (an indirect measurement of toxicity), which were the same for vehicle and isradipine-treated animals. Isradipine treatment also showed lowering trend for insoluble Aβ1-40 and Aβ1-42, no effect on soluble Aβ1-40, and undetectable soluble Aβ1-42 [93]. Histological Aβ burden in these animals reduced non-significantly in the hippocampus and isradipine significantly lowered IHC-detectable ptau burden in the hippocampus [93]. In a second cohort of 22-month-old female 3xTgAD (N=8) and age-matched wild type (N=8) mice, a similar isradipine or vehicle pellet implantation showed significantly upregulated LC3B protein, a marker for late-stage autophagy (unpublished results) in isradipine-treated mice. These animal studies together strongly suggest that protective functions of isradipine are similarly conserved across evolutionarily divergent species and these functions are mutually synergistic.

2.2 LTCC blocking with isradipine: implications for future research

Our novel findings show that isradipine can modulate AD pathology by influencing Aβ production upstream as well as components of the cav1.2-ptau-autophagy pathway. This will have much larger implications in developing a unique treatment strategy for AD. Isradipine appears to bind strongly to cav1.2 in the hippocampus and perhaps short-circuits the downstream pathway [3, 96, 97] (Figure 1; section 3). Mechanisms that promote the clearance of rapidly cluttering autophagosomal vacuoles in AD brains have been elusive for the past several decades, and they now appear to be an important bottleneck in moving forward with effective treatment strategies for AD. Fortunately, calcium channel blockers are emerging as modulators of autophagy function [75, 98]. We believe that an approach to use CCB to short-circuit the entire downstream pathway can overcome current hurdles in the clinical treatment of AD. We argue that isradipine is an appropriate candidate for future studies, as it appears to possess additional functions capable of modulating both upstream and downstream protective functions besides its calcium channel blocking ability.

Calcium channel blocking will have a many implications for future research and clinical trials: (1) Suppression of Cav1.2 expression through β2AR/cAMP/PKA or mitochondrial PKA-dependent pathways can defuse the upstream toxic effects of Aβ or reactive oxygen species [30, 31]. (2) PKA-dependent suppression of LC3 function in autophagy can be neutralized by blocking Cav1.2 [99, 100]. (3) Attenuated Ca2+-dependent calpain levels can downregulate ptau and caspase-cleaved tau production. It is possible that LTCC blocking may provide effective protection strategies for Parkinson’s disease, Huntington’s disease, ALS, axotomy, brain ischemia, and stroke, where autophagy or isradipine is known to play key protective roles. It is important to recognize that there may be some risk in modulating Cav1.2 expression specifically in the hippocampal CA1 pyramidal neurons, since these receptors also directly influence long-term potentiation, synaptic plasticity, and spatial memory function [3, 5, 28, 101]. However, several lines of evidence discussed at the top of this section suggest that isradipine predominantly modulates Cav1.2 rather than adversely effecting the memory function associated with normal function of Cav1.2 [27, 80, 86].

3. Bioavailability of calcium channel blockers

In the past three decades, several bioanalytical methods (radioactive labeling, gas chromatography, high pressure liquid chromatography) have been developed for assessing isradipine pharmacokinetics and pharmacodynamics in animal and human tissues [6, 86, 89, 90, 102, 103]. The most important discovery from these early methods is that isradipine undergoes extensive first-pass metabolism and nearly 90% of orally administered isradipine is absorbed in the digestive tract, limiting its bioavailability to about 17-28% in plasma. For an immediate release (IR) formulation of isradipine, a sharp peak concentration occurs in plasma about 1.5-2 h after administration with a terminal half-life of 8 hours [89]. Sustained-release (SR) formulations show superior bioavailability with an initial lag period for 2-3 hrs and then a slow increase in concentration reaching a plateau between 7 to 18 hrs after dosing. Thus, SR formulation shows a lower maximal plasma concentration (Cmax) and an extended maximal tissue concentration (Tmax) as well as extended mean elimination half-life (T½) compared to IR formulation [89]. However, many of the early methods showed low sensitivity, longer analytical time, and involved pre-column derivatization protocols affected by interferences, making them prohibitively expensive for large-scale rapid screening of clinical samples. Recently, a liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) has been developed as sensitive, accurate, and rapid method for assessment of isradipine bioavailability in the plasma samples from healthy volunteers [104]. This method showed an excellent linearity between 10 to 5000 pg/ml of isradipine in plasma, with the lower limit of quantitation (LLOQ) at 10 pg/ml plasma and a short retention time [104]. We have recently applied this method successfully to a 3xTgAD mice [29]. In our studies with 3xTgAD mice, we detected 33±7 ng/ml of isradipine in the plasma and 47±1 ng/g brain tissue of the animals implanted with carrier-bound isradipine pellets (3 μg/g/day, 60-day release, Innovative Research of America, Sarasota, FL) and none in animals implanted with placebo control pellets [29].

