You are on page 1of 7

Experimental Neurology 360 (2023) 114287

Contents lists available at ScienceDirect

Experimental Neurology
journal homepage: www.elsevier.com/locate/yexnr

Research paper

Chronic pain in Alzheimer’s disease: Endocannabinoid system


Henry Blanton a, P. Hemachandra Reddy a, b, Khalid Benamar a, *
a
Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA
b
Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Chronic pain, one of the most common reasons adults seek medical care, has been linked to restrictions in
Pain mobility and daily activities, dependence on opioids, anxiety, depression, sleep deprivation, and reduced quality
Endocannabinoid of life. Alzheimer’s disease (AD), a devastating neurodegenerative disorder (characterized by a progressive
Alzheimer
impairment of cognitive functions) in the elderly, is often co-morbid with chronic pain. AD is one of the most
Aging
common neurodegenerative disorders in the aged population. The reported prevalence of chronic pain is 45.8%
of the 50 million people with AD. As the population ages, the number of older people who experience AD and
chronic pain will also increase. The current treatment options for chronic pain are limited, often ineffective, and
have associated side effects. This review summarizes the role of the endocannabinoid system in pain, its potential
role in chronic pain in AD, and addresses gaps and future directions.

1. Introduction hyperphosphorylated tau.


Among the aging population, cognitive decline, and chronic pain are
Pain is a complex phenomenon that comprises sensory, affective, and two significant healthcare burdens affecting the aging population and
cognitive components. Pain is defined as an unpleasant sensory and the healthcare systems and caregivers that support these individuals.
emotional experience associated with or resembling that associated with (van Kooten et al., 2017). The reported prevalence of chronic pain in AD
actual or potential tissue damage. (IASP, 2020). Chronic pain is defined patients is 45.8% of the 50 million people worldwide (van Kooten et al.,
as pain persisting beyond three months. It is a significant healthcare 2016).
burden affecting approximately 50 million adults in the US alone This review will provide a brief overview of chronic pain in aging
(Dahlhamer et al., 2018), with an estimated annual healthcare burden of with and without AD, shared pathology and mechanisms seen in older
$600 billion in the United States alone (Gaskin and Richard, 2012). adults with AD and chronic pain, and explore the therapeutic potential
Chronic pain, one of the most common reasons adults seek medical care, of drugs targeting the endocannabinoid system as a novel treatment
has been linked to restrictions in mobility and daily activities, depen­ avenue.
dence on opioids, anxiety, depression, sleep deprivation, and reduced
quality of life. 2. Age, AD, and pain
Alzheimer’s disease (AD) is the most common form of dementia,
accounting for 60–80% of dementia cases (Association, A. S., 2021). In In 2020, the population aged 60 years and overreach 1.4 billion.
2020, the estimated cost of caring for and treating people with Alz­ (WHO, 2021) By 2050, the world’s population of people aged 60 years
heimer’s disease was $305 billion. By 2050, these costs are projected to and older will double (2.1 billion). (WHO, 2021) In the United States,
be more than $1.1 trillion. (CDC, 2020) AD is a devastating neurode­ the population of older individuals (age 65+) is projected to double in
generative disorder and one of the most common causes of neurode­ the coming decades, from 49 million in 2016 to 99 million in 2060
generative dementia in the elderly, and adults over 65 comprise (Vespa et al., 2018). Chronic pain in the elderly is a growing concern as
approximately 95% of AD cases (Zhu et al., 2015). It is characterized by the world’s population ages (Domenichiello and Ramsden, 2019).
a progressive impairment of cognitive functions. AD is characterized Indeed, old age (age 65+) is significantly correlated with an increased
pathologically by extracellular amyloid β (Aβ) plaques and intracellular incidence of chronic pain conditions (Dahlhamer et al., 2018; Yang and
neurofibrillary tangles (NFTs) composed of abnormally Chang, 2019), The most common forms of chronic pain among older

* Corresponding author.
E-mail address: khalid.benamar@ttuhsc.edu (K. Benamar).

https://doi.org/10.1016/j.expneurol.2022.114287
Received 8 August 2022; Received in revised form 9 November 2022; Accepted 23 November 2022
Available online 29 November 2022
0014-4886/© 2022 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
H. Blanton et al. Experimental Neurology 360 (2023) 114287

