Skip to main content

SYSTEMATIC REVIEW article

Front. Neurosci., 09 May 2019
Sec. Neurodegeneration
This article is part of the Research Topic Forms of Degeneration and Dysregulation in the Central Nervous System View all 8 articles

Molecular Targets of Bis (7)-Cognitin and Its Relevance in Neurological Disorders: A Systematic Review

  • 1Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
  • 2Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China

Background: The exact mechanisms involved in the pathogenesis of neurodegenerative conditions are not fully known. The design of drugs that act on multiple targets represents a promising approach that should be explored for more effective clinical options for neurodegenerative disorders. B7C is s synthetic drug that has been studied for over 20 years and represents a promising multi-target drug for the treatment of neurodegenerative disorders, such as AD.

Aims: The present systematic review, thus, aims at examining existing studies on the effect of B7C on different molecular targets and at discussing the relevance of B7C in neurological disorders.

Methods: A list of predefined search terms was used to retrieve relevant articles from the databases of Embase, Pubmed, Scopus, and Web of Science. The selection of articles was done by two independent authors, who were considering articles concerned primarily with the evaluation of the effect of B7C on neurological disorders. Only full-text articles written in English were included; whereas, systematic reviews, meta-analyses, book chapters, conference subtracts, and computational studies were excluded.

Results: A total of 2,266 articles were retrieved out of which 41 articles were included in the present systematic review. The effect of B7C on molecular targets, including AChE, BChE, BACE-1, NMDA receptor, GABA receptor, NOS, and Kv4.2 potassium channels was evaluated. Moreover, the studies that were included assessed the effect of B7C on biological processes, such as apoptosis, neuritogenesis, and amyloid beta aggregation. The animal studies examined in the review focused on the effect of B7C on cognition and memory.

Conclusions: The beneficial effects observed on different molecular targets and biological processes relevant to neurological conditions confirm that B7C is a promising multi-target drug with the potential to treat neurological disorders.

Introduction

Neurodegenerative disorders are not only disabling but incurable conditions characterized by the loss of neurons leading to chronic degeneration and deterioration of the brain (Pérez-Hernández et al., 2016). Alzheimer's disease (AD), Parkinson's disease, Multiple sclerosis, Huntington's disease, and Amyotrophic lateral sclerosis are the major causes of death in the elderly population, and they have been projected to be the second leading cause of death by 2040 (Valera and Masliah, 2016). AD is the most common cause of dementia and the most frequent neurodegenerative disorder manifesting in the decline in cognition, memory ability, and language and problem-solving skills due to failure in the synaptic signal transfer, decreased number of synapses, and neuronal death (Baquero and Martín, 2015; Teipel et al., 2016).

The molecular mechanisms behind neurodegenerative diseases remain elusive (Santiago et al., 2017). However, it is clear that the multifactorial pathogenic nature of neurodegenerative disorders requires the use of several drugs to tackle the multiple symptoms present in these disorders (Li et al., 2009). Another therapeutic strategy to address the multifactorial complexity of neurodegenerative disorders is the search for compounds that interact with multiple targets known as multi-target design ligands (Van der Schyf, 2011; Ramsay et al., 2016). This strategy requires biological screening at the early stages of drug discovery and lead optimization to screen for drugs that can interact with two or more desired targets (Ramsay et al., 2016). The design of drugs that act on multiple targets represents a promising approach that should be explored for more effective clinical options for neurodegenerative disorders (Trippier et al., 2013). Drugs with multi-target properties have the potential to provide a more significant effect by acting on different brain regions relevant to the mechanism of disease and symptomatology of the disorder (Li et al., 2007b; Ramsay et al., 2016). The lack of effective treatment options that stop the progression of the disease and alleviate the multiple symptoms observed in this kind of disorders is driving the search for novel therapeutic alternatives.

Bis (7)-cognitin was first synthesized as part of a two-step prototype optimization strategy using computer modeling of ligand docking with target proteins. B7C showed to be highly potent, selective and cheap acetylcholinesterase inhibitor with promising therapeutic application in Alzheimer's disease (Pang et al., 1996). In order to evaluate the multi-target potential of B7C for the treatment of neurodegenerative disorders, a detailed analysis of the existing studies on B7C is inevitable.

Since its synthesis, many studies have been conducted to evaluate the mechanism of action of B7C using different cell-based platforms and animal models focusing the neurotherapeutic effect of B7C. In order to evaluate the potential clinical applications of B7C and assess its usefulness as a multi-target drug, it is crucial to carry out an in-depth analysis of the studies conducted of B7C. Therefore, the present systematic review aims at examining existing studies on the effect of B7C on different molecular targets and at discussing the relevance of B7C in neurological disorders. The research questions used as guidance for the study include: What molecular targets in the central nervous system have been identified for B7C? What is the relevance of B7C on neurological disorders based on the evidence from pre-clinical studies?

Methods

Search Strategy and Selection of Studies

Search terms were pre-defined, and a search strategy was established as shown in Table 1. The search terms were used to retrieve articles using the following databases: Embase, Pubmed, Scopus, and Web of Science. The settings for the database search included no restriction in terms of the year of publication, the advance search option was utilized for the combined search, and no filter was set in the database search. The retrieved articles from all the databases were pooled. The title of the articles was checked, and duplicate articles were removed. The titles of the remaining articles were screened to preselect relevant articles based on predefined inclusion and exclusion criteria described in the next section. Articles that did not fulfilled the inclusion criteria were removed. The full text of the preselected articles was read through for further article selection. Only articles that fulfilled the inclusion criteria were included in the systematic review. The database search and selection of the articles were carried out by two independent authors. Discrepancies in the selection were resolved by discussion, and a third author was involved as adjudicator where it was deemed necessary.

TABLE 1
www.frontiersin.org

Table 1. Pre-defined search terms and database search strategy.

Inclusion and Exclusion Criteria

Included are full-text studies written in English focusing on the effect of B7C on neurological disorders. Excluded are systematic reviews, meta-analyses, book chapters, conference abstracts, and computational studies.