To be able to apply the above method to AD patients, we need to establish relationships between plasma and brain bioavailability of isradipine first in animal models of AD. This approach needs some practical considerations. First, there is a possibility that the plasma-to-brain relationship for isradipine may be a non-linear function. Because isradipine can easily cross the blood-brain barrier [105], we expect isradipine will reach the brain readily. Thus the non-linearity argument may be somewhat muted. Second, there is a possibility that Tmax and T½ for isradipine may differ from mouse to human. Interestingly, cross-species pharmacokinetics appears to be remarkably comparable between mouse and human, including for isradipine [89, 103, 106]. So, these parameters are expected to remain similar in mouse and human brains; thus, information obtained on mouse models can be easily translatable to humans. Third, transgenic animal models do not fully account for the genetic heterogeneity prevalent in the late-onset AD population. This is a serious issue and one way to maximize heterogeneity in animal populations is by using different genotypes/animal models and age groups. Alternatively, pharmacokinetic measures such as area under curve (AUC) can be determined for plasma and brain to ensure a greater accuracy. Such an approach will require multiple time points of measurements after isradipine dosing and will increase the number of animals required for the study. In live humans, brain function or cerebrospinal fluid levels rather that brain levels of isradipine can be modeled with plasma levels of isradipine to develop clinically useful predictive functions.

4. Conclusions

Despite their long-standing presence and well-defined safety records, the usefulness of CCBs for Alzheimer’s disease has been unclear. The reluctance to use CCBs to AD may be attributed to the initial failure of nimodipine in an AD clinical trial. However, recent experimental evidence presented in this review demonstrates that CCBs such as isradipine possess multiple beneficial effects besides their primary L-type calcium blocking ability. Rigorous, well-designed pre-clinical and clinical studies are expected to provide proof-of-concept on the effectiveness of CCBs for treatment of Alzheimer’s disease.

Acknowledgments

This work was supported by NIH/NEI 5R21EY018708-02 (TSA), Department of Veterans Affairs Merit Review Grant (JFQ), and NIA-R21AG027445A (JFQ).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].White JA, McKinney BC, John MC, Powers PA, Kamp TJ, Murphy GG. Conditional forebrain deletion of the L-type calcium channel Ca V 1.2 disrupts remote spatial memories in mice. Learn Mem. 2008;15:1–5. doi: 10.1101/lm.773208. [DOI] [PubMed] [Google Scholar]
  • [2].Tippens AL, Pare JF, Langwieser N, Moosmang S, Milner TA, Smith Y, Lee A. Ultrastructural evidence for pre- and postsynaptic localization of Cav1.2 L-type Ca2+ channels in the rat hippocampus. J Comp Neurol. 2008;506:569–583. doi: 10.1002/cne.21567. [DOI] [PubMed] [Google Scholar]
  • [3].Lacinova L, Moosmang S, Langwieser N, Hofmann F, Kleppisch T. Cav1.2 calcium channels modulate the spiking pattern of hippocampal pyramidal cells. Life Sci. 2008;82:41–49. doi: 10.1016/j.lfs.2007.10.009. [DOI] [PubMed] [Google Scholar]
  • [4].Oliveria SF, Dell’Acqua ML, Sather WA. AKAP79/150 anchoring of calcineurin controls neuronal L-type Ca2+ channel activity and nuclear signaling. Neuron. 2007;55:261–275. doi: 10.1016/j.neuron.2007.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Moosmang S, Haider N, Klugbauer N, Adelsberger H, Langwieser N, Muller J, Stiess M, Marais E, Schulla V, Lacinova L, Goebbels S, Nave KA, Storm DR, Hofmann F, Kleppisch T. Role of hippocampal Cav1.2 Ca2+ channels in NMDA receptor-independent synaptic plasticity and spatial memory. J Neurosci. 2005;25:9883–9892. doi: 10.1523/JNEUROSCI.1531-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Uchida S, Yamada S, Nagai K, Deguchi Y, Kimura R. Brain pharmacokinetics and in vivo receptor binding of 1,4-dihydropyridine calcium channel antagonists. Life Sci. 1997;61:2083–2090. doi: 10.1016/s0024-3205(97)00881-3. [DOI] [PubMed] [Google Scholar]
  • [7].Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, Cuervo AM. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol. 2005;64:113–122. doi: 10.1093/jnen/64.2.113. [DOI] [PubMed] [Google Scholar]
