OBM Neurobiology is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc. By design, the scope of OBM Neurobiology is broad, so as to reflect the multidisciplinary nature of the field of Neurobiology that interfaces biology with the fundamental and clinical neurosciences. As such, OBM Neurobiology embraces rigorous multidisciplinary investigations into the form and function of neurons and glia that make up the nervous system, either individually or in ensemble, in health or disease. OBM Neurobiology welcomes original contributions that employ a combination of molecular, cellular, systems and behavioral approaches to report novel neuroanatomical, neuropharmacological, neurophysiological and neurobehavioral findings related to the following aspects of the nervous system: Signal Transduction and Neurotransmission; Neural Circuits and Systems Neurobiology; Nervous System Development and Aging; Neurobiology of Nervous System Diseases (e.g., Developmental Brain Disorders; Neurodegenerative Disorders).

OBM Neurobiology  publishes a variety of article types (Original Research, Review, Communication, Opinion, Comment, Conference Report, Technical Note, Book Review, etc.). Although the OBM Neurobiology Editorial Board encourages authors to be succinct, there is no restriction on the length of the papers. Authors should present their results in as much detail as possible, as reviewers are encouraged to emphasize scientific rigor and reproducibility.

Publication Speed (median values for papers published in 2023): Submission to First Decision: 7.5 weeks; Submission to Acceptance: 15.9 weeks; Acceptance to Publication: 7 days (1-2 days of FREE language polishing included)

Current Issue: 2024  Archive: 2023 2022 2021 2020 2019 2018 2017
Open Access Commentary

Targeting Neuroplasticity for the Management of Pain and Agitation in Alzheimer’s Disease via Bergamot Nanotherapy

Damiana Scuteri 1,2,*, Paolo Tonin 2, Maria Tiziana Corasaniti 3, Giacinto Bagetta 1

  1. Pharmacotechnology Documentation and Transfer Unit, Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy

  2. Regional Center for Serious Brain Injuries, S. Anna Institute, Crotone, Italy

  3. Department of Health Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy

Correspondence: Damiana Scuteri

Academic Editor: Ping K. Yip

Special Issue: Neuroprotection, Neuroregeneration and Neuroplasticity

Received: July 05, 2022 | Accepted: September 05, 2022 | Published: September 14, 2022

OBM Neurobiology 2022, Volume 6, Issue 3, doi:10.21926/obm.neurobiol.2203133

Recommended citation: Scuteri D, Tonin P, Corasaniti MT, Bagetta G. Targeting Neuroplasticity for the Management of Pain and Agitation in Alzheimer’s Disease via Bergamot Nanotherapy. OBM Neurobiology 2022;6(3):12; doi:10.21926/obm.neurobiol.2203133.

© 2022 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Alzheimer’s disease (AD) accounts for 50–70% of cases of dementia worldwide and is a social burden to the affected population. Although several pathogenetic hypotheses have been proposed, evidence favoring the role of aberrant neuroplasticity in the development of the neuropsychiatric symptoms associated with dementia is increasing. Specifically, agitation is resistant to treatment and affects the quality of life, also because of the lack of safe and effective treatment for AD. Alterations in pain processing due to plastic modifications occur during aging and neurodegeneration. Up to 80% of AD patients have chronic pain due to age-related comorbidities that are misdiagnosed and remain unattended due to a lack of self-reporting because of communication hindrance, which also contributes to the development of agitation. Here, we reported a strategy to target altered neuroplasticity for treating pain and agitation by applying bergamot essential oil with evidence for in-vivo analgesic effects on neuropathic and inflammatory pain preclinical models. Bergamot was engineered in a nanotechnology delivery system, NanoBEO, which provides the opportunity to investigate its efficacy in the NCT04321889 randomized, double-blind, placebo-controlled clinical trial BRAINAID. This trial can provide a rational basis for safe and effective treatment to alleviate agitation and pain, thus improving the quality of life of people suffering from AD.

Keywords

Neuroplasticity; Alzheimer’s disease; dementia; agitation; neuropsychiatric symptoms; pain; bergamot essential oil; NanoBEO; BRAINAID

1. Mechanisms of Neurodegeneration in Dementia

Dementia is an umbrella term and includes neurological diseases characterized by memory loss and cognitive impairment. It mainly affects people over 65 years old. Alzheimer’s disease (AD) accounts for 50–70% of cases of dementia worldwide among dementia with Lewy bodies, vascular dementia, frontotemporal dementia, and mixed dementia [1]. The prevalence of AD is increasing, with 55 million people affected and around 41 million people undiagnosed [2]. AD is estimated to affect 131 million patients globally by 2050 [3]. During the pandemic, the risk of death among AD patients increased, which further indicates its severity [4,5]. Altered mitochondria can perform the activity of the inflammasome and the signaling of pro-interleukin (IL)-1β and pro-IL-18 [6]. The immune system is involved in the pathophysiology of AD [7], since AD is preceded by brain hypometabolism, oxidative stress, and accumulation of dysfunctional mitochondria from impaired mitophagy, causing aberrant inflammatory responses [6]. Moreover, aging is associated with brain inflammation and priming of microglia through IL-1β, tumor necrosis factor (TNF)-α, IL-4, IL-6, IL-9, IL-12, and IL-23, which also affect the levels of neurotrophins [8]. Proinflammatory cytokines are associated with cognitive decline in AD [9]. The risk factors that increase the susceptibility to the development of sporadic AD include age and mutations of apolipoprotein E4 (APOE4), along with mutations of other genes (e.g., apolipoprotein J and receptor 1 of complement component (3b/4b)), while amyloid precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2) cause familial AD [6,10,11,12,13]. The exact etiology of AD is not known. The available treatment methods are not effective, and better therapeutic options are necessary, although aducanumab was recently approved for treating AD [14].

