Open access peer-reviewed chapter

Current Therapeutic Approaches from Imidazoline and Opioid Receptors Modulators in Neuroprotection

Written By

Liliana Mititelu-Tartau, Maria Bogdan, Victor Gheorman, Liliana Foia, Ancuta Goriuc, Gabriela Rusu, Beatrice Buca, Liliana Pavel, Ana Cristofor, Cosmin-Gabriel Tartau and Gratiela Eliza Popa

Submitted: 13 May 2018 Reviewed: 10 October 2018 Published: 03 April 2019

DOI: 10.5772/intechopen.81951

From the Edited Volume

Neuroprotection

Edited by Raymond Chuen-Chung Chang and Yuen-Shan Ho

Chapter metrics overview

1,150 Chapter Downloads

View Full Metrics

Abstract

Due to brain plasticity, the nervous system is capable of manifesting behavioral variations, adapted to the influences from both external and internal environment. Multiple neurotransmitters are involved in the mediation of pathological processes at the molecular, cellular, regional, and interregional levels participating in cerebral plasticity, their intervention being responsible for various structural, functional, and behavioral disturbances. The current therapeutic strategies in neuroprotection aim at blocking on different levels, the molecular cascades of the pathophysiological mechanisms responsible for neuronal dysfunctions and ultimately for neuronal death. Different agents influencing these neurotransmitters have demonstrated beneficial effects in neurogenesis and neuroprotection, proved in experimental animal models of focal and global ischemic injuries. Serotonin, dopamine, glutamate, N-methyl-D-aspartate, and nitric oxide have been shown to play a significant role in modulating nervous system injuries. The imidazoline system is one of the important systems involved in human brain functioning. Experimental investigations have revealed the cytoprotective effects of imidazoline I2 receptor ligands against neuronal injury induced by hypoxia in experimental animals. The neuroprotective effects were also highlighted for kappa and delta receptors, whose agonists demonstrated the ability to reduce architectural lesions and to recover neuronal functions of animals with experimentally induced brain ischemia.

Keywords

  • neuroprotection
  • neurodegenerative diseases
  • ischemic stroke
  • imidazoline
  • opioids
  • nitric oxide

1. Introduction

Increase in life expectancy has led to aging of the population and consequently to an expansion of the prevalence of neurodegenerative diseases (NDDs) [1]. Neurodegeneration represents a loss of neurons and their structural components (dendrites, axons, and synapses) with a corresponding gradual atrophy in neuronal function [2].

NDDs (e.g., Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, dementia with Lewy bodies) determine cognitive and memory deterioration or alteration of the ability to move, speak, and breathe. These chronic and progressive disorders are an important cause of reduced quality of life, morbidity, caregiver burden and also, of the increase in total healthcare expenditure [3, 4, 5]. Table 1 lists the main clinical types of NDDs [6].

Clinical type Disease/disorder
Signs of progressive dementia with no other neurological signs (absent/inconspicuous)
  • Alzheimer’s disease

  • Frontotemporal dementias

  • Some cases of Lewy-body disease

  • Posterior cortical atrophy (visuospatial dementia)

Signs of progressive dementia accompanied by other neurological abnormalities
  • Huntington’s disease (chorea)

  • Lewy-body disease (Parkinsonian features)

  • Some cases of Parkinson’s disease

  • Corticobasal ganglionic degeneration

  • Cortical-striatal-spinal degeneration (Jakob’s disease)

  • Dementia-Parkinson-amyotrophic lateral sclerosis complex

  • Cerebrocerebellar degeneration

  • Familial dementia with spastic paraparesis, amyotrophy, or myoclonus

  • Polyglucosan body disease

  • Frontotemporal dementia with parkinsonism or ALS

Signs of movement disorders or other posture abnormalities
  • Parkinson’s disease

  • Multiple system atrophy

  • Essential tremor

  • Progressive supranuclear palsy

  • Dystonia musculorum deformans

  • Huntington’s disease (chorea)

  • Acanthocytosis with chorea

  • Corticobasal ganglionic degeneration

  • Lewy-body disease

  • Restricted dystonia

Signs of progressive ataxia
  • Spinocerebellar ataxias

  • Cerebellar cortical ataxias

  • Complicated hereditary and sporadic cerebellar ataxias

Signs of slowly developing muscular weakness and atrophy
  • Motor disorders with amyotrophy

  • Spastic paraplegia without amyotrophy

Sensory and sensorimotor disorders
  • Hereditary sensorimotor neuropathies

  • Pure or predominantly sensory or motor neuropathic

  • Riley-Day autonomic degeneration

Signs of progressive blindness with or without other neurological disorders
  • Pigmentary degeneration of retina

  • Stargardt’s disease

  • Age-related macular degeneration

Signs characterized by degenerative neurosensory deafness
  • Pure neurosensory deafness

  • Hereditary hearing loss with retinal diseases

  • Hereditary hearing loss with system atrophies of the nervous system

Table 1.

Neurodegenerative diseases: main clinical types.

Adapted from Ropper A, Samuels M, Klein J. Adams and Victor?s Principles of Neurology. 10th ed. McGraw-Hill Education; 2014.


Neuroprotection can be defined as a “relative preservation of neuronal structure and/or function” or as an action that aims “to prevent neuronal damage over time (either acute or chronic)” [7]. Neuroprotective action is primary if it is exerted directly on the neuron, or secondary if it appears from an activity on an intermediary that endangers neuronal function [8].

The mechanisms by which most agents with efficiency in NDDs act are not fully elucidated, requiring multiple and in-depth experimental and clinical studies.

Several experimental investigations highlight the multiple and various interrelations between adrenergic, serotoninergic, dopaminergic, glutamatergic, opioid, imidazoline systems, and the nitric oxide pathways, which may elucidate the effects of different compounds involved in the mediation of pathogenic mechanisms responsible for numerous structural, functional, and behavioral disturbances.

This chapter presents a brief overview of the most studied mechanisms related to neuroprotection and details the possibilities to pharmacologically influence through the main known neurotransmitters the pathophysiological mechanisms linked to various NDDs.

Advertisement

2. The imidazoline system

Imidazoline receptors are located not only in the mammalian central nervous system (CNS) cells but also in the peripheral nervous system [9], being involved in the mediation of various physiological processes in the body. It is currently known that there are four types of imidazoline receptors: I1, I2, I3, I4 (non I1-non I2), from which the first three have been mostly studied [10].