  • [8].Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430:631–639. doi: 10.1038/nature02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Cheung KH, Shineman D, Muller M, Cardenas C, Mei L, Yang J, Tomita T, Iwatsubo T, Lee VM, Foskett JK. Mechanism of Ca2+ disruption in Alzheimer’s disease by presenilin regulation of InsP3 receptor channel gating. Neuron. 2008;58:871–883. doi: 10.1016/j.neuron.2008.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].LaFerla FM. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat Rev Neurosci. 2002;3:862–872. doi: 10.1038/nrn960. [DOI] [PubMed] [Google Scholar]
  • [11].Dreses-Werringloer U, Lambert JC, Vingtdeux V, Zhao H, Vais H, Siebert A, Jain A, Koppel J, Rovelet-Lecrux A, Hannequin D, Pasquier F, Galimberti D, Scarpini E, Mann D, Lendon C, Campion D, Amouyel P, Davies P, Foskett JK, Campagne F, Marambaud P. A polymorphism in CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer’s disease risk. Cell. 2008;133:1149–1161. doi: 10.1016/j.cell.2008.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Green KN, LaFerla FM. Linking calcium to Abeta and Alzheimer’s disease. Neuron. 2008;59:190–194. doi: 10.1016/j.neuron.2008.07.013. [DOI] [PubMed] [Google Scholar]
  • [13].Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci. 2008;31:454–463. doi: 10.1016/j.tins.2008.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Arispe N, Pollard HB, Rojas E. beta-Amyloid Ca(2+)-channel hypothesis for neuronal death in Alzheimer disease. Mol Cell Biochem. 1994;140:119–125. doi: 10.1007/BF00926750. [DOI] [PubMed] [Google Scholar]
  • [15].Guo Q, Furukawa K, Sopher BL, Pham DG, Xie J, Robinson N, Martin GM, Mattson MP. Alzheimer’s PS-1 mutation perturbs calcium homeostasis and sensitizes PC12 cells to death induced by amyloid beta-peptide. Neuroreport. 1996;8:379–383. doi: 10.1097/00001756-199612200-00074. [DOI] [PubMed] [Google Scholar]
  • [16].Green KN, Demuro A, Akbari Y, Hitt BD, Smith IF, Parker I, LaFerla FM. SERCA pump activity is physiologically regulated by presenilin and regulates amyloid beta production. J Cell Biol. 2008;181:1107–1116. doi: 10.1083/jcb.200706171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Chan SL, Mayne M, Holden CP, Geiger JD, Mattson MP. Presenilin-1 mutations increase levels of ryanodine receptors and calcium release in PC12 cells and cortical neurons. J Biol Chem. 2000;275:18195–18200. doi: 10.1074/jbc.M000040200. [DOI] [PubMed] [Google Scholar]
  • [18].Lopez JR, Lyckman A, Oddo S, Laferla FM, Querfurth HW, Shtifman A. Increased intraneuronal resting [Ca2+] in adult Alzheimer’s disease mice. J Neurochem. 2008;105:262–271. doi: 10.1111/j.1471-4159.2007.05135.x. [DOI] [PubMed] [Google Scholar]
  • [19].Chakroborty S, Goussakov I, Miller MB, Stutzmann GE. Deviant ryanodine receptor-mediated calcium release resets synaptic homeostasis in presymptomatic 3xTg-AD mice. J Neurosci. 2009;29:9458–9470. doi: 10.1523/JNEUROSCI.2047-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Goussakov I, Miller MB, Stutzmann GE. NMDA-mediated Ca(2+) influx drives aberrant ryanodine receptor activation in dendrites of young Alzheimer’s disease mice. J Neurosci. 2010;30:12128–12137. doi: 10.1523/JNEUROSCI.2474-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Smith IF, Hitt B, Green KN, Oddo S, LaFerla FM. Enhanced caffeine-induced Ca2+ release in the 3xTg-AD mouse model of Alzheimer’s disease. J Neurochem. 2005;94:1711–1718. doi: 10.1111/j.1471-4159.2005.03332.x. [DOI] [PubMed] [Google Scholar]
  • [22].Mattson MP. Calcium and neurodegeneration. Aging Cell. 2007;6:337–350. doi: 10.1111/j.1474-9726.2007.00275.x. [DOI] [PubMed] [Google Scholar]
  • [23].Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:101–112. doi: 10.1038/nrm2101. [DOI] [PubMed] [Google Scholar]
  • [24].Ueda K, Shinohara S, Yagami T, Asakura K, Kawasaki K. Amyloid beta protein potentiates Ca2+ influx through L-type voltage-sensitive Ca2+ channels: a possible involvement of free radicals. J Neurochem. 1997;68:265–271. doi: 10.1046/j.1471-4159.1997.68010265.x. [DOI] [PubMed] [Google Scholar]
  • [25].Willis M, Kaufmann WA, Wietzorrek G, Hutter-Paier B, Moosmang S, Humpel C, Hofmann F, Windisch M, Knaus HG, Marksteiner J. L-type calcium channel CaV 1.2 in transgenic mice overexpressing human AbetaPP751 with the London (V717I) and Swedish (K670M/N671L) mutations. J Alzheimers Dis. 2010;20:1167–1180. doi: 10.3233/JAD-2010-091117. [DOI] [PubMed] [Google Scholar]