adults affect the back, neck, and joints (Husky et al., 2018; Guez et al., chronic pain via central sensitization, which can be induced and main­
2002; Pleis et al., 2009). Despite the increased prevalence of chronic tained by cytokines, chemokines, and other glia-produced mediators.
pain in the elderly, some studies have found increased pain thresholds to Neuroinflammation has a key role in the induction and maintenance of
experimentally induced pain in older adults (Lautenbacher et al., 2017), chronic pain (Ji et al., 2014; Ellis and Bennett, 2013; White et al., 2005),
while other studies show mixed effects in pain thresholds between (Ji et al., 2018).
younger versus older adults (El Tumi et al., 2017). Differences in pain Neuroinflammation not only serves as a driving force for chronic
progression, ages, gender, sample size, modalities of evoked pain, in­ pain but is also implicated in neurodegenerative diseases such as AD
tensity, and outcome measures used may account for the discrepancy. (Heneka et al., 2015). Clinical (Hamelin et al., 2016; Fan et al., 2017;
AD is associated with increased chronic pain prevalence (van Kooten Zhou et al., 2021; Leng and Edison, 2021) and preclinical (Martin et al.,
et al., 2016). Similarly showing varied responses to experimental pain 2017) data have shown neuroinflammation in the brains with AD. In AD,
thresholds, including increased sensitivity (van Kooten et al., 2016; neuroinflammation, instead of being a mere bystander activated by
Kunz et al., 2009; Kunz et al., 2007; Jensen-Dahm et al., 2014; Beach emerging senile plaques and neurofibrillary tangles, contributes as much
et al., 2015; Beach et al., 2016) and decreased sensitivity (Benedetti to the pathogenesis as the plaques and tangles themselves (Heneka et al.,
et al., 1999; Gibson et al., 2001; Monroe et al., 2017), which may vary 2015; Zhang et al., 2013). In AD, microglia can bind to soluble Aβ
based on the type of painful stimuli and its intensity. The most longi­ oligomers and Aβ fibrils, which are thought to be part of the inflam­
tudinal studies of pain in people with dementia revealed that at the time matory reaction in AD (Heneka et al., 2015). Studies showed that
of diagnosis, individuals with dementia reported significantly more pain microglial activation contributes to Tau pathology during AD patho­
than individuals without dementia. (Kumaradev et al., 2021). Pain genesis (Hopp et al., 2018; Asai et al., 2015; Maphis et al., 2015; Bhaskar
prevalence and the increase in its intensity are of particular concern. et al., 2010).
Currently, a considerable proportion of people with AD and chronic pain
likely may not be receiving adequate treatment compared to those 3.3. Mitochondria
without AD. Additionally, pain is a key trigger for behavioral and psy­
chological symptoms of dementia (Atee et al., 2021; Flo et al., 2014) Mitochondria play a critical role in ATP synthesis, redox balance, and
such as agitation and mood disorders, and poorly managed pain in AD many other biological processes, including ion homeostasis, nuclear
can result in overprescribing harmful antipsychotic medications. Addi­ gene expression, protein turnover, and post-translational modification
tionally, inadequately treated chronic pain can lead to restrictions on and apoptosis (Bozi et al., 2020). In terms of pain, both neuropathic and
daily activity and psychological problems, including depression and chronic inflammatory pain models have been associated with neuronal
anxiety. mitochondrial dysfunction, including impaired bioenergetics, calcium
Below we summarized some potential mechanisms that may account overload, oxidative stress, and aldehydic burden (Zambelli et al., 2014;
for the enhanced pain in AD. Flatters, 2015).
Many lines of evidence suggest that mitochondria have a central role
3. AD and chronic pain: shared disease features in aging-related neurodegenerative diseases (Lin and Beal, 2006).
Abnormal mitochondrial dynamics have been found to be early events in
3.1. Brain structures the AD disease process in studies using post-mortem AD brains, AD cell
cultures, and AD mouse models (Reddy and McWeeney, 2006; Reddy
Given that AD is a brain disease, there is a concern that AD may affect et al., 2005; Manczak et al., 2011; Manczak and Reddy, 2012a; Kandi­
some brain areas involved in pain and, therefore, pain response and the malla et al., 2016; Manczak et al., 2016; Manczak et al., 2018; Reddy
effect of analgesics. AD produces structural and functional brain alter­ et al., 2018). Increased mitochondria fission and reduced fusion can
ations, including changes in gray matter volume and disrupted func­ impair mitochondrial dynamics in AD-affected neurons (Manczak et al.,
tional connectivity between brain networks (Palesi et al., 2016). Among 2019; Manczak and Reddy, 2012b; Knott and Bossy-Wetzel, 2008).
the areas disrupted by AD are brain areas critical for processing pain Other studies using AD neurons revealed that Aβ interacts with The
information (Lawn et al., 2021). Among the earliest sites affected by AD dynamin-related protein (Drp1), with a subsequent increase in free
are the basal forebrain and medial temporal lobes, areas involved in pain radical production, which in turn activates Drp1 and Mitochondrial
processing, while the sensory cortices responsible for pain perception fission protein 1 (Fis1), causing excessive mitochondrial fragmentation,
remain intact (Lawn et al., 2021). defective transport of mitochondria to synapses, provides low synaptic
Neuroimaging studies showed that when exposed to experimental ATP and ultimately leads to synaptic dysfunction (Morton et al., 2021).
pain, subjects with early-stage AD had sustained activity in pain pro­ Further studies revealed that phosphorylated tau (p-tau) interacts with
cessing regions such as the dorsolateral prefrontal cortex (dlPFC), sug­ Drp1, enhances GTPase Drp1 enzymatic activity, and leads to excessive
gesting AD patients may have greater difficulty in the cognitive and fragmentation of mitochondria and mitochondrial dysfunction in AD.
emotional appraisal of pain, and pain may be more distressing (Cole Substantial amounts of data have demonstrated mitochondrial
et al., 2006). Similarly, fMRI analysis of networks in experimental pain dysfunction in chronic pain (Bozi et al., 2020; Flatters, 2015; Doyle and
reveals greater activation of Salience and Default Mode Networks in AD Salvemini, 2021; Hernandez-Beltran et al., 2013; Lagos-Rodriguez et al.,
patients compared to healthy controls, which may correlate to greater 2020).
attention to pain, and increased emotional responsiveness to pain Taken together, there is a significant overlap in AD and chronic pain
(Beach et al., 2017). In addition to the brain structure changes, other and shared mechanisms summarized in Fig. 1 that may account for the
shared features between AD and chronic pain include neuro­ enhanced pain in AD.
inflammation and mitochondria dysfunction. While the current treatment options for chronic pain are limited,
often ineffective, and have associated side effects, the simultaneous
3.2. Neuroinflammation presence of chronic pain and AD could pose an additional challenge in
managing chronic pain (please see section 3). In Fig. 2, we summarize
Neuroinflammation is a term used to describe the broad range of the potential mechanism(s) by which pain sensitivity is increased in AD
immune responses of the central nervous system (Lyman et al., 2014). with chronic pain.
Four features of neuroinflammation include increased vascular perme­ Based on this background (section 3), it is unlikely to assume that
ability, leukocyte infiltration, glial cell activation, and increased pro­ pain response and analgesic efficacy are the same in older adults with
duction of inflammatory mediators such as cytokines and chemokines AD versus non-AD. Therefore, chronic pain management in the aged
(Ji et al., 2014). Mechanistically, neuroinflammation drives widespread population needs to be addressed with and without AD. This will ensure

2
H. Blanton et al. Experimental Neurology 360 (2023) 114287

Fig. 1. Chronic pain and AD: commons disease features.

Fig. 2. Potential mechanism(s) by which pain severity is increased in AD.