It has been emphasized that these receptors play an essential role in cell proliferation, regulation of adipose tissue formation, body temperature maintenance, mediation of gastrointestinal motility, neuroprotection, inflammation, nociceptive sensitivity, and some neurological or psychiatric disorders (such as depression) [11]. Moreover, it is known that these imidazoline receptor subtypes exert control over the activity of the hypothalamic-pituitary-adrenal and noradrenergic axis [12, 13].

A number of different endogenous ligands have been characterized: agmatine, the best known and largely studied, harmane and harmalane (derivatives of the beta-carboline group), and the newly discovered ribotide (acetic acid imidazole). Agmatine, the potent neurotransmitter of the imidazoline system, has an important role in the mediation of body’s response to stress, analgesia, drug addiction, and abstinence syndrome, in modulation of seizures development, and in neuroprotection [14, 15].

Endogenous agmatine is produced in response to stress (in conditions of ischemia, prolonged exposure to cold) and/or to inflammation [16]. It is assumed that agmatine is also an effective neurotransmitter, due to its concentration in the brain similar to classical neurotransmitters [17, 18].

Literature data have revealed that agmatine stimulates the activity of endothelial nitric oxide synthase [16], this effect being also proved by its level in the rat brain after cerebral ischemia [19, 20].

Along with evidence of its neuromodulatory and neuroprotective properties, there are numerous preclinical studies demonstrating the beneficial effects of exogenous administration of agmatine in depression, anxiety, hypoxic ischemia, pain, morphine tolerance, memory impairment, Parkinson’s disease, Alzheimer’s disease, epilepsy, and other related conditions with traumatic brain injuries (Figure 1) [21, 22, 23]. All these are arguments in favor of the potential of agmatine as a new pharmacological agent for the treatment of various neurological diseases and NDDs [24].

Figure 1.

Neuroprotective effects of agmatine and their first discovery.

Advertisement

3. The involvement of the imidazoline system in the mediation of cognitive functions

Electrophysiological studies involving various brain areas, performed on laboratory animals with experimentally induced cerebral alterations, have demonstrated the neurotropic effects of agmatine [25].

In vitro experimental researches have shown that activation of I2 receptors via the agmatine endogenous ligand exerts neuroprotective effects by increasing the expression of glial fibrillary acidic protein in astrocyte cultures and by inhibiting MAO activity [26, 27]. Moreover, the beneficial effects of agmatine have been observed on ischemic-hypoxic lesions, on glutamate-induced neurotoxicity by activating the imidazoline receptors [28, 29]. It was also demonstrated that agmatine administration improves learning activity and memory of rats in experimental models of Alzheimer’s disease and streptozotocin-induced type two diabetes mellitus [22, 30].

Other experimental investigations highlight the neuroprotective effect of intranasal administration of agmatine in elderly female rats, with a significant improvement of neurological status and increase of survival rate [28, 29].

The neuroprotective effects of agmatine on the morphological changes determined by repeated induced stress on medial prefrontal cortex and hippocampus of the rat were also investigated [31]. It was emphasized that under constant stress conditions, morphological alterations of the brain are associated with the reduction of endogenous agmatine levels [measured by high-performance liquid chromatography (HPLC)] and with an increase of arginine-decarboxylase level in the prefrontal cortex, hippocampus, striatum, and hypothalamus [32].

The exogenous administration of agmatine lowers brain morphological impairment, suggesting thus its neuroprotective effects against structural changes in the rat brain, under recurrent stress circumstances [32]. Moreover, elevated levels of agmatine have been evidenced in the blood, cortex, hippocampus, and hypothalamus, immediately after brain hypoxic ischemia. Other studies emphasize the neuroprotective influences of agmatine, highlighted by the increase of its brain levels, in rats subjected to prolonged cold-exposure stress conditions [33].

The neuroprotective potential of agmatine was also highlighted in the experimental model of 1-methyl-4-phenyltetrahydropyridine (MPTP)-induced Parkinson’s disease in mice [29]. The use of agmatine attenuates the loss of cellular dopamine from the black substance and repeated treatment improves short-term memory impairment induced by MPTP in elderly mice. The behavioral benefits of agmatine are associated with the decrease in MPTP-induced glutamate capture in the hippocampal area, suggesting thus its involvement in modulation of glutamate recapture, the possible mechanisms responsible for lowering glutamate extracellular levels, thereby alleviating its neurotoxicity [29].

It is known that alteration of spatial memory in Parkinson’s disease and schizophrenia is attributed to several factors, including hypofunction of glutamate and reduction of hippocampal volume [34]. Literature data report that the administration of the N-methyl-D-aspartate (NMDA) receptor antagonists (phencyclidine, also coded MK801) frequently impairs the late alternation performance in a standardized behavioral model of cognitive functions alteration similar to schizophrenia in laboratory animals [34]. The use of the glutamate/NMDA receptor antagonist phencyclidine induces a spectrum of behavioral, neurochemical, and anatomical changes, manifested by locomotor hyperactivity, motor-negative deficits, and cognition alterations (with memory impairment and visual attention) in laboratory animals. This substance was used to induce the experimental schizophrenia in laboratory animals [35, 36]. Agmatine attenuates cognitive and behavioral deficiency in rats with experimental phencyclidine-induced schizophrenic manifestations [37].

The effects of agmatine on memory alterations similar to those found in Alzheimer’s disease have been evaluated in rats; in the pathogenesis of this degenerative disorder (which causes cognitive deficits in rodents), the fragment beta amyloid Aβ2 25–35 plays an essential role. Studies have shown that agmatine significantly reduces the alterations in memory and spatial learning induced by the beta amyloid Aβ2 25–35 fragment (the neurotoxic component of beta amyloid Aβ 1–42) in various behavioral experimental models, such as: the swimming test, the radial arm maze test, and the object recognition test [38].

It has been revealed that agmatine diminishes the activation of hippocampal caspase-3 (the early indicator of neuronal apoptosis) and prevents the alteration of spatial memory induced by lipopolysaccharides, in the swimming test in rat [39], suggesting its neuroprotective effects.

