Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 1.
Published in final edited form as: Exp Neurol. 2017 Aug 2;298(Pt B):202–209. doi: 10.1016/j.expneurol.2017.08.001

Calcium, mitochondrial dysfunction and slowing the progression of Parkinson’s disease

D James Surmeier 1, Glenda M Halliday 2,3, Tanya Simuni 4
PMCID: PMC6037988  NIHMSID: NIHMS900371  PMID: 28780195

Abstract

Parkinson’s disease is characterized by progressively distributed Lewy pathology and neurodegeneration. The motor symptoms of cPD are unequivocally linked to the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc). Several features of these neurons appear to make them selectively vulnerable to factors thought to cause cPD, like aging, genetic mutations and environmental toxins. Among these features, Ca2+ entry through Cav1 channels is particularly amenable to pharmacotherapy in early stage cPD patients. This review outlines the linkage between these channels, mitochondrial oxidant stress and cPD pathogenesis. It also summarizes considerations that went into the design and execution of the ongoing Phase 3 clinical trial with an inhibitor of these channels – isradipine.

Determinants of pathogenesis in Parkinson’s disease

Parkinson’s disease (PD) is the most common form of a broad class of movement disorders called parkinsonism defined by the the appearance of bradykinesia, rigidity or tremor. The cardinal motor manifestations of clinical cPD (cPD) are attributable to the progressive loss of dopaminergic (DA) neurons in the SNc that innervate the basal ganglia (Berg et al., 2014; Hornykiewicz, 2002). In addition to the loss of SNc DA neurons, a hallmark of cPD is the appearance of Lewy pathology (LP) — proteinaceous inclusions exclusively found in neurons. In cPD, LP is found in a number of brain regions outside the SNc, particularly within the brainstem (Goedert et al., 2012). Braak and others have argued that this distribution of LP evolves over time (or is staged) from well-defined starting points in the caudal medulla or olfactory bulb (Beach et al., 2009; Del Tredici et al., 2002; Kosaka et al., 1984). Moreover, it was hypothesized that the progressive accumulation of LP led to neuronal loss, including that in the SNc that results in the motor symptoms of cPD.

Recent work showing that misfolded alpha-synuclein (aSYN) fibrils, which are a major component of LP, can spread from a site of brain injection through synaptically coupled networks has garnered the Braak hypothesis new adherents (Luk et al., 2012; Peelaerts et al., 2015; Volpicelli-Daley et al., 2014; 2011). Moreover, because aSYN fibrils appear to be capable of templating the misfolding of endogenous aSYN, the hypothetical spreading of LP has been likened to a prion-like process (Brundin et al., 2010; Olanow and Brundin, 2013). This hypothesis has obvious translational implications. If transynaptic spreading of LP is the driving force in cPD, then the goal of disease modifying therapies should be focused on strategies to slow or stop this spread. Indeed, this effort is underway (NCT02459886).

Although attractive in its simplicity, there are compelling reasons to think that the neuropathology in cPD is not simply a consequence of a prion-like spreading of misfolded aSYN in the brain. Many of the issues surrounding this hypothesis have been recently outlined (Uchihara and Giasson, 2016; Walsh and Selkoe, 2016), including one by us (Surmeier et al., 2017). There are four basic problems with the simple prion variant of the Braak model. First, the pattern of LP in cPD brains is variable, with only about half the reliably diagnosed brains conforming to the Braak model, raising questions about the nature of the seeding event (Halliday et al., 2012; Kalaitzakis et al., 2008). Second, the distribution of LP in cPD cases is not predicted by what is known about the brain connectome alone; in particular, the strength of synaptic connections between sites of early LP and the rest of the brain does not predict the distribution of LP later in the disease (Surmeier et al., 2017). The pattern of LP also is not consistent with a spread to nearest neighbors, even within the same nucleus (Kingsbury et al., 2010).. Third, the relationship between LP and neurodegeneration is uncertain. In humans, neuronal loss, which can cleanly be linked to symptoms, evolves with a very different spatiotemporal pattern than does LP. For example, neuronal loss in the SNc precedes any discernible LP. Moreover, some patients with cPD have no discernible LP, whereas others who do, fail to manifest cPD symptoms (Dijkstra et al., 2014). The question is not whether aSYN fibrils or monomers in sufficient quantities can kill neurons; they clearly can. The issue is whether this is what happens in the human brain. And lastly, there is no compelling longitudinal data from cPD patients to support a spreading pathology. Adherents of the spreading hypothesis would argue that this is simply because there aren’t good biomarkers for disease progression. While it is true that there aren’t validated progression markers, particularly for the early, ‘presymptomatic’ phases of the disease, the absence of longitudinal data is a shortcoming of the case for the prion hypothesis and should promote a healthy skepticism.

The rationale for a Cav1 Ca2+ channel inhibitor

What other factors might contribute to cPD pathology? One approach to this question is to look at the properties of vulnerable neurons to determine if they have common features that might be affected by risk-factors associated with cPD: age, exposure to environmental toxins and a collection of genetic mutations (Surmeier et al., 2017). Indeed, many of the neurons that are most profoundly affected in cPD have a loosely connected functional role in the brain. They are principal neurons in neuromodulatory control networks, contrasting them with neurons in brain networks responsible for epicritic sensation and precise motor control. The SNc, locus ceruleus (LC), raphe nuclei (RN), pedunculopontine nucleus (PPN), basal forebrain nuclei (BFN), gigantocellularis nucleus (GCN), lateral hypothalamus (LH) and thalamic intralaminar nuclei (ILN) are involved in arousal or mobilization of sensorimotor networks necessary for rapid and effective action, which is critical to vigilance, escape and attack (Alexandre et al., 2013; Aston-Jones and Waterhouse, 2016; Palmiter, 2011; Pfaff et al., 2012; Saper et al., 2005; Sara and Bouret, 2012). The dorsal motor nuclei (DMV) and nucleus tractus solitarius (NTS) – two caudal brainstem nuclei manifesting LP in cPD patients – share a similar role through their control of the autonomic nervous system (Saper, 2003; Silvani et al., 2016). In the forebrain, the amygdala and limbic cortices, which have LP early in cPD, are part of a default activity network in the forebrain that regulates fear and affective behaviors – again, key parts of coordinating appropriate action in response to salient events in a threatening environment (Miskovic and Schmidt, 2012).

Although much remains to be done, it appears that these neurons share a number of traits that might put them at-risk. The most notable and best characterized of these is a long and highly branched axon with a large number of transmitter release sites. This diffuse axonal arbor helps these neurons coordinate the activity in large networks, like the basal ganglia or the spinal cord. For example, SNc DA neurons in the rodent have axons that branch profusely in the striatum and possess as many as 200,000 vesicular release sites (Matsuda et al., 2009). Although less well characterized, neurons in the DMV, GCN, RN, LC, PPN, BFN, LH and ILN all share this feature (large, diffuse axonal projections) to varying degrees, distinguishing them from the vast majority of sensory or motor neurons in the brain, which typically have spatially focused, modestly branched axons that conform to topographic maps (Aston-Jones and Waterhouse, 2016; Baufreton et al., 2009; Hornung, 2003; Hu et al., 2015; Liu et al., 2015; Martinez-Gonzalez et al., 2011; Pfaff et al., 2012; Ratcliffe et al., 2011). Why might a long and highly branched axon increase vulnerability? There are several theories that have been proposed (Bolam and Pissadaki, 2012; Hunn et al., 2015; Pacelli et al., 2015). But, not all neurons with long, branched axons are vulnerable in cPD (e.g., striatal cholinergic interneurons (Zhou et al., 2002)), suggesting that some other factor is in play.

Another shared feature of at-risk neurons appears to be their distinctive physiology. In vivo, at-risk neurons that have been studied have slow tonic activity (Surmeier et al., 2017). The best studied member of this class is the SNc DA neuron. The action potential of these neurons is slow and broad, which maximizes Ca2+ entry and promotes slow rhythmic activity (Bean, 2007). The slow, rhythmic activity (2–10 Hz) in these neurons is autonomously generated and accompanied by slow oscillations in intracellular Ca2+ concentration that are triggered by the opening of plasma membrane Cav1 (Cav1.2, Cav1.3) Ca2+ channels and release of Ca2+ from intracellular, endoplasmic reticulum (ER) stores (Guzman et al., 2010; Morikawa and Paladini, 2011; Nedergaard et al., 1993; Puopolo et al., 2007). Once in the cytoplasm, Ca2+ is relatively free to interact with other proteins as the abundance of Ca2+ buffering proteins, like calbindin, is low (Anderegg et al., 2015; Brichta et al., 2015; Poulin et al., 2014; Sulzer and Surmeier, 2013; Surmeier et al., 2016). This combination of features – broad spikes, pacemaking, low intrinsic Ca2+ buffering and cytosolic Ca2+ oscillations – (not any one) is what distinguishes SNc DA neurons. For example, VTA DA neurons, which are significantly less vulnerable than SNc DA neurons (see above), are autonomous pacemakers with broad spikes, but have smaller Cav1 channel currents and strong intrinsic Ca2+ buffering (Guzman et al., 2009; Khaliq et al., 2010). Although there have been very few studies that have examined these features in other at-risk neurons, those that have (LC, DMV, PPN) show that this phenotype is largely shared (Goldberg et al., 2012; Kang and Kitai, 1990; Sánchez-Padilla et al., 2014).