2. Pain and Neuropsychiatric Symptoms in AD

Multifactorial neuropsychiatric symptoms are the most challenging symptoms associated with dementia [15]. They compromise the quality of life of the patients and are often related, at least partly, to misdiagnosed and underestimated pain, besides cognitive decline [16,17]. These symptoms fluctuate and occur in 97% of AD cases. [18]. Patients develop at least one neuropsychiatric symptom, and up to 85% of them show the symptoms within five years of diagnosis [18]. Aberrant neuroplasticity is associated with chronic pain [19]. Cognitive functions and behavioral disturbances in dementia might improve after non-pharmacological treatment; non-pharmacological treatment is associated with modifications in brain activity and indicates the role of neuroplasticity in the effectiveness of this intervention [20]. Also, pain-induced neuroplasticity is related to the risk of AD (Figure 1) [21]. Around 20% of community patients [22,23] and up to 60% of patients in nursing homes [23,24] suffer from agitation, which is an extremely intractable neuropsychiatric symptom and is associated with unrelieved pain [25,26]. The other common neuropsychiatric symptoms associated with AD, as assessed by the European Alzheimer's disease consortium (EADC), include apathy, depression, anxiety, irritability, hallucinations, aberrant motor behavior, delusions, disinhibition, elation, and appetite and sleep disturbances [27]. These symptoms are based on psychosis, psychomotor factors, mood liability factors, and instinctual factors, which can be grouped into different behavioral syndromes [27]. Agitation includes inappropriate verbal, vocal, or motor activity and might be aggressive, inappropriate in frequency, or related to the social context [28]. With age, AD patients are more likely to experience chronic pain due to musculoskeletal pain, which is the leading cause of disability among the elderly [29,30], rheumatic conditions [31,32,33], neuropathies with up to 60% prevalence, such as diabetes [34] and shingles due to herpes Zoster infection [35] or spinal surgery [36], injury [37], and stroke [38]. Unfortunately, people suffering from AD who receive pharmacological treatment for pain are fewer than their peers with intact cognitive abilities in the general population of the elderly [39]. A one-year retrospective cross-sectional analysis showed that pain is highly prevalent in all types of dementia, and patients who experience pain develop more neuropsychiatric symptoms [40], confirming the finding that agitation is associated with pain and can be reduced by analgesia [15,41,42]. Our group found that the diagnosis and treatment of AD patients are poor; these patients also have limited access to the treatment of chronic pain, especially, neuropathic pain. They are often administered off-label antipsychotics and antidepressants for the management of neuropsychiatric symptoms [43,44,45]. This is partly due to communication hindrance and inefficient self-reporting of pain due to cognitive decline. This might spread the misconception that patients suffering from dementia feel less pain than their cognitively intact counterparts [25]. These patients might be administered unnecessary antipsychotics, antidepressants, and benzodiazepines, which might be harmful as these medications have cerebrocardiovascular side effects and anti-cholinergic properties, which might cause further cognitive decline.

Click to view original image

Figure 1 A possible mechanism of chronic pain induced in Alzheimer’s disease pathogenesis by the dysfunction of the locus coeruleus (LC)-noradrenaline (NE) system and microglial neuroinflammation. Chronic pain induces pathological activation of the LC-NE system and increases NE release in brain areas, such as the prefrontal cortex and hippocampus, which might be a mechanism of chronic pain-induced microglial proinflammatory activation. Proinflammatory activation might exacerbate AD pathogenesis by decreasing Aβ phagocytosis, increasing tau seeding, and promoting the loss of synaptic function and cytokine-induced neuron death in these brain regions. Reproduced with permission from [21].

