Elsevier

Neurobiology of Disease

Volume 43, Issue 2, August 2011, Pages 364-371
Neurobiology of Disease

The L-type channel antagonist isradipine is neuroprotective in a mouse model of Parkinson's disease

https://doi.org/10.1016/j.nbd.2011.04.007Get rights and content

Abstract

The motor symptoms of Parkinson's disease (PD) are due to the progressive loss of dopamine (DA) neurons in substantia nigra pars compacta (SNc). Nothing is known to slow the progression of the disease, making the identification of potential neuroprotective agents of great clinical importance. Previous studies using the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD have shown that antagonism of L-type Ca2+ channels protects SNc DA neurons. However, this was not true in a 6-hydroxydopamine (6-OHDA) model. One potential explanation for this discrepancy is that protection in the 6-OHDA model requires greater antagonism of Cav1.3 L-type Ca2+ channels thought to underlie vulnerability and this was not achievable with the low affinity dihydropyridine (DHP) antagonist used. To test this hypothesis, the DHP with the highest affinity for Cav1.3 L-type channels—isradipine—was systemically administered and then the DA toxin 6-OHDA injected intrastriatally. Twenty-five days later, neuroprotection and plasma concentration of isradipine were determined. This analysis revealed that isradipine produced a dose-dependent sparing of DA fibers and cell bodies at concentrations achievable in humans, suggesting that isradipine is a potentially viable neuroprotective agent for PD.

Research highlights

► Using a progressive model of Parkinson's disease created by intrastriatal 6-hydroxydopamine, the neuroprotective potential of the L-type Ca2+ channel antagonist isradipine was tested. ► Isradipine is dihydropyridine approved for human use in the treatment of hypertension. ► Systemic administration of isradipine protected both striatal dopaminergic terminals and parent cell bodies. ► Protection was dose-dependent and afforded by plasma concentrations of isradipine achievable in humans within FDA guidelines.

Introduction

PD is a slowly progressing neurodegenerative condition whose presenting motor symptoms—bradykinesia, rigidity and tremor—are attributable to the loss of DA neurons (Riederer and Wuketich, 1976). Postmortem analysis of PD patient brains has revealed a striking loss of tyrosine hydroxylase (TH) immunoreactive, DA neurons in the SNc and relative sparing of DA neurons in the neighboring ventral tegmental area (VTA) (Hirsch et al., 1988). As nothing is known to slow the progression of the disease, the identification of neuroprotective agents in PD is of considerable importance.

One potential target for neuroprotective therapies in PD is the L-type Ca2+ channel with a Cav1.3 pore-forming subunit. The rationale for targeting these channels comes from an appreciation of the potentially deleterious consequences of elevations in intracellular Ca2+ (Gleichmann and Mattson, 2010) and the recent discovery that vulnerable SNc DA neurons have an usually strong engagement of Cav1.3 L-type Ca2+ channels during autonomous pacemaking (Guzman et al., 2010, Khaliq and Bean, 2010). This influx has been shown to increase mitochondrial oxidant stress in SNc DA neurons and this stress is exacerbated in a genetic model of PD (Guzman et al., 2010). In principle, this stress should increase the sensitivity to mitochondrial toxins used to create animal models of PD. In agreement with this inference, previous work has shown that antagonizing L-type Ca2+ channels by systemic administration of the DHP nimodipine increased the resistance of SNc DA neurons to both acute and chronic challenge with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration (Chan et al., 2007, Kupsch et al., 1995, Kupsch et al., 1996). However, systemic administration of nimodipine did not protect SNc DA neurons against a challenge with 6-hydroxydopamine (6-OHDA) (Sautter et al., 1997).

The reason for this apparent discrepancy is unclear. Although both 6-OHDA and MPTP disrupt mitochondrial function, they do so through different mechanisms (Bove et al., 2005). As a consequence, it is possible that protection against 6-OHDA toxicity requires a greater reduction in Ca2+ influx through L-type channels. The efficacy of systemically administered DHPs in antagonizing Cav1.3 Ca2+ channels in SNc DA neurons depends upon their bioavailability and potency. In this regard, DHPs are heterogeneous (Eisenberg et al., 2004). Although nimodipine has good brain bioavailability (Kupsch et al., 1996), it has a relatively low affinity for Cav1.3 Ca2+ channels (Sinnegger-Brauns et al., 2009). In contrast, isradipine has much higher (> 40 fold) affinity for Cav1.3 channels as well as good brain bioavailability (Bonci et al., 1998, Fitton and Benfield, 1990, Sinnegger-Brauns et al., 2009). Another relevant factor is DHP potency at Cav1.2 Ca2+ channels found in the cardiovascular system (Simuni et al., 2010). The antagonism of these channels limits the dose of DHPs that can be used for neuroprotective purposes. Theoretically, the ideal DHP for protecting SNc DA neurons would be one that was selective for Cav1.3 channels. Although none of the known DHPs have a higher affinity for Cav1.3 channels than Cav1.2 channels, the DHP that comes the closest is isradipine, which has nearly equal potency at Cav1.2 and Cav1.3 channels (Sinnegger-Brauns et al., 2009).