The slow Ca2+ oscillations in at-risk neurons sub-serve two complementary functions. First, because they are electrogenic, the oscillations help maintain the slow tonic spiking in these neurons (Nedergaard et al., 1993; Puopolo et al., 2007; Putzier et al., 2009a). Ca1.3 channels, because they activate at sub-threshold membrane potentials, are critical to this function (Guzman et al., 2009; Helton et al., 2005; Puopolo et al., 2007). Second, although less well established, they promote Ca2+ entry into mitochondria, oxidative phosphorylation (OXPHOS) and the production of ATP (Guzman et al., 2010; Llorente-Folch et al., 2015; Sánchez-Padilla et al., 2014). In principle, this feed-forward control of OXPHOS helps to ensure that bioenergetic needs are met (Balaban, 2009; Nicholls, 1998) and that intracellular ATP levels do not fall into a range that would trigger protective activation of K-ATP channels and cessation of on-going activity (Dragicevic et al., 2015). Even temporary cessation of activity in neuronal networks necessary to mobilize sensory and motor systems directing escape or attack behavior would lessen the chances of survival in an unpredictable environment. As a consequence, there should have been strong evolutionary pressure to design neurons in these ‘too important to fail’ networks with this type of feed-forward control mechanism.

There are two obvious downsides of this design. First, stimulating OXPHOS when mitochondria are hyperpolarized in the absence of strong ATP demand increases the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) (Goldberg et al., 2012; Guzman et al., 2010; Sánchez-Padilla et al., 2014; Votyakova and Reynolds, 2001). ROS and RNS damage proteins, lipids and DNA, particularly in mitochondria. Sustained oxidant stress could be a major factor underlying declining mitochondrial function in at-risk neurons with age (Reeve et al., 2014). ROS and RNS also can exacerbate the impact of genetic mutations and environmental toxins affecting mitochondria (Gegg and Schapira, 2016), as well as increase the propensity of aSYN to aggregate (Gupta et al., 2009). The second downside is that this mechanism results in sustained elevations in cytosolic Ca2+ concentration. Ca2+ promotes aSYN aggregation both directly (Rcom-H'cheo-Gauthier et al., 2014) and indirectly through activation of calpain and calcineurin (Caraveo et al., 2014; Diepenbroek et al., 2014; Dufty et al., 2007). Elevated cytosolic Ca2+ also impairs lysosomal function and turnover of misfolded proteins (Gómez-Sintes et al., 2016; Medina and Ballabio, 2015), potentially synergizing with other defects in proteasomal/autophagic function to increase the likelihood of LP (Wong and Cuervo, 2010). Thus, these vulnerable neurons appear to reside close to mitochondrial and degradative ‘tipping points’.

Do other at-risk neurons conform to this model? In-depth analysis has only been performed in SNc, LC and DMV neurons. While much of the brainstem data is consistent with a shared phenotype, more in-depth phenotyping needs to be done. However, healthy, young telencephalic neurons are not phenocopies of SNc dopaminergic neurons. That said, many of the telencephalic regions at-risk in cPD (and AD) are part of a ‘default’ network that manifests high resting activity, albeit of synaptic origin (Andrews-Hanna et al., 2007). It is possible that in aged, late stage cPD patients, network dysfunction (Hammond et al., 2007; Ko et al., 2013) triggers adaptations that bring these neurons and networks phenotypically closer to other at-risk neurons. Cav1 Ca2+ channels, which are key determinants of the SNc phenotype, could be a major factor in this process. Sustained Ca2+ entry through Cav1 channels in forebrain neurons has long been associated with aging-related cognitive decline and AD (Disterhoft et al., 1994; Thibault et al., 2007). Moreover, in cPD patients, Cav1 Ca2+ channels are up-regulated in limbic and motor cortices (Hurley et al., 2013; 2014).

Can the phenotype of at-risk neurons account for LP staging? The simple answer is no. From what we currently know about cell autonomous risk factors, LP should appear in the SNc before it does in the DMV. Barring the emergence of some other cell autonomous factor that drives LP, the most parsimonious explanation of the LP pattern in cPD is that there is spreading of aSYN pathology – as posited by Braak et al. and the proponents of the prion model – but that spreading is limited to a subset of neurons whose phenotype renders them susceptible to spreading – a proposition that is very consistent with the phenotype outlined above.

What is better explained by cell autonomous factors is the sequence of cell death in cPD. The earliest known loss of neurons in cPD is the SNc (Surmeier et al., 2017). These neurons are at one extreme of the anatomical, physiological and molecular spectrum of vulnerable neurons as we currently understand it (Anderegg et al., 2015; Brichta et al., 2015; Poulin et al., 2014; Sulzer and Surmeier, 2013; Surmeier et al., 2016), exhibiting the highest basal levels of mitochondrial oxidant stress and free cytosolic Ca2+ of any cell examined. Mitochondria and intracellular Ca2+ are linchpins of all three major death cascades (apoptotic, autophagic and necrotic) (Nagley et al., 2010). In human SNc, there are telltale signs of sustained mitochondrial oxidant stress with aging and cPD, such as mitochondrial DNA deletions (Bender et al., 2006; 2008). Against this backdrop, it makes sense that genetic mutations that compromise mitochondrial oxidant defenses, biogenesis or quality control cause the preferential loss of SNc dopaminergic neurons and early onset forms of cPD (Kumaran and Cookson, 2015; Lin and Farrer, 2014; Mullin and Schapira, 2015). The tipping point for these neurons also could be reached by other genetic mutations that indirectly compromise mitochondrial function (Brini et al., 2014; Gegg and Schapira, 2016; Guardia-Laguarta et al., 2015; McCoy and Cookson, 2012; Mullin and Schapira, 2013).

Elevated cytosolic Ca2+, aSYN and DA in SNc DA neurons could be a particularly toxic combination, especially in axon terminals and dendrites (Brimblecombe et al., 2015; Caraveo et al., 2014; Dryanovski et al., 2013; Mosharov et al., 2009). Indeed, striatal DA axon terminals appear to be lost early in the development of cPD, preceding the loss of DA cell bodies (Kordower et al., 2013). In this regard, the inference that levodopa therapy does not accelerate disease progression (Fahnand the Parkinson Study Group, 2005) might be wrong if the primary site of DA toxicity is the axon terminal – terminals that are largely gone by the time levodopa therapy is usually started.

If cell autonomous factors are critical to the evolution of cPD, then ‘normalizing’ one or more of these factors should slow disease progression. As outlined above, Ca2+ entry through Cav1 – particularly Cav1.3 – Ca2+ channels appears to be a major driver of mitochondrial oxidant stress in the at-risk neurons examined to date. Moreover, these channels can be targeted. Dihydropyridines (DHPs) are FDA-approved, selective negative allosteric modulators (NAMs) of Cav1 channels that have good brain bioavailability (Anekonda et al., 2011a; Striessnig et al., 1998; Surmeier et al., 2016). Epidemiological studies have consistently found that the use of DHPs is associated with a decreased risk of developing cPD (Becker et al., 2008; Gudala et al., 2015; Lee et al., 2014; Pasternak et al., 2012; Ritz et al., 2010); their use even seems to slow progression after diagnosis (Marras et al., 2012). The combination of preclinical and clinical data implicating Cav1 channels in cPD pathogenesis provide the rationale for testing DHPs as potential diseas modifying agents in cPD.

One of the shortcomings of DHPs as a disease modifying therapy is their ‘off-target’ inhibition of Cav1 channels in vascular smooth muscle (and elsewhere). These off-target effects, which are primarily mediated by Cav1.2 channels, limit dosing. Because DHPs are voltage-dependent NAMs that bind preferentially to channels that are relatively depolarized (Bean, 1984), some of these off-target effects will be diminished by the fact that most neurons reside primarily at relatively hyperpolarized membrane potentials, where DHPs have a lower affinity; this should limit DHP action to cells that are depolarized for prolonged periods of time, like pacemaking neurons at-risk in cPD. Another translational consideration is that Cav1.3 Ca2+ channels, rather than the more common Cav1.2 channels, are likely to be the most important drivers of risk in PD. Most DHPs preferentially inhibit Cav1.2 channels; isradipine differs from most DHP in that it has nearly the same affinity for Cav1.2 and Cav1.3 channels in membrane binding assays (Sinnegger-Brauns et al., 2009). This consideration motivated the use of isradipine in preclinical studies and subsequent clinical trials (see below).