3. Management of Pain and Neuropsychiatric Symptoms Through Neuroplasticity

The pharmacological treatment of neuropsychiatric symptoms includes the use of atypical antipsychotics, such as quetiapine and aripiprazole. Among these drugs, risperidone is only approved for up to 6–12 weeks of administration because it increases the risk of death due to cerebrovascular accidents [46]. Several pathological alterations occur in neurodegeneration, affecting the cholinergic nucleus basalis [47] and the areas involved in pain modulation, such as the periaqueductal gray [48] and the locus coeruleus [47], associated with the descending inhibitory pain pathway. The level of expression of 5-HT2A receptors was also lower in the frontal and temporal cortices of the post-mortem brain in severe AD [49]. The T102C polymorphism of the 5-HT2A receptor gene is a possible risk factor for the development of neuropsychiatric symptoms [50,51]. Neurodegeneration and aging not only alter pain processing [52] but also enhance neuronal vulnerability and aberrant responsiveness to stress pathways [53,54] because of abnormal neuronal and synaptic plasticity that occur in depression and AD through inflammaging [55]. Aging and neurodegeneration induce primed microglia to produce proinflammatory cytokines [56,57] and decrease the levels of brain-derived neurotrophic factor (BDNF) responsible for neurogenesis [58]. Primed glia alters glutamatergic neurotransmission [59], which is fundamental in pain and dementia, through perisynaptic excitatory amino acid transporters (EAATs) [60,61]. A study on rats that were subjected to arthritic pain showed that the presynaptic group III metabotropic glutamate receptors (mGluRs) are involved in the synaptic plasticity of the amygdala [62]. The central nucleus of the amygdala (CeA) is known as the nociceptive amygdala since it receives nociceptive inputs from the spinal cord, brainstem, thalamus, and cortex. The CeA integrates the cognitive and conscious perception of painful stimuli in these areas [63]. Altered neuroplasticity is affected by the mechanism of action of the glutamatergic modulator ketamine, which has fast antidepressant effects [64]. A single administration of the N-methyl-D-aspartate (NMDA) antagonist ketamine (and its S-enantiomer approved by the FDA for resistant depression) can induce fast-onset antidepressant action that lasts two weeks, and the mechanism might involve the synthesis of BDNF [65,66,67]. Therefore, targeting glutamatergic neurotransmission can modulate neuroplasticity, which affects pain processing and neuropsychiatric symptoms, such as depression.

4. Novel Therapeutic Approaches for Pain and Neuropsychiatric Symptoms

The treatments administered currently, such as the use of acetylcholinesterase inhibitors (AChEI) and memantine, are not proven to effectively treat neuropsychiatric symptoms [68]. Specifically, memantine might only delay the onset of agitation [69]. The clinical trials in which psychotropic drugs were administered to treat agitation failed to offer an effective and safe strategy for treating the spectrum of neuropsychiatric symptoms. Complementary therapies are efficacious in the management of these symptoms, but strong evidence in clinical trials is lacking due to methodological biases [70,71]. Various novel non-pharmacological treatment strategies might be used, including music therapy, light therapy, technology-assisted therapy, exercise-based therapy, Snoezelen therapy, positive image therapy, and animal-assisted therapy [72]. These interventions might satisfy unmet needs, reduce overreactions, and increase functional connectivity. Here, we targeted altered neuroplasticity for pain and agitation treatment in AD by administering bergamot essential oil (BEO), which has strong analgesic effects in vivo on neuropathic and inflammatory pain preclinical models [73] and probably does not interact with other drugs [74]. Microdialysis experiments and studies on synaptosomes have highlighted its capability to induce glutamate release through exocytosis or via a Ca2+-independent carrier-mediated process depending on its concentration [75]. This is fundamental since aberrant glutamatergic neurotransmission has been experimentally shown to influence pain and related behavioral symptoms [76,77]. Moreover, BEO does not have sedative actions [78,79], and this is fundamental for AD patients. Finally, BEO and its fractions can be effectively administered through inhalation and the transdermal route [80,81]. The only reported toxicity of BEO consists of phototoxicity caused by furocoumarins [82]. The nonvolatile residue of BEO contains about 0.2% bergapten, which is responsible for the phototoxicity of BEO. Therefore, a bergapten-free extract of the essence (BEO-BF) was encapsulated in a nanotechnology delivery system called NanoBEO [83]. NanoBEO was added to a cream formulation for transdermal administration. The cream consists of solid lipid nanoparticles enriched with α-tococopherylstearate to confer anti-oxidant properties, in which BEO-BF was encapsulated. This nanotechnology delivery system is able to prevent the methodological biases of the clinical trials conducted in aromatherapy up to now. NanoBEO can entrap the smell allowing masking in clinical trials, prevent components from degrading, permit titration of the dose, and can be applied through an airless dispenser affording feasibility of administration [84]. NanoBEO (recently patented (EP 4003294)) retains the antinociceptive and anti-allodynic actions of BEO, to which it adds efficacy to scratching behavior, which is a typical neuropsychiatric symptom [83]. Its efficacy and safety in treating agitation and pain in over 65 patients affected by severe AD will be investigated in the NCT04321889 randomized, double-blind, placebo-controlled clinical trial BRAINAID [85]. The primary endpoint is represented by the reduction of agitation assessed through the Cohen-Mansfield agitation inventory (CMAI), according to the clinical trials for reduction of agitation in AD [86]. Adequate pain assessment in non-communicative AD patients often prevents the administration of appropriate treatment. In the NCT04321889 trial, pain assessment will be performed using the recently translated, adapted, and validated scale in the Italian setting, Italian Mobilization-Observation-Behavior-Intensity-Dementia (I-MOBID2), for non-verbal patients with severe dementia [87]. This method elucidates even concealed musculoskeletal and visceral pain [88]. This study can provide valuable insights into this population which is usually excluded from clinical trials [89]. Patients suffering from AD are often excluded from clinical trials that assess the efficacy of therapy for painful conditions, such as migraine [90,91,92]. This prevents researchers from determining the best therapeutic options [93], which also occurs in the case of polypharmacy [94].

