Elsevier

Brain Research

Volume 893, Issues 1–2, 2 March 2001, Pages 121-134
Brain Research

Research report
μ-, δ- and κ-opioid receptor populations are differentially altered in distinct areas of postmortem brains of Alzheimer’s disease patients

https://doi.org/10.1016/S0006-8993(00)03302-3Get rights and content

Abstract

The putative role of the opioid system in cognitive and memory functions prompted us to search for possible changes in the cohort of the major opioid receptors, μ, δ and κ, in Alzheimer’s disease. The present study examines alterations in opioid receptor levels by quantitative autoradiography. These experiments were carried out on coronal sections of postmortem brains from Alzheimer’s disease patients and from aged-matched, dementia-free individuals. Brain sections were labeled with the tritiated forms of μ-, δ- and κ-opioid ligands; DAMGO ([d-Ala2,N-Me-Phe4,Gly-ol5]-enkephalin), DPDPE ([d-Pen2,5]-enkephalin) and bremazocine (in the presence of μ- and δ-ligands), respectively. Nonspecific binding was determined in the presence of naloxone (10 μM). Brain areas analyzed were caudate, putamen, amygdaloid complex, hippocampal formation and various cerebral and cerebellar cortices. Image analyses of autoradiographs show, that in comparison to the same areas in control brain, statistically significant reductions in μ-opioid receptor binding occur in the subiculum and hippocampus of Alzheimer’s disease brains. Binding of δ-opioid receptors is also decreased in the amygdaloid complex and ventral putamen of Alzheimer’s disease brains. In contrast, large increases of κ-opioid receptor binding are found in the dorsal and ventral putamen as well as in the cerebellar cortex of Alzheimer’s disease brains. Levels of μ-, δ- and κ-opioid receptor binding are unaltered in the caudate, parahippocampal gyrus and occipito–temporal gyrus. These results may suggest an involvement of the endogenous opioid system in some of the multitude of effects that accompany this dementia.

Introduction

Alzheimer’s disease (AD) is the most common cause of dementia in the geriatric population and the prevalence of this progressive brain disease almost doubles every 5 years after the age of 65, reaching 24% of people over the age of 85 years [31]. Common symptoms of AD include memory and cognitive deficits such as language and judgment impairments, difficulties with complex motor tasks, impaired interpretation of sensory stimuli, hallucination and delusion. The complex processes controlling these physiological functions reside in numerous brain regions and are associated with several neurotransmitter systems that are most likely implicated in this dementia [3], [47], [50].

A unique pattern of distribution of the individual types of opioid receptors, μ, δ, and κ, exists in the human brain [4], [18], [37], [38], [45], [46], [48], [49], [60]. While these receptors are widely distributed in gray matter, they are preferentially found in limbic and limbic-associated brain structures. These receptors play important roles in a broad range of functions and behaviors, such as nociception, pleasure and dysphoria, neuroendocrine regulation, motor control, learning and memory [33], [38], [62]. Despite a large spectrum of affected physiological actions, only a few studies have specifically examined the endogenous opioid system in postmortem human brain and its possible alteration in pathological conditions such as suicide [13], [16], [23], addiction [12], [14], [56], Parkinson’s disease [11], [52], [68], schizophrenia [53], [59] and Alzheimer’s disease [2], [15], [18], [25], [29], [30], [54].

A possible linkage between AD and the endogenous opioid system is supported by recognition of the role of the opioid system in cognitive and memory functions. In animal models showing deficits in these functions, the integrity of the septohippocampal cholinergic system is a major factor. Thus, in rodents, the acute administration of κ-opioid receptor agonists [dynorphin A-(1-13), U50488H, U69,593] was apparently beneficial in: decreasing the degree of impairment of learning and memory processes after lesioning of cholinergic basal forebrain neurons [64], reducing cholinergic drug-induced deficits [21], [22], ameliorating other models of memory dysfunctions [20], [27], [28], [41], [65] and in the enhancement of spontaneous alternation memory in Y-maze testing [66]. Under some conditions, μ- and δ-opioid receptor ligands may have effects opposite to those of κ-opioid agonists, but they are also fully involved in memory processes in rodents [26], [28], [43]. In nonhuman primates, facilitation of learning was found after low doses of an enkephalin analog [42]. In humans, results of studies on the effects of selective opioid or opiate agonists on memory function are still inconclusive [70]. In studies of the effects of naloxone administration to AD patients, initial reports of improvement of cognitive deficit [51] were not supported by later findings [17], [44], [63].

