Elsevier

Biochemical Pharmacology

Volume 74, Issue 2, 15 July 2007, Pages 177-190
Biochemical Pharmacology

Commentary
Norepinephrine: The redheaded stepchild of Parkinson's disease

https://doi.org/10.1016/j.bcp.2007.01.036Get rights and content

Abstract

Parkinson's disease (PD) affects approximately 1% of the world's aging population. Despite its prevalence and rigorous research in both humans and animal models, the etiology remains unknown. PD is most often characterized by the degeneration of dopamine (DA) neurons in the substantia nigra pars compacta (SNc), and models of PD generally attempt to mimic this deficit. However, PD is a true multisystem disorder marked by a profound but less appreciated loss of cells in the locus coeruleus (LC), which contains the major group of noradrenergic neurons in the brain. Historic and more recent experiments exploring the role of norepinephrine (NE) in PD will be analyzed in this review. First, we examine the evidence that NE is neuroprotective and that LC degeneration sensitizes DA neurons to damage. The second part of this review focuses on the potential contribution of NE loss to the behavioral symptoms associated with PD. We propose that LC loss represents a crucial turning point in PD progression and that pharmacotherapies aimed at restoring NE have important therapeutic potential.

Introduction

Parkinson's disease (PD) is one of the most common diseases among the aging and affects approximately 1% of the population worldwide [1]. Although PD is characterized by the degeneration of nigrostriatal dopamine (DA) neurons, PD pathology also involves the serotinergic and cholinergic systems as well as a profound loss of neurons from the locus coeruleus (LC), the major noradrenergic nucleus in the brain [2], [3], [4], [5]. Other brain regions may be relevant to pathology and treatment of PD, this review will focus on the relationship between the dopaminergic and noradrenergic systems in PD. The cardinal symptoms of PD include resting tremor, bradykinesia, rigidity, and postural instability. These motor impairments do not become apparent until ∼80% of the DA terminals have been lost, which suggests the existence of an impressive compensatory mechanism in the earlier stages of disease. Several independent investigators have found that experimental lesions of the LC exacerbate PD pathology and behavioral symptomology in animal models, indicating that NE may play a key role in this compensation [6], [7], [8]. Postmortem examinations have shown that the LC in PD patients almost always demonstrates either cell loss or Lewy body inclusions, and most often both [9]. In fact, degeneration of the LC may precede and ultimately even surpass DA neuron degeneration in the substantia nigra par compacta (SNc) [2], [10], [11], [12], [13], [14]. NE has profound effects on brain inflammation, oxidative stress, and the function of other proteins implicated in PD, and in general confers potent neuroprotective properties. The first part of this review weighs the evidence that LC loss contributes directly to the demise of DA neurons in PD.

Apart from its importance for the survival of DA neurons, NE could also play an independent role in the behavioral symptoms of PD. Despite the popularity and utility of animal models of PD, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced DA neuron degeneration, it has proved difficult to simultaneously recapitulate the neuropathological and behavioral symptoms of PD, especially in mice. One limitation of these techniques is that they tend to model PD as a pure DA deficit, rather than as a multisystem disorder involving both the NE and DA systems. However, when researchers have lesioned both DA and NE neurons, the behavioral manifestations reminiscent of PD become more apparent, suggesting that NE loss exacerbates the motor deficits of PD. In addition to the spectrum of movement abnormalities, many PD patients experience neuropsychiatric symptoms, such as depression and sleep disturbances, which may be related to LC degeneration [15]. For example, PD patients with comorbid depression tend to exhibit more pronounced PD symptoms, and their depression can be alleviated by the selective NE transporter (NET) blocker reboxetine [16], [17]. Finally, the death of NE neurons may modulate the plastic changes and behavioral pathology associated with long-term l-dihydroxyphenylalanine (l-DOPA) therapy. Although DA replacement with l-DOPA remains one of the most effective treatments for PD, long-term treatment results in the development of dyskinesias (unwanted choreic involuntary movements), which complicate rather than alleviate the PD symptoms [18]. Human and animal studies have shown that α2-adrenoreceptor (α2AR) antagonists provide relief from l-DOPA-induced dyskinesias (LID) [19], [20]. While the mechanism of LID and the basis of the relief provided by blockade of α2ARs remains unknown, the data imply that NE continues to function in modulating the plasticity and activity of the basal ganglia during the progression of PD with l-DOPA treatment. The second part of this review will focus on the role of NE in the motor symptoms of PD.