5. Conclusions

The development of neuropsychiatric syndromes, in general, and agitation, in particular, is challenging for patients suffering from AD. An effective and safe treatment is unavailable. Misdiagnosed and inappropriately treated pain related to the development of neuropsychiatric symptoms need to be targeted for managing these resistant symptoms. The glutamatergic transmission is closely associated with neuroplasticity of pain processing and with the mechanism of action of pharmacological and non-pharmacological treatments, which affect neuropsychiatric symptoms. Therefore, an essential oil with analgesic activity and the ability to modulate glutamatergic neurotransmission might be used for treating AD. Here, we highlighted BEO as it has antinociceptive and anti-allodynic properties related to glutamate modulation. After removing furocoumarins to avoid phototoxicity, BEO was engineered in a nanotechnology delivery system to prevent the methodological biases of clinical trials that use aromatherapy. The efficacy and safety of the nanotechnology delivery system NanoBEO in the treatment of agitation and pain were is object of investigation of the NCT04321889 randomized, double-blind, placebo-controlled clinical trial BRAINAID.

Author Contributions

Conceptualization: D.S., P.T., M.T.C., G.B. All Authors have read and agreed to the final version of the manuscript.

Funding

This research is coordinated by D.S. and received partial financial support from: (1) MISE “Prima Vera Azione” prot. INVITALIA 37600 21/02/2021 and (2) Phase 2 RIABEO Funding (Executive Decree n.6790 of 22/06/2022) Progetto Ingegno POR Calabria FESR 2014/2020—Azione 1 1 5—Sostegno all’Avanzamento tecnologico delle Imprese Attraverso il Finanziamento di Linee Pilota e Azioni di Validazione Precoce di Prodotti e di Dimostrazione su Larga Scala (DDG N. 12814 DEL 17/10/2019).

Competing Interests

The authors have declared that no competing interests exist.