The neurotropic activity of agmatine has been also evidenced in the structural and cognitive alterations after the administration of NMDA (N-methyl-D-aspartate) in rats [40]. The use of high performance liquid chromatography (HPLC) and electrochemical detection allowed highlighting that the treatment with NMDA is associated with low concentrations of monoamines (epinephrine, norepinephrine, dopamine, and serotonin) in rat PC12 cells [29, 40]. In this experimental model (swimming test), agmatine protects against NMDA-induced PC12 cell lesions, augmenting the levels of epinephrine, norepinephrine, and dopamine, but not influencing serotonin values, together with lowering intracellular Ca2+ overload. These results indicate that the neuroprotective action of agmatine may be related to NMDA-receptor modulation and/or to controlling the decrease in monoamine content and NMDA-induced intracellular Ca2+ overload [40].

Immunohistochemical studies and electrophysiological investigations performed on the brain have validated the neuroprotective actions of both imidazoline receptor antagonists idazoxan and efaroxan in rats with cerebral damages caused by the use of quinolinic acid [41], and also in mice with experimentally induced autoimmune encephalomyelitis, confirming the improvement of brain structural alterations and blood brain barrier lesions curtail [42].

A new (+)2-(ethyl-2,3-dihydrobenzofuranyl)-2-imidazoline derivative—dexefaroxan—the (+) enantiomer of efaroxan has been characterized. It has a potent and selective α2 antagonist activity, with facultative effects on cognitive functions in the passive avoidance test in rats with memory-deficiency induced by scopolamine, diazepam, or by the 2-adrenergic agonist UK 14,304.

Dexefaroxan improves the cognitive performances in the passive avoidance test, facilitates spatial memory in the Morris swimming test in rats, and increases the object recognition ability in the specific behavioral test in mice [43, 44]. It has also proved to ameliorate the animal’s memory deficits in these tests, particularly by attenuation of spontaneous memory loss, and to improve their spatial recognition ability, rather than through the acquisition skills or various other non-cognitive effects.

After subcutaneous administration of dexefaroxan, its pharmacodynamic effects persist for about 21–25 days, indicating that tolerance does not occur during prolonged treatment. Moreover, it was emphasized that dexefaroxan exerts protective effects on the spatial memory deficit caused by cortical devascularization in the Morris swimming test in rats [43].

Dexefaroxan has also been shown to exhibit neuroprotective effects on the de-vascularization-induced neurodegeneration, to ameliorate the structural changes in the hippocampus, and to remove the cognitive deficits induced by cerebral ischemia in rats [45, 46]. Its neuroprotective effects were present also in the excitotoxic lesions produced at the region of the basal magnocellular nucleus, increasing the olfactory discriminative capacity of rats, suggesting thus the possibility of its use in the treatment of memory disturbances in Alzheimer’s disease [43].

Studies performed on genetically modified animals revealed that dexefaroxan improves cognitive performance in knockout mice with Alzheimer’s disease [47].

Literature data regarding the neuroprotective action of imidazoline agonist and antagonist agents in human studies are only few, and the mechanisms involved in these effects are not completely deciphered.

Some investigations suggested that agmatine manifests protective activity against brain cell injury in different in vivo models of Parkinson’s disease, as well as in vitro studies, performed on human-derived dopaminergic neuroblastoma cell lines. It was postulated that the neuromodulatory properties of agmatine are related to the protective effects on the dopaminergic neurons, to NMDA receptor blocking, and to the decrease in oxidative stress, due to the inhibition of nitric oxide synthase (NOS) activity [48, 23].

Other clinical trials highlighted that the treatment with agmatine was associated with cytoprotective actions, in patients with spinal cord injury, proved by lessening the glial weal construction, decreasing the collagen scar zone, relieving the neuronal alterations, and recovering remyelination [49]. Moreover, the beneficial effects of agmatine have been demonstrated in various CNS lesions such as: cerebrovascular accident, brain trauma, neuropathic pain, lumbar degenerative disc disease, and different other types of neuropathy [50, 51, 52].

The administration of dexmedetomidine has also shown neuroprotective effects in humans with acute cerebral lesions [53].

In patients with dementia due to brain frontal lesions, idazoxan alleviates attentional and executive dysfunctions evoked by classical cognitive function tests [54].

Advertisement

4. The interrelation between the adrenergic and the imidazoline systems in the mediation of cognitive functions

Clonidine, both a non-specific α2 adrenergic and imidazoline receptor agonist, decreases the cognitive function alterations induced by phencyclidine and MK801, facilitating spatial memory in the radial arm maze test in rats [35], but does not influence the behavioral and cognitive deficits in the experimental NMDA-induced excitotoxic dorsal hippocampal lesions [35]. Such findings indicate that clonidine improves memory alterations caused by glutamate hypofunction, but not by hippocampal injury, implying that multiple and distinct mechanisms are involved in the development of memory disorders.

The administration of the α2 adrenergic receptor agonist clonidine or guanfacine prevents some of the behavioral effects of NMDA antagonists, proving that the monoaminergic system mediates a number of aspects of the cognitive deficit. Clonidine and guanfacine improve the lack of visual attention and spatial memory induced by phencyclidine in rats [34, 36]. It was demonstrated that low doses of clonidine recover the animal’s ability to accurately choose the object and prevent the performance deficit induced by phencyclidine. At high doses, clonidine decreases the response time and induces a lack of the choice accuracy. These results indicate that clonidine treatment can alleviate phencyclidine-induced deficit of attention and of working memory, probably by preventing some of the neurochemical and anatomical effects of this psychotomimetic drug [34].

On the other hand, the use of only the selective α2 adrenergic receptor antagonist does not impair the animal’s spatial memory, but dramatically aggravates the phencyclidine-induced memory deficit [36]. These data demonstrate that α2 adrenergic receptors mediate the inhibition of spatial memory disturbances, suggesting their important role in cognitive deficits associated to NMDA receptor hypofunction [34, 36].

The role of moxonidine (an α2 adrenergic imidazoline I1 receptor agonist) on cognitive function in rats with Huntington’s disease experimentally induced with 3-nitropropionic acid (3-NPA) was investigated in the Morris swimming test and in the elevated plus maze test. The administration of 3-NPA induces degenerative brain damage, progressive motor dysfunction, loss of grip force, emotional disturbances, weight loss, anxiety, and impairment of learning activity and memory. An increase in cerebral acetylcholinesterase level, enhancement of oxidative stress, and impairment of the activity of mitochondrial enzyme complexes I, II, and IV were also noted [28]. The treatment with moxonidine resulted in the alleviation of disturbances caused on animal weight, motor activity, gripping ability, anxiety, impairment of learning ability and memory, and biochemical disturbances, thus indicating that substances modifying the activity of I1 receptors may be potential pharmacological agents for the treatment of degenerative brain disorders [55].