that 45.8% of the 50 million people worldwide (van Kooten et al., 2016) 1996; Dinh et al., 2002).
with chronic pain and AD are receiving adequate treatment compared to
those without AD. 4.2. The endocannabinoid system and pain
In the next section, we will explore the endocannabinoid system’s
role in pain and AD. The endocannabinoid system is expressed throughout pain pathways
in the CNS, PNS, and neuroimmune system, and cannabinoids have
4. The endocannabinoid system’s role in pain and AD shown beneficial effects in various preclinical models of acute and
chronic pain of mixed origins (Finn et al., 2021). Cannabinoids may
4.1. The cannabinoids and endocannabinoid System produce analgesic effects through inhibition of pain signaling at the
periphery, spinal cord, and brain by either dampening the ascending
The endocannabinoid system serves as a broadly acting modulatory nociceptive information that initiates the process of pain or producing
system, influencing various processes in the body, with key roles in in­ descending modulation of pain. In the periphery, evidence for inhibition
flammatory processes and pain modulation. The endocannabinoid sys­ of ascending nociceptive signaling by cannabinoids has been demon­
tem at its core is defined by three main components: 1) the endogenous strated via local administration routes such as intraplantar injection.
receptors, 2) the ligands for those receptors known as endocannabi­ Indeed, in animal pain models, local administration of cannabinoids has
noids, and 3) the enzymes responsible for the synthesis and degradation been demonstrated to be antinociceptive via both CB1 and CB2 re­
of those endocannabinoids. The two primary cannabinoid receptors, the ceptors. (Calignano et al., 1998)
CB1 and CB2 receptors are G-protein coupled receptors (GPCR) (Felder In the brain, CB1 receptor agonists (e.g., Δ9-THC, CP 55,940, WIN
et al., 1995; Glass and Northup, 1999). 55,212–2) activate descending (inhibitory) pain circuits in multiple
Activation of cannabinoid receptors produces an inhibitory effect on pain-modulatory regions, including the amygdala (Manning et al.,
cell activity via multiple signaling pathways, including inhibition of 2003), periaqueductal gray (Lichtman et al., 1996) and rostral ventro­
adenylyl cyclase-mediated cAMP formation and modulation of potas­ medial medulla (Martin et al., 1998). Cannabinoids may also relieve
sium and calcium channels (Felder et al., 1995). CB1 is expressed pain through their anti-inflammatory effects via CB1, CB2, and other
throughout the central and peripheral nervous systems, and CB1 acti­ receptors. Selective CB2 receptor agonists show analgesic efficacy in
vation on CNS neurons is responsible for the psychoactive effects asso­ animal inflammatory and neuropathic pain models by inhibiting pro-
ciated with delta-9-tetrahydrocannabinol (Δ-9-THC), the primary inflammatory signaling (Malanjr et al., 2003). Additionally, orphan
psychoactive component in Cannabis sativa (Devane et al., 1992). In GPCRs such as GPR55 and GPR18 have also been implicated as targets
contrast, CB2 shows low expression in CNS neurons under normal for cannabinoid-mediated antinociception (Armin et al., 2021; Nar­
conditions but is highly expressed in immune cells and peripheral tissues asimhan et al., 2020).
(Matsuda et al., 1990; Munro et al., 1993).
The ligands acting on these receptors, known as endocannabinoids,
4.3. The endocannabinoid system in AD and AD mouse models
are fatty acid derivatives synthesized on demand and acting in a retro­
grade manner (Wilson and Nicoll, 2001). The two best-characterized
The endocannabinoid system has been demonstrated to be altered in
endocannabinoids are 2-arachidonoylglycerol (2-AG) and 2-arachido­
AD, and these changes may reflect potential druggable targets to relieve
noylethanolamide, also known as Anandamide (AEA) (Devane et al.,
symptomology and alter disease progression. Post-mortem analyses of
1992; Mechoulam et al., 1995).
the frontal cortex from AD patients found a decrease in CB1 expression,
Levels of these endogenous cannabinoids may be elevated to produce
an increase in protein nitration of CB1 and CB2—thought to reflect
therapeutic effects through the use of inhibitors of the enzymes
oxidative damage, as well as a decrease in G-protein coupling at the CB1
responsible for their degradation, monoacylglycerol lipase (MAGL) for
receptor produced by the agonist 3HWIN55,212–2 (Ramirez et al.,
2-AG, and fatty acid amide hydrolase (FAAH) for AEA (Cravatt et al.,
2005). The CB2 receptor was found to be upregulated in activated

3
H. Blanton et al. Experimental Neurology 360 (2023) 114287

microglia in neuritic plaques in the parahippocampal cortex, in addition relationship has been proposed between worsening sleep quality and
to an increase in FAAH protein levels and activity in plaque-associated worsening AD symptomatology (Ju et al., 2014; Guarnieri and Sorbi,
astrocytes (Benito et al., 2003). Endocannabinoid levels have also 2015; Villa et al., 2015). In AD patients, Δ9THC-containing cannabis oil
been found to be altered in AD patients. Post-mortem studies of AD (Shelef et al., 2016) and dronabinol (Walther et al., 2006) have shown
brains found an increase in the 2-AG synthesizing and degradative en­ beneficial effects on sleep in addition to decreasing agitation and
zymes, DAGL and MAGL, in the hippocampus (Mulder et al., 2011), and improving mood.
the prefrontal and temporal cortex, lower levels of anandamide were
found, which correlated to an increase in amyloid beta (Aβ) levels (Ju 6. Conclusions, gaps, and future directions
et al., 2014).
Exogenous cannabinoids have shown beneficial effects in AD models, The reported prevalence of chronic pain is 45.8% of the 50 million
improving behavioral symptoms through improved clearance of plaques people with AD worldwide. (van Kooten et al., 2016) Pain may be
and decreasing neuroinflammation (Abate et al., 2021). In addition, underestimated in AD patients as they may be unable to communicate
centrally acting CB1 receptor agonists have shown beneficial effects in their pain, (Achterberg et al., 2013) particularly in more severe stages of
preclinical AD studies, reducing the accumulation of Aβ (Cao et al., dementia. As the population ages, the number of older people who