A fundamental question is whether isradipine can be given to humans at doses high enough to produce a clinically significant inhibition of Cav1.3 channels in SNc DA neurons and other neurons at-risk. There are several reasons to think this is achievable. First, peripheral administration of isradipine, at doses that were well tolerated in mice, protected SNc DA neurons against a mild, distributed striatal injection of the toxin 6-OHDA (Ilijic et al., 2011). Second, unpublished work by our group has shown that systemic administration of isradipine suppresses cytosolic Ca2+ transients, elevates mitochondrial mass and lowers mitophagy in SNc DA neurons (Guzman et al., unpublished results). Third, epidemiological data have consistently found that DHPs lower PD risk (see above).

A recent study has challenged this conclusion (Ortner et al., 2017). The authors claim that systemic administration of isradipine does not protect SNc DA neurons against striatal injection of 6-OHDA. As pointed out by our earlier work (Ilijic et al., 2011), the ability of isradipine to protect against a 6-OHDA challenge requires that the insult be modest and spatially distributed. Nevertheless, in their attempt to test isradipine, Ortner et al. used a 6-OHDA concentration that Ilijic et al. (2011) had shown to be refractory to systemic isradipine treatment; this choice makes their negative result problematic. Also, it is worth noting that other groups have reported that systemic administration of DHPs affords protection in both PD and Alzheimer’s disease models (Anekonda et al., 2011b; Kupsch et al., 1996). In an attempt to explain their result, the authors estimate the isradipine IC50s of Cav1.2 and Cav1.3 Ca2+ channels using a voltage trajectory recorded from SNc DA neurons; this interesting set of expeirments showed that in this circumstance Cav1.2 channels and Cav1.3 channels had IC50 values that were less than 10 nM, with the IC50 value of Cav1.2 channels being somewhat lower than that of Cav1.3 channels (cf., Sinnegger-Brauns et al., 2009). The authors take this information and then use imaging to assess the ability of isradipine – at more than three times the IC50 value for both Cav1.2 and Cav1.3 channels (30 nM) – to reduce the intracellular Ca2+ transient associated with pacemaking in SNc DA neurons. Paradoxically, the authors found no inhibition, contradicting a large literature showing the rich expression of Cav1.2 and Cav1.3 Ca2+ channels in these neurons throughout their somatodendritic membrane (e.g., Guzman et al., 2009; Nedergaard et al., 1993; Puopolo et al., 2007; Putzier et al., 2009b). The authors provide no plausible explanation for this apparent contradiction. On the basis of these two negative results, the authors assert that isradipine is unlikely to produce a significant inhibition of Cav1.3 channels in patients. Not only do we disagree with this conclusion because of the shortcomings in the work, their IC50 estimates suggest the opposite conclusion; namely, that at clinically relevant doses of isradipine where brain levels should reach or exceed 5–10 nM, there should be significant (~50%) inhibition of both Cav1.2 and Cav1.3 channels in SNc DA neurons. That said, it is unclear whether this will be enough to slow PD pathogenesis. Nevertheless, the caveats of using non-selective DHPs are real and underscores the need to develop a more selective Cav1.3 channel inhibitor.

From the bench to clinical trial

The section below outlines the pathway for clinical translation of the preclinical data, summarizes the design of the on-going Phase III clinical trial of isradipine and highlights the challenges of development of cPD disease modifying inetrventions in absence of validated biomarkers of disease progression.

As outlined above, DHPs are selective NAMs of Cav1 Ca2+ channels. They are Food and Drug Administration (FDA) approved for use as anti-hypertensive agents (Zhang et al., 2007). Isradipine has been FDA approved since 1990 and has extensive safety data in the hypertensive patient population. Isradipine was selected for clinical testing because it has a high affinity for both types of Cav1 channels expressed in SNc DA neurons, having roughly the same binding affinity for Cav1.2 and Cav1.3 channels (Sinnegger-Brauns et al., 2009) (see above). In contrast, nifedipine has a roughly five times higher affinity for Cav1.2 channels. Isradipine also has excellent brain penetration (Anekonda et al., 2011a; Ilijic et al., 2011).

As a first step toward a disease modification trial, the safety of isradipine was tested in Phase Ib and Phase II studies in early stage cPD patients. These studies found that isradipine had acceptable safety and tolerability at doses of 10mg/day or less (Parkinson Study Group, 2013; Simuni et al., 2010). The combination of the preclinical rationale, epidemiological data linking DHPs to reduced risk of cPD and the Phase Ib/II safety data motivated NIH to mount a 5 year, Phase III, disease modification clinical trial of isradipine in early stage cPD (STEADY-PD III, clintrials.gov NCT02168842). The study is being conducted at 57 Parkinson Study Group (PSG) sites in North America and is funded by the National Institute of Neurological Disorders and Stroke (NINDS) and the Michael J. Fox Foundation.

Design and execution of the Phase III trial

STEADY-PD III (NCT02168842) is an ongoing 36-month double-blind randomized, placebo-controlled study of isradipine in 336 participants with early stage cPD, who at enrollment were not receiving or requiring symptomatic therapy (ST). The study is testing the hypothesis that individuals given isradipine will have slower progression of cPD disability over the 36 month trial period, as determined by the change in the total Unified Parkinson Disease Rating Scale (UPDRS) score. Isradipine (5 mg) is being given twice daily for a total daily dose of 10 mg.

Trial participants had to fulfill several inclusion and exclusion criteria. Inclusion criteria were: 1) they had to be diagnosed with early stage, idiopathic cPD based upon the presence of two out of three cardinal manifestations of cPD (Hughes et al., 1992); 2) they had to be older than 29 years of age at the time of diagnosis; 3) they had to be Hoehn and Yahr stage less than or equal to 2 (Hoehn and Yahr 1967); 4) they had to have been diagnosed less than 3 years prior to enrollment; 5) they were not to have received symptomatic medication (levodopa, dopamine agonist or MAO-B inhibitors) and not projected to require medication for at least 3 months from enrollment. Use of amantadine and/or anticholinergics was allowed prior to enrollment, as was use of anti-hypertensives other than Ca2+ channel inhibitors. The key exclusion criteria were: 1) a diagnosis of atypical parkinsonism; 2) prior exposure to symptomatic medication; 3) a history of orthostatic hypotension, bradycardia, congestive heart failure or other cardiac and other systemic diseases; 4) abnormalities on the screening labs or ECG that might preclude safe participation in the study; 5) the presence of cognitive dysfunction defined by a Montreal Cognitive assessment (MOCA) score of less than 26 (Nasreddine et al., 2005); 6) clinically significant depression as determined by a Beck Depression Inventory II (BDI-II) score greater than 15 (Beck and Beamesderfer, 1974).

The primary outcome measure of the trial is the change in total UPDRS score during the 36 month trial period. Majority of participants were expected to start taking symptomatic medications during the trial and as such the study was designed and powered to test isradipine efficacy by comparing the baseline UPDRS with the end-of-trial UPDRS, measured in the medication on-state, accounting for the effect of the symptomatic treatment. The UPDRS is a valid and reliable measure of cPD disability that has been effectively used in a number of cPD trials. The study is powered to detect a 25% slowing of functional decline with isradipine above the benefit from symptomatic therapy, a difference that would be sufficient to influence clinical practice and may suggest the likelihood of longer term benefit.

The secondary outcomes of clinical importance include: 1) time to initiation of symptomatic therapy, which has been used as a primary outcome measure in several previous studies of putative disease modifying agents and reflects progression early in disease not obscured by symptomatic therapy (Parkinson Study Group, 1996; Parkinson Study Group PRECEPT Investigators, 2007); 2) time to and severity of motor complications, which is a another measure of disease progression (Rascol et al., 2000) ; 3) reduced need for symptomatic therapy (as measured by the levodopa equivalent dosages between treatment groups (Tomlinson et al., 2010); 4) the incidence and severity of non-motor symptoms, as these contribute disproportionately to quality of life and reflect clinically relevant outcomes in cPD.

Finally, there are exploratory outcome measures that include global measures of functional disability, quality of life, ambulatory capacity and cognitive function as measured by MOCA. The trajectory of UPDRS change before and after initiation of symptomatic therapy also will be modeled.

Biosamples are obtained from patients. Blood is obtained at enrollment and at 3 and 6 month follow-up visits to confirm isradipine pharmokinetic profiles. In addition, blood samples obtained at enrollment will be used to extract DNA for genetic testing and samples obtained at the conclusion of the study will be stored for future reserach.