One of the characteristics of the AD brain is the presence of an over-abundance of extraneuronal deposits of β-A4 amyloid protein (senile plaques) and intraneuronal inclusions of cytoskeletal elements (neurofibrillary tangles). These pathological hallmarks are found in the neocortex, hippocampus and the amygdala, and with a lower frequency in the basal ganglia, cerebellum and other areas [1], [7], [58]. These same anatomical regions contain, in the nondemented human brain, high levels of opioid receptors [18], [32], [49]. At present, it is unknown whether, in the AD brain, there exists a preferential specificity for the development of the lesions on or near neurons containing a specific receptor or a specific neurotransmitter. Nevertheless, the overlap between the distribution of opioid receptors and the location of lesions characteristic of AD led us to investigate possible alterations of binding levels of the three opioid receptors in AD in key areas of the brain thought to be involved in the pathology of AD.

Section snippets

Human brain postmortem tissue

All human brain specimens in this study were obtained from Dr. D.C. Mash, director of the Miami Brain Endowment Bank (School of Medicine, University of Miami, FL, USA). The postmortem samples used in this study were taken from a group of 11 clinically and neuropathologically confirmed AD patients and another group of 10 age-matched controls that had no history of dementia and presented normal results in neurological testing. The clinical diagnosis of our control individuals included

Assessment of neuropathological lesions in brain specimens

Examination of thioflavin S stained sections of CA1 and subiculum areas of hippocampus and parahippocampal gyrus of all AD brains with fluorescent microscopy revealed pronounced densities of neurofibrillary tangles and of senile plaques corresponding to a CERAD scale score of ‘severe’ and ‘moderate to severe’, respectively, thus confirming the diagnosis of AD. In contrast, there were an absence of neurofibrillary tangles and a paucity of senile plaques in the control subjects. Further,

Discussion

The present study examines the levels of μ-, δ- and κ-opioid receptors in various areas of coronal sections of postmortem human brain from AD patients and age-matched nondemented individuals using quantitative receptor autoradiography. This standard methodology, when used, as here, with saturating concentrations of tritiated ligands, allowed us to examine μ-, δ- and κ-opioid receptor levels with excellent reproducibility and high selectivity for a single type of opioid receptor, or under

Acknowledgements

This work was supported in part by DA00017 to E.J.S. and DA05130, DA10223 to M.J.K. Support from Pfizer Corp. is gratefully acknowledged.

References (70)

  • J.M. Hiller et al.

    Selective changes in μ, δ and κ opioid receptor binding in certain limbic regions of the brain in Alzheimer’s disease patients

    Brain Res.

    (1987)
  • M. Hiramatsu et al.

    Effects of dynorphin A (1-13) on carbon monoxide-induced delayed amnesia in mice

    Pharmacol. Biochem. Behav.

    (1997)
  • M. Ikeda et al.

    Differential alterations in adenosine A1 and κ 1 opioid receptors in the striatum in Alzheimer’s disease

    Brain Res.

    (1993)
  • R.Y. Ilyutchenok et al.

    Memory retrieval enhancement by κ opioid agonist and μ, δ antagonists

    Pharmacol. Biochem. Behav.

    (1995)
  • J. Itoh et al.

    Dynorphin A-(1-13) potently prevents memory dysfunctions induced by transient cerebral ischemia in mice

    Eur. J. Pharmacol.

    (1993)
  • K.L. Jansen et al.

    Alzheimer’s disease: changes in hippocampal N-methyl-d-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors – an autoradiographic study

    Neuroscience

    (1990)
  • R. Maurer et al.