2014; Janefjord et al., 2014), inhibiting neuroinflammation (Currais experience AD and pain will also increase (Prince et al., 2013). The
et al., 2016; Schubert et al., 2019), and improving cognition (Aso et al., current treatment options for chronic pain are limited, often ineffective,
2016). However, centrally acting CB1 agonists have known downsides; and have associated side effects. Despite rapidly shifting policies
their ability to impair learning and memory is particularly relevant to affecting access to cannabinoids, the use of cannabis continues to in­
AD. Impairments of CB1 agonists in memory are attributed to the high crease among older adults nationally (Han and Palamar, 2020) and the
density of CB1 receptors in the hippocampus (Lichtman et al., 1995; high prevalence of chronic pain in AD (van Kooten et al., 2016). To date,
Clarke et al., 2008; Kosiorek et al., 2003). Indeed, in the hippocampus, there are no data on whether and how the endocannabinoid system is
CB1 agonists may reduce long-term potentiation (Terranova et al., 1995; regulated in individuals with AD who have chronic pain. Therefore,
Hill et al., 2004), alter firing patterns (Robinson et al., 2007; Goona­ there is an urgent need to address these knowledge gaps.
wardena et al., 2011; Barbieri et al., 2016), and decrease acetylcholine
release (Gessa et al., 1997; Braida and Sala, 2000). It is possible, how­ Author contribution
ever, that some of these adverse effects are a function of dose and
perhaps age that can be avoided. Indeed, in old mice, Δ9THC has been All authors equally contributed to the conception and writing of the
found to have beneficial effects on memory when administered in low article.
doses (Aso et al., 2016; Bilkei-Gorzo et al., 2017; Sarne et al., 2018).
CB2 receptor expression has been found to be increased in the post- Declaration of Competing Interest
mortem brains of AD patients (Benito et al., 2003; Grunblatt et al.,
2007), while preclinical studies have shown increased CB2 receptor The authors declare that they have no known competing financial
expression in the brain and spinal cord in states of chronic pain and interests or personal relationships that could have appeared to.
inflammation (Hsieh et al., 2011; Ghosh et al., 2022). The potential role
of CB2 receptors as therapeutic targets in AD models has been explored Data availability
limitedly but with promising results. For example, selective targeting of
CB2 receptors with the agonist JWH133 improved cognitive perfor­ The data that has been used is confidential.
mance, decreased microglial reactivity, and reduced inflammatory cy­
tokines (Aso et al., 2013). Additionally, in rats injected in the
Acknowledgements
hippocampus with Aβ, selective targeting of CB2 has been shown to
improve cognitive performance by improving Aβ clearance (Wu et al.,
This work was supported by grants DA053824-02S1 and DA053824
2013). Moreover, genetic knockout of the CB2 receptor has been found
from NIH.
to increase Aβ42 and plaque deposition and increase plaque-associated
microglia in the J20 APP AD mouse model (Koppel et al., 2013). Thus,
References
CB2 could be a target to slow or reverse AD pathology and improve
cognition. Abate, G., Uberti, D., Tambaro, S., 2021. Potential and limits of cannabinoids in
Alzheimer’s disease therapy. Biol. (Basel) 10 (6).
5. Potential therapeutic role of cannabinoids in older adults Achterberg, W.P., Pieper, M.J., van Dalen-Kok, A.H., de Waal, M.W., Husebo, B.S.,
Lautenbacher, S., Kunz, M., Scherder, E.J., Corbett, A., 2013. Pain management in
with AD and chronic pain patients with dementia. Clin. Interv. Aging 8, 1471–1482.
Armin, S., Muenster, S., Abood, M., Benamar, K., 2021. GPR55 in the brain and chronic
Older adults also comprise one of the fastest-growing age groups neuropathic pain. Behav. Brain Res. 406, 113248.
Asai, H., Ikezu, S., Tsunoda, S., Medalla, M., Luebke, J., Haydar, T., Wolozin, B.,
using cannabis and cannabinoids for medical purposes (Han and Pala­ Butovsky, O., Kugler, S., Ikezu, T., 2015. Depletion of microglia and inhibition of
mar, 2020). Among older adults using cannabis/cannabinoids for exosome synthesis halt tau propagation. Nat. Neurosci. 18 (11), 1584–1593.
medical purposes, relief from chronic pain is overwhelmingly reported Aso, E., Juves, S., Maldonado, R., Ferrer, I., 2013. CB2 cannabinoid receptor agonist
ameliorates Alzheimer-like phenotype in AbetaPP/PS1 mice. J. Alzheimers Dis. 35
as a primary reason for use (Reynolds et al., 2018; Yang et al., 2021). (4), 847–858.
Currently, there is no direct information about the use of cannabis/ Aso, E., Andres-Benito, P., Carmona, M., Maldonado, R., Ferrer, I., 2016. Cannabinoid
cannabinoids for the treatment of chronic pain in older individuals with receptor 2 participates in amyloid-beta processing in a mouse model of Alzheimer’s
disease but plays a minor role in the therapeutic properties of a Cannabis-based
AD. However, there is some evidence showing that the endocannabinoid medicine. J. Alzheimers Dis. 51 (2), 489–500.
system may still function in AD and thus has the potential to be a target Association, A. S, 2021. Alzheimer’s disease facts and figures. Alzheimers Dement. 17
for therapeutic strategy. For example, the beneficial effects of cannabis/ (3), 327–406.
Atee, M., Morris, T., Macfarlane, S., Cunningham, C., 2021. Pain in dementia: prevalence
cannabinoids on the physiological, behavioral, and neuropsychiatric
and association with neuropsychiatric behaviors. J. Pain Symptom Manag. 61 (6),
symptoms associated with AD have been reported. In particular, in in­ 1215–1226.
dividuals with AD, decreased agitation was seen in studies investigating Barbieri, M., Ossato, A., Canazza, I., Trapella, C., Borelli, A.C., Beggiato, S., Rimondo, C.,
the effects of dronabinol (Volicer et al., 1997), nabilone (Herrmann Serpelloni, G., Ferraro, L., Marti, M., 2016. Synthetic cannabinoid JWH-018 and its
halogenated derivatives JWH-018-Cl and JWH-018-Br impair novel object
et al., 2019), and Δ9THC-containing cannabis oil (Shelef et al., 2016). recognition in mice: behavioral, electrophysiological and neurochemical evidence.
Sleep disturbances are also prevalent in AD, where a bidirectional Neuropharmacology 109, 254–269.