  • [26].Langwieser N, Christel CJ, Kleppisch T, Hofmann F, Wotjak CT, Moosmang S. Homeostatic switch in hebbian plasticity and fear learning after sustained loss of Cav1.2 calcium channels. J Neurosci. 2010;30:8367–8375. doi: 10.1523/JNEUROSCI.4164-08.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Ritz B, Rhodes SL, Qian L, Schernhammer E, Olsen JH, Friis S. L-type calcium channel blockers and Parkinson disease in Denmark. Ann Neurol. 2010;67:600–606. doi: 10.1002/ana.21937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Zhang H, Fu Y, Altier C, Platzer J, Surmeier DJ, Bezprozvanny I. Ca1.2 and CaV1.3 neuronal L-type calcium channels: differential targeting and signaling to pCREB. Eur J Neurosci. 2006;23:2297–2310. doi: 10.1111/j.1460-9568.2006.04734.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Anekonda TS, Quinn JF, Harris C, Frahler K, Wadsworth TL, Woltjer RL. L-type voltage-gated calcium channel blockade with isradipine as a therapeutic strategy for Alzheimer’s disease. Neurobiol Dis. 2011;41:62–70. doi: 10.1016/j.nbd.2010.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Wang D, Govindaiah G, Liu R, De Arcangelis V, Cox CL, Xiang YK. Binding of amyloid beta peptide to beta2 adrenergic receptor induces PKA-dependent AMPA receptor hyperactivity. Faseb J. 2010;24:3511–3521. doi: 10.1096/fj.10-156661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Hall DD, Davare MA, Shi M, Allen ML, Weisenhaus M, McKnight GS, Hell JW. Critical role of cAMP-dependent protein kinase anchoring to the L-type calcium channel Cav1.2 via A-kinase anchor protein 150 in neurons. Biochemistry. 2007;46:1635–1646. doi: 10.1021/bi062217x. [DOI] [PubMed] [Google Scholar]
  • [32].Yamamoto H, Hiragami Y, Murayama M, Ishizuka K, Kawahara M, Takashima A. Phosphorylation of tau at serine 416 by Ca2+/calmodulin-dependent protein kinase II in neuronal soma in brain. J Neurochem. 2005;94:1438–1447. doi: 10.1111/j.1471-4159.2005.03307.x. [DOI] [PubMed] [Google Scholar]
  • [33].Scott CW, Spreen RC, Herman JL, Chow FP, Davison MD, Young J, Caputo CB. Phosphorylation of recombinant tau by cAMP-dependent protein kinase. Identification of phosphorylation sites and effect on microtubule assembly. J Biol Chem. 1993;268:1166–1173. [PubMed] [Google Scholar]
  • [34].Honson NS, Kuret J. Tau aggregation and toxicity in tauopathic neurodegenerative diseases. J Alzheimers Dis. 2008;14:417–422. doi: 10.3233/jad-2008-14409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH. Mitochondria are a direct site of Abeta accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet. 2006;15:1437–1449. doi: 10.1093/hmg/ddl066. [DOI] [PubMed] [Google Scholar]
  • [36].Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci. 2007;120:4081–4091. doi: 10.1242/jcs.019265. [DOI] [PubMed] [Google Scholar]
  • [37].Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature. 2006;441:885–889. doi: 10.1038/nature04724. [DOI] [PubMed] [Google Scholar]
  • [38].Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature. 2006;441:880–884. doi: 10.1038/nature04723. [DOI] [PubMed] [Google Scholar]
  • [39].Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH, Mohan PS, Mercken M, Farmery MR, Tjernberg LO, Jiang Y, Duff K, Uchiyama Y, Naslund J, Mathews PM, Cataldo AM, Nixon RA. Macroautophagy--a novel Beta-amyloid peptide-generating pathway activated in Alzheimer’s disease. J Cell Biol. 2005;171:87–98. doi: 10.1083/jcb.200505082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Nixon RA. Autophagy in neurodegenerative disease: friend, foe or turncoat? Trends Neurosci. 2006;29:528–535. doi: 10.1016/j.tins.2006.07.003. [DOI] [PubMed] [Google Scholar]
  • [41].Dolan PJ, Johnson GV. A caspase cleaved form of tau is preferentially degraded through the autophagy pathway. J Biol Chem. 2010;285:21978–21987. doi: 10.1074/jbc.M110.110940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Lee JH, Yu WH, Kumar A, Lee S, Mohan PS, Peterhoff CM, Wolfe DM, Martinez-Vicente M, Massey AC, Sovak G, Uchiyama Y, Westaway D, Cuervo AM, Nixon RA. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141:1146–1158. doi: 10.1016/j.cell.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Cotman CW, Poon WW, Rissman RA, Blurton-Jones M. The role of caspase cleavage of tau in Alzheimer disease neuropathology. J Neuropathol Exp Neurol. 2005;64:104–112. doi: 10.1093/jnen/64.2.104. [DOI] [PubMed] [Google Scholar]