References

  1. Winblad B, Amouyel P, Andrieu S, Ballard C, Brayne C, Brodaty H, et al. Defeating Alzheimer's disease and other dementias: A priority for European science and society. Lancet Neurol. 2016; 15: 455-532. [CrossRef]
  2. Gauthier S, Rosa-Neto P, Morais JA, Webster C. World Alzheimer Report 2021: Journey through the diagnosis of dementia. London: Alzheimer’s Disease International; 2021.
  3. Prince MJ, Wimo A, Guerchet MM, Ali GC, Wu YT, Prina M. World Alzheimer Report 2015: The global impact of dementia: An analysis of prevalence, incidence, cost and trends. London: Alzheimer’s Disease International; 2015.
  4. Scuteri D, Matamala-Gomez M, Bottiroli S, Corasaniti MT, De Icco R, Bagetta G, et al. Pain assessment and treatment in dementia at the time of coronavirus disease COVID-19. Front Neurol. 2020; 11: 890. [CrossRef]
  5. Scuteri D, Contrada M, Tonin P, Corasaniti MT, Nicotera P, Bagetta G. Dementia and COVID-19: A case report and literature review on pain management. Pharmaceuticals (Basel). 2022; 15: 199. [CrossRef]
  6. Onyango IG. Modulation of mitochondrial bioenergetics as a therapeutic strategy in Alzheimer's disease. Neural Regen Res. 2018; 13: 19-25. [CrossRef]
  7. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015; 14: 388-405. [CrossRef]
  8. Scheiblich H, Trombly M, Ramirez A, Heneka MT. Neuroimmune connections in aging and neurodegenerative diseases. Trends Immunol. 2020; 41: 300-312. [CrossRef]
  9. Taipa R, das Neves SP, Sousa AL, Fernandes J, Pinto C, Correia AP, et al. Proinflammatory and anti-inflammatory cytokines in the CSF of patients with Alzheimer's disease and their correlation with cognitive decline. Neurobiol Aging. 2019; 76: 125-132. [CrossRef]
  10. Arun S, Liu L, Donmez G. Mitochondrial biology and neurological diseases. Curr Neuropharmacol. 2016; 14: 143-154. [CrossRef]
  11. Gibson GE, Shi Q. A mitocentric view of Alzheimer's disease suggests multi-faceted treatments. J Alzheimers Dis. 2010; 20 Suppl 2: S591-607. [CrossRef]
  12. Chen JX, Yan SS. Role of mitochondrial amyloid-beta in Alzheimer's disease. J Alzheimers Dis. 2010; 20 Suppl 2: S569-578. [CrossRef]
  13. Calsolaro V, Edison P. Alterations in glucose metabolism in Alzheimer's disease. Recent Pat Endocr Metab Immune Drug Discov. 2016; 10: 31-39. [CrossRef]
  14. Dunn B, Stein P, Cavazzoni P. Approval of aducanumab for Alzheimer disease–The FDA's perspective. JAMA Intern Med. 2021; 181: 1276-1278. [CrossRef]
  15. Husebo BS, Ballard C, Sandvik R, Nilsen OB, Aarsland D. Efficacy of treating pain to reduce behavioural disturbances in residents of nursing homes with dementia: Cluster randomised clinical trial. BMJ. 2011; 343: d4065. [CrossRef]
  16. Sengstaken EA, King SA. The problems of pain and its detection among geriatric nursing home residents. J Am Geriatr Soc. 1993; 41: 541-544. [CrossRef]
  17. Sampson EL, White N, Lord K, Leurent B, Vickerstaff V, Scott S, et al. Pain, agitation, and behavioural problems in people with dementia admitted to general hospital wards: A longitudinal cohort study. Pain. 2015; 156: 675-683. [CrossRef]
  18. Steinberg M, Shao H, Zandi P, Lyketsos CG, Welsh-Bohmer KA, Norton MC, et al. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: The Cache County Study. Int J Geriatr Psychiatry. 2008; 23: 170-177. [CrossRef]
  19. Sator-Katzenschlager S. Pain and neuroplasticity. Rev Méd Clín Las Condes. 2014; 25: 699-706. [CrossRef]
  20. Shigihara Y, Hoshi H, Shinada K, Okada T, Kamada H. Non-pharmacological treatment changes brain activity in patients with dementia. Sci Rep. 2020; 10: 6744. [CrossRef]
  21. Cao S, Fisher DW, Yu T, Dong H. The link between chronic pain and Alzheimer's disease. J Neuroinflammation. 2019; 16: 204. [CrossRef]
  22. Lyketsos CG, Steinberg M, Tschanz JT, Norton MC, Steffens DC, Breitner JC. Mental and behavioral disturbances in dementia: Findings from the Cache County Study on Memory in Aging. Am J Psychiatry. 2000; 157: 708-714. [CrossRef]
  23. Ballard C, Corbett A. Agitation and aggression in people with Alzheimer's disease. Curr Opin Psychiatry. 2013; 26: 252-259. [CrossRef]
  24. Margallo-Lana M, Swann A, O'Brien J, Fairbairn A, Reichelt K, Potkins D, et al. Prevalence and pharmacological management of behavioural and psychological symptoms amongst dementia sufferers living in care environments. Int J Geriatr Psychiatry. 2001; 16: 39-44. [CrossRef]
  25. Scherder E, Herr K, Pickering G, Gibson S, Benedetti F, Lautenbacher S. Pain in dementia. Pain. 2009; 145: 276-278. [CrossRef]
  26. Husebo BS, Ballard C, Aarsland D. Pain treatment of agitation in patients with dementia: A systematic review. Int J Geriatr Psychiatry. 2011; 26: 1012-1018. [CrossRef]