4
H. Blanton et al. Experimental Neurology 360 (2023) 114287

Beach, P.A., Huck, J.T., Miranda, M.M., Bozoki, A.C., 2015. Autonomic, behavioral, and Gibson, S.J., Voukelatos, X., Ames, D., Flicker, L., Helme, R.D., 2001. An examination of
subjective pain responses in Alzheimer’s disease. Pain Med. 16 (10), 1930–1942. pain perception and cerebral event-related potentials following carbon dioxide laser
Beach, P.A., Huck, J.T., Miranda, M.M., Foley, K.T., Bozoki, A.C., 2016. Effects of stimulation in patients with Alzheimer’s disease and age-matched control
Alzheimer disease on the facial expression of pain. Clin. J. Pain 32 (6), 478–487. volunteers. Pain Res. Manag. 6 (3), 126–132.
Beach, P.A., Huck, J.T., Zhu, D.C., Bozoki, A.C., 2017. Altered behavioral and autonomic Glass, M., Northup, J.K., 1999. Agonist selective regulation of G proteins by cannabinoid
pain responses in Alzheimer’s disease are associated with dysfunctional affective, CB(1) and CB(2) receptors. Mol. Pharmacol. 56 (6), 1362–1369.
self-reflective and salience network resting-state connectivity. Front. Aging Neurosci. Goonawardena, A.V., Riedel, G., Hampson, R.E., 2011. Cannabinoids alter spontaneous
9, 297. firing, bursting, and cell synchrony of hippocampal principal cells. Hippocampus 21
Benedetti, F., Vighetti, S., Ricco, C., Lagna, E., Bergamasco, B., Pinessi, L., Rainero, I., (5), 520–531.
1999. Pain threshold and tolerance in Alzheimer’s disease. Pain 80 (1–2), 377–382. Grunblatt, E., Zander, N., Bartl, J., Jie, L., Monoranu, C.M., Arzberger, T., Ravid, R.,
Benito, C., Nunez, E., Tolon, R.M., Carrier, E.J., Rabano, A., Hillard, C.J., Romero, J., Roggendorf, W., Gerlach, M., Riederer, P., 2007. Comparison analysis of gene
2003. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively expression patterns between sporadic Alzheimer’s and Parkinson’s disease.
overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains. J. Alzheimers Dis. 12 (4), 291–311.
J. Neurosci. 23 (35), 11136–11141. Guarnieri, B., Sorbi, S., 2015. Sleep and cognitive decline: a strong bidirectional
Bhaskar, K., Konerth, M., Kokiko-Cochran, O.N., Cardona, A., Ransohoff, R.M., Lamb, B. relationship. It is time for specific recommendations on routine assessment and the
T., 2010. Regulation of tau pathology by the microglial fractalkine receptor. Neuron Management of Sleep Disorders in patients with mild cognitive impairment and
68 (1), 19–31. dementia. Eur. Neurol. 74 (1–2), 43–48.
Bilkei-Gorzo, A., Albayram, O., Draffehn, A., Michel, K., Piyanova, A., Oppenheimer, H., Guez, M., Hildingsson, C., Nilsson, M., Toolanen, G., 2002. The prevalence of neck pain:
Dvir-Ginzberg, M., Racz, I., Ulas, T., Imbeault, S., Bab, I., Schultze, J.L., Zimmer, A., a population-based study from northern Sweden. Acta Orthop. Scand. 73 (4),
2017. A chronic low dose of Delta(9)-tetrahydrocannabinol (THC) restores cognitive 455–459.
function in old mice. Nat. Med. 23 (6), 782–787. Hamelin, L., Lagarde, J., Dorothee, G., Leroy, C., Labit, M., Comley, R.A., de Souza, L.C.,
Bozi, L.H.M., Campos, J.C., Zambelli, V.O., Ferreira, N.D., Ferreira, J.C.B., 2020. Corne, H., Dauphinot, L., Bertoux, M., Dubois, B., Gervais, P., Colliot, O., Potier, M.
Mitochondrially-targeted treatment strategies. Mol. Asp. Med. 71, 100836. C., Bottlaender, M., Sarazin, M., Clinical, I.T., 2016. Early and protective microglial
Braida, D., Sala, M., 2000. Cannabinoid-induced working memory impairment is activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET
reversed by a second generation cholinesterase inhibitor in rats. Neuroreport 11 (9), imaging. Brain 139 (Pt 4), 1252–1264.
2025–2029. Han, B.H., Palamar, J.J., 2020. Trends in Cannabis use among older adults in the United
Calignano, A., La Rana, G., Giuffrida, A., Piomelli, D., 1998. Control of pain initiation by States, 2015-2018. JAMA Intern. Med. 180 (4), 609–611.
endogenous cannabinoids. Nature 394 (6690), 277–281. Heneka, M.T., Carson, M.J., El Khoury, J., Landreth, G.E., Brosseron, F., Feinstein, D.L.,
Cao, C., Li, Y., Liu, H., Bai, G., Mayl, J., Lin, X., Sutherland, K., Nabar, N., Cai, J., 2014. Jacobs, A.H., Wyss-Coray, T., Vitorica, J., Ransohoff, R.M., Herrup, K., Frautschy, S.
The potential therapeutic effects of THC on Alzheimer’s disease. J. Alzheimers Dis. A., Finsen, B., Brown, G.C., Verkhratsky, A., Yamanaka, K., Koistinaho, J., Latz, E.,
42 (3), 973–984. Halle, A., Petzold, G.C., Town, T., Morgan, D., Shinohara, M.L., Perry, V.H.,
CDC, https://www.cdc.gov/aging/publications/features/Alz-Greater-Risk.html. 2020. Holmes, C., Bazan, N.G., Brooks, D.J., Hunot, S., Joseph, B., Deigendesch, N.,
Clarke, J.R., Rossato, J.I., Monteiro, S., Bevilaqua, L.R., Izquierdo, I., Cammarota, M., Garaschuk, O., Boddeke, E., Dinarello, C.A., Breitner, J.C., Cole, G.M., Golenbock, D.
2008. Posttraining activation of CB1 cannabinoid receptors in the CA1 region of the T., Kummer, M.P., 2015. Neuroinflammation in Alzheimer’s disease. Lancet Neurol.
dorsal hippocampus impairs object recognition long-term memory. Neurobiol. 14 (4), 388–405.
Learn. Mem. 90 (2), 374–381. Hernandez-Beltran, N., Moreno, C.B., Gutierrez-Alvarez, A.M., 2013. Contribution of
Cole, L.J., Farrell, M.J., Duff, E.P., Barber, J.B., Egan, G.F., Gibson, S.J., 2006. Pain mitochondria to pain in diabetic neuropathy. Endocrinol. Nutr. 60 (1), 25–32.
sensitivity and fMRI pain-related brain activity in Alzheimer’s disease. Brain 129 (Pt Herrmann, N., Ruthirakuhan, M., Gallagher, D., Verhoeff, N., Kiss, A., Black, S.E.,
11), 2957–2965. Lanctot, K.L., 2019. Randomized placebo-controlled trial of Nabilone for agitation in
Cravatt, B.F., Giang, D.K., Mayfield, S.P., Boger, D.L., Lerner, R.A., Gilula, N.B., 1996. Alzheimer’s disease. Am. J. Geriatr. Psychiatry 27 (11), 1161–1173.
Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid Hill, M.N., Froc, D.J., Fox, C.J., Gorzalka, B.B., Christie, B.R., 2004. Prolonged
amides. Nature 384 (6604), 83–87. cannabinoid treatment results in spatial working memory deficits and impaired long-