The effects of clonidine have also been evaluated in mice with subacute brain ischemia obtained after permanent ligation of common carotid arteries. The subsequent brain damages consisted of expansion of cerebral infarction areas, assessed by computed tomography scans. This experimentally induced chronic cerebral hypoperfusion was associated with a significant impairment of animal’s learning ability and memory in the Morris swimming test [25, 28]. Subacute treatment with clonidine for 7 days increases the expression of neuronal nuclei, glutamic acid-decarboxylase-67, and gamma-aminobutyric acid (GABA) B receptor (GABAB1) in hippocampal subregion cornu amonis (CA1) but does not influence the level of these elements in the hippocampal area CA3, nor in the dentate gyrus. These data support the idea that clonidine exerts neuroprotective effects on chronic cerebral ischemic lesions, by regulating GABAB1 receptors and the activity of glutamic acid-decarboxylase-67 [25].

Additionally, the decrease in superoxide dismutase (SOD), catalase (CAT), and glutathione levels as well as the increase of both malondialdehyde (MDA) level and cerebral acetylcholinesterase activity were noted in animals with brain ischemic lesions [28].

Both moxonidine and clonidine have shown a decrease in histopathological changes, oxidative stress, central cholinesterase activity, as well as a reduction in memory disturbances and learning deficits in mice with vascular dementia induced by subacute ischemia after permanent bilateral cerebral artery ligation [28, 40].

In vitro cell culture studies from the rat frontal cortex with glutamate-induced neurotoxicity revealed the partial neuroprotective effects of moxonidine, with a significant decrease in the number of dead cells [26]. Moxonidine has shown beneficial effects on cerebral spasm in an experimental rabbit model of subarachnoid hemorrhage [40, 56].

Advertisement

5. The interrelations between the imidazoline system and the oxidative stress in the mediation of cognitive functions

Different pathological conditions of the body, as well as the physiological process of aging, can cause cognitive impairment and free oxygen radicals production, being responsible for abnormal functioning and cell death. Subsequently, a new idea has emerged claiming that nitric oxide (NO), along with the free radicals, plays a key role in the aging process, due to neurotoxic effects on the brain exerted by its excessive levels [57]. Nitric oxide is generated from L-arginine under the action of nitric oxide synthase (NOS). The three isoforms of NOS have different roles in the body: neuronal NOS (nNOS) is responsible for synaptic plasticity, learning and memory processes; endothelial NOS (eNOS) provides stabilization and regulation of vascular micro-environment and contributes to neuroplasticity [58]; and inducible NOS (iNOS) is involved in various pathophysiological conditions [57].

Numerous experimental researches reveal that NOS activity is significantly elevated in the brain of elderly rats, being associated with existing cognitive alterations [57, 59]. Mediated by the competitive inhibition of nNOS and iNOS, and correlated with the stimulation of NOS, agmatine contributes to the improvement of cognitive functions [19, 60, 61], while exhibiting neuroprotective effects [38, 39].

Literature data have shown that agmatine eliminates neuroinflammation and lipopolysaccharide-induced memory impairment (which is known to stimulate iNOS activity and, implicitly, the NO production) in laboratory animals. It prevents cognitive alterations, probably as a result of inhibition of iNOS activity [39]. Other researchers have disproved these results by showing that agmatine can cause cognitive impairment due to the inhibition of NMDA receptors and of NO, important elements in the modulation of learning and memory processes [62, 63].

On the other hand, it is known that the central cholinergic system plays a crucial role in the mediation of cognitive functions. Cognitive deficits have been induced in laboratory animals by using an anticholinergic agent, scopolamine, its administration producing a significant reduction in NOS activity, and an increase in arginase activity, of L-ornithine and putrescine levels in the hippocampus [51]. It has been observed that agmatine eliminates the scopolamine-induced alterations of memory and learning capacity [64, 65].

Although glutamatergic activity is required for cognitive processes, it is assumed that the increase of glutamate levels or of NMDA activity would also be responsible for the scopolamine-induced cognitive disturbances [66].

Knowing that agmatine blocks NMDA receptors and also interferes with the pathways of NO, NOS, and L-arginine, it was assumed that the removal of scopolamine-induced cognitive deficits can be attributed to its modulating effect on NO/NOS activity, on L-arginine, and also to the antagonization of NMDA receptors, with subsequent suppression of excessive glutamatergic activity [61, 65].

Abnormal release and disturbances of neuromodulatory activities due to variation in cerebral agmatine levels may be correlated to different CNS diseases (such as schizophrenia). Interactions of agmatine with other central neurotransmitter systems (such as glutamatergic and nitrergic) appear to be particularly important in the pathophysiological mechanisms of CNS disorders associated with brain damage and cognitive functions deficit.

Advertisement

6. The opioid system

Neurodegeneration can be caused by chronic disease progression or by acute injury (cerebral ischemia—stroke or trauma) [67]. Ischemic stroke represents a vascular ailment with neurological consequences produced by the obstruction of the arteries in a part of the brain, therefore by blood supply privation [68]. Stroke can determine long-term neurological and psychiatric impairments, its therapy being focused on confining secondary injury processes [67].

In a recent review article, Chamorro presented that ischemic stroke is “the first cause of permanent disability in adult people, the second single most frequent cause of death for people older than 60 years, the second most common cause of dementia, representing approximately 3% to 7% of the total health-care expenditure in high income countries” [69].

A superpose of pathologies in different neurological disorders was proposed since NDDs and ischemic stroke are frequently concomitant, hence the neuroprotective therapy could be similar [70].

Opioids are substances with morphine-like action binding to specific opioid receptors (ORs). In the early 1990s, three important opioid receptor families [μ (MOR), κ (KOR), and δ (DOR)] were identified, and in 1994 another opioid receptor was discovered [nociceptin, orphanin FQ receptor (NOP), or the opioid receptor-like orphan receptor (ORL)]. ORs are found in the nervous system, lungs, heart, liver, and gastrointestinal and reproductive tracts. They have been intensively studied and it was emphasized that they not only are related to antinociceptive action, but also have a role in cell proliferation, ionic homeostasis, emotional response, immune function, epileptic seizures, feeding, obesity, respiratory and cardiovascular control, hibernation, and neuroprotection [71, 72].