    Multiple opiate receptor in human brain: an autoradiographic investigation

    Life Sci.

    (1983)
  • M. Minami et al.

    Molecular biology of the opioid receptors: structures, functions and distributions

    Neurosci. Res.

    (1995)
  • M. Ohno et al.

    Effect of the κ-receptor agonist, U-50,488H, on cerebral ischemia-induced impairment of working memory assessed in rats by a three-panel runway task

    Eur. J. Pharmacol.

    (1991)
  • G.A. Olson et al.

    Effects of an enkephalin analog on complex learning in the Rhesus monkey

    Pharmacol. Biochem. Behav.

    (1979)
  • Z.Z. Pan

    μ-Opposing actions of the κ-opioid receptor

    Trends Pharmacol. Sci.

    (1998)
  • D. Peckys et al.

    Expression of μ, κ, and δ opioid receptor messenger RNA in the human CNS: A 33P in situ hybridization study

    Neuroscience

    (1999)
  • C. Pilapil et al.

    Autoradiographic distribution of multiple classes of opioid receptor binding sites in human forebrain

    Brain Res. Bull.

    (1987)
  • A.W. Procter et al.

    Topographical distribution of neurochemical changes in Alzheimer’s disease

    J. Neurol. Sci.

    (1988)
  • R. Quirion et al.

    Distribution of multiple opioid receptors in the human brain

  • K.J. Reinikainen et al.

    A post-mortem study of noradrenergic, serotonergic and GABAergic neurons in Alzheimer’s disease

    J. Neurol. Sci.

    (1988)
  • T.D. Reisine et al.

    Alterations in brain opiate receptors in Parkinson’s disease

    Brain Res.

    (1979)
  • J.O. Rinne et al.

    Brain methionine- and leucine-enkephalin receptors in patients with dementia

    Neurosci. Lett.

    (1993)
  • D. Risser et al.

    Endogenous opioids in frontal cortex of patients with Down syndrome

    Neurosci. Lett.

    (1996)
  • T. Ritchie et al.

    [3H]Naloxone binding in the human brain: alcoholism and the TaqI A D2 dopamine receptor polymorphism

    Brain Res.

    (1996)
  • M.C. Royston et al.

    Analysis of laminar distribution of κ opiate receptor in human cortex: comparison between schizophrenia and normal

    J. Neurosci. Methods

    (1991)
  • M. Ukai et al.

    Dynorphin A-(1-13) attenuates basal forebrain-lesion-induced amnesia in rats

    Brain Res.

    (1993)
  • M. Ukai et al.

    Systemic administration of dynorphin A-(1-13) markedly improves cycloheximide-induced amnesia in mice

    Eur. J. Pharmacol.

    (1996)
  • P.M. Wall et al.

    U-69,593 microinjection in the infralimbic cortex reduces anxiety and enhances spontaneous alternation memory in mice

    Brain Res.

    (2000)
  • J. Wegiel et al.

    Cerebellar atrophy in Alzheimer’s disease – clinicopathological correlations

    Brain Res.

    (1999)
  • Cited by (109)

    • G protein-coupled receptors in neurodegenerative diseases

      2019, GPCRs: Structure, Function, and Drug Discovery
    • Can crosstalk between DOR and PARP reduce oxidative stress mediated neurodegeneration?

      2018, Neurochemistry International
      Citation Excerpt :

      It was observed that DOR's were unchanged in the hippocampus and the occipito-temporal gyrus in both the groups while the DOR's decreased in both the groups in the amygdala, which could imply the role of DOR in the early pathophysiology of AD. Also, there was a decrease in DOR-binding levels in the putamen which suggested the possibility of changes in the pathology of the disease as it progressed further (Mathieu-Kia et al., 2001). Aβ, which is a major pathological hallmark of AD, is generated from Aβ-protein precursor (AβPP) via sequential cleavage by β- (BACE1) and γ-secretase whereas α-secretase prevents Aβ generation by cleaving within the middle of the Aβ domain.

    View all citing articles on Scopus
    View full text