Currais, A., Quehenberger, O., Armando, O.M.A., Daugherty, D., Maher, P., Schubert, D., term potentiation in the CA1 region of the hippocampus in vivo. Eur. J. Neurosci. 20
2016. Amyloid proteotoxicity initiates an inflammatory response blocked by (3), 859–863.
cannabinoids. NPJ. Aging. Mech. Dis. 2 (1), 16012. Hopp, S.C., Lin, Y., Oakley, D., Roe, A.D., DeVos, S.L., Hanlon, D., Hyman, B.T., 2018.
Dahlhamer, J., Lucas, J., Zelaya, C., Nahin, R., Mackey, S., DeBar, L., Kerns, R., Von The role of microglia in processing and spreading of bioactive tau seeds in
Korff, M., Porter, L., Helmick, C., 2018. Prevalence of chronic pain and high-impact Alzheimer’s disease. J. Neuroinflammation 15 (1), 269.
chronic pain among adults - United States, 2016. MMWR Morb. Mortal. Wkly Rep. Hsieh, G.C., Pai, M., Chandran, P., Hooker, B.A., Zhu, C.Z., Salyers, A.K., Wensink, E.J.,
67 (36), 1001–1006. Zhan, C., Carroll, W.A., Dart, M.J., Yao, B.B., Honore, P., Meyer, M.D., 2011. Central
Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G., and peripheral sites of action for CB(2) receptor mediated analgesic activity in
Gibson, D., Mandelbaum, A., Etinger, A., Mechoulam, R., 1992. Isolation and chronic inflammatory and neuropathic pain models in rats. Br. J. Pharmacol. 162
structure of a brain constituent that binds to the cannabinoid receptor. Science 258 (2), 428–440.
(5090), 1946–1949. Husky, M.M., Ferdous Farin, F., Compagnone, P., Fermanian, C., Kovess-Masfety, V.,
Dinh, T.P., Carpenter, D., Leslie, F.M., Freund, T.F., Katona, I., Sensi, S.L., Kathuria, S., 2018. Chronic back pain and its association with quality of life in a large French
Piomelli, D., 2002. Brain monoglyceride lipase participating in endocannabinoid population survey. Health Qual. Life Outcomes 16 (1), 195.
inactivation. Proc. Natl. Acad. Sci. U. S. A. 99 (16), 10819–10824. IASP, 2020. IASP Announces Revised Definition of Pain.
Domenichiello, A.F., Ramsden, C.E., 2019. The silent epidemic of chronic pain in older Janefjord, E., Maag, J.L., Harvey, B.S., Smid, S.D., 2014. Cannabinoid effects on beta
adults. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 93, 284–290. amyloid fibril and aggregate formation, neuronal and microglial-activated
Doyle, T.M., Salvemini, D., 2021. Mini-review: mitochondrial dysfunction and neurotoxicity in vitro. Cell. Mol. Neurobiol. 34 (1), 31–42.
chemotherapy-induced neuropathic pain. Neurosci. Lett. 760, 136087. Jensen-Dahm, C., Werner, M.U., Dahl, J.B., Jensen, T.S., Ballegaard, M., Hejl, A.M.,
El Tumi, H., Johnson, M.I., Dantas, P.B.F., Maynard, M.J., Tashani, O.A., 2017. Age- Waldemar, G., 2014. Quantitative sensory testing and pain tolerance in patients with
related changes in pain sensitivity in healthy humans: a systematic review with mild to moderate Alzheimer disease compared to healthy control subjects. Pain 155
meta-analysis. Eur. J. Pain 21 (6), 955–964. (8), 1439–1445.
Ellis, A., Bennett, D.L., 2013. Neuroinflammation and the generation of neuropathic Ji, R.R., Xu, Z.Z., Gao, Y.J., 2014. Emerging targets in neuroinflammation-driven chronic
pain. Br. J. Anaesth. 111 (1), 26–37. pain. Nat. Rev. Drug Discov. 13 (7), 533–548.
Fan, Z., Brooks, D.J., Okello, A., Edison, P., 2017. An early and late peak in microglial Ji, R.R., Nackley, A., Huh, Y., Terrando, N., Maixner, W., 2018. Neuroinflammation and
activation in Alzheimer’s disease trajectory. Brain 140 (3), 792–803. central sensitization in chronic and widespread pain. Anesthesiology 129 (2),
Felder, C.C., Joyce, K.E., Briley, E.M., Mansouri, J., Mackie, K., Blond, O., Lai, Y., Ma, A. 343–366.
L., Mitchell, R.L., 1995. Comparison of the pharmacology and signal transduction of Ju, Y.E., Lucey, B.P., Holtzman, D.M., 2014. Sleep and Alzheimer disease pathology–a
the human cannabinoid CB1 and CB2 receptors. Mol. Pharmacol. 48 (3), 443–450. bidirectional relationship. Nat. Rev. Neurol. 10 (2), 115–119.
Finn, D.P., Haroutounian, S., Hohmann, A.G., Krane, E., Soliman, N., Rice, A.S.C., 2021. Kandimalla, R., Manczak, M., Fry, D., Suneetha, Y., Sesaki, H., Reddy, P.H., 2016.
Cannabinoids, the endocannabinoid system, and pain: a review of preclinical studies. Reduced dynamin-related protein 1 protects against phosphorylated tau-induced
Pain 162 (Suppl. 1), S5–S25. mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum. Mol.
Flatters, S.J., 2015. The contribution of mitochondria to sensory processing and pain. Genet. 25 (22), 4881–4897.
Prog. Mol. Biol. Transl. Sci. 131, 119–146. Knott, A.B., Bossy-Wetzel, E., 2008. Impairing the mitochondrial fission and fusion
Flo, E., Gulla, C., Husebo, B.S., 2014. Effective pain management in patients with balance: a new mechanism of neurodegeneration. Ann. N. Y. Acad. Sci. 1147,
dementia: benefits beyond pain? Drugs Aging 31 (12), 863–871. 283–292.
Gaskin, D.J., Richard, P., 2012. The economic costs of pain in the United States. J. Pain Koppel, J., Vingtdeux, V., Marambaud, P., d’Abramo, C., Jimenez, H., Stauber, M.,
13 (8), 715–724. Friedman, R., Davies, P., 2013. CB(2) receptor deficiency increases amyloid
Gessa, G.L., Mascia, M.S., Casu, M.A., Carta, G., 1997. Inhibition of hippocampal pathology and alters tau processing in a transgenic mouse model of Alzheimer’s
acetylcholine release by cannabinoids: reversal by SR 141716A. Eur. J. Pharmacol. disease. Mol. Med. 19, 357–364.
327 (1), R1–R2. Kosiorek, P., Hryniewicz, A., Bialuk, I., Zawadzka, A., Winnicka, M.M., 2003.
Ghosh, K., Zhang, G.F., Chen, H., Chen, S.R., Pan, H.L., 2022. Cannabinoid CB2 receptors Cannabinoids alter recognition memory in rats. Pol. J. Pharmacol. 55 (5), 903–910.
are upregulated via bivalent histone modifications and control primary afferent Kumaradev, S., Fayosse, A., Dugravot, A., Dumurgier, J., Roux, C., Kivimaki, M., Singh-
input to the spinal cord in neuropathic pain. J. Biol. Chem. 298 (6), 101999. Manoux, A., Sabia, S., 2021. Timeline of pain before dementia diagnosis: a 27-year
follow-up study. Pain 162 (5), 1578–1585.