In the last decades, researches have pointed out that the opioid system can be promising to get neuroprotective treatment in the event of stroke, through OR agonists at lower doses, to avoid tolerance and/or physical dependency. DOR agonists followed by KOR agonists have revealed the most intense neuroprotective efficacy [64]. Major OR agonists tested for neuroprotection are listed in Table 2 [71, 72].

Opioid receptor Agonist
δ DADLE [D-Ala2, D-Leu5]-enkephalin
DPDPE (D-Pen2,D-Pen5)-enkephalin
SNC80
Tan-67
Remifentanil
κ BRL 52537
CI-977
GR89696
Salvinorin A
U-50,488H
Dynorphin
μ DAMGO [D-Ala2,N-MePhe4,Gly-ol]-enkephalin
Endorphin 1 and 2 (EM 1/2),
Morphine

Table 2.

Opioid receptors and their agonists tested for neuroprotective action.

DOR activation is beneficial against ischemic, hypoxic, and excitotoxic injuries [73]; recent studies promote DOR and especially DADLE (an analog of endogenous delta-opioid enkephalin) as promising targets for treating NDDs like stroke and PD [74, 75, 76].

DADLE alleviates apoptotic pathways, supports not only cell survival of peripheral organs (such as lung, heart, kidney, and liver) but also neuronal survival, and protects neurons and glial cells from ischemia-induced cell death [76, 77, 78].

In a cellular model of PD, DADLE administration augmented cell survivability with concurrent downregulation of the unfolded protein response stress sensors and protein aggregates [79].

Findings from a rat middle cerebral artery occlusion (MCAO) stroke model proposed that neuroprotection of DADLE treatment was based on the activation of PI3K-Akt pathway by reducing nerve cell apoptosis [80].

Non-selective opioid receptor agonists were also tested: LYS739 (fluorinated enkephalin-fentanyl derivative) and the most promising compound—biphalin—which proved to be effective both in vitro and in vivo stroke models [72].

The latter is a dimeric enkephalin analog (Tyr-D-Ala-Gly-Phe-NH-)2 with high potency and affinity for MOR and DOR and low affinity for KOR. Biphalin crosses blood-brain barrier reaching spinal and supraspinal sites expressing OR and produces less physical dependence and tolerance compared to morphine [72, 81, 82, 83, 84].

Different studies using a mouse MCAO stroke model reported that biphalin reduced brain edema and infarction, ROS production, and NMDA-induced excitotoxicity. It also increased locomotor activities and neurological score after stroke resembled to saline-treated animals [72, 81, 85, 86]. Biphalin notably diminished penumbral expression of Na+, K+, 2Cl cotransporter (NKCC), and the translocation of the conventional isoforms of protein kinase C [81].

It has been hypothesized that biphalin’s neuroprotective effects are more intense compared to subtype-selective agonists due to concomitant activation of the three types of OR [72, 85].

Advertisement

7. Conclusion

Various pharmacological substances influencing the pathways of the main neurotransmitters have confirmed valuable effects in neurogenesis and neuroprotection, being validated in different in vitro researches and in vivo experimental animal models of limited or extensive ischemic brain lesions.

Deciphering the roles of the neurotransmitters in central nervous system activity other than the signaling function will represent a starting point to deepen the knowledge about the complex mechanisms of the brain functions and to obtain new agents useful for protection of ischemic neurons and for preventing their irreversible damage.