  • [44].Khurana V, Elson-Schwab I, Fulga TA, Sharp KA, Loewen CA, Mulkearns E, Tyynela J, Scherzer CR, Feany MB. Lysosomal dysfunction promotes cleavage and neurotoxicity of tau in vivo. PLoS Genet. 2010;6:e1001026. doi: 10.1371/journal.pgen.1001026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Cataldo AM, Nixon RA. Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain. Proc Natl Acad Sci U S A. 1990;87:3861–3865. doi: 10.1073/pnas.87.10.3861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Hamano T, Gendron TF, Causevic E, Yen SH, Lin WL, Isidoro C, Deture M, Ko LW. Autophagic-lysosomal perturbation enhances tau aggregation in transfectants with induced wild-type tau expression. Eur J Neurosci. 2008;27:1119–1130. doi: 10.1111/j.1460-9568.2008.06084.x. [DOI] [PubMed] [Google Scholar]
  • [47].Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, Small S, Spencer B, Rockenstein E, Levine B, Wyss-Coray T. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest. 2008;118:2190–2199. doi: 10.1172/JCI33585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J Biol Chem. 2010;285:13107–13120. doi: 10.1074/jbc.M110.100420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Ma T, Hoeffer CA, Capetillo-Zarate E, Yu F, Wong H, Lin MT, Tampellini D, Klann E, Blitzer RD, Gouras GK. Dysregulation of the mTOR pathway mediates impairment of synaptic plasticity in a mouse model of Alzheimer’s disease. PLoS ONE. 2010;5:e12845. doi: 10.1371/journal.pone.0012845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen D, Richardson A, Strong R, Galvan V. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS ONE. 2010;5:e9979. doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Cardoso SM, Pereira CF, Moreira PI, Arduino DM, Esteves AR, Oliveira CR. Mitochondrial control of autophagic lysosomal pathway in Alzheimer’s disease. Exp Neurol. 2009;223:294–298. doi: 10.1016/j.expneurol.2009.06.008. [DOI] [PubMed] [Google Scholar]
  • [52].Braak H, Braak E, Strothjohann M. Abnormally phosphorylated tau protein related to the formation of neurofibrillary tangles and neuropil threads in the cerebral cortex of sheep and goat. Neurosci Lett. 1994;171:1–4. doi: 10.1016/0304-3940(94)90589-4. [DOI] [PubMed] [Google Scholar]
  • [53].Koson P, Zilka N, Kovac A, Kovacech B, Korenova M, Filipcik P, Novak M. Truncated tau expression levels determine life span of a rat model of tauopathy without causing neuronal loss or correlating with terminal neurofibrillary tangle load. Eur J Neurosci. 2008;28:239–246. doi: 10.1111/j.1460-9568.2008.06329.x. [DOI] [PubMed] [Google Scholar]
  • [54].Zhang Q, Zhang X, Sun A. Truncated tau at D421 is associated with neurodegeneration and tangle formation in the brain of Alzheimer transgenic models. Acta Neuropathol. 2009;117:687–697. doi: 10.1007/s00401-009-0491-6. [DOI] [PubMed] [Google Scholar]
  • [55].Siddiqua A, Margittai M. Three- and four-repeat Tau coassemble into heterogeneous filaments: an implication for Alzheimer disease. J Biol Chem. 2010;285:37920–37926. doi: 10.1074/jbc.M110.185728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Zilkova M, Zilka N, Kovac A, Kovacech B, Skrabana R, Skrabanova M, Novak M. Hyperphosphorylated truncated protein tau induces caspase-3 independent apoptosis-like pathway in the Alzheimer’s disease cellular model. J Alzheimers Dis. 2011;23:161–169. doi: 10.3233/JAD-2010-101434. [DOI] [PubMed] [Google Scholar]
  • [57].Alonso AC, Grundke-Iqbal I, Iqbal K. Alzheimer’s disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nat Med. 1996;2:783–787. doi: 10.1038/nm0796-783. [DOI] [PubMed] [Google Scholar]
  • [58].Hung KS, Hwang SL, Liang CL, Chen YJ, Lee TH, Liu JK, Howng SL, Wang CH. Calpain inhibitor inhibits p35-p25-Cdk5 activation, decreases tau hyperphosphorylation, and improves neurological function after spinal cord hemisection in rats. J Neuropathol Exp Neurol. 2005;64:15–26. doi: 10.1093/jnen/64.1.15. [DOI] [PubMed] [Google Scholar]
  • [59].Yang LS, Ksiezak-Reding H. Calpain-induced proteolysis of normal human tau and tau associated with paired helical filaments. Eur J Biochem. 1995;233:9–17. doi: 10.1111/j.1432-1033.1995.009_1.x. [DOI] [PubMed] [Google Scholar]