The rationale for the study design

PD is a slowly progressing neurodegenerative disease. Previously conducted disease-modification studies enrolled participants with newly diagnosed cPD not yet requiring symptomatic treatment and followed them for a relatively short period of time (12–24 months) and data were censored at the time of initiation of symptomatic treatment (Hart et al., 2009). These previous trial designs were driven by lack of objective biomarkers of cPD progression and the significant impact of symptomatic treatment on standard clinical outcome measures. However, they do not address the “real life scenario” in which the disease modification strategy might slow but not stop disease progression, requiring that all patients ultimately be treated for cPD symptoms. On average 50% of de novo cPD patients initiate symptomatic treatment within one year of diagnosis and nearly all patients require therapy within three years (Parashos et al., 2009)(Ravina et al., 2009). If isradipine slows this progression, it would be an important advance. With our trial design, this can be assessed by the secondary outcome measures (time to initiation of symptomatic treatment and differential use of symptomatic therapy). It also is unclear whether an intervention that is effective early in the course of the disease will affect progression later in the disease; that is, will the effects of treatment persist? Previous trial designs don’t allow this question to be answered.

STEADY-PDIII attempts to address these limitations by extending the drug treatment period to 36 months and allowing symptomatic medication to be initiated early in the trial. At 36 months post diagnosis, nearly all participants are expected to be treated with symptomatic therapies. The primary outcome measure is the change in UPDRS score from the time of enrollement to that obtained in the “on-state” at the end of the trial 36 months later. This will allow us to identify the benefit of isradipine “on top” of the benefit conferred by symptomatic therapy – an outcome with “real world” relevance to patients and clinicians. The UPDRS “off-state” was considered as an alternative primary outcome. While it may be argued that this assessment is a better representation of dopaminergic deficit, this is not supported by the clinical data. Both levodopa and dopamine agonists have shown long duration effects on UPDRS, lasting for days and even weeks. So, the traditional 12 hours off medication for “off-state” assessment would not reflect the true dopaminergic deficit (Fahn et al., 2004; Stocchi et al., 2001). Nevertheless, this assessment is of value and the “off-state” motor UPDRS will be used as a secondary outcome measure once symptomatic treatment has been initiated.

In regard to the study duration, 36 months was chosen as a compromise between the attempt to assess long-term efficacy of isradipine (if it exists) and feasibility. While 36 months is still a short period for a slowly progressing disease like cPD, and long-term cPD complications are not expected (e.g., like postural instability and dementia) a longer study was fiscally problematic and would have been compromised by retention issues. Nevertheless, this is the longest duration ever proposed for a study in baseline de-novo untreated cPD population and is likely long enough to provide insight into the effects of isradipine on relevant motor and nonmotor outcomes. It is also short enough to maximize participant retention. Thus, the study design is novel in that it allows us to use a relatively small cohort of patients to better test the hypothesis that isradipine will slow disease progression and add to the benefit derived from symptomatic treatment.

Several alternative study designs were considered. For example, a ”simple long duration study” design (LS-1) was considered, but it would have required in excess of 1500 participants and 7–8 years to complete (Elm,NINDS NET-PD Investigators, 2012). Another design utilized in cPD disease modification trials is the delayed-start design (Olanow et al., 2009). The arguments against a delayed start design in our case are 1) the lack of demonstrable symptomatic benefit of isradipine, 2) the requirement of > 1000 participants for sufficient power, and 3) controversy on its ability to demonstrate disease modification in cPD. Another design to consider would enroll individuals at the time of initiation of symptomatic therapy (like CALM-PD) (Parkinson Study Group CALM Cohort Investigators, 2009). However, this would not allow to test the impact of isradipine on progression early in disease (before symptomatic treatment). In addition, enrolling subjects as early as possible in the disease process would allow the neuroprotective benefit of isradipine (if such an effect exists) to be maximized. A prolonged wash out at the end of study or at the time of initiation of symptomatic treatment was considered to reassess for the evidence of symptomatic benefit, but there are strong arguments against such design, including lack of obvious symptomatic effect of isradipine in our Phase II STEADY-PDII study; the participant burden is also a drawback. Moreover, there is no consensus on the necessary duration of the washout that would be required for isradipine (Holford and Nutt, 2011). Therefore, although not without shortcomings, our design appears to be the most valid currently available approach to study the effect of isradipine on progression of disability in cPD. It should be highlighted that as of today based on expert concensus and discussions with the FDA, there is no single “preferred” study design to test disease modification in cPD.

There are a number of study limitations to be considered. The major limitation of our trial – and all others at this point – is lack of validated biomarkers of disease progression. This has two consequences. One is that our study is limited to patients who have already progressed to the point that symptoms are manifest corresponding to more advanced stages of neurodegeneration. Disease modification with isradipine or other drugs may only be feasible in the premotor phases of the disease. The other is that we use a clinical scale, UPDRS, as the primary outcome measure. UPDRS, while validated and a widely used scale in cPD clinical trials, does not directly assess biological disease progression. DNA and plasma samples are being collected for future analyses of novel biomarkers that could address these limitations.

Another limitation of our study is that it is not clear that the dose of isradipine chosen is high enough. At present, there is no good way to assess the relevant pharmacokinetic features of isradipine in humans, particularly target (Cav1 channel) engagement in the SNc. The tested dose (10 mg/day) was chosen based on the tolerability in the Phase II study and while that dose achieves serum concentrations that were neuroprotective in animal models, it is not clear that it is sufficient in humans. Again, pharmacokinetic samples will help determine whether isradipine concentrations that were effective in animal models were achieved and whether variability in clinical response was related to variations in serum concentrations.

Our study uses a novel primary outcomer measure, UPDRS in the medications ON state. While such approach has a number of advantages as discussed above, the study may be criticized for lack of definitive way to exclude symptomatic effect of isradipine which could account for the benefit compared to placebo (if the study was positive). Our rationale for such a design are data from the phase II study that demonstrated lack of symptomatic effect of isradipine based on the short term change in UPDRS after initiation of treatment (assessed every 2 weeks up to 3 months) and after 2 weeks taper between active treatment arms and placebo. In addition, the mechanism of action of isradipine preclinical data do not point to a potential symptomativc effect of the compound. However, such effects can’t be completely excluded short of conducitng a study with a prolonged wash out which is also subject to pitfalls as discussed above. Even if a symptomatic effect of isradipine existed and persisted long term ON TOP of exisiting symptomatic therapy, that in conjunction with the compelling preclinical data of neuroprotecitve effect of NAMs, would be a significant contribution to the existing armamentarium of cPD therapeutics.

Lastly, it should be noted that STEADY cPD III is conducted under the FDA Investigational New Drug (IND) path with no plans to license isradipine for cPD use. Such a path was chosen based on the fact that isradipine is an FDA approved drug that is generic and provided the study showed positive results, it wil be readily available to the cPD community.

Current status of the study

Enrollment of the 336 participants began in November 2014 and was completed in October 2015 at 55 of the 57 active PSG sites. Enrollment was completed 6 months ahead of the expected timeline. The final subject is expected to complete the study in November 2018. As of May 2017, 330 participants remain active in the study.

Acknowledgments

This work was supported by NIH grant NS047085 and grants from the JPB and IDP Foundations to DJS. The STEADY-PD III is supported by NIH grant U01NS080818 and biomarker substudy by the MJFF. GMH is a National Health and Medical Research Council of Australia Senior Principal Research Fellow (grant #1079679).

Dr. Simuni has served as a consultant received consulting fees from Acadia, Abbvie, Allergan, Anavex, Avid, GE Medical, Eli Lilly and Company, Harbor, Ibsen, IMPAX, Lundbeck, Merz, Inc., the National Parkinson Foundation, Navidea, Pfizer, TEVA Pharmaceuticals, UCB Pharma, Voyager, US World Meds, and the Michael J. Fox Foundation for Parkinson’s Research; Dr. Simuni has served as a speaker and received an honorarium from Acadia, IMPAX, Lundbeck, TEVA Pharmaceuticals, and UCB Pharma; Dr Simuni is on the Scientific advisory board for Anavex, Sanofi, MJFF. Dr. Simuni sits on the Advisory Board for IMPAX; Dr. Simuni has received research funding from the NINDS, MJFF, NPF, TEVA Pharmaceuticals, Auspex, Biotie, Civitas, Acorda, Lundbeck, Neuroderm, NINDS, National Institutes of Health, Northwestern Foundation, and the Michael J. Fox Foundation for Parkinson’s Research; Dr. Simuni received funding support for educational programs from GE Medical, TEVA, and Lundbeck.

Dr. Halliday serves on the scientific advisory board of DANDRITE; has been a consultant for the NHMRC, GSK and Lundbeck Neuroscience; received conference travel funds from AAIC, Int Soc Neurochemistry, Int DLB Conference, AAN, Int MSA Conference, NHMRC National Institute for Dementia Research, 2nd Chinese Brain Banking Meeting, Japanese Neuroscience Soc; receives royalties from Academic press, Elsevier & Oxford University Press; receives research grant funding from NHMRC (1008307, 1037746 & 1079679), MJ Fox Foundation, Shake-it-up Australia, Parkinson’s NSW, and the Universities of Sydney & NSW (infrastructure & equipment).