To elucidate the way NE could modulate DA system function and survival directly and play a role in the progression and expression of PD, it is critical to summarize the intimate molecular, functional, and anatomical relationships between NE and DA. First, NE and DA share a biosynthetic pathway, and DA is in fact the direct precursor of NE [21]. In DA neurons, tyrosine is converted to l-DOPA by tyrosine hydroxylase (TH), which is subsequently converted to DA by aromatic acid decarboxylase (AADC), whereupon the DA is transported into synaptic vesicles by the vesicular monoamine transporter (VMAT). In NE neurons dopamine β-hydroxylase (DBH) acts within the synaptic vesicles to convert DA to NE. Second, noradrenergic neurons directly innervate midbrain DA neurons and the striatum. Stimulation of the LC facilitates burst firing of SNc neurons, while administration of the α1AR antagonist prazosin attenuates firing [22]. DBH, NE, and NET can be detected in the midbrain and striatum [23], [24], [25], [26] and, as in other regions of the brain, striatal NE release is controlled by both NET and α2AR autoreceptors [27], [28], [29]. Finally, either lesions of the LC or chronic NE depletion decrease striatal DA release [30], [31] and result in the compensatory upregulation of striatal D2 receptors [32], [33]. Combined, these results establish a functional connection between the noradrenergic and dopaminergic systems, which may form the basis for the influence of NE on PD neuropathology and motor deficits.

Section snippets

The neuroprotective properties of NE in PD

The first hints that NE promotes DA neuron survival came from MPTP studies in nonhuman primates and mice. Both Mavridis et al. [7] and Fornai et al. [34] noticed that the MPTP-induced damage to nigrostriatal DA neurons was potentiated by pretreatment with DSP4, which is a selective LC neurotoxin. Later, Kilbourn and colleagues found that the tottering mouse, which has noradrenergic hyperinnervation and increased levels of NE throughout the forebrain, was protected from MPTP toxicity [35]. We

The role of NE in motor function in PD

Animal models of PD have been established in an attempt to recapitulate the salient features of human PD. MPTP treatment is considered the gold-standard PD model, because of its abilities to faithfully produce SN neuronal death and motor impairments in nonhuman primates and to cause PD in humans [90], [91]. Although MPTP used in mouse models has successfully created the characteristic nigrostriatal loss seen in PD, its ability to induce PD-like behavioral symptoms has been inconsistent, at best

Future directions

Understanding the physiological interactions between NE and DA pathology in PD could lead to novel co-therapies with the potential to preserve and protect nigrostriatal DA neurons, improve l-DOPA efficacy, and prevent LID. Many issues will first need to be addressed in order to move forward with testing noradrenergic drugs as potential PD treatments.

Summary

There is no known cure for PD, and understanding the cause and progression of the neurodegenerative process is as challenging as it is necessary. Historically, DA has been the focus of most PD research, which led to the current first-line treatment for PD, l-DOPA. However, l-DOPA treatment does not slow the degeneration of DA neurons, and its benefits are temporary, because of an eventual loss of efficacy and the development of LID. In addition, l-DOPA does not reverse some motor symptoms such

References (134)

  • A. Gobert et al.

    Quantification and pharmacological characterization of dialysate levels of noradrenaline in the striatum of freely-moving rats: release from adrenergic terminals and modulation by alpha2-autoreceptors

    J Neurosci Meth

    (2004)
  • L. Yavich et al.

    Alpha2-adrenergic control of dopamine overflow and metabolism in mouse striatum

    Eur J Pharmacol

    (1997)
  • L. Yavich et al.

    Atipamezole, an alpha2-adrenoceptor antagonist, augments the effects of l-DOPA on evoked dopamine release in rat striatum

    Eur J Pharmacol

    (2003)
  • A.J. Lategan et al.

    Effects of locus coeruleus lesions on the release of endogenous dopamine in the rat nucleus accumbens and caudate nucleus as determined by intracerebral microdialysis.

    Brain Res

    (1990)
  • A.J. Lategan et al.

    Suppression of nigrostriatal and mesolimbic dopamine release in vivo following noradrenaline depletion by DSP-4: a microdialysis study

    Life Sci

    (1992)
  • J. Harro et al.

    Effect of denervation of the locus coeruleus projections by DSP-4 treatment on [3H]-raclopride binding to dopamine D(2) receptors and D(2) receptor-G protein interaction in the rat striatum

    Brain Res

    (2003)
  • J. Lahdesmaki et al.

    Behavioral and neurochemical characterization of alpha(2A)-adrenergic receptor knockout mice

    Neuroscience

    (2002)
  • E. MacDonald et al.

    Gene targeting – homing in on alpha 2-adrenoceptor-subtype function

    Trends Pharmacol Sci

    (1997)
  • R. Cash et al.

    Adrenergic receptors in Parkinson's disease

    Brain Res

    (1984)
  • J.S. Schneider et al.

    Volume transmission of dopamine over large distances may contribute to recovery from experimental parkinsonism

    Brain Res

    (1994)
  • D.L. Feinstein et al.

    Noradrenergic regulation of inflammatory gene expression in brain

    Neurochem Int

    (2002)
  • S. Fahn et al.

    Neurodegeneration and neuroprotection in Parkinson disease

    NeuroRx

    (2004)
  • H. Akiyama et al.