  27. Petrovic M, Hurt C, Collins D, Burns A, Camus V, Liperoti R, et al. Clustering of behavioural and psychological symptoms in dementia (BPSD): A European Alzheimer's disease consortium (EADC) study. Acta Clin Belg. 2007; 62: 426-432. [CrossRef]
  28. Cohen-Mansfield J, Marx MS, Rosenthal AS. Dementia and agitation in nursing home residents: How are they related? Psychol Aging. 1990; 5: 3-8. [CrossRef]
  29. Molton IR, Terrill AL. Overview of persistent pain in older adults. Am Psychol. 2014; 69: 197-207. [CrossRef]
  30. Harvey MP, Martel M, Houde F, Daguet I, Riesco E, Léonard G. Relieving chronic musculoskeletal pain in older adults using transcranial direct current stimulation: Effects on pain intensity, quality, and pain-related outcomes. Front Pain Res (Lausanne). 2022; 3: 817984. [CrossRef]
  31. Burckhardt CS. The use of the mcgill pain questionnaire in assessing arthritis pain. Pain. 1984; 19: 305-314. [CrossRef]
  32. Roche PA, Klestov AC, Heim HM. Description of stable pain in rheumatoid arthritis: A 6 year study. J Rheumatol. 2003; 30: 1733-1738.
  33. Koop SM, ten Klooster PM, Vonkeman HE, Steunebrink LM, van de Laar MA. Neuropathic-like pain features and cross-sectional associations in rheumatoid arthritis. Arthritis Res Ther. 2015; 17: 237. [CrossRef]
  34. Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care. 2011; 34: 2220-2224. [CrossRef]
  35. Devor M. Rethinking the causes of pain in herpes zoster and postherpetic neuralgia: The ectopic pacemaker hypothesis. Pain Rep. 2018; 3: e702. [CrossRef]
  36. Bajwa SJ, Haldar R. Pain management following spinal surgeries: An appraisal of the available options. J Craniovertebr Junction Spine. 2015; 6: 105-110. [CrossRef]
  37. Masri R, Keller A. Chronic pain following spinal cord injury. Adv Exp Med Biol. 2012; 760: 74-88. [CrossRef]
  38. Scuteri D, Mantovani E, Tamburin S, Sandrini G, Corasaniti MT, Bagetta G, et al. Opioids in post-stroke pain: A systematic review and meta-analysis. Front Pharmacol. 2020; 11: 587050. [CrossRef]
  39. Ballard C, Smith J, Husebo B, Aarsland D, Corbett A. The role of pain treatment in managing the behavioural and psychological symptoms of dementia (BPSD). Int J Palliat Nurs. 2011; 17: 420, 422, 424. [CrossRef]
  40. Atee M, Morris T, Macfarlane S, Cunningham C. Pain in dementia: Prevalence and association with neuropsychiatric behaviors. J Pain Symptom Manage. 2021; 61: 1215-1226. [CrossRef]
  41. Husebo BS, Vislapuu M, Cyndecka MA, Mustafa M, Patrascu M. Understanding pain and agitation through system analysis algorithms in people with dementia. A novel explorative approach by the DIGI.PAIN Study. Front Pain Res (Lausanne). 2022; 3: 847578. [CrossRef]
  42. Husebo BS, Ballard C, Fritze F, Sandvik RK, Aarsland D. Efficacy of pain treatment on mood syndrome in patients with dementia: A randomized clinical trial. Int J Geriatr Psychiatry. 2014; 29: 828-836. [CrossRef]
  43. Scuteri D, Piro B, Morrone LA, Corasaniti MT, Vulnera M, Bagetta G. The need for better access to pain treatment: Learning from drug consumption trends in the USA. Funct Neurol. 2017; 22: 229-230. [CrossRef]
  44. Scuteri D, Garreffa MR, Esposito S, Bagetta G, Naturale MD, Corasaniti MT. Evidence for accuracy of pain assessment and painkillers utilization in neuropsychiatric symptoms of dementia in Calabria region, Italy. Neural Regen Res. 2018; 13: 1619-1621. [CrossRef]
  45. Scuteri D, Vulnera M, Piro B, Bossio RB, Morrone LA, Sandrini G, et al. Pattern of treatment of behavioural and psychological symptoms of dementia and pain: Evidence on pharmacoutilization from a large real-world sample and from a centre for cognitive disturbances and dementia. Eur J Clin Pharmacol. 2021; 77: 241-249. [CrossRef]
  46. Ballard CG, Gauthier S, Cummings JL, Brodaty H, Grossberg GT, Robert P, et al. Management of agitation and aggression associated with Alzheimer disease. Nat Rev Neurol. 2009; 5: 245-255. [CrossRef]
  47. Zarow C, Lyness SA, Mortimer JA, Chui HC. Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch Neurol. 2003; 60: 337-341. [CrossRef]
  48. Parvizi J, Van Hoesen GW, Damasio A. Selective pathological changes of the periaqueductal gray matter in Alzheimer's disease. Ann Neurol. 2000; 48: 344-353. [CrossRef]
  49. Lai MK, Tsang SW, Alder JT, Keene J, Hope T, Esiri MM, et al. Loss of serotonin 5-HT2A receptors in the postmortem temporal cortex correlates with rate of cognitive decline in Alzheimer's disease. Psychopharmacology (Berl). 2005; 179: 673-677. [CrossRef]
  50. Nacmias B, Tedde A, Forleo P, Piacentini S, Guarnieri BM, Bartoli A, et al. Association between 5-HT(2A) receptor polymorphism and psychotic symptoms in Alzheimer's disease. Biol Psychiatry. 2001; 50: 472-475. [CrossRef]