Data Extraction and Analysis

The data extracted from the selected studies included the type of study, the molecular target or behavioral test evaluated, and the conclusions. The cell line, in vitro assay, and concentration of B7C was extracted from the in vitro studies; whereas, the animal species, strain, and B7C dose used were extracted from the in vivo studies. The evidence of the effect of B7C on the respective molecular target was analyzed and the relevance on neurological disorders discussed.

Results

A total of 2266 articles were pooled from all the databases. The ratio was as follows, 194 articles from Embase, 61 from Pubmed, 1,909 from Scopus, and 102 from Web of Science. An overview of the flow chart with regard to the selection of articles is illustrated in Figure 1. A total of 349 articles were removed from the list due to their being duplicates. In the preselection stage, 1805 articles were omitted as they did not focus on B7C (n = 1744), were not concerned with neurological disorders (n = 4), or were review articles (n = 57). Out of the remaining 112 articles, 69 were excluded as they did not focus on B7C (n = 26), were not written in English (n = 3), did not focus on neurological disorders (n = 6), were review articles (n = 17), computational studies (n = 12), or conference abstracts (n = 5). Two of the preselected articles were an erratum linked to two studies that were included. The errata were checked and found to be referring to amendments in the author list, article title, or acknowledgments. Since the errata did not refer to relevant content in terms of the present systematic review, they were eliminated. A total of 41 articles fulfilled both the inclusion and exclusion criteria and thus made it into the final stage.

FIGURE 1
www.frontiersin.org

Figure 1. Study selection flow chart.

The data extracted from the selected studies are summarized in Table 2. Most of the studies that were included evaluated the effect of B7C on in vitro platforms (n = 25); whereas 13 studies evaluated the effect in vivo. Three studies included both in vitro and in vivo approaches. The molecular target that was primarily studied was AChE in 11 studies (Li et al., 1999; Wang et al., 1999b; Ros et al., 2001; Hu et al., 2002, 2015b; Fu et al., 2006; Yu et al., 2008; Pan et al., 2009; Bolognesi et al., 2010; Rizzo et al., 2011; Qian et al., 2014), followed by the NMDA receptor (n = 8) (Bai-fang et al., 2001; Li et al., 2005, 2007b; Luo et al., 2007; Liu et al., 2008a,b; Zhang et al., 2011; Liu and Li, 2012). The BChE was under scrutiny in five studies (Wang et al., 1999b; Hu et al., 2002; Bolognesi et al., 2010; Rizzo et al., 2011; Qian et al., 2014), the BACE-1 in four studies (Fu et al., 2008, 2009; Bolognesi et al., 2010; Rizzo et al., 2011), the GABA receptor in three studies (Li et al., 1999, 2007a; Zhou et al., 2009), the nitric oxide synthase (NOS) in two studies (Li et al., 2006, 2007b), the Kv4.2 potassium channels also in two studies (Nie et al., 2007; Li et al., 2010), the serotonin receptor 5-HT3 in one study (Luo et al., 2004), the α-secretase also in one study (Fu et al., 2009), the L-type voltage-dependent Ca2+ channels in one study (Fu et al., 2006), and the choline acetyl transferase in one study as well (Liu et al., 2000). Apoptosis (Han et al., 2000; Fu et al., 2007; Zhao et al., 2008; Fang et al., 2010), neuritogenesis, and neurite outgrowth (Chang et al., 2015; Hu et al., 2015a), long-term potentiation (Chang et al., 2015), amyloid beta (Aβ) aggregation and toxicity (Chang et al., 2015; Hu et al., 2015a), cell toxicity (Xiao et al., 2000), and retinal ischaemia (Li et al., 2014) were those biological processes where the effect of B7C was examined. The effect of B7C on spatial learning and memory was evaluated using the Morris water maze test in six studies (Wang et al., 1999a; Liu et al., 2000; Han et al., 2012; Shu et al., 2012; Chang et al., 2015; Hu et al., 2015b), the step-through task was used to measure the passive-avoidance response in two studies (Pan et al., 2007, 2009), the open field test was used also in two studies (Pan et al., 2009, 2011), and the novel object recognition test was used in one study (Han et al., 2012).

TABLE 2
www.frontiersin.org

Table 2. Description of in vitro and in vivo studies on B7C.

The results of the studies that evaluated the effect of B7C on AChE and BChE demonstrated a clear inhibition of both enzymes with IC50 values ranging from 0.81 to 5.1 nM for AChE and from 2.6 to 328.9 nM for BChE, respectively. The antagonistic effect of B7C on the NMDA receptor was demonstrated in several studies. For example, treatment with B7C reduced the NMDA-mediated activity and showed a protective effect against glutamate-induced excitotoxicity (Bai-fang et al., 2001). Prevention of neuronal apoptosis by B7C blockade of the NMDA receptor was also observed (Li et al., 2005). Inhibition of the NMDA receptor by B7C was reported by Li et al. (2007b), Luo et al. (2007), Liu et al. (2008a,b), Zhang et al. (2011), and Liu and Li (2012). An inhibitory effect of B7C on BACE-1 led to a decrease in the generation of Aβ by activation of α-secretase (Fu et al., 2008, 2009; Bolognesi et al., 2010; Rizzo et al., 2011). Furthermore, B7C acted as a competitive antagonist of the GABA receptor (Li et al., 1999, 2007a; Zhou et al., 2009). Finally, treatment with B7C at doses of 0.18-0.89 μM/kg and 0.1–0.2 mg/kg showed improved cognitive performance in several animal models in which learning and memory deficits were induced, demonstrating the restorative effect of B7C (Wang et al., 1999a; Liu et al., 2000; Han et al., 2012; Shu et al., 2012; Chang et al., 2015; Hu et al., 2015b).