    Inflammation and Alzheimer's disease

    Neurobiol Aging

    (2000)
  • J.Y. Chang et al.

    Catecholamines inhibit microglial nitric oxide production.

    Brain Res Bull

    (2000)
  • K. Mori et al.

    Effects of norepinephrine on rat cultured microglial cells that express alpha1, alpha2, beta1 and beta2 adrenergic receptors

    Neuropharmacology

    (2002)
  • C.P. Zabetian et al.

    A quantitative-trait analysis of human plasma-dopamine beta-hydroxylase activity: evidence for a major functional polymorphism at the DBH locus

    Am J Hum Genet

    (2001)
  • J. Liu et al.

    Monoamine metabolism provides an antioxidant defense in the brain against oxidant- and free radical-induced damage

    Arch Biochem Biophys

    (1993)
  • D. Weinshenker et al.

    Regulation of norepinephrine transporter abundance by catecholamines and desipramine in vivo.

    Brain Res

    (2002)
  • M.R. Issidorides et al.

    Noradrenaline storage function of species-specific protein bodies, markers of monoamine neurons in human locus coeruleus demonstrated by dopamine-beta-hydroxylase immunogold localization

    Prog Neuropsychopharmacol Biol Psychiatry

    (2004)
  • E. Castren et al.

    Brain-derived neurotrophic factor messenger RNA is expressed in the septum, hypothalamus and in adrenergic brain stem nuclei of adult rat brain and is increased by osmotic stimulation in the paraventricular nucleus

    Neuroscience

    (1995)
  • M. Sun et al.

    Comparison of the capability of GDNF, BDNF, or both, to protect nigrostriatal neurons in a rat model of Parkinson's disease

    Brain Res

    (2005)
  • V.M. Lee et al.

    Mechanisms of Parkinson's disease linked to pathological alpha-synuclein: new targets for drug discovery

    Neuron

    (2006)
  • N.A. Seniuk et al.

    Dose-dependent destruction of the coeruleus-cortical and nigral-striatal projections by MPTP

    Brain Res

    (1990)
  • J.L. Tillerson et al.

    Detection of behavioral impairments correlated to neurochemical deficits in mice treated with moderate doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

    Exp Neurol

    (2002)
  • D.C. German et al.

    Pharmacological inactivation of the vesicular monoamine transporter can enhance 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurodegeneration of midbrain dopaminergic neurons, but not locus coeruleus noradrenergic neurons

    Neuroscience

    (2000)
  • E. Rousselet et al.

    Behavioral changes are not directly related to striatal monoamine levels, number of nigral neurons, or dose of parkinsonian toxin MPTP in mice

    Neurobiol Dis

    (2003)
  • D.C. German et al.

    Disease-specific patterns of locus coeruleus cell loss

    Ann Neurol

    (1992)
  • D.M. Mann et al.

    Pathological basis for neurotransmitter changes in Parkinson's disease

    Neuropathol Appl Neurobiol

    (1983)
  • A.H. Schapira et al.

    Novel pharmacological targets for the treatment of Parkinson's disease

    Nat Rev Drug Discov

    (2006)
  • J. Srinivasan et al.

    Potentiation of parkinsonian symptoms by depletion of locus coeruleus noradrenaline in 6-hydroxydopamine-induced partial degeneration of substantia nigra in rats

    Eur J Neurosci

    (2003)
  • L.S. Forno et al.

    Similarities and differences between MPTP-induced parkinsonsim and Parkinson's disease. Neuropathologic considerations

    Adv Neurol

    (1993)
  • V. Chan-Palay

    Locus coeruleus and norepinephrine in Parkinson's disease

    Jpn J Psychiatry Neurol

    (1991)
  • K. Del Tredici et al.

    Where does parkinson disease pathology begin in the brain?

    J Neuropathol Exp Neurol

    (2002)
  • S. Patt et al.

    A Golgi study of human locus coeruleus in normal brains and in Parkinson's disease

    Neuropathol Appl Neurobiol

    (1993)
  • C. Zarow et al.

    Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases

    Arch Neurol

    (2003)
  • O. Rascol et al.

    Idazoxan, an alpha-2 antagonist, and l-DOPA-induced dyskinesias in patients with Parkinson's disease

    Mov Disord

    (2001)
  • T. Archer et al.

    An antihypokinesic action of alpha2-adrenoceptors upon MPTP-induced behaviour deficits in mice

    J Neural Transm

    (2003)
  • R. Grondin et al.

    Noradrenoceptor antagonism with idazoxan improves l-DOPA-induced dyskinesias in MPTP monkeys

    Naunyn Schmiedebergs Arch Pharmacol

    (2000)
  • P.B. Molinoff et al.

    Biochemistry of catecholamines

    Annu Rev Biochem

    (1971)
  • S. Udenfriend et al.

    Localization of dopamine-beta-oxidase in brain

    J Neurochem

    (1959)
  • Cited by (0)

    View full text