Voltage-gated calcium channels and Parkinson's disease

https://doi.org/10.1016/j.pharmthera.2011.11.006Get rights and content

Abstract

A complex interaction of environmental, genetic and epigenetic factors combine with ageing to cause the most prevalent of movement disorders Parkinson's disease. Current pharmacological treatments only tackle the symptoms and do not stop progression of the disease or reverse the neurodegenerative process. While some incidences of Parkinson's disease arise through heritable genetic defects, the cause of the majority of cases remains unknown. Likewise, why some neuronal populations are more susceptible to neurodegeneration than others is not clear, but as the molecular pathways responsible for the process of cell death are unravelled, it is increasingly apparent that disrupted cellular energy metabolism plays a central role. Precise control of cellular calcium concentrations is crucial for maintenance of energy homeostasis. Recently, differential cellular expression of neuronal voltage-gated calcium channel (CaV) isoforms has been implicated in the susceptibility of vulnerable neurons to neurodegeneration in Parkinson's disease. CaV channels are also involved in the synaptic plasticity response to the denervation that occurs in Parkinson's disease and following chronic treatment with anti-parkinsonian drugs. This review will examine the putative role neuronal CaV channels have in the pathogenesis and treatment of Parkinson's disease.

Introduction

Parkinson's disease is a progressive hypokinetic neurodegenerative neurological disorder characterised by bradykinesia, rigidity, akinesia, abnormal posture and resting tremor. In the later stages of the disease autonomic and sensorimotor dysfunction, cognitive decline, depression and sleep disturbances also occur and become clinically relevant (Marsden, 1990). Non-motor functional deficits often precede the major motor symptoms by a number of years and it has been suggested that they are indicative of neurodegeneration that originates in the brain stem and progresses throughout the brain (Braak et al., 2003).

Calcium (Ca2+) plays a central role in the normal functioning of neurons and is also involved in the many cellular processes (e.g. oxidative stress, mitochondrial impairment, proteasomal dysfunction, excitotoxicity, neuroinflammation, apoptosis) that can lead to cell death in Parkinson's disease (Berridge et al., 2000, Büeler, 2010, Lau and Tymianski, 2010, Witte et al., 2010, Hegde and Upadhya, 2011). However, it is not clear whether Ca2+ dysfunction is causative or secondary to the disease process. Similarly, the reason why some neurons within brain regions degenerate more in affected areas of brain (e.g. ventral–lateral zone > dorsal tier of the substantia nigra (SN)) in Parkinson's disease while other regions containing similar neuron types are less affected (e.g. ventral tegmental area) is unknown. The observation that degenerating neurons in the SN were mainly in areas with low levels of the Ca2+ binding protein calbindin-D28k suggested a role for Ca2+ in neuronal susceptibility (Yamada et al., 1990, German et al., 1992, Damier et al., 1999). Another common feature of neurons which degenerate in Parkinson's disease, irrespective of the neurotransmitter type, is that they are poorly myelinated with long fine axons that connect different brain regions (Braak et al., 2004). This, together with the large axonal field of long projection neurons means they have high energy requirements, which necessitates efficient mitochondrial function and a good Ca2+ buffering capacity if toxicity through oxidative stress or excitotoxicity is to be avoided (Surmeier et al., 2011).