  • [60].Fujino Y, Wang DS, Thomas N, Espinoza M, Davies P, Dickson DW. Increased frequency of argyrophilic grain disease in Alzheimer disease with 4R tau-specific immunohistochemistry. J Neuropathol Exp Neurol. 2005;64:209–214. doi: 10.1093/jnen/64.3.209. [DOI] [PubMed] [Google Scholar]
  • [61].Quintanilla RA, Matthews-Roberson TA, Dolan PJ, Johnson GV. Caspase-cleaved tau expression induces mitochondrial dysfunction in immortalized cortical neurons: implications for the pathogenesis of Alzheimer disease. J Biol Chem. 2009;284:18754–18766. doi: 10.1074/jbc.M808908200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Zilka N, Filipcik P, Koson P, Fialova L, Skrabana R, Zilkova M, Rolkova G, Kontsekova E, Novak M. Truncated tau from sporadic Alzheimer’s disease suffices to drive neurofibrillary degeneration in vivo. FEBS Lett. 2006;580:3582–3588. doi: 10.1016/j.febslet.2006.05.029. [DOI] [PubMed] [Google Scholar]
  • [63].de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, Hyman BT. Caspase activation precedes and leads to tangles. Nature. 2010;464:1201–1204. doi: 10.1038/nature08890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Zempel H, Thies E, Mandelkow E, Mandelkow EM. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J Neurosci. 2010;30:11938–11950. doi: 10.1523/JNEUROSCI.2357-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Forette F, Seux ML, Staessen JA, Thijs L, Birkenhager WH, Babarskiene MR, Babeanu S, Bossini A, Gil-Extremera B, Girerd X, Laks T, Lilov E, Moisseyev V, Tuomilehto J, Vanhanen H, Webster J, Yodfat Y, Fagard R. Prevention of dementia in randomised double-blind placebo-controlled Systolic Hypertension in Europe (Syst-Eur) trial. Lancet. 1998;352:1347–1351. doi: 10.1016/s0140-6736(98)03086-4. [DOI] [PubMed] [Google Scholar]
  • [66].Fritze J, Walden J. Clinical findings with nimodipine in dementia: test of the calcium hypothesis. J Neural Transm Suppl. 1995;46:439–453. [PubMed] [Google Scholar]
  • [67].Tollefson GD. Short-term effects of the calcium channel blocker nimodipine (Bay-e-9736) in the management of primary degenerative dementia. Biol Psychiatry. 1990;27:1133–1142. doi: 10.1016/0006-3223(90)90050-c. [DOI] [PubMed] [Google Scholar]
  • [68].Lopez-Arrieta BJ. Nimodipine for primary degenerative, mixed and vascular dementia. Cochrane Database of Systematic Reviews. 2002 doi: 10.1002/14651858.CD000147. article no CD000147. [DOI] [PubMed] [Google Scholar]
  • [69].Morich F, Bieber F, Lewis JM, Kaiser L, Cutler NR, Escobar JI, Willmer J, Petersen RC, Reisberg B. Nimodipine in the Treatment of Probably Alzheimer’s Disease Results of Two Multicentre Trials. Clin Drug Invest. 1996;11:185–196. [Google Scholar]
  • [70].Facchinetti F, Fasolato C, Del Giudice E, Burgo A, Furegato S, Fusco M, Basso E, Seraglia R, D’Arrigo A, Leon A. Nimodipine selectively stimulates beta-amyloid 1-42 secretion by a mechanism independent of calcium influx blockage. Neurobiol Aging. 2006;27:218–227. doi: 10.1016/j.neurobiolaging.2005.02.006. [DOI] [PubMed] [Google Scholar]
  • [71].Bellew KM, Pigeon JG, Stang PE, Fleischman W, Gardner RM, Baker WW. Hypertension and the rate of cognitive decline in patients with dementia of the Alzheimer type. Alzheimer Dis Assoc Disord. 2004;18:208–213. [PubMed] [Google Scholar]
  • [72].Hanyu H, Hirao K, Shimizu S, Sato T, Kiuchi A, Iwamoto T. Nilvadipine prevents cognitive decline of patients with mild cognitive impairment. Int J Geriatr Psychiatry. 2007;22:1264–1266. doi: 10.1002/gps.1851. [DOI] [PubMed] [Google Scholar]
  • [73].Khachaturian AS, Zandi PP, Lyketsos CG, Hayden KM, Skoog I, Norton MC, Tschanz JT, Mayer LS, Welsh-Bohmer KA, Breitner JC. Antihypertensive medication use and incident Alzheimer disease: the Cache County Study. Arch Neurol. 2006;63:686–692. doi: 10.1001/archneur.63.5.noc60013. [DOI] [PubMed] [Google Scholar]
  • [74].Yasar S, Corrada M, Brookmeyer R, Kawas C. Calcium channel blockers and risk of AD: the Baltimore Longitudinal Study of Aging. Neurobiol Aging. 2005;26:157–163. doi: 10.1016/j.neurobiolaging.2004.03.009. [DOI] [PubMed] [Google Scholar]