References

  1. 1. Jaafaru MS, Abd Karim NA, Enas ME, Rollin P, Mazzon E, Abdull Razis AF. Protective effect of glucosinolates hydrolytic products in neurodegenerative diseases (NDDs). Nutrients. 2018;10(5), 580:1-15
  2. 2. Cummings J. Disease modification and neuroprotection in neurodegenerative disorders. Translational Neurodegeneration. 2017;6:25
  3. 3. Lauterbach EC, Victoroff J, Coburn KL, Shillcutt SD, Doonan SM, Mendez MF. Psychopharmacological neuroprotection in neurodegenerative disease: Assessing the preclinical data. The Journal of Neuropsychiatry and Clinical Neurosciences. 2010;22:8-18
  4. 4. Gitler AD, Dhillon P, Shorter J. Neurodegenerative disease: Models, mechanisms, and a new hope. Disease Models & Mechanisms. 2017;10:499-502
  5. 5. Gao HM, Hong JS. Why neurodegenerative diseases are progressive: Uncontrolled inflammation drives disease progression. Trends in Immunology. 2008;29:357-365
  6. 6. Ropper A, Samuels M, Klein J. Adams and Victor’s Principles of Neurology. 10th ed. Boston: McGraw-Hill Education; 2014. pp. 1060-1131
  7. 7. Wiendl H, Elger C, Förstl H, Hartung H-P, Oertel W, Reichmann H, et al. Gaps between aims and achievements in therapeutic modification of neuronal damage ("Neuroprotection"). Neurotherapeutics. 2015;12:449-454
  8. 8. Cummings J, Fox N. Defining disease modifying therapy for Alzheimer's disease. The Journal of Prevention of Alzheimer's Disease. 2017;4:109-115
  9. 9. Ferrari F, Fiorentino S, Mennuni L, Garofalo P, Letari O, Mandelli S, et al. Analgesic efficacy of CR4056, a novel imidazoline-2 receptor ligand, in rat models of inflammatory and neuropathic pain. Journal of Pain Research. 2011;4:111-125
  10. 10. Nechifor M. Imidazoline receptors—Normal and pathological factors. Revista Medico-Chirurgicala a Societatii de Medici Si Naturalisti din Iasi. 2001;105:438-443
  11. 11. Dardonville C, Rozas I, Callado LF, Meana JJ. I(2)-imidazoline binding site affinity of a structurally different type of ligands. Bioorganic & Medicinal Chemistry. 2002;10:1525-1533
  12. 12. Pypendop BH, Silverstein D, Hopper K. 2 agonists and antagonists. In: Small Animal Critical Care Medicine. 2nd ed. Missouri: Saunders Inc.; 2015. pp. 866-871.Ch 165. SRC - BaiduScholar
  13. 13. Thorn DA, An X-F, Zhang Y, Pigini M, Li J-X. Characterization of the hypothermic effects of imidazoline I2 receptor agonists in rats. British Journal of Pharmacology. 2012;166:1936-1945
  14. 14. Smith KL, Jessop DS, Finn DP. Modulation of stress by imidazoline binding sites: Implications for psychiatric disorders. Stress (Amsterdam, Netherlands). 2009;12:97-114
  15. 15. Ciubotariu D, Nechifor M. Involvement of imidazoline system in drug addiction. Revista Medico-Chirurgicala a Societatii de Medici Si Naturalisti din Iasi. 2012;116:1118-1122
  16. 16. Halaris A, Plietz J. Agmatine: Metabolic pathway and spectrum of activity in brain. CNS Drugs. 2007;21:885-900
  17. 17. Haenisch B, von Kügelgen I, Bönisch H, Göthert M, Sauerbruch T, Schepke M, et al. Regulatory mechanisms underlying agmatine homeostasis in humans. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2008;295:G1104-G1110
  18. 18. Reis DJ, Regunathan S. Is agmatine a novel neurotransmitter in brain? Trends in Pharmacological Sciences. 2000;21:187-193
  19. 19. Mun CH, Lee WT, Park KA, Lee JE. Regulation of endothelial nitric oxide synthase by agmatine after transient global cerebral ischemia in rat brain. Anatomy & Cell Biology. 2010;43:230-240
  20. 20. Nissim I, Horyn O, Daikhin Y, Chen P, Li C, Wehrli SL, et al. The molecular and metabolic influence of long term agmatine consumption. The Journal of Biological Chemistry. 2014;289:9710-9729
  21. 21. Mancinelli F, Ragonese F, Cataldi S, Ceccarini MR, Iannitti RG, Arcuri C, et al. Inhibitory effects of agmatine on monoamine oxidase (MAO) activity: Reconciling the discrepancies. The EuroBiotech Journal. 2018;2:121-127
  22. 22. Sirvanci-Yalabik M, Sehirli AO, Utkan T, Aricioglu F. Agmatine, a metabolite of arginine, improves learning and memory in streptozotocin-induced Alzheimer's disease model in rats. Bulletin of Clinical Psychopharmacology. 2016;26(4):342-354
  23. 23. Neis VB, Rosa PB, Olescowicz G, Rodrigues ALS. Therapeutic potential of agmatine for CNS disorders. Neurochemistry International. 2017;108:318e331
  24. 24. Moretti M, Neis VB, Matheus FC, Cunha MP, Rosa PB, Ribeiro CM, et al. Effects of agmatine on depressive-like behavior induced by intracerebroventricular administration of 1-methyl-4-phenylpyridinium (MPP(+)). Neurotoxicity Research. 2015;28:222-231
  25. 25. Lu Y, Li C, Zhou M, Luo P, Huang P, Tan J, et al. Clonidine ameliorates cognitive impairment induced by chronic cerebral hypoperfusion via up-regulation of the GABABR1 and GAD67 in hippocampal CA1 in rats. Pharmacology, Biochemistry, and Behavior. 2015;132:96-102
  26. 26. Bakuridze K, Savli E, Gongadze N, Baş DB, Gepdiremen A. Protection in glutamate-induced neurotoxicity by imidazoline receptor agonist moxonidine. The International Journal of Neuroscience. 2009;119:1705-1717
  27. 27. Head GA, Mayorov DN. Imidazoline receptors, novel agents and therapeutic potential. Cardiovascular & Hematological Agents in Medicinal Chemistry. 2006;4:17-32
  28. 28. Gupta S, Sharma B. Pharmacological modulation of I(1)-imidazoline and α2-adrenoceptors in sub acute brain ischemia induced vascular dementia. European Journal of Pharmacology. 2014;723:80-90
  29. 29. Matheus FC, Aguiar AS, Castro AA, Villarinho JG, Ferreira J, Figueiredo CP, et al. Neuroprotective effects of agmatine in mice infused with a single intranasal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Behavioural Brain Research. 2012;235:263-272
  30. 30. Kang S, Kim C-H, Jung H, Kim E, Song H-T, Lee JE. Agmatine ameliorates typ. 2 diabetes induced-Alzheimer's disease-like alterations in high-fat diet-fed mice via reactivation of blunted insulin signalling. Neuropharmacology. 2017;113:467-479
  31. 31. Aricioglu F, Ercil E, Dulger G. Agmatine inhibits naloxone-induced contractions in morphine-dependent Guinea pig ileum. Annals of the New York Academy of Sciences. 2003;1009:147-151