Dr. Surmeier serves on scientific advisory boards of Pfizer and Adamas Pharmaceuticals. He serves as a consultant for Lundbeck Neuroscience. Dr. Surmeier receives research funding from NIH, MJFF, the JPB Foundation, CHDI, IDP Foundation and Adamas Pharmaceuticals.

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.

Author Financial Disclosures: Neither DJS, GH, or TS have any financial interests linked to the topic of this review.

References

  1. Alexandre C, Andermann ML, Scammell TE. Control of arousal by the orexin neurons. Curr Opin Neurobiol. 2013;23:752–759. doi: 10.1016/j.conb.2013.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Anderegg A, Poulin J-F, Awatramani R. Molecular heterogeneity of midbrain dopaminergic neurons - Moving toward single cell resolution. FEBS Lett. 2015;589:3714–3726. doi: 10.1016/j.febslet.2015.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Andrews-Hanna JR, Snyder AZ, Vincent JL, Lustig C, Head D, Raichle ME, Buckner RL. Disruption of large-scale brain systems in advanced aging. Neuron. 2007;56:924–935. doi: 10.1016/j.neuron.2007.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. 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. 2011a;41:62–70. doi: 10.1016/j.nbd.2010.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. 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. 2011b;41:62–70. doi: 10.1016/j.nbd.2010.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Aston-Jones G, Waterhouse B. Locus coeruleus: From global projection system to adaptive regulation of behavior. Brain Res. 2016 doi: 10.1016/j.brainres.2016.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Balaban RS. The role of Ca2+ signaling in the coordination of mitochondrial ATP production with cardiac work. Biochimica et Biophysica Acta (BBA) - Bioenergetics. 2009;1787:1334–1341. doi: 10.1016/j.bbabio.2009.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Baufreton J, Kirkham E, Atherton JF, Menard A, Magill PJ, Bolam JP, Bevan MD. Sparse but selective and potent synaptic transmission from the globus pallidus to the subthalamic nucleus. J Neurophysiol. 2009;102:532–545. doi: 10.1152/jn.00305.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, Sasse J, Boyer S, Shirohi S, Brooks R, Eschbacher J, White CL, Akiyama H, Caviness J, Shill HA, Connor DJ, Sabbagh MN, Walker DG Arizona Parkinson's Disease Consortium. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009;117:613–634. doi: 10.1007/s00401-009-0538-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bean BP. The action potential in mammalian central neurons. Nat Rev Neurosci. 2007;8:451–465. doi: 10.1038/nrn2148. [DOI] [PubMed] [Google Scholar]
  11. Bean BP. Nitrendipine block of cardiac calcium channels: high-affinity binding to the inactivated state. Proc Natl Acad Sci U S A. 1984;81:6388–6392. doi: 10.1073/pnas.81.20.6388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Beck AT, Beamesderfer A. Assessment of depression: the depression inventory. Mod Probl Pharmacopsychiatry. 1974;7:151–169. doi: 10.1159/000395074. [DOI] [PubMed] [Google Scholar]
  13. Becker C, Jick SS, Meier CR. Use of antihypertensives and the risk of Parkinson disease. Neurology. 2008;70:1438–1444. doi: 10.1212/01.wnl.0000303818.38960.44. [DOI] [PubMed] [Google Scholar]
  14. Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK, Perry RH, Jaros E, Hersheson JS, Betts J, Klopstock T, Taylor RW, Turnbull DM. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet. 2006;38:515–517. doi: 10.1038/ng1769. [DOI] [PubMed] [Google Scholar]
  15. Bender A, Schwarzkopf RM, McMillan A, Krishnan KJ, Rieder G, Neumann M, Elstner M, Turnbull DM, Klopstock T. Dopaminergic midbrain neurons are the prime target for mitochondrial DNA deletions. J Neurol. 2008;255:1231–1235. doi: 10.1007/s00415-008-0892-9. [DOI] [PubMed] [Google Scholar]
  16. Berg D, Postuma RB, Bloem B, Chan P, Dubois B, Gasser T, Goetz CG, Halliday GM, Hardy J, Lang AE, Litvan I, Marek K, Obeso J, Oertel W, Olanow CW, Poewe W, Stern M, Deuschl G. Time to redefine PD? Introductory statement of the MDS Task Force on the definition of Parkinson's disease. Mov Disord. 2014;29:454–462. doi: 10.1002/mds.25844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bolam JP, Pissadaki EK. Living on the edge with too many mouths to feed: Why dopamine neurons die. Mov Disord. 2012;27:1478–1483. doi: 10.1002/mds.25135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Brichta L, Shin W, Jackson-Lewis V, Blesa J, Yap E-L, Walker Z, Zhang J, Roussarie J-P, Alvarez MJ, Califano A, Przedborski S, Greengard P. Identification of neurodegenerative factors using translatome-regulatory network analysis. Nat Neurosci. 2015;18:1325–1333. doi: 10.1038/nn.4070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Brimblecombe KR, Gracie CJ, Platt NJ, Cragg SJ. Gating of dopamine transmission by calcium and axonal N-, Q-, T- and L-type voltage-gated calcium channels differs between striatal domains. J Physiol (Lond) 2015;593:929–946. doi: 10.1113/jphysiol.2014.285890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brini M, Calì T, Ottolini D, Carafoli E. Neuronal calcium signaling: function and dysfunction. Cell. Mol. Life Sci. 2014;71:2787–2814. doi: 10.1007/s00018-013-1550-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol. 2010;11:301–307. doi: 10.1038/nrm2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Caraveo G, Auluck PK, Whitesell L, Chung CY, Baru V, Mosharov EV, Yan X, Ben Johny M, Soste M, Picotti P, Kim H, Caldwell KA, Caldwell GA, Sulzer D, Yue DT, Lindquist S. Calcineurin determines toxic versus beneficial responses to α-synuclein. Proceedings of the National Academy of Sciences. 2014;111:E3544–52. doi: 10.1073/pnas.1413201111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Del Tredici K, Rub U, de Vos RA, Bohl JR, Braak H. Where does parkinson disease pathology begin in the brain? J Neuropathol Exp Neurol. 2002;61:413–426. doi: 10.1093/jnen/61.5.413. [DOI] [PubMed] [Google Scholar]
  24. Diepenbroek M, Casadei N, Esmer H, Saido TC, Takano J, Kahle PJ, Nixon RA, Rao MV, Melki R, Pieri L, Helling S, Marcus K, Krueger R, Masliah E, Riess O, Nuber S. Overexpression of the calpain-specific inhibitor calpastatin reduces human alpha-Synuclein processing, aggregation and synaptic impairment in [A30P]αSyn transgenic mice. Hum Mol Genet. 2014;23:ddu112–3989. doi: 10.1093/hmg/ddu112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Dijkstra AA, Voorn P, Berendse HW, Groenewegen HJ, Netherlands Brain Bank. Rozemuller AJM, van de Berg WDJ. Stage-dependent nigral neuronal loss in incidental Lewy body and Parkinson's disease. Mov Disord. 2014;29:1244–1251. doi: 10.1002/mds.25952. [DOI] [PubMed] [Google Scholar]
  26. Disterhoft JF, Moyer JR, Thompson LT. The calcium rationale in aging and Alzheimer's disease. Evidence from an animal model of normal aging. Ann N Y Acad Sci. 1994;747:382–406. doi: 10.1111/j.1749-6632.1994.tb44424.x. [DOI] [PubMed] [Google Scholar]
  27. Dragicevic E, Schiemann J, Liss B. Dopamine midbrain neurons in health and Parkinson's disease: Emerging roles of voltage-gated calcium channels and ATP-sensitive potassium channels. Neuroscience. 2015;284C:798–814. doi: 10.1016/j.neuroscience.2014.10.037. [DOI] [PubMed] [Google Scholar]