  • [75].Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, Zhu H, Yu AD, Xie X, Ma D, Yuan J. Small molecule regulators of autophagy identified by an image-based high-throughput screen. Proc Natl Acad Sci U S A. 2007;104:19023–19028. doi: 10.1073/pnas.0709695104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Mai A, Valente S, Meade S, Carafa V, Tardugno M, Nebbioso A, Galmozzi A, Mitro N, De Fabiani E, Altucci L, Kazantsev A. Study of 1,4-dihydropyridine structural scaffold: discovery of novel sirtuin activators and inhibitors. J Med Chem. 2009;52:5496–5504. doi: 10.1021/jm9008289. [DOI] [PubMed] [Google Scholar]
  • [77].Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos M, Alt FW, Finkel T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci U S A. 2008;105:3374–3379. doi: 10.1073/pnas.0712145105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Bachmeier C, Beaulieu-Abdelahad D, Mullan M, Paris D. Selective dihydropyiridine compounds facilitate the clearance of beta-amyloid across the blood-brain barrier. Eur J Pharmacol. 2011;659:124–129. doi: 10.1016/j.ejphar.2011.03.048. [DOI] [PubMed] [Google Scholar]
  • [79].Paris D, Bachmeier C, Patel N, Quadros A, Volmar CH, Laporte V, Ganey J, Beaulieu-Abdelahad D, Ait-Ghezala G, Crawford F, Mullan MJ. Selective Antihypertensive Dihydropyridines Lower Abeta Accumulation by Targeting both the Production and the Clearance of Abeta across the Blood-Brain Barrier. Mol Med. 2011;17:149–162. doi: 10.2119/molmed.2010.00180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Simuni T, Borushko E, Avram MJ, Miskevics S, Martel A, Zadikoff C, Videnovic A, Weaver FM, Williams K, Surmeier DJ. Tolerability of isradipine in early Parkinson’s disease: A pilot dose escalation study. Mov Disord. 2010 doi: 10.1002/mds.23308. [DOI] [PubMed] [Google Scholar]
  • [81].Chang CC, Cao S, Kang S, Kai L, Tian X, Pandey P, Dunne SF, Luan CH, Surmeier DJ, Silverman RB. Antagonism of 4-substituted 1,4-dihydropyridine-3,5-dicarboxylates toward voltage-dependent L-type Ca2+ channels Ca V 1.3 and Ca V 1.2. Bioorg Med Chem. 2010;18:3147–3158. doi: 10.1016/j.bmc.2010.03.038. [DOI] [PubMed] [Google Scholar]
  • [82].Chan CS, Gertler TS, Surmeier DJ. A molecular basis for the increased vulnerability of substantia nigra dopamine neurons in aging and Parkinson’s disease. Mov Disord. 2010;25(Suppl 1):S63–70. doi: 10.1002/mds.22801. [DOI] [PubMed] [Google Scholar]
  • [83].Singh A, Gebhart M, Fritsch R, Sinnegger-Brauns MJ, Poggiani C, Hoda JC, Engel J, Romanin C, Striessnig J, Koschak A. Modulation of voltage- and Ca2+-dependent gating of CaV1.3 L-type calcium channels by alternative splicing of a C-terminal regulatory domain. J Biol Chem. 2008;283:20733–20744. doi: 10.1074/jbc.M802254200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Lenhard SC, Strittmatter R, Price WJ, Chandra S, White RF, Barone FC. Brain MRI and neurological deficit measurements in focal stroke: rapid throughput validated with isradipine. Pharmacology. 2008;81:1–10. doi: 10.1159/000107661. [DOI] [PubMed] [Google Scholar]
  • [85].Campbell CA, Mackay KB, Patel S, King PD, Stretton JL, Hadingham SJ, Hamilton TC. Effects of isradipine, an L-type calcium channel blocker on permanent and transient focal cerebral ischemia in spontaneously hypertensive rats. Exp Neurol. 1997;148:45–50. doi: 10.1006/exnr.1997.6611. [DOI] [PubMed] [Google Scholar]
  • [86].Coon AL, Wallace DR, Mactutus CF, Booze RM. L-type calcium channels in the hippocampus and cerebellum of Alzheimer’s disease brain tissue. Neurobiol Aging. 1999;20:597–603. doi: 10.1016/s0197-4580(99)00068-8. [DOI] [PubMed] [Google Scholar]
  • [87].Ikeda M, Dewar D, McCulloch J. A correlative study of calcium channel antagonist binding and local neuropathological features in the hippocampus in Alzheimer’s disease. Brain Res. 1992;589:313–319. doi: 10.1016/0006-8993(92)91292-m. [DOI] [PubMed] [Google Scholar]
  • [88].Barhwal K, Hota SK, Baitharu I, Prasad D, Singh SB, Ilavazhagan G. Isradipine antagonizes hypobaric hypoxia induced CA1 damage and memory impairment: Complementary roles of L-type calcium channel and NMDA receptors. Neurobiol Dis. 2009;34:230–244. doi: 10.1016/j.nbd.2009.01.008. [DOI] [PubMed] [Google Scholar]
  • [89].Sica DA. Calcium channel blocker class heterogeneity: select aspects of pharmacokinetics and pharmacodynamics. J Clin Hypertens (Greenwich) 2005;7:21–26. doi: 10.1111/j.1524-6175.2006.04482.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Christensen HR, Antonsen K, Simonsen K, Lindekaer A, Bonde J, Angelo HR, Kampmann JP. Bioavailability and pharmacokinetics of isradipine after oral and intravenous administration: half-life shorter than expected? Pharmacol Toxicol. 2000;86:178–182. doi: 10.1034/j.1600-0773.2000.d01-32.x. [DOI] [PubMed] [Google Scholar]