Discussion

Tacrine (9-amino-1,2,3,4-tetrahydroacridine, under the trade name Cognex®, was the first drug approved for the treatment of AD in 1993 (Han et al., 2012). Other AChE inhibitors, such as donepezil (Aricept®), rivastigmine (Exelon®) and galantamine (Reminyl®) introduced in 1996, 2000, and 2001, respectively, in addition to the N-methyl-D-aspartate (NMDA) receptor antagonist memantine (Namenda®) followed the release of tacrine (Han et al., 2012; Lopes et al., 2017). Tacrine binds in a reversible mode to AChE and is considered a classical AChE pharmacophore (Lopes et al., 2017). Due to its side effects, such as hepatotoxicity and myopathy as well as its poor pharmacokinetic properties, including low bioavailability and narrow therapeutic index, a series of new tacrine-based compounds have been developed (Luo et al., 2004; Han et al., 2012). B7C, a product of the structure-activity-relationship drug design, is a dimer formed by two tacrine molecules linked by a spacer containing 7 methylene groups (Hu et al., 2015,a). This particular molecule has caught the attention of researchers, especially as a treatment option for AD. An advantageous feature of the B7C molecule is that it easily crosses the brain blood barrier due to its highly lipophilic profile making it a promising drug candidate for central nervous system disturbances (Hu et al., 2015a).

Although the pathological processes in AD are not well-understood, it is clear that disturbances in the cholinergic system and other neurotransmitters play a pivotal role in the pathogenesis of this neurodegenerative disorder (Han et al., 2012). Strategies to develop drugs for AD have focused on acetylcholinesterase (AChE) as a target for drug design based on the cholinergic hypothesis for AD (Ros et al., 2001; Lopes et al., 2017). The cholinergic hypothesis states that increased levels of acetylcholine in the brain alleviate the cognitive deficiencies observed in AD (Ros et al., 2001). Although a series of AChE inhibitors have been extensively studied, none of them represent a real cure for AD (Ros et al., 2001; Lopes et al., 2017).

The enzyme AChE has 2 sub-active sites within the binding pocket, namely the catalytic anionic site (CAS) and the peripheral anionic site (PAS) (Li et al., 2009). Using computational tools, molecular docking simulations were carried out to design and optimize the synthesis of tacrine analogs (Li et al., 2009). New compounds have been developed using the dual active sites in AChE as a basic hypothesis to increase the therapeutic action of tacrine analogs (Li et al., 2009). Alkylene-linked tacrine dimers interact with the CAS and PAS and potently inhibit AChE being B7C one of the promising alkylene dimers analogs (Lopes et al., 2017). As shown in Table 2, the multi-target activity of B7C has been evaluated in cell-based platforms and in vivo. The molecular targets studied included the AChE, BChE, NMDA receptor, ChAT, GABA receptor, BACE-1, Kv4.2 potassium channels, NOS, and the 5-HT3 receptor. B7C has been found to be 150 times more potent and 250 times more selective to inhibit AChE when compared to tacrine (Ros et al., 2001; Li et al., 2009). The studies included in the present systematic review not only demonstrated the inhibitory effect of B7C on AChE, but also supported the promising action of B7C on neurodegenerative disorders based on the AChE hypothesis. The biological properties of B7C are superior to those of tacrine because B7C interacts simultaneously with the CAS and PAS of the enzyme (Li et al., 2009). The addition of the heptylene chain to the two tacrine molecules allows the dual interaction with the AChE binding sites, which explains its superior activity compared to tacrine (Bolognesi et al., 2010). B7C is a multi-target compound that shows promising biological activity, including inhibition of AChE, prevention of the aggregation of the β-amyloid (Aβ) protein, regulation of the downstream signaling mediated by the NMDA receptor, and inhibition of the nitric oxide synthase (NOS) signaling pathway (Zhang et al., 2011).

AChE is an important element of the cholinergic system that acts in the synaptic cleft by hydrolyzing the neurotransmitter acetylcholine (ACh) at central and peripheral levels (Colović et al., 2013). In addition to its function in cholinergic synapses, AChE plays a very crucial role of AChE relevant for the disease mechanism of AD because it accelerates the formation of Aβ formation in the Alzheimer's brain (Inestrosa et al., 1996). AChE is an important target in neurodegenerative disorders as its inhibition leads to accumulation of ACh by decreasing the ACh breakdown rate (Colović et al., 2013). Also, AChE stimulates Aβ fibrillogenesis through the formation of AChE-Aβ complexes, which is a characteristic feature in AD patients (Muñoz-Ruiz et al., 2005). Due to the nature of the AChE enzyme, dual binding is a highly desirable property for the design of AChE inhibitors, such as B7C (Li et al., 2009).

Several studies (see Table 2) have already demonstrated the effect of B7C on the inhibition of AChE in a selective manner and at lower concentration than tacrine (Li et al., 1999; Wang et al., 1999b; Ros et al., 2001; Hu et al., 2002, 2015b; Fu et al., 2006; Yu et al., 2008; Pan et al., 2009; Bolognesi et al., 2010; Rizzo et al., 2011; Qian et al., 2014). Drugs that inhibit AChE keep the ACh levels high in the synaptic cleft, thereby stimulating cholinergic transmission in regions of the forebrain that compensate for the loss of cells (Muñoz-Ruiz et al., 2005; Colović et al., 2013).

B7C has also been evaluated on the NMDA receptor and has been found to show an antagonistic effect (Mattson, 2000; Bai-fang et al., 2001; Li et al., 2005, 2007b; Luo et al., 2007; Liu et al., 2008b; Zhang et al., 2011; Liu and Li, 2012). Excitotoxicity significantly contributes to neuronal cell damage and death in neurodegenerative disorders (Lipton, 2004). Excitotoxicity is the result of overactivation of the NMDA glutamate receptor that leads to an excessive Ca2+ influx in the cell (Newcomer et al., 2000). Glutamate, the major excitatory neurotransmitter in the brain, is a crucial mediator involved in the normal functioning of the nervous system (Lipton, 2004). It is hypothesized that chronic exposure to elevated levels of glutamate or glutamate receptor hyperactivity triggers apoptotic pathways, a phenomenon of clinical relevance in disorders, such as Huntington's disease, Parkinson's disease, Multiple sclerosis, HIV-associated dementia, Amyotrophic lateral sclerosis, Glaucoma, and Alzheimer's disease (Lipton, 2004).