  28. Dryanovski DI, Guzman JN, Xie Z, Galteri DJ, Volpicelli-Daley LA, Lee VM-Y, Miller RJ, Schumacker PT, Surmeier DJ. Calcium Entry and α-Synuclein Inclusions Elevate Dendritic Mitochondrial Oxidant Stress in Dopaminergic Neurons. Journal of Neuroscience. 2013;33:10154–10164. doi: 10.1523/JNEUROSCI.5311-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dufty BM, Warner LR, Hou ST, Jiang SX, Gomez-Isla T, Leenhouts KM, Oxford JT, Feany MB, Masliah E, Rohn TT. Calpain-cleavage of alpha-synuclein: connecting proteolytic processing to disease-linked aggregation. The American Journal of Pathology. 2007;170:1725–1738. doi: 10.2353/ajpath.2007.061232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Elm JJ NINDS NET-PD Investigators. Design innovations and baseline findings in a long-term Parkinson“s trial: the National Institute of Neurological Disorders and Stroke Exploratory Trials in Parkinson”s Disease Long-Term Study-1. Movement Disorders. 2012;27:1513–1521. doi: 10.1002/mds.25175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Fahn S the Parkinson Study Group. Does levodopa slow or hasten the rate of progression of Parkinson’s disease? J Neurol. 2005;252:iv37–iv42. doi: 10.1007/s00415-005-4008-5. [DOI] [PubMed] [Google Scholar]
  32. Fahn S, Oakes D, Shoulson I, Kieburtz K, Rudolph A, Lang A, Olanow CW, Tanner C, Marek K Parkinson Study Group. Levodopa and the progression of Parkinson's disease. N Engl J Med. 2004;351:2498–2508. doi: 10.1056/NEJMoa033447. [DOI] [PubMed] [Google Scholar]
  33. Gegg ME, Schapira AHV. Mitochondrial dysfunction associated with glucocerebrosidase deficiency. Neurobiol Dis. 2016;90:43–50. doi: 10.1016/j.nbd.2015.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of Lewy pathology. Nat Rev Neurol. 2012;9:13–24. doi: 10.1038/nrneurol.2012.242. [DOI] [PubMed] [Google Scholar]
  35. Goldberg JA, Guzman JN, Estep CM, Ilijic E, Kondapalli J, Sánchez-Padilla J, Surmeier DJ. Calcium entry induces mitochondrial oxidant stress in vagal neurons at risk in Parkinson's disease. Nat Neurosci. 2012;15:1414–1421. doi: 10.1038/nn.3209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gómez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev. 2016 doi: 10.1016/j.arr.2016.02.009. [DOI] [PubMed] [Google Scholar]
  37. Guardia-Laguarta C, Area-Gomez E, Schon EA, Przedborski S. A new role for α-synuclein in Parkinson's disease: Alteration of ER-mitochondrial communication. Movement Disorders. 2015;30:1026–1033. doi: 10.1002/mds.26239. [DOI] [PubMed] [Google Scholar]
  38. Gudala K, Kanukula R, Bansal D. Reduced Risk of Parkinson's Disease in Users of Calcium Channel Blockers: A Meta-Analysis. Int J Chronic Dis. 2015:697404–7. doi: 10.1155/2015/697404. 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gupta A, Dawson VL, Dawson TM. What causes cell death in Parkinson's disease? Ann Neurol. 2009;64:S3–S15. doi: 10.1002/ana.21573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Guzman JN, Sanchez-Padilla J, Chan CS, Surmeier DJ. Robust Pacemaking in Substantia Nigra Dopaminergic Neurons. Journal of Neuroscience. 2009;29:11011–11019. doi: 10.1523/JNEUROSCI.2519-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Guzman JN, Sánchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, Surmeier DJ. Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature. 2010;468:696–700. doi: 10.1038/nature09536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Halliday G, McCann H, Shepherd C. Evaluation of the Braak hypothesis: how far can it explain the pathogenesis of Parkinson's disease? Expert Rev Neurother. 2012;12:673–686. doi: 10.1586/ern.12.47. [DOI] [PubMed] [Google Scholar]
  43. Hammond C, Bergman H, Brown P. Pathological synchronization in Parkinson's disease: networks, models and treatments. Trends Neurosci. 2007;30:357–364. doi: 10.1016/j.tins.2007.05.004. [DOI] [PubMed] [Google Scholar]
  44. Hart RG, Pearce LA, Ravina BM, Yaltho TC, Marler JR. Neuroprotection trials in Parkinson's disease: systematic review. Movement Disorders. 2009;24:647–654. doi: 10.1002/mds.22432. [DOI] [PubMed] [Google Scholar]
  45. Helton T, Xu W, Lipscombe D. Neuronal L-type calcium channels open quickly and are inhibited slowly. J Neurosci. 2005;25:10247–10251. doi: 10.1523/JNEUROSCI.1089-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Holford NHG, Nutt JG. Interpreting the results of Parkinson's disease clinical trials: time for a change. Mov Disord. 2011;26:569–577. doi: 10.1002/mds.23555. [DOI] [PubMed] [Google Scholar]
  47. Hornung J-P. The human raphe nuclei and the serotonergic system. J Chem Neuroanat. 2003;26:331–343. doi: 10.1016/j.jchemneu.2003.10.002. [DOI] [PubMed] [Google Scholar]
  48. Hornykiewicz O. Dopamine miracle: from brain homogenate to dopamine replacement. Mov Disord. 2002;17:501–508. doi: 10.1002/mds.10115. [DOI] [PubMed] [Google Scholar]
  49. Hu B, Yang N, Qiao Q-C, Hu Z-A, Zhang J. Roles of the orexin system in central motor control. Neuroscience & Biobehavioral Reviews. 2015;49:43–54. doi: 10.1016/j.neubiorev.2014.12.005. [DOI] [PubMed] [Google Scholar]
  50. Hughes AJ, Ben-Shlomo Y, Daniel SE, Lees AJ. What features improve the accuracy of clinical diagnosis in Parkinson's disease: a clinicopathologic study. Neurology. 1992;42:1142–1146. doi: 10.1212/wnl.42.6.1142. [DOI] [PubMed] [Google Scholar]
  51. Hunn BHM, Cragg SJ, Bolam JP, Spillantini MG, Wade-Martins R. Impaired intracellular trafficking defines early Parkinson's disease. Trends Neurosci. 2015;38:178–188. doi: 10.1016/j.tins.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hurley MJ, Brandon B, Gentleman SM, Dexter DT. Parkinson's disease is associated with altered expression of CaV1 channels and calcium-binding proteins. Brain. 2013;136:2077–2097. doi: 10.1093/brain/awt134. [DOI] [PubMed] [Google Scholar]
  53. Hurley MJ, Gentleman SM, Dexter DT. Calcium CaV1 Channel Subtype mRNA Expression in Parkinson's Disease Examined by In Situ Hybridization. J Mol Neurosci. 2014;55:1–10. doi: 10.1007/s12031-014-0410-8. [DOI] [PubMed] [Google Scholar]
  54. Ilijic E, Guzman JN, Surmeier DJ. The L-type channel antagonist isradipine is neuroprotective in a mouse model of Parkinson's disease. Neurobiol Dis. 2011;43:364–371. doi: 10.1016/j.nbd.2011.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RKB. The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson's disease: a critical analysis of α-synuclein staging. Neuropathol Appl Neurobiol. 2008;34:284–295. doi: 10.1111/j.1365-2990.2007.00923.x. [DOI] [PubMed] [Google Scholar]
  56. Kang Y, Kitai ST. Electrophysiological properties of pedunculopontine neurons and their postsynaptic responses following stimulation of substantia nigra reticulata. Brain Res. 1990;535:79–95. doi: 10.1016/0006-8993(90)91826-3. [DOI] [PubMed] [Google Scholar]
  57. Khaliq ZM, Khaliq ZM, Bean BP, Bean BP. Pacemaking in dopaminergic ventral tegmental area neurons: depolarizing drive from background and voltage-dependent sodium conductances. Journal of Neuroscience. 2010;30:7401–7413. doi: 10.1523/JNEUROSCI.0143-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kingsbury AE, Bandopadhyay R, Silveira-Moriyama L, Ayling H, Kallis C, Sterlacci W, Maeir H, Poewe W, Lees AJ. Brain stem pathology in Parkinson's disease: An evaluation of the Braak staging model. Mov Disord. 2010;25:2508–2515. doi: 10.1002/mds.23305. [DOI] [PubMed] [Google Scholar]
  59. Ko JH, Mure H, Tang CC, Ma Y, Dhawan V, Spetsieris P, Eidelberg D. Parkinson's disease: increased motor network activity in the absence of movement. Journal of Neuroscience. 2013;33:4540–4549. doi: 10.1523/JNEUROSCI.5024-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kordower JH, Olanow CW, Dodiya HB, Chu Y, Beach TG, Adler CH, Halliday GM, Bartus RT. Disease duration and the integrity of the nigrostriatal system in Parkinson's disease. Brain. 2013;136:2419–2431. doi: 10.1093/brain/awt192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy body disease: progressive dementia with abundant cortical Lewy bodies and senile changes of varying degree--a new disease? Clin. Neuropathol. 1984;3:185–192. [PubMed] [Google Scholar]
  62. Kumaran R, Cookson MR. Pathways to Parkinsonism Redux: convergent pathobiological mechanisms in genetics of Parkinson's disease. Hum Mol Genet. 2015;24:R32–44. doi: 10.1093/hmg/ddv236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kupsch A, Sautter J, Schwarz J, Riederer P, Gerlach M, Oertel WH. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in non-human primates is antagonized by pretreatment with nimodipine at the nigral, but not at the striatal level. Brain Res. 1996;741:185–196. doi: 10.1016/s0006-8993(96)00917-1. [DOI] [PubMed] [Google Scholar]