  • [91].Sopher BL, Fukuchi K, Kavanagh TJ, Furlong CE, Martin GM. Neurodegenerative mechanisms in Alzheimer disease. A role for oxidative damage in amyloid beta protein precursor-mediated cell death. Mol Chem Neuropathol. 1996;29:153–168. doi: 10.1007/BF02814999. [DOI] [PubMed] [Google Scholar]
  • [92].Sopher BL, Fukuchi K, Smith AC, Leppig KA, Furlong CE, Martin GM. Cytotoxicity mediated by conditional expression of a carboxyl-terminal derivative of the beta-amyloid precursor protein. Brain Res Mol Brain Res. 1994;26:207–217. doi: 10.1016/0169-328x(94)90092-2. [DOI] [PubMed] [Google Scholar]
  • [93].Copenhaver PF, Anekonda TS, Musashe D, Robinson KM, Swanson TL, Wadsworth TL, Kretzschmar D, Woltjer RL, Quinn JF. A translational continuum of model systems for evaluating treatment strategies in Alzheimer’s disease: Isradipine as a candidate drug. Disease Models & Mechanisms. 2011 doi: 10.1242/dmm.006841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM. Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol Aging. 2003;24:1063–1070. doi: 10.1016/j.neurobiolaging.2003.08.012. [DOI] [PubMed] [Google Scholar]
  • [95].Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, Metherate R, Mattson MP, Akbari Y, LaFerla FM. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
  • [96].Sinnegger-Brauns MJ, Huber IG, Koschak A, Wild C, Obermair GJ, Einzinger U, Hoda JC, Sartori SB, Striessnig J. Expression and 1,4-dihydropyridine-binding properties of brain L-type calcium channel isoforms. Mol Pharmacol. 2009;75:407–414. doi: 10.1124/mol.108.049981. [DOI] [PubMed] [Google Scholar]
  • [97].Lacinova L, Klugbauer N, Hofmann F. State- and isoform-dependent interaction of isradipine with the alpha1C L-type calcium channel. Pflugers Arch. 2000;440:50–60. doi: 10.1007/s004249900244. [DOI] [PubMed] [Google Scholar]
  • [98].Diaz JC, Simakova O, Jacobson KA, Arispe N, Pollard HB. Small molecule blockers of the Alzheimer Abeta calcium channel potently protect neurons from Abeta cytotoxicity. Proc Natl Acad Sci U S A. 2009;106:3348–3353. doi: 10.1073/pnas.0813355106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Cherra SJ, 3rd, Dagda RK, Chu CT. Review: autophagy and neurodegeneration: survival at a cost? Neuropathol Appl Neurobiol. 2010;36:125–132. doi: 10.1111/j.1365-2990.2010.01062.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Cherra SJ, 3rd, Kulich SM, Uechi G, Balasubramani M, Mountzouris J, Day BW, Chu CT. Regulation of the autophagy protein LC3 by phosphorylation. J Cell Biol. 2010;190:533–539. doi: 10.1083/jcb.201002108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Striessnig J, Bolz HJ, Koschak A. Channelopathies in Cav1.1, Cav1.3, and Cav1.4 voltage-gated L-type Ca2+ channels. Pflugers Arch. 2010;460:361–374. doi: 10.1007/s00424-010-0800-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Clifton GD, Blouin RA, Dilea C, Schran HF, Hassell AE, Gonasun LM, Foster TS. The pharmacokinetics of oral isradipine in normal volunteers. J Clin Pharmacol. 1988;28:36–42. doi: 10.1002/j.1552-4604.1988.tb03098.x. [DOI] [PubMed] [Google Scholar]
  • [103].Sen AP, Boksa P, Quirion R. Brain calcium channel related dihydropyridine and phenylalkylamine binding sites in Alzheimer’s, Parkinson’s and Huntington’s diseases. Brain Res. 1993;611:216–221. [PubMed] [Google Scholar]
  • [104].Park JH, Park YS, Rhim SY, Jhee OH, Kim SH, Yang SC, Lee MH, Shaw LM, Kang JS. Quantification of isradipine in human plasma using LC-MS/MS for pharmacokinetic and bioequivalence study. J Chromatogr B Analyt Technol Biomed Life Sci. 2009;877:59–64. doi: 10.1016/j.jchromb.2008.11.021. [DOI] [PubMed] [Google Scholar]
  • [105].Gaggi R, Dall’Olio R, Roncada P, Gianni AM. Peculiar effects of isradipine and darodipine on the rat brain dopaminergic system. Gen Pharmacol. 1995;26:303–308. doi: 10.1016/0306-3623(94)00196-t. [DOI] [PubMed] [Google Scholar]
  • [106].Sanftner LM, Gibbons JA, Gross MI, Suzuki BM, Gaeta FC, Johnson KW. Cross-species comparisons of the pharmacokinetics of ibudilast. Xenobiotica. 2009;39:964–977. doi: 10.3109/00498250903254340. [DOI] [PubMed] [Google Scholar]

RESOURCES