The link between Aβ formation, deposition, and AD pathogenesis has been well-established (Murphy and LeVine, 2010). The findings of the effect of B7C on AChE have also demonstrated inhibition of the Aβ fibrils formation and stimulated the disaggregation of pre-formed Aβ fibrils and improved memory impairment induced by Aβ (Fu et al., 2006; Bolognesi et al., 2010; Rizzo et al., 2011; Hu et al., 2015b). The protective effect of B7C against Aβ challenge reported in the studies included demonstrating the promising potential of B7C as treatment of AD. The BACE-1 is the enzyme responsible for the onset of the generation of Aβ. Therefore, it represents a very promising target for AD (Vassar et al., 2009). Aβ is the major component that occurs in neuritic plates found in AD (Chen et al., 2012). BACE-1 overexpression or hyperactivity is associated with the pathogenesis of AD while the opposite scenario has been found to have a neuroprotective effect (Chen et al., 2012). The findings of two studies, shown in Table 2, demonstrated an inhibitory effect of B7C on BACE-1 leading to a decreased generation of Aβ (Fu et al., 2008, 2009). Therefore, B7C showed a protective effect for the treatment of AD.

Cognitive impairment is a common symptom of neurological diseases, including AD (Bland, 2016), which warrants potential treatment options that act on improving represents a promising drug for the treatment of neurological disorders in which cognition is affected.

An important mediator of cell death is the N the cognitive impairment induced by a neuropathological condition as valuable alternatives. B7C improved cognition, special learning, and memory observed in several animal models (Pan et al., 2007, 2011; Han et al., 2012; Shu et al., 2012). Consequently, it OS, which has also been evaluated in B7C studies (Li et al., 2006, 2007b). In these studies, B7C was shown to have an inhibitory effect on NOS (Li et al., 2006, 2007a). Hyperactivity of NOS leads to excitotoxicity-mediated cell death. The enzyme is tethered to the NMDA receptor and gets activated by the influx of Ca2+ which increases the levels of NO associated with stroke and neurodegenerative diseases (Lipton, 2004). As B7C has demonstrated NMDA and NOS inhibitory activity, it represents a valuable candidate for the treatment of degenerative disorders.

Several studies on B7C focused on apoptosis as a target physiological mechanism (Han et al., 2000; Xiao et al., 2000; Fu et al., 2007; Zhao et al., 2008; Fang et al., 2010). Abnormal regulation of neuronal cell death has been associated with many neurological disorders (Mattson, 2000). Excessive death of one or more populations of neurons occurs as a result of disease or injury (Mattson, 2000). In AD, loss of hippocampal and cortical neurons is responsible for the symptomatology observed in this neurodegenerative disease (Mattson, 2000). The regulation of apoptosis involves several mediators, such as neurotrophic factors (inhibition) or glutamate (activation) (Mattson, 2000). Excessive glutamate-mediated activity increases the influx of Ca2+ leading to excitotoxicity and ultimately cell death (Mattson, 2000). As shown in Table 2, B7C provided protection against glutamate-induced excitotoxicity and free radical-induced damage (Han et al., 2000; Xiao et al., 2000; Fu et al., 2007; Zhao et al., 2008; Fang et al., 2010).

Another target for B7C is the GABA receptor (Li et al., 1999, 2007a; Zhou et al., 2009). GABA is the major inhibitory neurotransmitter in the central nervous system and functions primarily as a metabolite and a neurotransmitter (Zhou et al., 2009; Best et al., 2014). Together with the excitatory neurotransmitter glutamate, GABA is an important modulator of the inhibitory-excitatory balance that is essential for the proper functioning of the brain (Wu and Sun, 2015). Dysfunctions in the GABA system have been closely linked with neurological disorders, such as Huntington's chorea, epilepsy, AD, anxiety, and depression (Krogsgaard-Larsen, 1992; Kim and Yoon, 2017). The GABAA receptor is a ligand-gated ion channel that regulates the influx of chloride ions, causing hyperpolarization in the postsynaptic neuron (Kim and Yoon, 2017) and mediating the fast inhibitory neurotransmission in the brain (Best et al., 2014). Changes in the concentration of endogenous modulator or in the composition of the GABAA receptor lead to downregulation of the neuronal inhibition seen in pathological states (Nuss, 2015). B7C has shown to bind to the GABA receptor in a potent but reversable manner displaying a competitive antagonistic role (Li et al., 2007a; Zhou et al., 2009). Since innhibition of GABA has been linked to improvements in pathological states, drugs that regulate the GABA system, such as B7C are highly relevant in the treatment of neurological disorders.

Limitations

Only one study out of the 41 studies included in the present systematic review reported adverse effect related to the treatment with B7C, namely hepatotoxicity. Due to the lack of information on side effects linked to B7C in the studies included, an overview of the safety of the drug could not be carried out. Thus, it is recommended to analyze the safety profile of B7C based on pre-clinical assessment in further studies.

Conclusions

B7C is a computationally designed drug that has shown promising effects on several in vitro and in vivo platforms for AD and other neurodegenerative disorders. In the last two decades, numerous studies have focused on the evaluation of B7C on different targets. From the analysis presented, it is clear that B7C shows a superior activity when compared to its basic structure tacrine. Also, the beneficial effects observed on different molecular targets and biological processes relevant to neurological conditions confirm B7C's potency as a multi-target drug for the treatment of neurological disorders.

Author Contributions

DIS-V was responsible for the database search, article selection, and manuscript writing. JKWC was responsible for the database search and article selection. BWML resolved discrepancies in the selection process, reviewed, and approved the manuscript, and SQH and Y-FH reviewed the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We would like to immensely thank Dr. Anna Laszlo for the careful proofreading of the manuscript, and for making suggestions that greatly improved upon the use of language in the paper.