  51. Holmes C, Arranz MJ, Powell JF, Collier DA, Lovestone S. 5-HT2A and 5-HT2C receptor polymorphisms and psychopathology in late onset Alzheimer's disease. Hum Mol Genet. 1998; 7: 1507-1509. [CrossRef]
  52. Scuteri D, Berliocchi L, Rombolà L, Morrone LA, Tonin P, Bagetta G, et al. Effects of aging on formalin-induced pain behavior and analgesic activity of gabapentin in C57BL/6 mice. Front Pharmacol. 2020; 11: 663. [CrossRef]
  53. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, et al. Dementia prevention, intervention, and care: 2020 report of the lancet commission. Lancet. 2020; 396: 413-446. [CrossRef]
  54. Hoffman TE, Hanneman WH, Moreno JA. Network simulations reveal molecular signatures of vulnerability to age-dependent stress and Tau accumulation. Front Mol Biosci. 2020; 7: 590045. [CrossRef]
  55. Scheiblich H, Trombly M, Ramirez A, Heneka MT. Neuroimmune connections in aging and neurodegenerative diseases. Trends Immunol. 2020; 41: 300-312. [CrossRef]
  56. Taipa R, das Neves SP, Sousa AL, Fernandes J, Pinto C, Correia AP, et al. Proinflammatory and anti-inflammatory cytokines in the CSF of patients with Alzheimer's disease and their correlation with cognitive decline. Neurobiol Aging. 2019; 76: 125-132. [CrossRef]
  57. Tejera D, Mercan D, Sanchez-Caro JM, Hanan M, Greenberg D, Soreq H, et al. Systemic inflammation impairs microglial Aβ clearance through NLRP3 inflammasome. EMBO J. 2019; 38: e101064. [CrossRef]
  58. Jones GH, Vecera CM, Pinjari OF, Machado-Vieira R. Inflammatory signaling mechanisms in bipolar disorder. J Biomed Sci. 2021; 28: 45. [CrossRef]
  59. Haroon E, Miller AH, Sanacora G. Inflammation, glutamate, and glia: A trio of trouble in mood disorders. Neuropsychopharmacology. 2017; 42: 193-215. [CrossRef]
  60. McCullumsmith RE, Sanacora G. Regulation of extrasynaptic glutamate levels as a pathophysiological mechanism in disorders of motivation and addiction. Neuropsychopharmacology. 2015; 40: 254-255. [CrossRef]
  61. Bonfiglio T, Olivero G, Merega E, Di Prisco S, Padolecchia C, Grilli M, et al. Prophylactic versus therapeutic fingolimod: Restoration of presynaptic defects in mice suffering from experimental autoimmune encephalomyelitis. PLoS One. 2017; 12: e0170825. [CrossRef]
  62. Han JS, Bird GC, Neugebauer V. Enhanced group III mGLUR-mediated inhibition of pain-related synaptic plasticity in the amygdala. Neuropharmacology. 2004; 46: 918-926. [CrossRef]
  63. Ossipov MH, Dussor GO, Porreca F. Central modulation of pain. J Clin Invest. 2010; 120: 3779-3787. [CrossRef]
  64. Kadriu B, Farmer CA, Yuan P, Park LT, Deng ZD, Moaddel R, et al. The kynurenine pathway and bipolar disorder: Intersection of the monoaminergic and glutamatergic systems and immune response. Mol Psychiatry. 2021; 26: 4085-4095. [CrossRef]
  65. Price RB, Nock MK, Charney DS, Mathew SJ. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry. 2009; 66: 522-526. [CrossRef]
  66. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, et al. Nmda receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011; 475: 91-95. [CrossRef]
  67. Kim J, Farchione T, Potter A, Chen Q, Temple R. Esketamine for treatment-resistant depression - first fda-approved antidepressant in a new class. N Engl J Med. 2019; 381: 1-4. [CrossRef]
  68. McShane R, Westby MJ, Roberts E, Minakaran N, Schneider L, Farrimond LE, et al. Memantine for dementia. Cochrane Database Syst Rev. 2019; 3: CD003154. [CrossRef]
  69. Cummings JL, Schneider E, Tariot PN, Graham SM. Behavioral effects of memantine in Alzheimer disease patients receiving donepezil treatment. Neurology. 2006; 67: 57-63. [CrossRef]
  70. Forrester LT, Maayan N, Orrell M, Spector AE, Buchan LD, Soares-Weiser K. Aromatherapy for dementia. Cochrane Database Syst Rev. 2014: CD003150. [CrossRef]
  71. Ball EL, Owen-Booth B, Gray A, Shenkin SD, Hewitt J, McCleery J. Aromatherapy for dementia. Cochrane Database Syst Rev. 2020; 8: CD003150. [CrossRef]
  72. Wang G, Albayrak A, van der Cammen TJM. A systematic review of non-pharmacological interventions for bpsd in nursing home residents with dementia: From a perspective of ergonomics. Int Psychogeriatr. 2019; 31: 1137-1149. [CrossRef]
  73. Scuteri D, Hamamura K, Sakurada T, Watanabe C, Sakurada S, Morrone LA, et al. Efficacy of essential oils in pain: A systematic review and meta-analysis of preclinical evidence. Front Pharmacol. 2021; 12: 640128. [CrossRef]
  74. Rombolà L, Scuteri D, Marilisa S, Watanabe C, Morrone LA, Bagetta G, et al. Pharmacokinetic interactions between herbal medicines and drugs: Their mechanisms and clinical relevance. Life (Basel). 2020; 10: 106. [CrossRef]