Recent studies have implicated L-type CaV channels (CaV1) in the pathogenesis of Parkinson's disease. First, Chan et al. (2007) demonstrated that dopaminergic neurons vulnerable to neurodegeneration in the SN pars compacta of adult (but not juvenile) mice use the CaV1.3 subtype of Ca2+ channel for pacemaking activity. In contrast, dopaminergic neurons in the adjacent ventral tegmental area, that are less affected by neurodegeneration in Parkinson's disease, rely instead upon sodium ion entry for maintenance of conductance oscillations (Fujimura and Matsuda, 1989, Chan et al., 2007, Khaliq and Bean, 2010). The use of Ca2+ rather than sodium ions for pacemaking requires more energy expenditure in order to maintain a safe intracellular Ca2+ concentration. This is because extrusion of Ca2+ out of the cell and sequestration of Ca2+ into intracellular stores requires energy derived from mitochondrial oxidative phosphorylation and the electrochemical gradient for Ca2+ across the cellular membranes is much larger (1000×) than that of sodium ions. In Parkinson's disease where mitochondrial dysfunction is evident, the reliance on CaV1.3 channels may make the SN pars compacta cells more susceptible to Ca2+ mediated excitotoxicity (Chan et al., 2009). The reliance of the SN on CaV1.3 channels for pacemaking may also explain the decline in dopaminergic neurons of the SN pars compacta seen with advancing age (Chan et al., 2010). However, Parkinson's disease is not just accelerated ageing because the pattern of cell loss is different in post-mortem brain from non-parkinsonian elderly people to that seen in brain from patients dying with Parkinson's disease (Fearnley & Lees, 1991). Second, retrospective analysis of patients treated with CaV1 channel blocking drugs for hypertension and cardiac arrhythmias have indicated a decreased risk for Parkinson's disease in patients treated with dihydropyridines that cross the blood brain barrier, which suggested a neuroprotective effect of such drugs and implied a pathogenic role for the CaV1 channel subtype in Parkinson's disease (Becker et al., 2008, Ritz et al., 2010).

This review will examine the role brain CaV channels may have in the pathology and treatment of Parkinson's disease. For the above reasons the focus will on the CaV1 (L-type current) channel isoforms, but discussion of other subtypes of CaV channel will be included when it is pertinent to do so.

Section snippets

Epidemiology

Parkinson's disease is the most prevalent movement disorder and the second most frequently diagnosed neurodegenerative disease, with an overall incidence in industrialized countries of 0.15–0.3%. The greatest risk factor for Parkinson's disease is increasing age, such that it affects 1% of people over 60 years of age, rising to 2–4% for those aged over 80 (Ben-Shlomo and Sieradzan, 1995, Checkoway and Nelson, 1999, Nussbaum and Ellis, 2003, Leung and Mok, 2005, de Lau and Breteler, 2006). Though

Neuronal voltage-gated calcium channels

The electric potential across cellular membranes is controlled by the gradient of ions produced by membrane pumps, buffering molecules and the activity of the ion channels themselves, either in response to the electrical gradient (voltage sensing ion channels) or inter- and intra-cellular chemical messengers (ligand-gated ion channels) (Aston-Jones & Siggins, 2000). Voltage-gated channels respond to neuronal membrane depolarization by opening and allowing conductance of the ion down the

Pathways to cell death in Parkinson's disease

The pathogenesis of neurodegeneration in Parkinson's disease is multifactorial and theories concerning the origin of cell death processes centre on malfunctioning protein degradation, neuroinflammation and mitochondrial dysfunction occurring because of genetic susceptibility or/and exposure to an environmental toxin (Schapira & Jenner, 2011). The role CaV channels might have in these pathogenic processes is discussed below.

Calcium channel function and Parkinson's disease

The available evidence suggests that CaV1 channels contribute to cell death in Parkinson's disease and that blockade of midbrain CaV1 channels provides some degree of neuroprotection to dying neurons in the SN (Chan et al., 2007, Becker et al., 2008, Ritz et al., 2010). Although in the absence of selective ligands for each Cav isoform, the precise role played by Cav1.2 or Cav1.3 in neurodegeneration or/and neuroprotection cannot be discerned. However, the use of gene knockout models has allowed

Conclusions

The unforeseen connection between Parkinson's disease and CaV1 channel physiology has provided a new avenue of research for understanding the neurodegenerative process that occurs in Parkinson's disease and has also identified CaV1 channels as potential new therapeutic targets for treatment of the disorder.