  32. 32. Zhu M-Y, Wang W-P, Cai Z-W, Regunathan S, Ordway G. Exogenous agmatine has neuroprotective effects against restraint-induced structural changes in the rat brain. The European Journal of Neuroscience. 2008;27:1320-1332
  33. 33. Aricioglu F, Regunathan S. Agmatine attenuates stress- and lipopolysaccharide-induced fever in rats. Physiology & Behavior. 2005;85:370-375
  34. 34. Jeutsch JD, Anzivino LA. A low dose of the alpha2 agonist clonidine ameliorates the visual attention and spatial working memory deficits produced by phencyclidine administration to rats. Psychopharmacology (Berl). 2004;175:76-83
  35. 35. Bardget ME, Points M, Ramsey-Faulkner C. The effects of clonidine on discrete-trial delayed spatial alternation in two rat models of memory loss. Neuropsychopharmacology. 2008;33:1980-1991
  36. 36. Marrs W, Kuperman J, Avedian T, Roth RH, Jentsch JD. Alpha-2 adrenoceptor activation inhibits phencyclidine-induced deficits of spatial working memory in rats. Neuropsychopharmacology. 2005;30:1500-1510
  37. 37. Unal G, Ates A, Aricioglu F. Agmatine-attenuated cognitive and social deficits in subchronic MK-801 model of schizophrenia in rats. Psychiatry and Clinical Psychopharmacology. 2018:1-10
  38. 38. Bergin DH, Liu P. Agmatine protects against beta-amyloid25–35-induced memory impairments in the rat. Neuroscience. 2010;169:794-811
  39. 39. Zarifkar A, Choopani S, Ghasemi R, Naghdi N, Maghsoudi AH, Maghsoudi N, et al. Agmatine prevents LPS-induced spatial memory impairment and hippocampal apoptosis. European Journal of Pharmacology. 2010;634:84-88
  40. 40. Li YF, Gong ZH, Cao JB, Wang HL, Luo ZP, Li J. Antidepressant-like effect of agmatine and its possible mechanism. European Journal of Pharmacology. 2003;469(1–3):81-88
  41. 41. Martel J, Chopin P, Colpaert F, Marien M. Neuroprotective effects of the alpha2-adrenoceptor antagonists, (+)-efaroxan and (+/−)-idazoxan, against quinolinic acid-induced lesions of the rat striatum. Experimental Neurology. 1998;154:595-601
  42. 42. Wang X-S, Fang H-L, Chen Y, Liang S-S, Zhu Z-G, Zeng Q-Y, et al. Idazoxan reduces blood-brain barrier damage during experimental autoimmune encephalomyelitis in mouse. European Journal of Pharmacology. 2014;736:70-76
  43. 43. Chopin P, Debeir T, Raisman-Vozari R, Colpaert FC, Marien MR. Protective effect of the alpha2-adrenoceptor antagonist, dexefaroxan, against spatial memory deficit induced by cortical devascularization in the adult rat. Experimental Neurology. 2004;185:198-200
  44. 44. Pauwels PJ, Rauly I, Wurch T. Dissimilar pharmacological responses by a new series of imidazoline derivatives at precoupled and ligand-activated alpha 2A-adrenoceptor states: Evidence for effector pathway-dependent differential antagonism. The Journal of Pharmacology and Experimental Therapeutics. 2003;305:1015-1023
  45. 45. Rizk P, Salazar J, Raisman-Vozari R, Marien M, Ruberg M, Colpaert F, et al. The alpha2-adrenoceptor antagonist dexefaroxan enhances hippocampal neurogenesis by increasing the survival and differentiation of new granule cells. Neuropsychopharmacology. 2006;31:1146-1157
  46. 46. Debeir T, Marien M, Ferrario J, Rizk P, Prigent A, Colpaert F, et al. In vivo upregulation of endogenous NGF in the rat brain by the alpha2-adrenoreceptor antagonist dexefaroxan: Potential role in the protection of the basalocortical cholinergic system during neurodegeneration. Experimental Neurology. 2004;190:384-395
  47. 47. Francis BM, Yang J, Hajderi E, Brown ME, Michalski B, McLaurin J, et al. Reduced tissue levels of noradrenaline are associated with behavioral phenotypes of the TgCRND8 mouse model of Alzheimer's disease. Neuropsychopharmacology. 2012;37:1934-1944
  48. 48. Condello S, Monica Currò M, Ferlazzo N, Caccamo D, Satriano J, Lentile R. Agmatine effects on mitochondrial membrane potential and NF-κB activation protect against rotenone-induced cell damage in human neuronal-like SH-SY5Y cells. Journal of Neurochemistry. 2011;116(1):67-75
  49. 49. Park YM, Lee WT, Bokara KK, Seo SK, Park SH, Kim JH, et al. The multifaceted effects of agmatine on functional recovery after spinal cord injury through modulations of BMP-2/4/7 expressions in neurons and glial cells. PLoS One. 2013;8:e53911
  50. 50. Tohidi V, Hassanzadeh B, Sherwood B, Ma W, Rosenberg M, Gilad V, et al. Effect of agmatine sulfate on neuropathic pain. Neurology. 2014;82(Suppl. 10):P7.094
  51. 51. Piletz JE, Aricioglu F, Cheng JT, Fairbanks CA, Gilad VH, Haenisch B, et al. Agmatine: Clinical applications after 100 years in translation. Drug Discovery Today. 2013;18(17–18):880-893
  52. 52. Keynan O, Mirovsky Y, Dekel S, Gilad VH, Gilad GM. Safety and efficacy of dietary agmatine sulfate in lumbar disc-associated radiculopathy. An open-label, dose-escalating study followed by a randomized, double-blind, placebo-controlled trial. Pain Medicine. 2010;11(3):356-368
  53. 53. Himmelseher S, Kochs EF. Neuroprotection by dexmedetomidine. In: Gullo A, editor. Anaesthesia, Pain, Intensive Care and Emergency A.P.I.C.E. Italia: Springer-Verlag; 2006. pp. 633-639. Ch. 56
  54. 54. Coull JT, Sahakian BJ, Hodges JR. The α2 antagonist idazoxan remediates certain attentional and executive dysfunction in patients with dementia of frontal type. Psychopharmacology. 1996;123:239-249
  55. 55. Gupta S, Sharma B. Pharmacological benefit of I(1)-imidazoline receptors activation and nuclear factor kappa-B (NF-κB) modulation in experimental Huntington's disease. Brain Research Bulletin. 2014;102:57-68
  56. 56. Ilik MK, Kocaogullar Y, Koc O, Esen H. Beneficial effects of moxonidine on cerebral vasospasm after experimental subarachnoid. Turkish Neurosurgery. 2014;24(6):873-879
  57. 57. Liu P, Smith PF, Appleton I, Darlington CL, Bilkey DK. Potential involvement of NOS and arginase in age-related behavioural impairments. Experimental Gerontology. 2004;39:1207-1222
  58. 58. Rushaidhi M, Collie ND, Zhang H, Liu P. Agmatine selectively improves behavioural function in aged male Sprague-Dawley rats. Neuroscience. 2012;218:206-215
  59. 59. Gupta N, Jing Y, Collie ND, Zhang H, Liu P. Ageing alters behavioural function and brain arginine metabolism in male Sprague-Dawley rats. Neuroscience. 2012;226:178-196
