Elsevier

Neurobiology of Aging

Volume 29, Issue 3, March 2008, Pages 379-396
Neurobiology of Aging

Arrestins and two receptor kinases are upregulated in Parkinson's disease with dementia

https://doi.org/10.1016/j.neurobiolaging.2006.10.012Get rights and content

Abstract

Arrestins and G proteins-coupled receptor kinases (GRKs) regulate signaling and trafficking of G protein-coupled receptors. We investigated changes in the expression of arrestins and GRKs in the striatum of patients with Parkinson's disease without (PD) or with dementia (PDD) at postmortem using Western blotting and ribonuclease protection assay. Both PD and PDD groups had similar degree of dopamine depletion in all striatal regions. Arrestin proteins and mRNAs were increased in the PDD group throughout striatum. Protein and mRNA of GRK5, the major subtype in the human striatum, and GRK3 were also upregulated, whereas GRK2 and 6 were mostly unchanged. The PD group had lower concentration of arrestins and GRKs than the PDD group. There was no statistical link between the load of Alzheimer's pathology and the expression of these signaling proteins. Upregulation of arrestins and GRK in PDD may confer resistance to the therapeutic effects of levodopa often observed in these patients. In addition, increased arrestin and GRK concentrations may lead to dementia via perturbation of multiple signaling mechanisms.

Introduction

Parkinson's disease (PD) is an age-related neurodegenerative disorder caused by degeneration of dopaminergic neurons of the substantia nigra that provide dopamine (DA) to the brain, resulting in the depletion of DA in the striatum, the main recipient of dopaminergic input from the substantia nigra. Neurobiological mechanisms responsible for motor symptoms produced by the degeneration of dopaminergic neurons in PD are poorly understood. It is generally believed that dysregulation of DA receptor signaling in the basal ganglia plays a role in generating motor deficits (Guigoni et al., 2005, Joyce et al., 2001, Muriel et al., 1999, Ryoo et al., 1998), but the underlying molecular events are not known.

DA receptors belong to the G protein-coupled receptor (GPCR) superfamily. Upon persistent stimulation many GPCRs undergo desensitization via a two-step process: activation-dependent receptor phosphorylation by G protein-coupled receptor kinases (GRK) followed by the binding of arrestins that precludes further signaling via G proteins by shielding the cytoplasmic surface of receptors (Krupnick and Benovic, 1998). Arrestins promote receptor internalization via interaction with the internalization machinery of coated pits (reviewed in Gurevich and Gurevich, 2003, Gurevich and Gurevich, 2006 and Lefkowitz and Whalen (2004)). Internalized receptor can either be recycled back to the plasma membrane (resensitization) or degraded (receptor down-regulation). In addition to being key players in the GPCR desensitization and trafficking, arrestins are multifunctional signaling molecules translating GPCR activation into modulation of G protein-independent signaling pathways (Beaulieu et al., 2004, Beaulieu et al., 2005, Lefkowitz and Shenoy, 2005) and regulating the expression of specific genes (Kang et al., 2005). Two arrestin subtypes, arrestin2 and arrestin3, are ubiquitous (Attramadal et al., 1992, Bezard et al., 2005, Gurevich et al., 2002, Gurevich et al., 2004). Five out of seven GRK subtypes, GRK2, 3, 4, 5, and 6, are widely expressed in the brain with largely overlapping cellular distribution (Arriza et al., 1992, Benovic and Gomez, 1993, Bezard et al., 2005, Gurevich et al., 2004, Premont et al., 1994). In vitro data demonstrate that most GPCR subtypes can be phosphorylated by several GRKs (Ménard et al., 1996, Richardson and Hosey, 1993) and bind both arrestins equally well (Gurevich et al., 1995, Krupnick and Benovic, 1998, Richardson and Hosey, 1993). Conversely, in vivo studies suggest that receptors may be preferentially phosphorylated by specific GRKs (Gainetdinov et al., 1999, Iaccarino et al., 1998a, Iaccarino et al., 1998b, Koch et al., 1995, Rockman et al., 1996) and interact with specific arrestins (Kohout et al., 2001, Oakley et al., 2000). Importantly, GPCRs phosphorylated by different GRKs may signal differently (Kim et al., 2005, Ren et al., 2005). Thus, signaling via GPCRs may be differentially regulated by the changes in the cellular complement of arrestins and GRKs. Dysregulation of DA receptors following dopaminergic degeneration in PD may be linked to defects in their desensitization, trafficking, and signaling brought about by the alterations in the expression of specific arrestins and/or GRKs.