References

Bai-fang, Z., Fang-fang, P., Jiang-zhou, Z., and Dong-cheng, W. (2001). bis(7)-tacrine, a promising anti-Alzheimer's agent, attenuates glutamate-induced cell injury in primary cultured cerebrocortical neurons of rats. Wuhan Univ. J. Nat. Sci. 6, 737–741. doi: 10.1007/BF02830295

CrossRef Full Text | Google Scholar

Baquero, M., and Martín, N. (2015). Depressive symptoms in neurodegenerative diseases. World J. Clin. Cases 3, 682–693. doi: 10.12998/wjcc.v3.i8.682

PubMed Abstract | CrossRef Full Text | Google Scholar

Best, J. G., Stagg, C. J., and Dennis, A. (2014). Other significant metabolites: myo-inositol, gaba, glutamine, and lactate. Magn. Reson. Spectrosc. 2014, 122–138. doi: 10.1016/B978-0-12-401688-0.00010-0

CrossRef Full Text | Google Scholar

Bland, J. (2016). Mild cognitive impairment, neurodegeneration, and personalized lifestyle medicine. Integr. Med. 15, 12–14.

PubMed Abstract | Google Scholar

Bolognesi, M. L., Bartolini, M., Mancini, F., Chiriano, G., Ceccarini, L., Rosini, M., et al. (2010). bis(7)-tacrine Derivatives as multitarget-directed ligands: focus on anticholinesterase and antiamyloid activities. Chem. Med. Chem. 5, 1215–1220. doi: 10.1002/cmdc.201000086

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, L., Cui, W., Yang, Y., Xu, S., Zhou, W., Fu, H., et al. (2015). Protection against β-amyloid-induced synaptic and memory impairments via altering β-amyloid assembly by bis(heptyl)-cognitin. Sci. Rep. 5:10256. doi: 10.1038/srep10256

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Huang, X., Zhang, Y., Rockenstein, E., Bu, G., Golde, T. E., et al. (2012). Alzheimer's β-secretase (BACE1) regulates the cAMP/PKA/CREB pathway independently of β-amyloid. J. Neurosci. 32, 11390–11395. doi: 10.1523/JNEUROSCI.0757-12.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Colović, M. B., Krsti,ć, D. Z., Lazarević-Pašti, T. D., BondŽić, A. M., and Vasi,ć, V. M. (2013). Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr. Neuropharmacol. 11, 315–335. doi: 10.2174/1570159X11311030006

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, J. H., Wang, X. H., Xu, Z. R., and Jiang, F. G. (2010). Neuroprotective effects of bis(7)-tacrine against glutamate-induced retinal ganglion cells damage. BMC Neurosci. 11:31. doi: 10.1186/1471-2202-11-31

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, H., Dou, J., Li, W., Cui, W., Mak, S., Hu, Q., et al. (2009). Promising multifunctional anti-Alzheimer's dimer bis(7)-Cognitin acting as an activator of protein kinase C regulates activities of α-secretase and BACE-1 concurrently. Eur. J. Pharmacol. 623, 14–21. doi: 10.1016/j.ejphar.2009.09.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, H., Li, W., Lao, Y., Luo, J., Lee, N. T. K., Kan, K. K. W., et al. (2006). bis(7)-tacrine attenuates β amyloid-induced neuronal apoptosis by regulating L-type calcium channels. J. Neurochem. 98, 1400–1410. doi: 10.1111/j.1471-4159.2006.03960.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, H., Li, W., Liu, Y., Lao, Y., Liu, W., Chen, C., et al. (2007). Mitochondrial proteomic analysis and characterization of the intracellular mechanisms of bis(7)-tacrine in protecting against glutamate-induced excitotoxicity in primary cultured neurons. J. Proteome Res. 6, 2435–2446. doi: 10.1021/pr060615g

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, H., Li, W., Luo, J., Lee, N. T. K., Li, M., Tsim, K. W. K., et al. (2008). Promising anti-Alzheimer's dimer bis(7)-tacrine reduces β-amyloid generation by directly inhibiting BACE-1 activity. Biochem. Biophys. Res. Commun. 366, 631–636. doi: 10.1016/j.bbrc.2007.11.068

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, R., Zhang, R., Chang, M., Peng, Y., Wang, P., Hu, S., et al. (2012). Reversal of scopolamine-induced spatial and recognition memory deficits in mice by novel multifunctional dimers bis-cognitins. Brain Res. 1470, 59–68. doi: 10.1016/j.brainres.2012.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y.-F., Wu, D.-C., Xiao, X.-Q., Chen, P. M. Y., Chung, W., Lee, N. T. K., et al. (2000). Protection against ischemic injury in primary cultured astrocytes of mouse cerebral cortex by bis(7)-tacrine, a novel anti-Alzheimer's agent. Neurosci. Lett. 288, 95–98. doi: 10.1016/S0304-3940(00)01198-8

CrossRef Full Text | Google Scholar

Hu, M.-K., Wu, L.-J., Hsiao, G., and Yen, M.-H. (2002). Homodimeric tacrine congeners as acetylcholinesterase inhibitors. J. Med. Chem. 45, 2277–2282. doi: 10.1021/jm010308g

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, S., Cui, W., Mak, S., Xu, D., Hu, Y., Tang, J., et al. (2015). Substantial neuroprotective and neurite outgrowth-promoting activities by bis(propyl)-cognitin via the activation of alpha7-nAChR, a promising anti-alzheimer's dimer. ACS Chem. Neurosci. 6, 1536–1545. doi: 10.1021/acschemneuro.5b00108

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, S.-Q., Cui, W., Mak, S.-H., Choi, C.-L., Hu, Y.-J., Li, G., et al. (2015a). Robust neuritogenesis-promoting activity by bis(heptyl)-cognitin through the activation of alpha7-nicotinic acetylcholine receptor/ERK pathway. CNS Neurosci. Ther. 21, 520–529. doi: 10.1111/cns.12401