It will be important to determine whether CaV1 channels function normally in patients with Parkinson's disease. That is, do they contribute to the neurodegenerative process because they are

Conflict of Interest statement

The authors declare that there are no conflicts of interest.

Acknowledgment

No funding source was involved in this work.

References (187)

  • N.C. Day et al.

    Distribution of alpha 1A, alpha 1B and alpha 1E voltage-dependent calcium channel subunits in the human hippocampus and parahippocampal gyrus

    Neuroscience

    (1996)
  • L.M. de Lau et al.

    Epidemiology of Parkinson's disease

    Lancet Neurol

    (2006)
  • A.C. Dolphin

    Calcium channel diversity: multiple roles of calcium channel subunits

    Curr Opin Neurobiol

    (2009)
  • K.L. Double et al.

    Selective cell death in neurodegeneration: why are some neurons spared in vulnerable regions?

    Prog Neurobiol

    (2010)
  • E.A. Ertel et al.

    Nomenclature of voltage-gated calcium channels

    Neuron

    (2000)
  • J. Fuchs et al.

    The transcription factor PITX3 is associated with sporadic Parkinson's disease

    Neurobiol Aging

    (2009)
  • K. Fujimura et al.

    Autogenous oscillatory potentials in neurons of the guinea pig substantia nigra pars compacta in vitro

    Neurosci Lett

    (1989)
  • J. Hardy

    Genetic analysis of pathways to Parkinson disease

    Neuron

    (2010)
  • D. Haubenberger et al.

    Association of transcription factor polymorphisms PITX3 and EN1 with Parkinson's disease

    Neurobiol Aging

    (2011)
  • A.N. Hegde et al.

    Role of ubiquitin-proteasome-mediated proteolysis in nervous system disease

    Biochim Biophys Acta

    (2011)
  • E.C. Hirsch et al.

    Neuroinflammation in Parkinson's disease: a target for neuroprotection?

    Lancet Neurol

    (2009)
  • E. Ilijic et al.

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

    Neurobiol Dis

    (2011)
  • C.A. Ingham et al.

    Spine density on neostriatal neurones changes with 6-hydroxydopamine lesions and with age

    Brain Res

    (1989)
  • P.J. Kahle et al.

    DJ-1 and prevention of oxidative stress in Parkinson's disease and other age-related disorders

    Free Radic Biol Med

    (2009)
  • P. Anglade et al.

    Apoptosis and autophagy in nigral neurons of patients with Parkinson's disease

    Histol Histopathol

    (1997)
  • G.S. Aston-Jones et al.

    Electrophysiology

  • R.B. Banati et al.

    Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson's disease

    Mov Disord

    (1998)
  • A. Barbeau et al.

    Levodopa combined with peripheral decarboxylase inhibition in Parkinson's disease

    Can Med Assoc J

    (1972)
  • C. Barcia et al.

    Evidence of active microglia in substantia nigra pars compacta of parkinsonian monkeys 1 year after MPTP exposure

    Glia

    (2004)
  • C. Becker et al.

    Use of antihypertensives and the risk of Parkinson disease

    Neurology

    (2008)
  • Y. Ben-Shlomo et al.

    Idiopathic Parkinson's disease: epidemiology, diagnosis and management

    Br J Gen Pract

    (1995)
  • M.J. Berridge et al.

    The versatility and universality of calcium signalling

    Nat Rev Mol Cell Biol

    (2000)
  • D.L. Birx et al.

    The interference of T cell activation by calcium channel blocking agents

    J Immunol

    (1984)
  • F. Blandini et al.

    Calcium homeostasis is dysregulated in parkinsonian patients with L-DOPA-induced dyskinesias

    Clin Neuropharmacol

    (2009)
  • A. Bloch et al.

    Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects

    Neuropathol Appl Neurobiol

    (2006)
  • V. Bonifati et al.

    Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism

    Science

    (2003)
  • H. Braak et al.