5
H. Blanton et al. Experimental Neurology 360 (2023) 114287

Kunz, M., Scharmann, S., Hemmeter, U., Schepelmann, K., Lautenbacher, S., 2007. The Munro, S., Thomas, K.L., Abu-Shaar, M., 1993. Molecular characterization of a peripheral
facial expression of pain in patients with dementia. Pain 133 (1–3), 221–228. receptor for cannabinoids. Nature 365 (6441), 61–65.
Kunz, M., Mylius, V., Scharmann, S., Schepelman, K., Lautenbacher, S., 2009. Influence Narasimhan, M., Mahimainathan, L., Reddy, P.H., Benamar, K., 2020. GPR18-NAGly
of dementia on multiple components of pain. Eur. J. Pain 13 (3), 317–325. system in periaqueductal gray and chronic neuropathic pain. Biochim. Biophys. Acta
Lagos-Rodriguez, V., Martinez-Palma, L., Marton, S., Miquel, E., Escobar-Pintos, R., Mol. basis Dis. 1866 (12), 165974.
Cassina, A., Lago, N., Cassina, P., 2020. Mitochondrial bioenergetics, glial reactivity, Palesi, F., Castellazzi, G., Casiraghi, L., Sinforiani, E., Vitali, P., Gandini Wheeler-
and pain-related behavior can be restored by dichloroacetate treatment in rodent Kingshott, C.A., D’Angelo, E., 2016. Exploring patterns of alteration in Alzheimer’s
pain models. Pain 161 (12), 2786–2797. disease brain networks: a combined structural and functional connectomics analysis.
Lautenbacher, S., Peters, J.H., Heesen, M., Scheel, J., Kunz, M., 2017. Age changes in Front. Neurosci. 10, 380.
pain perception: a systematic-review and meta-analysis of age effects on pain and Pleis, J.R., Lucas, J.W., Ward, B.W., 2009. Summary health statistics for U.S. adults:
tolerance thresholds. Neurosci. Biobehav. Rev. 75, 104–113. national health interview survey, 2008. Vital Health Stat. 10 (242), 1–157.
Lawn, T., Aman, Y., Rukavina, K., Sideris-Lampretsas, G., Howard, M., Ballard, C., Ray Prince, M., Bryce, R., Albanese, E., Wimo, A., Ribeiro, W., Ferri, C.P., 2013. The global
Chaudhuri, K., Malcangio, M., 2021. Pain in the neurodegenerating brain: insights prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement.
into pharmacotherapy for Alzheimer disease and Parkinson disease. Pain 162 (4), 9 (1), 63–75 e2.
999–1006. Ramirez, B.G., Blazquez, C., Gomez del Pulgar, T., Guzman, M., de Ceballos, M.L., 2005.
Leng, F., Edison, P., 2021. Neuroinflammation and microglial activation in Alzheimer Prevention of Alzheimer’s disease pathology by cannabinoids: neuroprotection
disease: where do we go from here? Nat. Rev. Neurol. 17 (3), 157–172. mediated by blockade of microglial activation. J. Neurosci. 25 (8), 1904–1913.
Lichtman, A.H., Dimen, K.R., Martin, B.R., 1995. Systemic or intrahippocampal Reddy, P.H., McWeeney, S., 2006. Mapping cellular transcriptosomes in autopsied
cannabinoid administration impairs spatial memory in rats. Psychopharmacology Alzheimer’s disease subjects and relevant animal models. Neurobiol. Aging 27 (8),
119 (3), 282–290. 1060–1077.
Lichtman, A.H., Cook, S.A., Martin, B.R., 1996. Investigation of brain sites mediating Reddy, P.H., Mani, G., Park, B.S., Jacques, J., Murdoch, G., Whetsell Jr., W., Kaye, J.,
cannabinoid-induced antinociception in rats: evidence supporting periaqueductal Manczak, M., 2005. Differential loss of synaptic proteins in Alzheimer’s disease:
gray involvement. J. Pharmacol. Exp. Ther. 276 (2), 585–593. implications for synaptic dysfunction. J. Alzheimers Dis. 7 (2), 103–117 discussion
Lin, M.T., Beal, M.F., 2006. Mitochondrial dysfunction and oxidative stress in 173–80.
neurodegenerative diseases. Nature 443 (7113), 787–795. Reddy, P.H., Yin, X., Manczak, M., Kumar, S., Pradeepkiran, J.A., Vijayan, M., Reddy, A.
Lyman, M., Lloyd, D.G., Ji, X., Vizcaychipi, M.P., Ma, D., 2014. Neuroinflammation: the P., 2018. Mutant APP and amyloid beta-induced defective autophagy, mitophagy,
role and consequences. Neurosci. Res. 79, 1–12. mitochondrial structural and functional changes and synaptic damage in
Malanjr, T., Ibrahim, M., Lai, J., Vanderah, T., Makriyannis, A., Porreca, F., 2003. CB hippocampal neurons from Alzheimer’s disease. Hum. Mol. Genet. 27 (14),
cannabinoid receptor agonists: pain relief without psychoactive effects? Curr. Opin. 2502–2516.
Pharmacol. 3 (1), 62–67. Reynolds, I.R., Fixen, D.R., Parnes, B.L., Lum, H.D., Shanbhag, P., Church, S.,
Manczak, M., Reddy, P.H., 2012a. Abnormal interaction of VDAC1 with amyloid beta Linnebur, S.A., Orosz, G., 2018. Characteristics and patterns of marijuana use in
and phosphorylated tau causes mitochondrial dysfunction in Alzheimer’s disease. community-dwelling older adults. J. Am. Geriatr. Soc. 66 (11), 2167–2171.
Hum. Mol. Genet. 21 (23), 5131–5146. Robinson, L., Goonawardena, A.V., Pertwee, R.G., Hampson, R.E., Riedel, G., 2007. The
Manczak, M., Reddy, P.H., 2012b. Abnormal interaction between the mitochondrial synthetic cannabinoid HU210 induces spatial memory deficits and suppresses
fission protein Drp1 and hyperphosphorylated tau in Alzheimer’s disease neurons: hippocampal firing rate in rats. Br. J. Pharmacol. 151 (5), 688–700.
implications for mitochondrial dysfunction and neuronal damage. Hum. Mol. Genet. Sarne, Y., Toledano, R., Rachmany, L., Sasson, E., Doron, R., 2018. Reversal of age-
21 (11), 2538–2547. related cognitive impairments in mice by an extremely low dose of
Manczak, M., Calkins, M.J., Reddy, P.H., 2011. Impaired mitochondrial dynamics and tetrahydrocannabinol. Neurobiol. Aging 61, 177–186.
abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons Schubert, D., Kepchia, D., Liang, Z., Dargusch, R., Goldberg, J., Maher, P., 2019. Efficacy
from patients with Alzheimer’s disease: implications for neuronal damage. Hum. of cannabinoids in a pre-clinical drug-screening platform for Alzheimer’s disease.
Mol. Genet. 20 (13), 2495–2509. Mol. Neurobiol. 56 (11), 7719–7730.
Manczak, M., Kandimalla, R., Fry, D., Sesaki, H., Reddy, P.H., 2016. Protective effects of Shelef, A., Barak, Y., Berger, U., Paleacu, D., Tadger, S., Plopsky, I., Baruch, Y., 2016.
reduced dynamin-related protein 1 against amyloid beta-induced mitochondrial Safety and efficacy of medical Cannabis oil for behavioral and psychological
dysfunction and synaptic damage in Alzheimer’s disease. Hum. Mol. Genet. 25 (23), symptoms of dementia: an-open label, add-on, pilot study. J. Alzheimers Dis. 51 (1),
5148–5166. 15–19.