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, S.-Q., Wang, R., Cui, W., Mak, S.-H., Li, G., Hu, Y.-J., et al. (2015b). Dimeric bis (heptyl)-cognitin blocks alzheimer's β-amyloid neurotoxicity Via the inhibition of Aβ fibrils formation and disaggregation of preformed fibrils. CNS Neurosci. Ther. 21, 953–961. doi: 10.1111/cns.12472

PubMed Abstract | CrossRef Full Text | Google Scholar

Inestrosa, N. C., Alvarez, A., Pérez, C. A., Moreno, R. D., Vicente, M., Linker, C., et al. (1996). Acetylcholinesterase accelerates assembly of amyloid-beta-peptides into alzheimer's fibrils: possible role of the peripheral site of the enzyme. Neuron 16, 881–891. doi: 10.1016/S0896-6273(00)80108-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y. S., and Yoon, B.-E. (2017). Altered GABAergic signaling in brain disease at various stages of life. Exp. Neurobiol. 26, 122–131. doi: 10.5607/en.2017.26.3.122

PubMed Abstract | CrossRef Full Text | Google Scholar

Krogsgaard-Larsen, P. (1992). GABA and glutamate receptors as therapeutic targets in neurodegenerative disorders. Pharmacol. Toxicol. 70, 95–104. doi: 10.1111/j.1600-0773.1992.tb00436.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Carlier, P. R., Ren, H., Kan, K. K. W., Hui, K., Wang, H., et al. (2007a). Alkylene tether-length dependent γ-aminobutyric acid type A receptor competitive antagonism by tacrine dimers. Neuropharmacology 52, 436–443. doi: 10.1016/j.neuropharm.2006.07.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C. Y., Wang, H., Xue, H., Carlier, P. R., Hui, K. M., Pang, Y. P., et al. (1999). bis(7)-tacrine, a novel dimeric AChE inhibitor, is a potent GABA(A) receptor antagonist. Neuroreport 10, 795–800.

PubMed Abstract | Google Scholar

Li, J., Lu, Z., Xu, L., Wang, Q., Zhang, Z., and Fang, J. (2014). Neuroprotective Effects of bis(7)-tacrine in a rat model of pressure-induced retinal ischemia. Cell Biochem. Biophys. 68, 275–282. doi: 10.1007/s12013-013-9707-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Lee, N. T. K., Fu, H., Kan, K. K. W., Pang, Y., Li, M., et al. (2006). Neuroprotection via inhibition of nitric oxide synthase by bis(7)-tacrine. Neuroreport 17, 471–474. doi: 10.1097/01.wnr.0000209014.09094.72

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Mak, M., Jiang, H., Wang, Q., Pang, Y., Chen, K., et al. (2009). Novel anti-alzheimer's dimer bis(7)-cognitin: cellular and molecular mechanisms of neuroprotection through multiple targets. Neurotherapeutics 6, 187–201. doi: 10.1016/j.nurt.2008.10.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, W., Pi, R., Chan, H. H. N., Fu, H., Lee, N. T. K., Tsang, H. W., et al. (2005). Novel dimeric acetylcholinesterase inhibitor bis(7)-tacrine, but Not donepezil, prevents glutamate-induced neuronal apoptosis by blocking N-Methyl-d-aspartate receptors. J. Biol. Chem. 280, 18179–18188. doi: 10.1074/jbc.M411085200

CrossRef Full Text | Google Scholar

Li, W., Xue, J., Niu, C., Fu, H., Lam, C. S. C., Luo, J., et al. (2007b). Synergistic neuroprotection by bis(7)-tacrine via concurrent blockade of N-Methyl-D-aspartate receptors and neuronal nitric-oxide synthase. Mol. Pharmacol. 71, 1258–1267. doi: 10.1124/mol.106.029108

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X.-Y., Zhang, J., Dai, J.-P., Liu, X.-M., and Li, Z.-W. (2010). Actions of bis(7)-tacrine and tacrine on transient potassium current in rat DRG neurons and potassium current mediated by KV4.2 expressed in xenopus oocyte. Brain Res. 1318, 23–32. doi: 10.1016/j.brainres.2009.12.047

CrossRef Full Text | Google Scholar

Lipton, S. A. (2004). Failures and successes of NMDA receptor antagonists: molecular basis for the use of open-channel blockers like memantine in the treatment of acute and chronic neurologic insults. NeuroRx 1, 101–110. doi: 10.1602/neurorx.1.1.101

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Ho, W., Lee, N. T., Carlier, P. R., Pang, Y., and Han, Y. (2000). bis(7)-tacrine, a novel acetylcholinesterase inhibitor, reverses AF64A-induced deficits in navigational memory in rats. Neurosci. Lett. 282, 165–168. doi: 10.1016/S0304-3940(00)00905-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., and Li, C. (2012). Inhibition of N-methyl-D-aspartate-activated current by bis(7)-tacrine in HEK-293 cells expressing NR1/NR2A or NR1/NR2B receptors. J. Huazhong Univ. Sci. Technol. Medical Sci. 32, 793–797. doi: 10.1007/s11596-012-1036-0

CrossRef Full Text | Google Scholar

Liu, Y.-W., Li, C.-Y., Luo, J.-L., Li, W.-M., Fu, H.-J., Lao, Y.-Z., et al. (2008a). bis(7)-tacrine prevents glutamate-induced excitotoxicity more potently than memantine by selectively inhibiting NMDA receptors. Biochem. Biophys. Res. Commun. 369, 1007–1011. doi: 10.1016/j.bbrc.2008.02.133

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y.-W., Luo, J.-L., Ren, H., Peoples, R. W., Ai, Y.-X., Liu, L.-J., et al. (2008b). Inhibition of NMDA-gated ion channels by bis(7)-tacrine: whole-cell and single-channel studies. Neuropharmacology 54, 1086–1094. doi: 10.1016/j.neuropharm.2008.02.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, J., da Costa, J., Ceschi, M., Gonçalves, C., Konrath, E., Karl, A., et al. (2017). Chiral bistacrine analogues: synthesis, cholinesterase inhibitory activity and a molecular modeling approach. J. Braz. Chem. Soc. 28, 2218–2228. doi: 10.21577/0103-5053.20170074