  64. Lee Y-C, Lin C-H, Wu R-M, Lin J-W, Chang C-H, Lai M-S. Antihypertensive agents and risk of Parkinson's disease: a nationwide cohort study. PLoS ONE. 2014;9:e98961. doi: 10.1371/journal.pone.0098961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lin MK, Farrer MJ. Genetics and genomics of Parkinson's disease. Genome Med. 2014;6:48. doi: 10.1186/gm566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Liu AKL, Chang RC-C, Pearce RKB, Gentleman SM. Nucleus basalis of Meynert revisited: anatomy, history and differential involvement in Alzheimer“s and Parkinson”s disease. Acta Neuropathol. 2015;129:527–540. doi: 10.1007/s00401-015-1392-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Llorente-Folch I, Rueda CB, Pardo B, Szabadkai G, Duchen MR, Satrustegui J. The regulation of neuronal mitochondrial metabolism by calcium. J Physiol (Lond) 2015;593:3447–3462. doi: 10.1113/JP270254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Luk KC, Luk KC, Kehm V, Kehm V, Carroll J, Carroll J, Zhang B, Zhang B, O'Brien P, O'Brien P, Trojanowski JQ, Trojanowski JQ, Lee VMY, Lee VM-Y. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338:949–953. doi: 10.1126/science.1227157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Marras C, Gruneir A, Rochon P, Wang X, Anderson G, Brotchie J, Bell CM, Fox S, Austin PC. Dihydropyridine calcium channel blockers and the progression of parkinsonism. Ann Neurol. 2012;71:362–369. doi: 10.1002/ana.22616. [DOI] [PubMed] [Google Scholar]
  70. Martinez-Gonzalez C, Bolam JP, Mena-Segovia J. Topographical organization of the pedunculopontine nucleus. Front Neuroanat. 2011;5:22. doi: 10.3389/fnana.2011.00022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Matsuda W, Furuta T, Nakamura KC, Hioki H, Fujiyama F, Arai R, Kaneko T. Single Nigrostriatal Dopaminergic Neurons Form Widely Spread and Highly Dense Axonal Arborizations in the Neostriatum. Journal of Neuroscience. 2009;29:444–453. doi: 10.1523/JNEUROSCI.4029-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. McCoy MK, Cookson MR. Mitochondrial quality control and dynamics in Parkinson's disease. Antioxid Redox Signal. 2012;16:869–882. doi: 10.1089/ars.2011.4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Medina DL, Ballabio A. Lysosomal calcium regulates autophagy. Autophagy. 2015;11:970–971. doi: 10.1080/15548627.2015.1047130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Miskovic V, Schmidt LA. Social fearfulness in the human brain. Neuroscience & Biobehavioral Reviews. 2012;36:459–478. doi: 10.1016/j.neubiorev.2011.08.002. [DOI] [PubMed] [Google Scholar]
  75. Morikawa H, Paladini CA. Dynamic regulation of midbrain dopamine neuron activity: intrinsic, synaptic, and plasticity mechanisms. Neuroscience. 2011;198:95–111. doi: 10.1016/j.neuroscience.2011.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Mosharov EV, Larsen KE, Kanter E, Phillips KA, Wilson K, Schmitz Y, Krantz DE, Kobayashi K, Edwards RH, Sulzer D. Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons. Neuron. 2009;62:218–229. doi: 10.1016/j.neuron.2009.01.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Mullin S, Schapira A. The genetics of Parkinson's disease. Br. Med. Bull. 2015;114:39–52. doi: 10.1093/bmb/ldv022. [DOI] [PubMed] [Google Scholar]
  78. Mullin S, Schapira A. α-Synuclein and mitochondrial dysfunction in Parkinson's disease. Mol Neurobiol. 2013;47:587–597. doi: 10.1007/s12035-013-8394-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Nagley P, Higgins GC, Atkin JD, Beart PM. Multifaceted deaths orchestrated by mitochondria in neurones. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2010;1802:167–185. doi: 10.1016/j.bbadis.2009.09.004. [DOI] [PubMed] [Google Scholar]
  80. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, Cummings JL, Chertkow H. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53:695–699. doi: 10.1111/j.1532-5415.2005.53221.x. [DOI] [PubMed] [Google Scholar]
  81. Nedergaard S, Flatman JA, Engberg I. Nifedipine- and omega-conotoxin-sensitive Ca2+ conductances in guinea-pig substantia nigra pars compacta neurones. J Physiol (Lond) 1993;466:727–747. [PMC free article] [PubMed] [Google Scholar]
  82. Nicholls DG. Mitochondria in the life and death of neurons. Essays Biochem. 1998 doi: 10.1042/bse0330043. [DOI] [PubMed] [Google Scholar]
  83. Olanow CW, Brundin P. Parkinson“s Disease and Alpha Synuclein: Is Parkinson”s Disease a Prion-Like Disorder? Movement Disorders. 2013;28:31–40. doi: 10.1002/mds.25373. [DOI] [PubMed] [Google Scholar]
  84. Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, Langston W, Melamed E, Poewe W, Stocchi F, Tolosa E. A Double-Blind, Delayed-Start Trial of Rasagiline in Parkinson's Disease. N Engl J Med. 2009;361:1268–1278. doi: 10.1056/NEJMoa0809335. [DOI] [PubMed] [Google Scholar]
  85. Ortner NJ, Bock G, Dougalis A, Kharitonova M, Duda J, Hess S, Tuluc P, Pomberger T, Stefanova N, Pitterl F, Ciossek T, Oberacher H, Draheim HJ, Kloppenburg P, Liss B, Striessnig J. Lower Affinity of Isradipine for L-Type Ca(2+) Channels during Substantia Nigra Dopamine Neuron-Like Activity: Implications for Neuroprotection in Parkinson's Disease. Journal of Neuroscience. 2017;37:6761–6777. doi: 10.1523/JNEUROSCI.2946-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Pacelli C, Giguère N, Bourque M-J, Lévesque M, Slack RS, Trudeau L-E. Elevated Mitochondrial Bioenergetics and Axonal Arborization Size Are Key Contributors to the Vulnerability of Dopamine Neurons. Curr Biol. 2015;25:2349–2360. doi: 10.1016/j.cub.2015.07.050. [DOI] [PubMed] [Google Scholar]
  87. Palmiter RD. Dopamine signaling as a neural correlate of consciousness. Neuroscience. 2011;198:213–220. doi: 10.1016/j.neuroscience.2011.06.089. [DOI] [PubMed] [Google Scholar]
  88. Parashos SA, Swearingen CJ, Biglan KM, Bodis-Wollner I, Liang GS, Ross GW, Tilley BC, Shulman LM NET-PD Investigators. Determinants of the timing of symptomatic treatment in early Parkinson disease: The National Institutes of Health Exploratory Trials in Parkinson Disease (NET-PD) Experience. Arch Neurol. 2009;66:1099–1104. doi: 10.1001/archneurol.2009.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Parkinson Study Group. Phase II safety, tolerability, and dose selection study of isradipine as a potential disease-modifying intervention in early Parkinson's disease (STEADY-PD) Mov Disord. 2013;28:1823–1831. doi: 10.1002/mds.25639. [DOI] [PubMed] [Google Scholar]
  90. Parkinson Study Group. Impact of deprenyl and tocopherol treatment on Parkinson's disease in DATATOP subjects not requiring levodopa. Parkinson Study Group. Ann Neurol. 1996;39:29–36. doi: 10.1002/ana.410390106. [DOI] [PubMed] [Google Scholar]
  91. Parkinson Study Group CALM Cohort Investigators. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol. 2009;66:563–570. doi: 10.1001/archneur.66.1.nct90001. [DOI] [PubMed] [Google Scholar]
  92. Parkinson Study Group PRECEPT Investigators. Mixed lineage kinase inhibitor CEP-1347 fails to delay disability in early Parkinson disease. Neurology. 2007;69:1480–1490. doi: 10.1212/01.wnl.0000277648.63931.c0. [DOI] [PubMed] [Google Scholar]
  93. Pasternak B, Pasternak B, Svanström H, Svanstrom H, Nielsen NM, Nielsen NM, Fugger L, Fugger L, Melbye M, Melbye M, Hviid A, Hviid A. Use of calcium channel blockers and Parkinson's disease. American journal of epidemiology. 2012;175:627–635. doi: 10.1093/aje/kwr362. [DOI] [PubMed] [Google Scholar]
  94. Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, Van den Haute C, Melki R, Baekelandt V. α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522:340–344. doi: 10.1038/nature14547. [DOI] [PubMed] [Google Scholar]