Approximately 30–40% of PD patients develop dementia as the disease progresses (Emre, 2003 and references therein). Neuropathological basis of dementia in the idiopathic PD remains controversial. Many PD patients have substantial load of Alzheimer's histopathological features (Boller et al., 1980, Braak et al., 1996, Jellinger, 1997, Jellinger et al., 2002). There is evidence that dementia in PD is associated more closely with cortical deposition of the hallmark component of Lewy bodies, α-synuclein, than with Alzheimer's pathology (Aarsland et al., 2005, Hurtig et al., 2000, Mattila et al., 2000), although this conclusion is disputed by some authors (Parkkinen et al., 2005). A strong correlation between frequency of plaques and neurofibrillary tangles with neocortical Lewy bodies has been reported (Apaydin et al., 2002), suggesting common origin and/or cooperativity between the two types of pathology. Although dementia in idiopathic PD is very important for the disease's prognosis and management (e Lau et al., 2005, Jellinger et al., 2002, Poewe, 2005), underlying neurochemical mechanisms remain largely unexplored. Dementia associated with PD is resistant to L-DOPA treatment (Poewe, 2005). Importantly, dementia in PD strongly correlates with declining antiparkinsonian response to dopaminergic medication including direct DA agonists (Apaydin et al., 2002, Bonelli et al., 2004, Joyce et al., 2002), which may reflect specific changes in DA receptor signaling that hamper their responsiveness. It is conceivable that specific alterations in mechanisms governing signaling via DA receptors and/or other GPCRs brought about by concomitant dementia pathology may be responsible. The GPCR regulation machinery is positioned at the intersection of important signaling pathways and is shared by many GPCRs. Therefore, changes in availability and/or cellular complement of arrestins and/or GRKs induced by the activity of specific GPCRs may affect multiple GPCRs and signaling pathways, thereby propagating the pathology.

Here we report profound changes in the concentrations of arrestins and GRKs at postmortem in the basal ganglia of patients with PD and dementia as opposed to control and patients with PD only. Arrestin binding to GRK-phosphorylated GPCRs initiates second round of signaling via multiple mechanisms. Therefore, we investigated changes in expression of other signaling molecules that may be affected by the modifications in arrestin/GRK expression.

Section snippets

Postmortem samples

Human brain tissue from 16 control and 21 PD patients was used for these studies. Postmortem brains were obtained via Sun Health Research Institute Brain Donor Program in Sun City, AZ. This is a community-based program located in a retirement community with high incidence of PD. The staff of the Brain Donor Program keeps in contact with people signed up for the Program and is regularly informed by donors or family members about changes in the potential donor's health and family situation. Most

Cohort characterization

We have analyzed the expression levels of arrestins and GRKs at postmortem in the basal ganglia of control cases as compared to patients with Parkinson's disease. The cases of PD with dementia were analyzed separately from cases with PD only (designated as PDD group). We have grouped the cases based on clinical diagnosis rather than on any neuropathological feature, for two reasons: first, we were interested in the neurochemical mechanisms of dementia in PD; second, neuropathological substrate

Upregulation of arrestin and GRK expression in Parkinson's disease with dementia

Our data indicate that a subgroup of patients with PD that had been clinically diagnosed with dementia has higher levels of the two non-visual arrestins and two of GRK subtypes, GRK3, and GRK5, in the striatum when compared with control and PD patients without dementia. Within the striatum, upregulation of both mRNA and protein levels for the arrestins was the most pronounced in CN and VSTR. Increase in the arrestins’ and GRKs’ protein expression was accompanied by elevation in their mRNA

Acknowledgements

We are grateful to the Sun Health Research Institute Brain Donation Program of Sun City, Arizona (Dr. Thomas Beach, Director) for the provision of human brain material. The Brain Donation Program is supported by the National Institute on Aging (P30 AG19610), Arizona Alzheimer's Disease Core Center, the Arizona Department of Health Services (contract 211002, Arizona Alzheimer's Research Center) and the Arizona Biomedical Research Commission (contract 0011, Arizona Parkinson's Disease Center). We

References (103)

  • R.R. Gainetdinov et al.