CrossRef Full Text | Google Scholar

Luo, J., Li, W., Liu, Y., Zhang, W., Fu, H., Lee, N. T. K., et al. (2007). Novel dimeric bis(7)-tacrine proton-dependently inhibits NMDA-activated currents. Biochem. Biophys. Res. Commun. 361, 505–509. doi: 10.1016/j.bbrc.2007.07.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, J.-L., Zhang, J., Guan, B.-C., Pang, Y.-P., Han, Y.-F., and Li, Z.-W. (2004). Inhibition by bis(7)-tacrine of 5-HT-activated current in rat TG neurons. Neuroreport 15, 1335–1338. doi: 10.1097/01.WNR.0000127075.51445.3E

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattson, M. P. (2000). Apoptosis in neurodegenerative disorders. Nat. Rev. Mol. Cell Biol. 1, 120–130. doi: 10.1038/35040009

PubMed Abstract | CrossRef Full Text | Google Scholar

Muñoz-Ruiz, P., Rubio, L., García-Palomero, E., Dorronsoro, I., del Monte-Millán, M., Valenzuela, R., et al. (2005). design, synthesis, and biological evaluation of dual binding site acetylcholinesterase inhibitors: new disease-modifying agents for Alzheimer's disease. J. Med. Chem. 48, 7223–7233. doi: 10.1021/jm0503289

PubMed Abstract | CrossRef Full Text | Google Scholar

Murphy, M. P., and LeVine, H. III (2010). Alzheimer's disease and the amyloid-beta peptide. J. Alzheimers Dis. 19, 311–323. doi: 10.3233/JAD-2010-1221

PubMed Abstract | CrossRef Full Text | Google Scholar

Newcomer, J. W., Farber, N. B., and Olney, J. W. (2000). NMDA receptor function, memory, and brain aging. Dialogues Clin. Neurosci. 2, 219–232.

PubMed Abstract | Google Scholar

Nie, H., Yu, W.-J., Li, X.-Y., Yuan, C.-H., Pang, Y.-P., Li, C.-Y., et al. (2007). Inhibition by bis(7)-tacrine of native delayed rectifier and KV1.2 encoded potassium channels. Neurosci. Lett. 412, 108–113. doi: 10.1016/j.neulet.2006.10.047

PubMed Abstract | CrossRef Full Text | Google Scholar

Nuss, P. (2015). Anxiety disorders and GABA neurotransmission: a disturbance of modulation. Neuropsychiatr. Dis. Treat. 11, 165–175. doi: 10.2147/NDT.S58841

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, S.-Y., Yu, Z.-L., Xiang, C.-J., Dong, H., Fang, H.-Y., and Ko, K.-M. (2009). Comparison studies of tacrine and bis(7)-tacrine on the suppression of scopolamine-induced behavioral changes and inhibition of acetylcholinesterase in mice. Pharmacology 83, 294–300. doi: 10.1159/000211668

CrossRef Full Text | Google Scholar

Pan, S.-Y., Zhang, Y., Guo, B., Han, Y.-F., and Ko, K.-M. (2011). Tacrine and bis(7)-tacrine attenuate cycloheximide-induced amnesia in mice, with attention to acute toxicity. Basic Clin. Pharmacol. Toxicol. 109, 261–265. doi: 10.1111/j.1742-7843.2011.00715.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pan, S. Y., Yu, Z. L., Dong, H., Lee, N. T. K., Wang, H., Fong, W. F., et al. (2007). Evaluation of acute bis(7)-tacrine treatment on behavioral functions in 17-day-old and 30-day-old mice, with attention to drug toxicity. Pharmacol. Biochem. Behav. 86, 778–783. doi: 10.1016/j.pbb.2007.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Pang, Y. P., Quiram, P., Jelacic, T., Hong, F., and Brimijoin, S. (1996). Highly potent, selective, and low cost bis-tetrahydroaminacrine inhibitors of acetylcholinesterase. Steps toward novel drugs for treating Alzheimer's disease. J. Biol. Chem. 271, 23646–23649.

PubMed Abstract | Google Scholar

Pérez-Hernández, J., Zaldívar-Machorro, V. J., Villanueva-Porras, D., Vega-Ávila, E., and Chavarría, A. (2016). A potential alternative against Neurodegenerative diseases: phytodrugs. Oxid. Med. Cell. Longev. 2016, 1–19. doi: 10.1155/2016/8378613

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, S., He, L., Mak, M., Han, Y., Ho, C.-Y., and Zuo, Z. (2014). Synthesis, biological activity, and biopharmaceutical characterization of tacrine dimers as acetylcholinesterase inhibitors. Int. J. Pharm. 477, 442–453. doi: 10.1016/J.IJPHARM.2014.10.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramsay, R. R., Majekova, M., Medina, M., and Valoti, M. (2016). Key targets for multi-target ligands designed to combat neurodegeneration. Front. Neurosci. 10:375. doi: 10.3389/fnins.2016.00375

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizzo, S., Bisi, A., Bartolini, M., Mancini, F., Belluti, F., Gobbi, S., et al. (2011). Multi-target strategy to address Alzheimer's disease: design, synthesis and biological evaluation of new tacrine-based dimers. Eur. J. Med. Chem. 46, 4336–4343. doi: 10.1016/j.ejmech.2011.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ros, E., Aleu, J., Gomez De Aranda, I., Cant,í, C., Pang, Y.-P., Marsal, J., et al. (2001). Effects of bis(7)-tacrine on spontaneous synaptic activity and on the nicotinic ACh receptor of Torpedo electric organ. J. Neurophysiol. 86, 183–189. doi: 10.1152/jn.2001.86.1.183