The studies reported were designed to test the hypothesis that isradipine would protect SNc DA neurons and fibers in a progressive model of PD created by intrastriatal injection of 6-OHDA. We found that indeed isradipine produced a dose-dependent protection in this model. Moreover, it was found that the plasma concentrations of isradipine that led to protection were within those that can be safely achieved in humans.

Section snippets

Animals and surgical procedures

Male C57BL/6 mice (6–7 weeks old and 20 ± 1 g) were purchased from Jackson Laboratory (Bar Harbor, Maine). Animals were handled according to the guidelines established by the Northwestern University Animal Care and Use Committee, the National Institutes of Health and the Society for Neuroscience.

All of the experiments shown, isradipine was delivered by subcutaneous Alzet osmotic minipumps (model 2004; Alzet, Cupertino, CA). These minipumps were found to provide a more reliable, sustained elevation

Results

In preliminary experiments, unilateral striatal lesions were made with decreasing doses of 6-OHDA to determine a dose that would produce roughly a 60% loss of SNc DA neurons 3–4 weeks after injection. Injection of 3.5 μg 6-OHDA induced a complete loss of TH positive fibers in the whole dorsal part of the striatum (caudate–putamen) and near a complete loss of TH positive DA cell bodies in SNc (Fig. 1A). In contrast, 2.5 μg of 6-OHDA only partially lesioned SNc DA neurons (Fig. 1B). To ensure that

Discussion

The main finding of this study was that in mice, systemically, administration of the DHP L-type calcium channel antagonist isradipine protected striatal dopaminergic terminals and parent SNc cell bodies against intrastriatal injection of the toxin 6-OHDA (2.5 μg). In the absence of isradipine, this insult decreased striatal TH fiber density by ~ 90% and dopaminergic neurons in the SNc by ~ 70% within 25 days. The protection afforded by isradipine in this model of PD was evident at all levels of the

Conclusion

The data presented here show that systemic administration of isradipine is capable of protecting striatal dopaminergic terminals and SNc dopaminergic cell bodies against a slow, progressive insult created by intrastriatal injection of 6-OHDA. The effect of isradipine was dose-dependent spanning a range of plasma concentrations achievable within humans at doses that are approved for the treatment of hypertension. These data add to a growing pre-clinical and epidemiological collection of studies

Acknowledgments

This work was supported by grants from the MJFF, Hartman Foundation and the National Institutes of Health (NS047085). We thank Nicholas Schwarz, Karen Saporito, Sasha Ulrich and Michael Avram for help with LC-MS/MS analysis.

References (43)

  • S. Schuster

    Antagonizing L-type Ca2+ channel reduces development of abnormal involuntary movement in the rat model of L-3,4-dihydroxyphenylalanine-induced dyskinesia

    Biol. Psychiatry

    (2009)
  • N.A. Tatton et al.

    In situ detection of apoptotic nuclei in the substantia nigra compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using terminal deoxynucleotidyl transferase labelling and acridine orange staining

    Neuroscience

    (1997)
  • B.P. Bean

    Nitrendipine block of cardiac calcium channels: high-affinity binding to the inactivated state

    Proc Natl Acad Sci USA.

    (1984)
  • B.P. Bean

    Multiple types of calcium channels in heart muscle and neurons. Modulation by drugs and neurotransmitters

    Ann. N.Y. Acad. Sci.

    (1989)
  • C. Becker

    Use of antihypertensives and the risk of Parkinson disease

    Neurology

    (2008)
  • A. Bonci

    L-Type calcium channels mediate a slow excitatory synaptic transmission in rat midbrain dopaminergic neurons

    J. Neurosci.

    (1998)
  • C.S. Chan

    'Rejuvenation' protects neurons in mouse models of Parkinson's disease

    Nature

    (2007)
  • G.D. Clifton

    The pharmacokinetics of oral isradipine in normal volunteers

    J. Clin. Pharmacol.

    (1988)
  • A. Fitton et al.

    Isradipine. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in cardiovascular disease

    Drugs

    (1990)
  • K.B.J. Franklin et al.

    The mouse brain in stereotaxic coordinates

    (1997)
  • M. Ganz

    Comparison of blood pressure control with amlodipine and controlled-release isradipine: an open-label, drug substitution study

    J Clin Hypertens (Greenwich).

    (2005)
  • Cited by (208)

    • Striatal glutamatergic hyperactivity in Parkinson's disease

      2022, Neurobiology of Disease
      Citation Excerpt :

      If 6-OHDA is injected into the striatum, the lesion induces a slower and less severe loss of dopaminergic nigrostriatal neurons (≈60–70%), modeling a more progressive neuronal degeneration (Kirik et al., 1998). This modular paradigm is widely used to evaluate antiparkinsonian effects of pharmacological treatments and for neuroprotection studies to develop new drug therapies (Ilijic et al., 2011; Jiang et al., 1993), or surgical approaches with cell transplantation (Bjorklund et al., 2002; Roy et al., 2006). 6-OHDA fully-lesioned rats show increased neuronal excitability of striatal neurons that results from a more robust glutamatergic cortical input to the striatum, as confirmed by electrophysiology, electron microscopy, and in vivo microdialysis studies (Calabresi et al., 1993; Ingham et al., 1998; Lindefors and Ungerstedt, 1990; Meshul et al., 1999).

    View all citing articles on Scopus
    View full text