  60. 60. Liu P, Jing Y, Collie ND, Chary S, Zhang H. Memory-related changes in L-citrulline and agmatine in the rat brain. Hippocampus. 2009;19:597-602
  61. 61. Moosavi M, Khales GY, Abbasi L, Zarifkar A, Rastegar K. Agmatine protects against scopolamine-induced water maze performance impairment and hippocampal ERK and Akt inactivation. Neuropharmacology. 2012;62(5–6):2018-2023
  62. 62. Stewart LS, McKay BE. Acquisition deficit and time-dependent retrograde amnesia for contextual fear conditioning in agmatine-treated rats. Behavioural Pharmacology. 2000;11:93-97
  63. 63. McKay BE, Lado WE, Martin LJ, Galic MA, Fournier NM. Learning and memory in agmatine-treated rats. Pharmacology, Biochemistry, and Behavior. 2002;72:551-557
  64. 64. Utkan T, Gocmez SS, Regunathan S, Aricioglu F. Agmatine, a metabolite of L-arginine, reverses scopolamine-induced learning and memory impairment in rats. Pharmacology, Biochemistry, and Behavior. 2012;102:578-584
  65. 65. Knox LT, Jing Y, Fleete MS, Collie ND, Zhang H, Liu P. Scopolamine impairs behavioural function and arginine metabolism in the rat dentate gyrus. Neuropharmacology. 2011;61:1452-1462
  66. 66. Barber TA, Haggarty MK. Memantine ameliorates scopolamine-induced amnesia in chicks trained on taste-avoidance learning. Neurobiology of Learning and Memory. 2010;93:540-545
  67. 67. Nguyen L, Lucke-Wold BP, Mookerjee S, Kaushal N, Matsumoto RR. Sigma-1 receptors and neurodegenerative diseases: Towards a hypothesis of sigma-1 receptors as amplifiers of neurodegeneration and neuroprotection. Advances in Experimental Medicine and Biology. 2017;964:133-152
  68. 68. Xiong XY, Liu L, Yang QW. Refocusing neuroprotection in cerebral reperfusion era: New challenges and strategies. Frontiers in Neurology. 2018;9:249
  69. 69. Chamorro A. Neuroprotectants in the era of reperfusion therapy. Journal of Stroke. 2018;20(2):197-207
  70. 70. Wu Q J, Tymianski M. Targeting NMDA receptors in stroke: New hope in neuroprotection. Molecular Brain. 2018;11(1):15
  71. 71. Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y. Current research on opioid receptor function. Current Drug Targets. 2012;13:230-246
  72. 72. Vaidya B, Sifat AE, Karamyan VT, Abbruscato TJ. The neuroprotective role of the brain opioid system in stroke injury. Drug Discovery Today. 2018;23:1385-1395
  73. 73. Yang Y, Zhi F, He X, Moore ML, Kang X, Chao D, et al. Delta-opioid receptor activation and microRNA expression of the rat cortex in hypoxia. PLoS One. 2012;7:e51524
  74. 74. Grant Liska M, Crowley MG, Lippert T, Corey S, Borlongan CV. Delta opioid receptor and peptide: A dynamic therapy for stroke and other neurological disorders. In: Handbook of Experimental Pharmacology. Berlin, Heidelberg: Springer; 2017
  75. 75. Staples M, Acosta S, Tajiri N, Pabon M, Kaneko Y, Borlongan CV. Delta opioid receptor and its peptide: A receptor-ligand neuroprotection. International Journal of Molecular Sciences. 2013;14(9):17410-17419
  76. 76. Liu H, Chen B, Li S, Yao J. Dose-dependent neuroprotection of delta-opioid peptide [D-Ala(2), D-Leu(5)] enkephalin on spinal cord ischemia-reperfusion injury by regional perfusion into the abdominal aorta in rabbits. Journal of Vascular Surgery. 2016;63(4):1074-1081
  77. 77. Lee JY, Liska MG, Crowley M, Xu K, Acosta SA, Borlongan CV, et al. Multifaceted effects of delta opioid receptors and DADLE in diseases of the nervous system. Current Drug Discovery Technologies. 2018;15(2):94-108
  78. 78. Crowley MG, Liska MG, Lippert T, Corey S, Borlongan CV. Utilizing delta opioid receptors and peptides for cytoprotection: Implications in stroke and other neurological disorders. CNS & Neurological Disorders Drug Targets. 2017;16(4):414-424
  79. 79. Moghal ETB, Venkatesh K, Sen D. The delta opioid peptide D-Alanine 2, Leucine 5 Enkephaline (DADLE)-induces neuroprotection through cross-talk between the UPR and pro-survival MAPK-NGF-Bcl2 signaling pathways via modulation of several micro-RNAs in SH-SY5Y cells subjected to ER stress. Cell Biology International. 2018;42(5):543-569
  80. 80. Lv MR, Li B, Wang MG, Meng FG, Yu JJ, Guo F, et al. Activation of the PI3K-Akt pathway promotes neuroprotection of the delta-opioid receptor agonist against cerebral ischemia-reperfusion injury in rat models. Biomedicine & Pharmacotherapy. 2017;93:230-237
  81. 81. Yang L, Shah K, Wang H, Karamyan VT, Abbruscato TJ. Characterization of neuroprotective effects of biphalin, an opioid receptor agonist, in a model of focal brain ischemia. The Journal of Pharmacology and Experimental Therapeutics. 2011;339(2):499-508
  82. 82. Kawalec M, Kowalczyk JE, Beresewicz M, Lipkowski AW, Zablocka B. Neuroprotective potential of biphalin, multireceptor opioid peptide, against excitotoxic injury in hippocampal organotypic culture. Neurochemical Research. 2011;36(11):2091-2095
  83. 83. Remesic M, Macedonio G, Mollica A, Porreca F, Hruby V, Lee YS. Cyclic biphalin analogues with a novel linker lead to potent agonist activities at mu, delta, and kappa opioid receptors. Bioorganic & Medicinal Chemistry. 2018;26(12):3664-3667
  84. 84. Popiolek-Barczyk K, Piotrowska A, Makuch W, Mika J. Biphalin, a dimeric enkephalin, alleviates LPS-induced activation in rat primary microglial cultures in opioid receptor-dependent and receptor-independent manners. Neural Plasticity. 2017;2017:3829472
  85. 85. Yang L, Islam MR, Karamyan VT, Abbruscato TJ. In vitro and in vivo efficacy of a potent opioid receptor agonist, biphalin, compared to subtype-selective opioid receptor agonists for stroke treatment. Brain Research. 1609;2015:1-11
  86. 86. Islam MR, Yang L, Lee YS, Hruby VJ, Karamyan VT, Abbruscato TJ. Enkephalin-fentanyl multifunctional opioids as potential neuroprotectants for ischemic stroke treatment. Current Pharmaceutical Design. 2016;22:6459-6468

Notes

  • These authors contributed equally to this work.

Written By

Liliana Mititelu-Tartau, Maria Bogdan, Victor Gheorman, Liliana Foia, Ancuta Goriuc, Gabriela Rusu, Beatrice Buca, Liliana Pavel, Ana Cristofor, Cosmin-Gabriel Tartau and Gratiela Eliza Popa

Submitted: 13 May 2018 Reviewed: 10 October 2018 Published: 03 April 2019