    Muscarinic supersensitivity and impaired receptor desensitization in G protein-coupled receptor kinase 5-deficient mice

    Neuron

    (1999)
  • R.R. Gainetdinov et al.

    Dopaminergic supersensitivity in G protein-coupled receptor kinase 6-deficient mice

    Neuron

    (2003)
  • M. Grange-Midroit et al.

    Regulation of GRK 2 and 6, beta-arrestin-2 and associated proteins in the prefrontal cortex of drug-free and antidepressant drug-treated subjects with major depression

    Brain Res. Mol. Brain Res.

    (2003)
  • C. Guigoni et al.

    Pathogenesis of levodopa-induced dyskinesia: focus on D1 and D3 dopamine receptors

    Parkinsonism Relat. Disord.

    (2005)
  • E.V. Gurevich et al.

    Arrestin2 and arrestin3 are differentially expressed in the rat brain during postnatal development

    Neuroscience

    (2002)
  • V.V. Gurevich et al.

    Arrestin interaction with G protein-coupled receptors. Direct binding studies of wild type and mutant arrestins with rhodopsin, b2-adrenergic, and m2 muscarinic cholinergic receptors

    J. Biol. Chem.

    (1995)
  • V.V. Gurevich et al.

    The new face of active receptor bound arrestin attracts new partners

    Structure

    (2003)
  • V.V. Gurevich et al.

    Structural basis of arrestin regulation of G protein-coupled receptors

    Pharmacol. Ther.

    (2006)
  • S.M. Hanson et al.

    Visual arrestin binding to microtubules involves a distinct conformational change

    J. Biol. Chem.

    (2006)
  • K. Horie et al.

    Retrovirally mediated transfer of a G protein-coupled receptor kinase (GRK) dominant-negative mutant enhances endogenous calcitonin receptor signaling in chinese hamster ovary cells. GRK inhibition enhances expression of receptors and receptor mRNA

    J. Biol. Chem.

    (2000)
  • J.N. Joyce et al.

    Ventral striatal D(3) receptors and Parkinson's Disease

    Parkinsonism Relat. Disord.

    (2001)
  • J.N. Joyce et al.

    Loss of response to levodopa in Parkinson's disease and co-occurrence with dementia: role of D3 and not D2 receptors

    Brain Res.

    (2002)
  • J. Kang et al.

    A nuclear function of beta-arrestin1 in GPCR signaling: regulation of histone acetylation and gene transcription

    Cell

    (2005)
  • K.M. Kim et al.

    Differential regulation of the dopamine D2 and D3 receptors by G protein-coupled receptor kinases and beta-arrestins

    J. Biol. Chem.

    (2001)
  • P. Kunapuli et al.

    Expression, purification, and characterization of the G protein-coupled receptor kinase GRK5

    J. Biol. Chem.

    (1994)
  • R.J. Lefkowitz et al.

    Beta-arrestins: traffic cops of cell signaling

    Curr. Opin. Cell Biol.

    (2004)
  • R.P. Loudon et al.

    Expression, purification, and characterization of the G protein-coupled receptor kinase GRK6

    J. Biol. Chem.

    (1994)
  • A. Miralles et al.

    Acute treatment with the cyclic antidepressant desipramine, but not fluoxetine, increases membrane-associated G protein-coupled receptor kinases 2/3 in rat brain

    Neuropharmacology

    (2002)
  • K.S. Nair et al.

    Light-dependent redistribution of arrestin in vertebrate rods is an energy-independent process governed by protein-protein interactions

    Neuron

    (2005)
  • R.H. Oakley et al.

    Differential affinities of visual arrestin, barrestin1, and barrestin2 for G protein-coupled receptors delineate two major classes of receptors

    J. Biol. Chem.

    (2000)
  • M.J. Orsini et al.

    Characterization of dominant negative arrestins that inhibit beta-2-adrenergic receptor internalization by distinct mechanisms

    J. Biol. Chem.

    (1998)
  • L. Pan et al.

    The nature of the arrestin x receptor complex determines the ultimate fate of the internalized receptor

    J. Biol. Chem.

    (2003)
  • G. Parruti et al.

    Molecular analysis of human beta-arrestin-1: cloning, tissue distribution, and regulation of expression, Identification of two isoforms generated by alternative splicing

    J. Biol. Chem.

    (1993)
  • P. Penela et al.

    Mechanisms of regulation of the expression and function of G protein-coupled receptor kinases

    Cell Signal

    (2003)
  • R.T. Premont et al.

    Identification, purification, and characterization of GRK5, a member of the family of G protein-coupled receptor kinases

    J. Biol. Chem.

    (1994)
  • A.N. Pronin et al.

    Synucleins are a novel class of substrates for G protein-coupled receptor kinases

    J. Biol. Chem.

    (2000)
  • M. Sallese et al.

    G protein-coupled receptor kinase GRK4. Molecular analysis of the four isoforms and ultrastructural localization in spermatozoa and germinal cells

    J. Biol. Chem.

    (1997)
  • R. Sterne-Marr et al.

    Polypeptide variants of beta-arrestin and arrestin3

    J. Biol. Chem.

    (1993)
  • A. Tohgo et al.

    Beta-Arrestin scaffolding of the ERK cascade enhances cytosolic ERK activity but inhibits ERK-mediated transcription following angiotensin AT1a receptor stimulation

    J. Biol. Chem.

    (2002)
  • D. Aarsland et al.

    Neuropathology of dementia in Parkinson's disease: a prospective, community-based study

    Ann. Neurol.

    (2005)
  • S. Ahn et al.

    Desensitization, internalization, and signaling functions of beta-arrestins demonstrated by RNA interference

    Proc. Natl. Acad Sci. U.S.A.

    (2003)
  • H. Apaydin et al.

    Parkinson disease neuropathology: later-developing dementia and loss of the levodopa response

    Arch. Neurol.

    (2002)
  • J.L. Arriza et al.

    The G-protein-coupled receptor kinases bARK1 and bARK2 are widely distributed at synapses in rat brain

    J. Neurosci.

    (1992)
  • T. Bawa et al.

    Desensitization of alpha 2A-adrenoceptor signaling by modest levels of adrenaline is facilitated by beta 2-adrenoceptor-dependent GRK3 up-regulation

    Br. J. Pharmacol.

    (2003)
  • J.M. Beaulieu et al.

    Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • L.M. Bohn et al.

    Enhanced morphine analgesia in mice lacking beta-arrestin2

    Science

    (1999)
  • F. Boller et al.

    Parkinson disease, dementia, and Alzheimer disease: clinicopathological correlations

    Ann. Neurol.

    (1980)
  • S.B. Bonelli et al.

    L-dopa responsiveness in dementia with Lewy bodies, Parkinson disease with and without dementia

    Neurology

    (2004)
  • H. Braak et al.

    New aspects of pathology in Parkinson's disease with concomitant incipient Alzheimer's disease

    J. Neural. Transm.

    (1996)
  • H. Braak et al.

    Staging of Alzheimer-related cortical destruction

    Int. Psychogeriatr.

    (1997)
  • Cited by (58)

    • Targeting GRK2 and GRK5 for treating chronic degenerative diseases: Advances and future perspectives

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Several studies have demonstrated that GRK2 and GRK5 are essential for regulating cardiac functions [11–14], with the growing consensus on them being critical targets for the treatment of cardiovascular diseases. Extensive studies on the role of GRK2 and GRK5 in neurodegenerative diseases revealed that the increased levels of GRK2 and GRK5 are associated with Alzheimer's disease and Parkinson's disease and that both GRK2 and GRK5 affect the accumulation of beta-amyloid (Aβ) [15–18]. In addition, GRK2 and GRK5 have been shown to regulate tumor growth and progression in specific cancers [19–23].

    • G protein-coupled receptor kinases as regulators of dopamine receptor functions

      2016, Pharmacological Research
      Citation Excerpt :

      Four of these, including GRK2 and GRK3 belonging to the GRK2 subfamily (Fig. 1C), and GRK5, and GRK6 belonging to the GRK4 subfamily, are generally ubiquitous, and these proteins are responsible for regulating hundreds of known GPCR subtypes [49]. The striatal neurons express GRKs 2, 3, 5, and 6, albeit the expression levels and the cellular and subcellular distribution of these GRK isoforms vary substantially (Fig. 1A) [50⿿54]. It has been reported that phosphorylation of a receptor by different GRKs triggers differential downstream events leading to the receptor internalization or initiation of arrestin-mediated signaling [47,48].

    View all citing articles on Scopus
    View full text