    Stages in the development of Parkinson's disease-related pathology

    Cell Tissue Res

    (2004)
  • H. Braak et al.

    Parkinson's disease: lesions in dorsal horn layer I, involvement of parasympathetic and sympathetic pre- and postganglionic neurons

    Acta Neuropathol

    (2007)
  • H. Büeler

    Mitochondrial dynamics, cell death and the pathogenesis of Parkinson's disease

    Apoptosis

    (2010)
  • V.S. Burchell et al.

    Targeting mitochondrial dysfunction in neurodegenerative disease: Part I

    Expert Opin Ther Targets

    (2010)
  • W.A. Catterall et al.

    International Union of Pharmacology. XLVIII. Nomenclature and structure–function relationships of voltage-gated calcium channels

    Pharmacol Rev

    (2005)
  • V. Cavallucci et al.

    Matter of life and death: the pharmacological approaches targeting apoptosis in brain diseases

    Curr Pharm Des

    (2011)
  • C.S. Chan et al.

    A molecular basis for the increased vulnerability of substantia nigra dopamine neurons in aging and Parkinson's disease

    Mov Disord

    (2010)
  • C.S. Chan et al.

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

    Nature

    (2007)
  • H. Checkoway et al.

    Epidemiologic approaches to the study of Parkinson's disease etiology

    Epidemiology

    (1999)
  • H. Cheng et al.

    Calcium sparks

    Physiol Rev

    (2008)
  • P.J. Craig et al.

    Distribution of the voltage-dependent calcium channel alpha1G subunit mRNA and protein throughout the mature rat brain

    Eur J Neurosci

    (1999)
  • P. Damier et al.

    The substantia nigra of the human brain. I. Nigrosomes and the nigral matrix, a compartmental organization based on calbindin D(28K) immunohistochemistry

    Brain

    (1999)
  • M. Day et al.

    Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models

    Nat Neurosci

    (2006)
  • M. Day et al.

    Differential excitability and modulation of striatal medium spiny neuron dendrites

    J Neurosci

    (2008)
  • Cited by (37)

    • α-Synuclein Translocates to the Nucleus to Activate Retinoic-Acid-Dependent Gene Transcription

      2020, iScience
      Citation Excerpt :

      Of note, alterations in calcium levels are implicated in the pathophysiology of α-Syn. A key pathological feature caused by α-Syn aggregation is the disruption of calcium homeostasis (Caraveo et al., 2014; Goldberg et al., 2012; Guzman et al., 2010; Hurleyand Dexter, 2012; Surmeier et al., 2017), which is implicated in mechanism of PD. Calcium binding at the C-terminus of α-Syn accelerated its oligomerization and aggregation (Rcom-H'cheo-Gauthier et al., 2016).

    • The Cellular Environment Affects Monomeric α-Synuclein Structure

      2019, Trends in Biochemical Sciences
      Citation Excerpt :

      Two features of SNpc neurons have been suggested to play an important role in their sensitivity (i.e., frequent cytosolic calcium fluctuations in the cytosol due to calcium-mediated pace-making activity and the lower levels of calcium-buffering proteins, such as calbindin, calretinin, and parvalbumin) [67–71]. A dysregulation of calcium levels in certain neurons has been observed in cases of PD [72,73], while neurons that express high levels of calcium-buffering proteins are spared [67,74]. Several studies and reviews have investigated the interaction between aSyn and calcium in PD [75–77].

    • Emerging targets and new small molecule therapies in Parkinson's disease treatment

      2016, Bioorganic and Medicinal Chemistry
      Citation Excerpt :

      The expression of calbindin, a Ca2+ binding protein in midbrain dopaminergic neurons, is increased in PD patients and animal models. Moreover, activation of Ca2+ protease has been observed in isolated PD patients and animal models.105 Nath et al. found through in vitro and in vivo experiments that higher Ca2+ levels promoted α-synuclein aggregation.

    View all citing articles on Scopus
    View full text