PubMed Abstract | CrossRef Full Text | Google Scholar

Santiago, J. A., Bottero, V., and Potashkin, J. A. (2017). Dissecting the molecular mechanisms of neurodegenerative diseases through network biology. Front. Aging Neurosci. 9:166. doi: 10.3389/fnagi.2017.00166

PubMed Abstract | CrossRef Full Text | Google Scholar

Shu, X.-J., Liu, W., Zhang, L., Yang, R., Yi, H.-L., Li, C.-L., et al. (2012). Effect of bis(7)-tacrine on cognition in rats with chronic cerebral ischemia. Neurosci. Lett. 512, 103–108. doi: 10.1016/j.neulet.2012.01.068

PubMed Abstract | CrossRef Full Text | Google Scholar

Teipel, S., Grothe, M. J., Zhou, J., Sepulcre, J., Dyrba, M., Sorg, C., et al. (2016). Measuring cortical connectivity in Alzheimer's disease as a brain neural network pathology: toward clinical applications. J. Int. Neuropsychol. Soc. 22, 138–163. doi: 10.1017/S1355617715000995

PubMed Abstract | CrossRef Full Text | Google Scholar

Trippier, P. C., Jansen Labby, K., Hawker, D. D., Mataka, J. J., and Silverman, R. B. (2013). Target- and mechanism-based therapeutics for neurodegenerative diseases: strength in numbers. J. Med. Chem. 56, 3121–3147. doi: 10.1021/jm3015926

PubMed Abstract | CrossRef Full Text | Google Scholar

Valera, E., and Masliah, E. (2016). Therapeutic approaches in Parkinson's disease and related disorders. J. Neurochem. 139, 346–352. doi: 10.1111/jnc.13529

PubMed Abstract | CrossRef Full Text | Google Scholar

Van der Schyf, C. J. (2011). The use of multi-target drugs in the treatment of neurodegenerative diseases. Expert Rev. Clin. Pharmacol. 4, 293–298. doi: 10.1586/ecp.11.13

PubMed Abstract | CrossRef Full Text | Google Scholar

Vassar, R., Kovacs, D. M., Yan, R., and Wong, P. C. (2009). The beta-secretase enzyme BACE in health and Alzheimer's disease: regulation, cell biology, function, and therapeutic potential. J. Neurosci. 29, 12787–12794. doi: 10.1523/JNEUROSCI.3657-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., Carlier, P. R., Ho, W. L., Lee, N. T., Pang, Y. P., and Han, Y. F. (1999a). Attenuation of scopolamine-induced deficits in navigational memory performance in rats by bis(7)-tacrine, a novel dimeric AChE inhibitor. Zhongguo Yao Li Xue Bao 20, 211–217.

PubMed Abstract | Google Scholar

Wang, H., Carlier, P. R., Ho, W. L., Wu, D. C., Lee, N. T., Li, C. P., et al. (1999b). Effects of bis(7)-tacrine, a novel anti-Alzheimer's agent, on rat brain AChE. Neuroreport 10, 789–793.

PubMed Abstract | Google Scholar

Wu, C., and Sun, D. (2015). GABA receptors in brain development, function, and injury. Metab. Brain Dis. 30, 367–379. doi: 10.1007/s11011-014-9560-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, X. Q., Lee, N. T., Carlier, P. R., Pang, Y., and Han, Y. F. (2000). bis(7)-tacrine, a promising anti-Alzheimer's agent, reduces hydrogen peroxide-induced injury in rat pheochromocytoma cells: comparison with tacrine. Neurosci. Lett. 290, 197–200. doi: 10.1016/S0304-3940(00)01357-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, H., Li, W.-M., Kan, K. K. W., Ho, J. M. K., Carlier, P. R., Pang, Y.-P., et al. (2008). The physicochemical properties and the in vivo AChE inhibition of two potential anti-Alzheimer agents, bis(12)-hupyridone and bis(7)-tacrine. J. Pharm. Biomed. Anal. 46, 75–81. doi: 10.1016/j.jpba.2007.08.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Z.-H., Liu, Y.-W., Jiang, F.-G., Tian, X., Zhu, Y.-H., Li, J.-B., et al. (2011). bis(7)-tacrine protects retinal ganglion cells against excitotoxicity via NMDA receptor inhibition. Int. J. Ophthalmol. 4, 125–130. doi: 10.3980/j.issn.2222-3959.2011.02.03

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, Y., Li, W., Chow, P. C. Y., Lau, D. T. K., Lee, N. T. K., Pang, Y., et al. (2008). bis(7)-tacrine, a promising anti-Alzheimer's dimer, affords dose- and time-dependent neuroprotection against transient focal cerebral ischemia. Neurosci. Lett. 439, 160–164. doi: 10.1016/j.neulet.2008.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, L., Liu, Y.-W., Peoples, R. W., Yang, M., Tian, X., Ai, Y.-X., et al. (2009). Mechanism of bis(7)-tacrine inhibition of GABA-activated current in cultured rat hippocampal neurons. Neuropharmacology 57, 33–40. doi: 10.1016/j.neuropharm.2009.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: B7C, Alzheimer's disease, AChE inhibitor, cognitive impairment, NMDA antagonist

Citation: Sánchez-Vidaña DI, Chow JKW, Hu SQ, Lau BWM and Han Y-F (2019) Molecular Targets of Bis (7)-Cognitin and Its Relevance in Neurological Disorders: A Systematic Review. Front. Neurosci. 13:445. doi: 10.3389/fnins.2019.00445

Received: 16 October 2018; Accepted: 18 April 2019;
Published: 09 May 2019.

Edited by:

Dietrich Ernst Lorke, Florida International University, United States

Reviewed by:

Ghulam Md Ashraf, King Abdulaziz University, Saudi Arabia
Diego Muñoz-Torrero, University of Barcelona, Spain

Copyright © 2019 Sánchez-Vidaña, Chow, Hu, Lau and Han. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Benson Wui Man Lau, benson.lau@polyu.edu.hk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.