  75. Morrone LA, Rombolà L, Pelle C, Corasaniti MT, Zappettini S, Paudice P, et al. The essential oil of bergamot enhances the levels of amino acid neurotransmitters in the hippocampus of rat: Implication of monoterpene hydrocarbons. Pharmacol Res. 2007; 55: 255-262. [CrossRef]
  76. Boccella S, Marabese I, Guida F, Luongo L, Maione S, Palazzo E. The modulation of pain by metabotropic glutamate receptors 7 and 8 in the dorsal striatum. Curr Neuropharmacol. 2020; 18: 34-50. [CrossRef]
  77. Boccella S, Marabese I, Iannotta M, Belardo C, Neugebauer V, Mazzitelli M, et al. Metabotropic glutamate receptor 5 and 8 modulate the ameliorative effect of ultramicronized palmitoylethanolamide on cognitive decline associated with neuropathic pain. Int J Mol Sci. 2019; 20: 1757. [CrossRef]
  78. Rombolà L, Tridico L, Scuteri D, Sakurada T, Sakurada S, Mizoguchi H, et al. Bergamot essential oil attenuates anxiety-like behaviour in rats. Molecules. 2017; 22: 614. [CrossRef]
  79. Rombolà L, Scuteri D, Watanabe C, Sakurada S, Hamamura K, Sakurada T, et al. Role of 5-HT1A receptor in the anxiolytic-relaxant effects of bergamot essential oil in rodent. Int J Mol Sci. 2020; 21: 2597. [CrossRef]
  80. Scuteri D, Rombolà L, Crudo M, Watanabe C, Mizoguchi H, Sakurada S, et al. Preclinical characterization of antinociceptive effect of bergamot essential oil and of its fractions for rational translation in complementary therapy. Pharmaceutics. 2022; 14: 312. [CrossRef]
  81. Scuteri D, Rombolà L, Crudo M, Watanabe C, Mizoguchi H, Sakurada S, et al. Translational value of the transdermal administration of bergamot essential oil and of its fractions. Pharmaceutics. 2022; 14: 1006. [CrossRef]
  82. European Medicine Agency [EMA] SEHCoHMPH.
  83. Scuteri D, Cassano R, Trombino S, Russo R, Mizoguchi H, Watanabe C, et al. Development and translation of nanobeo, a nanotechnology-based delivery system of bergamot essential oil deprived of furocumarins, in the control of agitation in severe dementia. Pharmaceutics. 2021; 13: 379. [CrossRef]
  84. Scuteri D, Rombolà L, Hayashi T, Watanabe C, Sakurada S, Hamamura K, et al. Analgesic characteristics of NanoBEO released by an airless dispenser for the control of agitation in severe dementia. Molecules. 2022; 27: 4987. [CrossRef]
  85. Scuteri D, Sandrini G, Tamburin S, Corasaniti MT, Nicotera P, Tonin P, et al. Bergamot rehabilitation against agitation in dementia (BRAINAID): Study protocol for a randomized, double-blind, placebo-controlled trial to assess the efficacy of furocoumarin-free bergamot loaded in a nanotechnology-based delivery system of the essential oil in the treatment of agitation in elderly affected by severe dementia. Phytother Res. 2021; 35: 5333-5338. [CrossRef]
  86. Ballard CG, O'Brien JT, Reichelt K, Perry EK. Aromatherapy as a safe and effective treatment for the management of agitation in severe dementia: The results of a double-blind, placebo-controlled trial with melissa. J Clin Psychiatry. 2002; 63: 553-558. [CrossRef]
  87. Scuteri D, Contrada M, Loria T, Sturino D, Cerasa A, Tonin P, et al. Pain and agitation treatment in severe dementia patients: The need for italian mobilization-observation-behavior-intensity-dementia (I-MOBID2) pain scale translation, adaptation and validation with psychometric testing. Biomed Pharmacother. 2022; 150: 113013. [CrossRef]
  88. Scuteri D, Contrada M, Loria T, Tonin P, Sandrini G, Tamburin S, et al. Pharmacological treatment of pain and agitation in severe dementia and responsiveness to change of the italian mobilization-observation-behavior-intensity-dementia (I-MOBID2) pain scale: Study protocol. Brain Sci. 2022; 12: 573. [CrossRef]
  89. Bayer A, Tadd W. Unjustified exclusion of elderly people from studies submitted to research ethics committee for approval: Descriptive study. BMJ. 2000; 321: 992-993. [CrossRef]
  90. Scuteri D, Adornetto A, Rombolà L, Naturale MD, De Francesco AE, Esposito S, et al. Pattern of triptans use: A retrospective prescription study in Calabria, Italy. Neural Regen Res. 2020; 15: 1340-1343. [CrossRef]
  91. Scuteri D, Corasaniti MT, Tonin P, Bagetta G. Eptinezumab for the treatment of migraine. Drugs Today (Barc). 2019; 55: 695-703. [CrossRef]
  92. Scuteri D, Corasaniti MT, Tonin P, Nicotera P, Bagetta G. Role of CGRP pathway polymorphisms in migraine: A systematic review and impact on CGRP mAbs migraine therapy. J Headache Pain. 2021; 22: 87. [CrossRef]
  93. McLachlan AJ, Hilmer SN, Le Couteur DG. Variability in response to medicines in older people: Phenotypic and genotypic factors. Clin Pharmacol Ther. 2009; 85: 431-433. [CrossRef]
  94. Nørgaard A, Jensen-Dahm C, Gasse C, Hansen ES, Waldemar G. Psychotropic polypharmacy in patients with dementia: Prevalence and predictors. J Alzheimers Dis. 2017; 56: 707-716. [CrossRef]
Newsletter
Download PDF Download Citation
0 0

TOP