Manczak, M., Kandimalla, R., Yin, X., Reddy, P.H., 2018. Hippocampal mutant APP and Terranova, J.P., Michaud, J.C., Le Fur, G., Soubrie, P., 1995. Inhibition of long-term
amyloid beta-induced cognitive decline, dendritic spine loss, defective autophagy, potentiation in rat hippocampal slices by anandamide and WIN55212-2: reversal by
mitophagy and mitochondrial abnormalities in a mouse model of Alzheimer’s SR141716 A, a selective antagonist of CB1 cannabinoid receptors. Naunyn
disease. Hum. Mol. Genet. 27 (8), 1332–1342. Schmiedeberg’s Arch. Pharmacol. 352 (5), 576–579.
Manczak, M., Kandimalla, R., Yin, X., Reddy, P.H., 2019. Mitochondrial division van Kooten, J., Binnekade, T.T., van der Wouden, J.C., Stek, M.L., Scherder, E.J.,
inhibitor 1 reduces dynamin-related protein 1 and mitochondrial fission activity. Husebo, B.S., Smalbrugge, M., Hertogh, C.M., 2016. A review of pain prevalence in
Hum. Mol. Genet. 28 (2), 177–199. Alzheimer’s, vascular, frontotemporal and Lewy body dementias. Dement. Geriatr.
Manning, B.H., Martin, W.J., Meng, I.D., 2003. The rodent amygdala contributes to the Cogn. Disord. 41 (3–4), 220–232.
production of cannabinoid-induced antinociception. Neuroscience 120 (4), van Kooten, J., Smalbrugge, M., van der Wouden, J.C., Stek, M.L., Hertogh, C.M., 2017.
1157–1170. Prevalence of pain in nursing home residents: the role of dementia stage and
Maphis, N., Xu, G., Kokiko-Cochran, O.N., Jiang, S., Cardona, A., Ransohoff, R.M., dementia subtypes. J. Am. Med. Dir. Assoc. 18 (6), 522–527.
Lamb, B.T., Bhaskar, K., 2015. Reactive microglia drive tau pathology and contribute Vespa, J., Armstrong, D.M., Medina, L., 2018. Demographic Turning Points for the
to the spreading of pathological tau in the brain. Brain 138 (Pt 6), 1738–1755. United States: Population Projections for 2020 to 2060. US Department of
Martin, W.J., Tsou, K., Walker, J.M., 1998. Cannabinoid receptor-mediated inhibition of Commerce, Economics and Statistics Administration, US ….
the rat tail-flick reflex after microinjection into the rostral ventromedial medulla. Villa, C., Ferini-Strambi, L., Combi, R., 2015. The synergistic relationship between
Neurosci. Lett. 242 (1), 33–36. Alzheimer’s disease and sleep disorders: an update. J. Alzheimers Dis. 46 (3),
Martin, E., Boucher, C., Fontaine, B., Delarasse, C., 2017. Distinct inflammatory 571–580.
phenotypes of microglia and monocyte-derived macrophages in Alzheimer’s disease Volicer, L., Stelly, M., Morris, J., McLaughlin, J., Volicer, B.J., 1997. Effects of dronabinol
models: effects of aging and amyloid pathology. Aging Cell 16 (1), 27–38. on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int. J.
Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I., 1990. Structure of Geriatr. Psychiatry. 12 (9), 913–919.
a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346 Walther, S., Mahlberg, R., Eichmann, U., Kunz, D., 2006. Delta-9-tetrahydrocannabinol
(6284), 561–564. for nighttime agitation in severe dementia. Psychopharmacology 185 (4), 524–528.
Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N.E., Schatz, A.R., White, F.A., Bhangoo, S.K., Miller, R.J., 2005. Chemokines: integrators of pain and
Gopher, A., Almog, S., Martin, B.R., Compton, D.R., Pertwee, R.G., Griffin, G., inflammation. Nat. Rev. Drug Discov. 4 (10), 834–844.
Bayewitch, M., Barg, J., Vogel, Z., 1995. Identification of an endogenous 2-mono­ WHO, 2021. Ageing and Health.
glyceride, present in canine gut, that binds to cannabinoid receptors. Biochem. Wilson, R.I., Nicoll, R.A., 2001. Endogenous cannabinoids mediate retrograde signalling
Pharmacol. 50 (1), 83–90. at hippocampal synapses. Nature 410 (6828), 588–592.
Monroe, T.B., Beach, P.A., Bruehl, S.P., Dietrich, M.S., Rogers, B.P., Gore, J.C., Atalla, S. Wu, J., Bie, B., Yang, H., Xu, J.J., Brown, D.L., Naguib, M., 2013. Activation of the CB2
W., Cowan, R.L., 2017. The impact of Alzheimer’s disease on the resting state receptor system reverses amyloid-induced memory deficiency. Neurobiol. Aging 34
functional connectivity of brain regions modulating pain: a cross sectional study. (3), 791–804.
J. Alzheimers Dis. 57 (1), 71–83. Yang, S., Chang, M.C., 2019. Chronic pain: structural and functional changes in brain
Morton, H., Kshirsagar, S., Orlov, E., Bunquin, L.E., Sawant, N., Boleng, L., George, M., structures and associated negative affective states. Int. J. Mol. Sci. 20 (13).
Basu, T., Ramasubramanian, B., Pradeepkiran, J.A., Kumar, S., Vijayan, M., Yang, K.H., Kaufmann, C.N., Nafsu, R., Lifset, E.T., Nguyen, K., Sexton, M., Han, B.H.,
Reddy, A.P., Reddy, P.H., 2021. Defective mitophagy and synaptic degeneration in Kim, A., Moore, A.A., 2021. Cannabis: an emerging treatment for common symptoms
Alzheimer’s disease: focus on aging, mitochondria and synapse. Free Radic. Biol. in older adults. J. Am. Geriatr. Soc. 69 (1), 91–97.
Med. 172, 652–667. Zambelli, V.O., Gross, E.R., Chen, C.H., Gutierrez, V.P., Cury, Y., Mochly-Rosen, D.,
Mulder, J., Zilberter, M., Pasquare, S.J., Alpar, A., Schulte, G., Ferreira, S.G., Kofalvi, A., 2014. Aldehyde dehydrogenase-2 regulates nociception in rodent models of acute
Martin-Moreno, A.M., Keimpema, E., Tanila, H., Watanabe, M., Mackie, K., inflammatory pain. Sci. Transl. Med. 6 (251), 251ra118.
Hortobagyi, T., de Ceballos, M.L., Harkany, T., 2011. Molecular reorganization of Zhang, B., Gaiteri, C., Bodea, L.G., Wang, Z., McElwee, J., Podtelezhnikov, A.A.,
endocannabinoid signalling in Alzheimer’s disease. Brain 134 (Pt 4), 1041–1060. Zhang, C., Xie, T., Tran, L., Dobrin, R., Fluder, E., Clurman, B., Melquist, S.,
Narayanan, M., Suver, C., Shah, H., Mahajan, M., Gillis, T., Mysore, J.,

6
H. Blanton et al. Experimental Neurology 360 (2023) 114287

MacDonald, M.E., Lamb, J.R., Bennett, D.A., Molony, C., Stone, D.J., Gudnason, V., Zhu, X.C., Tan, L., Wang, H.F., Jiang, T., Cao, L., Wang, C., Wang, J., Tan, C.C., Meng, X.
Myers, A.J., Schadt, E.E., Neumann, H., Zhu, J., Emilsson, V., 2013. Integrated F., Yu, J.T., 2015. Rate of early onset Alzheimer’s disease: a systematic review and
systems approach identifies genetic nodes and networks in late-onset Alzheimer’s meta-analysis. Ann. Transl. Med. 3 (3), 38.
disease. Cell 153 (3), 707–720.
Zhou, R., Ji, B., Kong, Y., Qin, L., Ren, W., Guan, Y., Ni, R., 2021. PET imaging of
Neuroinflammation in Alzheimer’s disease. Front. Immunol. 12, 739130.

You might also like