  95. Pfaff DW, Martin EM, Faber D. Origins of arousal: roles for medullary reticular neurons. Trends Neurosci. 2012;35:468–476. doi: 10.1016/j.tins.2012.04.008. [DOI] [PubMed] [Google Scholar]
  96. Poulin J-F, Zou J, Drouin-Ouellet J, Kim K-YA, Cicchetti F, Awatramani RB. Defining midbrain dopaminergic neuron diversity by single-cell gene expression profiling. Cell Rep. 2014;9:930–943. doi: 10.1016/j.celrep.2014.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Puopolo M, Puopolo M, Raviola E, Raviola E, Bean BP, Bean BP. Roles of subthreshold calcium current and sodium current in spontaneous firing of mouse midbrain dopamine neurons. Journal of Neuroscience. 2007;27:645–656. doi: 10.1523/JNEUROSCI.4341-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Putzier I, Kullmann PHM, Horn JP, Levitan ES. Cav1.3 Channel Voltage Dependence, Not Ca2+ Selectivity, Drives Pacemaker Activity and Amplifies Bursts in Nigral Dopamine Neurons. Journal of Neuroscience. 2009a;29:15414–15419. doi: 10.1523/JNEUROSCI.4742-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Putzier I, Kullmann PHM, Horn JP, Levitan ES. Cav1.3 Channel Voltage Dependence, Not Ca2+ Selectivity, Drives Pacemaker Activity and Amplifies Bursts in Nigral Dopamine Neurons. J Neurosci. 2009b;29:15414–15419. doi: 10.1523/JNEUROSCI.4742-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson's disease who were treated with ropinirole or levodopa. 056 Study Group. N Engl J Med. 2000;342:1484–1491. doi: 10.1056/NEJM200005183422004. [DOI] [PubMed] [Google Scholar]
  101. Ratcliffe EM, Farrar NR, Fox EA. Development of the vagal innervation of the gut: steering the wandering nerve. Neurogastroenterol Motil. 2011;23:898–911. doi: 10.1111/j.1365-2982.2011.01764.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Ravina B, Tanner C, Dieuliis D, Eberly S, Flagg E, Galpern WR, Fahn S, Goetz CG, Grate S, Kurlan R, Lang AE, Marek K, Kieburtz K, Oakes D, Elliott R, Shoulson I Parkinson Study Group LABS-PD Investigators. A longitudinal program for biomarker development in Parkinson's disease: a feasibility study. Movement Disorders. 2009;24:2081–2090. doi: 10.1002/mds.22690. [DOI] [PubMed] [Google Scholar]
  103. Rcom-H'cheo-Gauthier A, Goodwin J, Pountney DL. Interactions between calcium and alpha-synuclein in neurodegeneration. Biomolecules. 2014;4:795–811. doi: 10.3390/biom4030795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Reeve A, Simcox E, Turnbull D. Ageing and Parkinson's disease: why is advancing age the biggest risk factor? Ageing Res Rev. 2014;14:19–30. doi: 10.1016/j.arr.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. 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]
  106. Saper CB. THE CENTRAL AUTONOMIC NERVOUS SYSTEM: Conscious Visceral Perception and Autonomic Pattern Generation. 2003;25:433–469. doi: 10.1146/annurev.neuro.25.032502.111311. doi: 10.1146/annurev.neuro.25.032502.111311. [DOI] [PubMed] [Google Scholar]
  107. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep and circadian rhythms. Nature. 2005;437:1257–1263. doi: 10.1038/nature04284. [DOI] [PubMed] [Google Scholar]
  108. Sara SJ, Bouret S. Orienting and reorienting: the locus coeruleus mediates cognition through arousal. Neuron. 2012;76:130–141. doi: 10.1016/j.neuron.2012.09.011. [DOI] [PubMed] [Google Scholar]
  109. Sánchez-Padilla J, Guzman JN, Ilijic E, Kondapalli J, Galtieri DJ, Yang B, Schieber S, Oertel W, Wokosin D, Schumacker PT, Surmeier DJ. Mitochondrial oxidant stress in locus coeruleus is regulated by activity and nitric oxide synthase. Nat Neurosci. 2014;17:832–840. doi: 10.1038/nn.3717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Silvani A, Calandra-Buonaura G, Dampney RAL, Cortelli P. Brain-heart interactions: physiology and clinical implications. Philos Trans A Math Phys Eng Sci. 2016;374:20150181. doi: 10.1098/rsta.2015.0181. [DOI] [PubMed] [Google Scholar]
  111. 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;25:2863–2866. doi: 10.1002/mds.23308. [DOI] [PubMed] [Google Scholar]
  112. 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]
  113. Stocchi F, Vacca L, Berardelli A, De Pandis F, Ruggieri S. Long-duration effect and the postsynaptic compartment: study using a dopamine agonist with a short half-life. Mov Disord. 2001;16:301–305. doi: 10.1002/mds.1070. [DOI] [PubMed] [Google Scholar]
  114. Striessnig J, Grabner M, Mitterdorfer J, Hering S, Sinnegger MJ, Glossmann H. Structural basis of drug binding to L Ca2+ channels. Trends Pharmacol Sci. 1998;19:108–115. doi: 10.1016/s0165-6147(98)01171-7. [DOI] [PubMed] [Google Scholar]
  115. Sulzer D. Multiple hit hypotheses for dopamine neuron loss in Parkinson's disease. Trends Neurosci. 2007;30:244–250. doi: 10.1016/j.tins.2007.03.009. [DOI] [PubMed] [Google Scholar]
  116. Sulzer D, Surmeier DJ. Neuronal vulnerability, pathogenesis, and Parkinson's disease. Mov Disord. 2013;28:41–50. doi: 10.1002/mds.25095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Surmeier DJ, Obeso JA, Halliday GM. Selective neuronal vulnerability in Parkinson disease. Nat Rev Neurosci. 2017;18:101–113. doi: 10.1038/nrn.2016.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Surmeier DJ, Schumacker PT, Guzman JD, Ilijic E, Yang B, Zampese E. Calcium and Parkinson's disease. 2016 doi: 10.1016/j.bbrc.2016.08.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Thibault O, Gant JC, Landfield PW. Expansion of the calcium hypothesis of brain aging and Alzheimer's disease: minding the store. Aging Cell. 2007;6:307–317. doi: 10.1111/j.1474-9726.2007.00295.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Tomlinson CL, Stowe R, Patel S, Rick C, Gray R, Clarke CE. Systematic review of levodopa dose equivalency reporting in Parkinson's disease. Movement Disorders. 2010;25:2649–2653. doi: 10.1002/mds.23429. [DOI] [PubMed] [Google Scholar]
  121. Uchihara T, Giasson BI. Propagation of alpha-synuclein pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol. 2016;131:49–73. doi: 10.1007/s00401-015-1485-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Volpicelli-Daley LA, Luk KC, Lee VM-Y. Addition of exogenous α-synuclein preformed fibrils to primary neuronal cultures to seed recruitment of endogenous α-synuclein to Lewy body and Lewy neurite–like aggregates. Nat Protoc. 2014;9:2135–2146. doi: 10.1038/nprot.2014.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, Meaney DF, Trojanowski JQ, Lee VM-Y. Exogenous α-Synuclein Fibrils Induce Lewy Body Pathology Leading to Synaptic Dysfunction and Neuron Death. Neuron. 2011;72:57–71. doi: 10.1016/j.neuron.2011.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Votyakova TV, Reynolds IJ. DeltaPsi(m)-Dependent and -independent production of reactive oxygen species by rat brain mitochondria. J Neurochem. 2001;79:266–277. doi: 10.1046/j.1471-4159.2001.00548.x. [DOI] [PubMed] [Google Scholar]
  125. Walsh DM, Selkoe DJ. A critical appraisal of the pathogenic protein spread hypothesis of neurodegeneration. Nat Rev Neurosci. 2016;17:251–260. doi: 10.1038/nrn.2016.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci. 2010;13:805–811. doi: 10.1038/nn.2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Zhang J, Berra-Romani R, Sinnegger-Brauns MJ, Striessnig J, Blaustein MP, Matteson DR. Role of Cav1.2 L-type Ca2+ channels in vascular tone: effects of nifedipine and Mg2+ AJP: Heart and Circulatory Physiology. 2007;292:H415–25. doi: 10.1152/ajpheart.01214.2005. [DOI] [PubMed] [Google Scholar]
  128. Zhou F-M, Wilson CJ, Dani JA. Cholinergic interneuron characteristics and nicotinic properties in the striatum. J Neurobiol. 2002;53:590–605. doi: 10.1002/neu.10150. [DOI] [PubMed] [Google Scholar]
  129. Zucca FA, Segura-Aguilar J, Ferrari E, Muñoz P, Paris I, Sulzer D, Sarna T, Casella L, Zecca L. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson's disease. Prog Neurobiol. 2015 doi: 10.1016/j.pneurobio.2015.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES