Next Article in Journal
A Comprehensive Review of Genetically Engineered Mouse Models for Prader-Willi Syndrome Research
Next Article in Special Issue
Pharmacological Effects of Guava (Psidium guajava L.) Seed Polysaccharides: GSF3 Inhibits PC-3 Prostate Cancer Cell Growth through Immunotherapy In Vitro
Previous Article in Journal
Analysis of Gene Expression Changes in Plants Grown in Salty Soil in Response to Inoculation with Halophilic Bacteria
Previous Article in Special Issue
Biomimetic Chromatographic Studies Combined with the Computational Approach to Investigate the Ability of Triterpenoid Saponins of Plant Origin to Cross the Blood–Brain Barrier
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Main Alkaloids in Uncaria rhynchophylla and Their Anti-Alzheimer’s Disease Mechanism Determined by a Network Pharmacology Approach

Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(7), 3612; https://doi.org/10.3390/ijms22073612
Submission received: 15 March 2021 / Revised: 27 March 2021 / Accepted: 28 March 2021 / Published: 31 March 2021
(This article belongs to the Special Issue Biological Properties of Medicinal Plants)

Abstract

:
Alzheimer’s disease (AD) is a growing concern in modern society, and effective drugs for its treatment are lacking. Uncaria rhynchophylla (UR) and its main alkaloids have been studied to treat neurodegenerative diseases such as AD. This study aimed to uncover the key components and mechanism of the anti-AD effect of UR alkaloids through a network pharmacology approach. The analysis identified 10 alkaloids from UR based on HPLC that corresponded to 90 anti-AD targets. A potential alkaloid target-AD target network indicated that corynoxine, corynantheine, isorhynchophylline, dihydrocorynatheine, and isocorynoxeine are likely to become key components for AD treatment. KEGG pathway enrichment analysis revealed the Alzheimers disease (hsa05010) was the pathway most significantly enriched in alkaloids against AD. Further analysis revealed that 28 out of 90 targets were significantly correlated with Aβ and tau pathology. These targets were validated using a Gene Expression Omnibus (GEO) dataset. Molecular docking studies were carried out to verify the binding of corynoxine and corynantheine to core targets related to Aβ and tau pathology. In addition, the cholinergic synapse (hsa04725) and dopaminergic synapse (hsa04728) pathways were significantly enriched. Our findings indicate that UR alkaloids directly exert an AD treatment effect by acting on multiple pathological processes in AD.

1. Introduction

Alzheimer’s disease (AD), the most common form of dementia in the elderly, is characterized by the progressive deterioration of cognitive function and memory [1]. Approximately 50 million people were affected by AD worldwide in 2019, and nearly 10 million new cases occur every year [2]. The Chinese Center for Disease Control and Prevention estimated that there are more than 9 million AD patients in China at present, and it is expected that 40 million could be affected by 2050. AD not only brings a heavy mental burden and economic pressure to the family members and friends of patients but also places a very large burden on the social economy [3]. At present, the typical events in AD pathogenesis are considered to be the formation of extracellular amyloid-β (Aβ) plaques, intracellular accumulation of abnormally phosphorylated tau, neuronal synaptic dysfunction, and neuronal loss [4,5,6,7]. However, over the last decade, all clinical trials targeting a single target, such as Aβ or tau, have failed, and there are also no effective drugs for the prevention or treatment of AD in the clinic [8]. The anti-AD drugs currently approved by the Food and Drug Administration are cholinesterase inhibitors: donepezil, rivastigmine, tacrine, galantamine, and the N-methyl D-aspartate (NMDA) receptor antagonist memantine [9]. These drugs are symptomatic treatments and cannot halt the disease course. Moreover, the above drugs also cause a variety of unfavorable effects, including hypertensive crisis, nausea, diarrhea, and vomiting [10]. Given that multiple pathologies are associated with AD, the development of novel therapeutics that target these multiple pathologies is desirable in AD treatment. Therefore, the search for safer and effective drugs for the treatment of AD is extremely urgent.
Chinese herbal medicine has been widely used for dementia [11]. Uncaria rhynchophylla (UR), known as “Gou-Teng” in Chinese, has been used in traditional medicine in Asia, Africa and South America. Traditional Chinese medicine products or formulae containing UR have been used mainly to treat cardiovascular and central nervous system diseases, such as neurodegenerative diseases, lightheadedness, dizziness, convulsions, numbness, and hypertension [12,13,14,15]. Clinically, phytochemicals containing UR, such as Yigan-san (Yokukansan in Japanese), Tianmagouteng granules, and Gouteng-san, have become better herbal medicines for the treatment of stroke, hypertension, and chronic headache in China and Japan. The effects of UR are largely attributed to its predominant active component alkaloids. Therefore, the protective effects of UR and its main alkaloids on the central nervous system diseases have become a focus of research in recent decades.
UR is enriched in alkaloids, including rhynchophylline, isorhynchophylline, corynoxeine, isocorynoxeine, and hirsutine [16,17]. The total alkaloid content in UR is approximately 0.2%, among which rhynchophylline accounts for 28–50%, and isorhynchophylline accounts for 15% [18]. In 5xFAD mice, a transgenic mouse model of AD, UR ethanol extract (including rhynchophylline) significantly reduced Aβ aggregation and ameliorated AD-related pathologies (neuronal loss, synaptic degeneration, neuroinflammation, and neurogenesis) [19]. Rhynchophylline (bilateral hippocampal injection, 100 μM, 2 μL) was shown to improve soluble Aβ1-42-induced impairment of spatial cognition function by inhibiting excessive activation of extrasynaptic NR2B-containing NMDA receptors [20]. Treatment with isorhynchophylline (20 or 40 mg/kg/day) by gavage for 21 days improved Aβ25-35-induced cognitive impairment in rats via inhibition of neuronal apoptosis and tau protein hyperphosphorylation [21]. A recent study suggested that isorhynchophylline (20 or 40 mg/kg/day) treatment for 4 months improved cognitive impairment in TgCRND8 transgenic mice by reducing Aβ generation and deposition, tau hyperphosphorylation and neuroinflammation [22]. Moreover, 8 weeks of oral treatment with isorhynchophylline (20 or 40 mg/kg/day) could ameliorate D-galactose-induced learning and memory impairments by enhancing the antioxidant status and anti-inflammatory effect of D-galactose in brain tissues via NF-κB signaling [23]. Most of the mentioned studies focus on the therapeutic effects of UR alkaloids, only a few reports have shown that UR alkaloids can affect MAPT, GSK3B, AKT, BCL2, ect. Despite a series of studies on the biological activities of alkaloids in UR, the potential targets and underlying molecular mechanism of UR alkaloids in AD remain largely unclear. Due to the good curative effects of UR alkaloids on AD in animal researches and a variety of pathological processes involved, we therefore hypothesized that UR alkaloids possibly exert an AD treatment effect by acting on multiple pathological processes in AD.
As an emerging approach for drug discovery, network pharmacology combines network biology and multipharmacology [24]. This study aimed to identify effective target proteins of UR alkaloids and uncover the mechanism of the anti-AD effect of UR alkaloids through a network pharmacology approach, microarray data analysis and molecular docking. Unlike other studies, we focus primarily on UR alkaloids, the predominant active ingredient of the UR. A scheme of the study protocol is shown in Figure 1. We obtained UR alkaloids from a recent study based on high-performance liquid chromatography (HPLC) [17], this increases the quality of this study significantly. In addition to enrichment analyses, an AD database was used to analyze the correlation between the targets of UR alkaloids against AD and AD pathology in this study. Furthermore, we also used the human high-throughput omics data to validate the targets of UR alkaloids against AD. Our study offers new insight into the mechanisms of UR alkaloids and provides a more specific and effective treatment for AD.

2. Results

2.1. The Chemical Structure and ADME Properties of the Main Alkaloids from UR

This study primarily focused on the therapeutic effect of UR alkaloids against AD. A recent study identified 10 alkaloids from UR based on HPLC [17]. The extraction rate of the UR alkaloids (100 g) was 9.4%. The chemical structures of the main alkaloids were obtained from the PubChem database and are shown in Figure 2. Next, we used an online tool SwissADME for an in-depth evaluation of the ADME-related properties of the main alkaloids from UR. SwissADME prediction showed that all the main alkaloids satisfied Lipinski’s rule of five, and other chemical and pharmacological properties, including topological polar surface area (TPSA) and solubility (LogS), were also evaluated (Table 1). These results indicate that these alkaloids may exhibit good permeability across cell membranes.

2.2. Screening of Targets of the Main Alkaloids from UR in AD

Potential targets of the 10 alkaloids were predicted via the SwissTargetPrediction database based on their structure, and a total of 365 targets were obtained. Results from the GeneCards, DrugBank, Therapeutic Target Database (TTD), and Chemogenomics Database for Alzheimer’s Disease identified a total of 622 targets relevant to AD. Ultimately, a Venn diagram was used to summarize 90 common targets associated with both UR alkaloids and AD for further analysis (Figure 3A). Detailed information about these common targets is provided in Table 2. Furthermore, these 90 target proteins of UR alkaloids against AD were categorized into 6 different classes based on their cellular functions, of which protein-modifying enzyme (PC00260, 34.4%) was the most enriched class (Figure 3B). Among these protein-modifying enzymes, AKT1, CDK1, CDK5, EIF2AK3, GSK3B, MAPK10, MTOR and DYRK1A are nonreceptor serine/threonine protein kinases, and ECE1, MME, MMP2, MMP3 and MMP9 are metalloproteases (Figure 3C). The above results indicated that alkaloids from UR can protect against AD through multiple targets and biological functions.

2.3. PPI Analysis of Targets of the Main Alkaloids against AD

To explore the relationship between these 90 targets of alkaloids against AD, protein–protein interaction (PPI) analysis was performed using the STRING database [25]. A PPI network with a total of 90 nodes and 610 edges and an average node degree of 13.6 was generated (Figure 3D). The darker the color and the larger the node were, the greater the degree was. AKT1, CASP3, MTOR, DRD2, HTR1A, MMP9, PTGS2, CCND1, GRIN2B, and DRD4, which are ranked by degree, were identified as core targets (Figure 3D). Among these, AKT1 showed the highest degree (49). This demonstrated that these core targets are closely related to other targets in the PPI network, suggesting that these targets may play important roles in AD treatment.

2.4. Clusters of Common Targets of the Main Alkaloids against AD

Six clusters were found after the PPI network of the main alkaloids against AD was analyzed through Molecular Complex Detection (MCODE) (k-core = 2). This demonstrates that these clusters may be the most relevant to AD treatment. The details are provided in Figure 4. Cluster 1 contains 13 nodes and 78 edges with a score of 13. The seed node of this cluster is somatostatin receptor 3 (SSTR3), which regulates diverse cellular functions, such as neurotransmission, cell proliferation, and endocrine signaling (Figure 4A). Cluster 2 contains 14 nodes and 53 edges with a score of 8.15. The seed node of this cluster is MET proto-oncogene receptor tyrosine kinase (MET), which encodes a member of the receptor tyrosine kinase family of proteins and the product of the proto-oncogene MET (Figure 4B). Cluster 3 contains 16 nodes and 30 edges with a score of 4. The seed node of this cluster is butyrylcholinesterase (BCHE), which contributes to inactivation of the neurotransmitter acetylcholine and degrades neurotoxic organophosphate esters (Figure 4C). Cluster 4 contains 6 nodes and 10 edges with a score of 4. The seed node of this cluster is neurotrophic receptor tyrosine kinase 1 (NTRK1) (Figure 4D). Cluster 5 contains 4 nodes and 5 edges with a score of 3.33. The seed node of this cluster is aph-1 homolog B (APH1B) (Figure 4E). Cluster 6 contains 7 nodes and 10 edges with a score of 3.33. The seed node of this cluster is cytochrome P450 family 2 subfamily D member 6 (CYP2D6) (Figure 4F). The respective MCODE scores are listed in Figure 4G.

2.5. Construction of a Main Potential Alkaloid Target-AD Target Network

To explore the therapeutic mechanism of the main alkaloids in the treatment of AD, 90 common targets and 10 main alkaloids from UR were used to construct a main potential alkaloid target-AD target network (Figure 5). All of these alkaloids are related to multiple targets, resulting in 295 component-target associations between 10 alkaloids and 90 targets. The average number of targets per alkaloid was 29.5, and the mean degree of components per target was 3.3. This clearly indicates that UR fits the multicomponent and multitarget characteristics of traditional Chinese medicine. Corynoxine (degree = 48) had the most targets, followed by corynantheine (degree = 39), isorhynchophylline (degree = 33), dihydrocorynatheine (degree = 29), isocorynoxeine (degree = 27), and hirsuteine (degree = 27), indicating that these alkaloids from UR are highly likely to become key components in AD treatment.

2.6. Potential Synergistic Mechanism of the 10 Alkaloids against AD

2.6.1. Gene Ontology (GO) Enrichment Analysis

The common targets between UR alkaloids and AD were further analyzed for functional prediction by Metascape. The primary enriched GO biological process (BP) terms were transsynaptic signaling (GO:0099537), positive regulation of kinase activity (GO:0033674), regulation of ion transport (GO:0043269), cellular response to nitrogen compound (GO:1901699), positive regulation of cell death (GO:0010942), response to inorganic substance (GO:0010035), and response to ammonium ion (GO:0060359) (Figure 6A). In particular, 7 out of 10 targets involved in transsynaptic signaling were core targets (AKT1, DRD2, DRD4, GRIN2B, HTR1A, MTOR, and PTGS2). These findings implicated the involvement of multiple targets and various biological processes in the multiple synergistic effects of UR alkaloids against AD.

2.6.2. Pathway Analysis to Explore the Therapeutic Mechanisms of the Main Alkaloids in AD

The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database is a collection of manually drawn pathway maps of molecular interactions. To identify signaling pathways associated with targets of the main alkaloids against AD, Metascape was used to identify pathways enriched in these 90 targets. The involved KEGG pathways were mainly the Alzheimer disease (hsa05010), serotonergic synapse (hsa04726), the calcium signaling pathway (hsa04020), the PI3K-Akt signaling pathway (hsa04151), dopaminergic synapse (hsa04728), the neurotrophin signaling pathway (hsa04722), the notch signaling pathway (hsa04330), and cholinergic synapse (hsa04725) (Figure 6B). Detailed information on the KEGG pathway enrichment analysis is shown in Table 3. The most significantly enriched pathway, the Alzheimer disease pathway (hsa05010, p = 2.4E-18), belongs to the Human Diseases category pathway, and is mainly related to the production and clearance of Aβ and aberrant tau hyperphosphorylation (Figure 6C). A network of the targets and pathways enriched in the 10 UR alkaloids against AD is shown in Figure 7 and contains 90 nodes, including the top 20 KEGG pathways associated with 70 targets and 163 edges. Based on the results of pathway analysis, it was found that these pathways are closely related to AD, axons, and synapses. In particular, the PI3K-Akt signaling pathway (hsa04151) exhibits the highest number of target connections (degree = 17), and its targets include AKT1, CCND1, ERBB2, MTOR, GSK3B, MET, NOS3, and PDGFRB. Next, the Alzheimer disease pathway (hsa05010) exhibits the second largest number of degrees (16), and the associated targets include ADAM17, BACE1, CASP3, CDK5, GSK3B, PSEN1, PSEN2, PSENEN, and NOS1.

2.7. Bioinformatics Analysis of Targets of the Alkaloids Related to Aβ and Tau Pathology

To further verify the relationship between the potential targets of the alkaloids and Aβ and tau pathology, the AlzData database [26] was used. Among the targets, 28 out of 90 were significantly correlated with tau, Aβ, or Aβ and tau (Figure 8A). Specifically, ADAM17, BACE1, CAPN1, EIF2AK3, GRIN2B, HTR4, MERTK, NOS2, and PDE4D were significantly correlated with Aβ pathology; CCND1, CHRM2, ECE1, HTR1A, and so on were significantly correlated with tau pathology; and ADRB2, CASP1, CCR5, CDK5, CHRNB2, CSF1R, GSK3B, MMP2, NTRK2, and P2RX7 were significantly associated with both Aβ and tau pathology. To construct a PPI network, the gene IDs of the 28 targets were input into the STRING database. A total of 25 nodes and 45 edges were found in the PPI network, and CCR5, CDK5, HTR1A, GSK3B, CCND1, NTRK2, MMP2, GRIN2B, MMP3, and CHRM2 were identified as core targets ranked by degree (Figure 8B).
The 28 targets were further analyzed for functional prediction by Metascape. GO BP terms primarily enriched in the targets included the response to amyloid-beta (GO:1904645), synaptic signaling (GO:0099536), the transmembrane receptor protein tyrosine kinase signaling pathway (GO:0007169), positive regulation of kinase activity (GO:0033674), second messenger-mediated signaling (GO:0019932), and positive regulation of cell death (GO:0010942) (Figure 8C). In particular, synaptic signaling (GO:0099536) exhibited the highest number of target connections (degree = 11), and the targets included ADRB2, BACE1, CDK5, CHRM2, CHRNB2, GRIN2B, GSK3B, HTR1A, HTR4, NTRK2, and P2RX7. In the KEGG pathway analysis, targets related to the MAPK signaling pathway (hsa04010), endocytosis (hsa04144), the NOD-like receptor signaling pathway (hsa04621), apoptosis (hsa04210), the Ras signaling pathway (hsa04014), and the PI3K-Akt signaling pathway (hsa04151) were highly enriched (Figure 8D). ADAM17, BACE1, CAPN1, CDK5, EIF2AK3, GRIN2B, and GSK3B were enriched in the Alzheimer disease pathway (hsa05010). Normalized expression targets of the main alkaloids against AD in the control and AD groups of the Gene Expression Omnibus (GEO) dataset were analyzed by the “Differential expression” module of the AlzData database (Figure 9). Of the targets, ADRB2, CASP1, CCR5, CSF1R, MERTK, MMP2, and P2RX7 were significantly upregulated, and CDK5, CHRNB2, GRIN2B, GSK3B, and MAPK10 were significantly downregulated in AD patients compared to controls. The above results indicated that the alkaloid targets are intimately associated with the pathology of Aβ and tau.

2.8. Molecular Docking Simulation

From the main potential alkaloid target-AD target network (Figure 5), corynoxine (degree = 48) and corynantheine (degree = 39) had the highest number of targets against AD. Therefore, molecular docking analysis was used to validate the binding of corynoxine and corynantheine to core targets related to AD pathology. Delta G is defined as the binding energy based on the ensemble free energy; the greater the absolute value of Delta G is, the more stable binding is. The results of molecular docking of corynoxine and corynantheine to core targets are shown in Table 4. Corynoxine and corynantheine exhibited strong binding to all core targets. Among these targets, corynoxine showed the highest binding energy with CCR5, MMP3, CCND1, and NTRK2, with delta G values of −9.28, −8.52, −8.17 and −8.10 kcal/mol, respectively; corynantheine showed the highest binding energy with CHRM2, MMP3, MMP2, GRIN2B, with delta G values of −9.38, −9.30, −9.24, and −9.02 kcal/mol, respectively.

3. Discussion

As the predominant active pharmacological components in UR, UR alkaloids have been widely used as an intervention for neurodegenerative diseases in animal models for several years [12,15,17,19,20,21]. UR alkaloids are mainly administered by oral administration of monomers in the experiments above mentioned. Unfortunately, the potential targets UR alkaloids are not clear at present. This study comprehensively investigated the therapeutic effect of UR alkaloids on AD.
Generally, network pharmacology studies use public databases to obtain the main components in traditional Chinese Medicine. Conventional screening methods of public databases do not take into account the content and distribution specificity of components, which will lead to a large number of non-specific components to be screened out. For instance, beta-sitosterol exists widely in 567 kinds of herbs and stigmasterol 284 kinds of herbs (data from HERB, http://herb.ac.cn/ (accessed on 30 March 2021)). UR alkaloids have significant pharmacological activities and are also representative active ingredients in UR. Accordingly, alkaloids in UR were obtained by HPLC based on a recent study [17], this greatly improved the quality of the data collected. In the present study, all of the 10 alkaloids in UR were in agreement with Lipinski’s rule of five, demonstrating that the alkaloids have acceptable pharmacokinetic properties. Furthermore, 90 potential targets of these 10 alkaloids against AD were obtained using network pharmacology strategies. A potential alkaloid target-AD target network indicated that corynoxine, corynantheine, isorhynchophylline, dihydrocorynatheine, isocorynoxeine, and hirsuteine are likely to become key components for AD treatment. Isorhynchophylline, isocorynoxeine, and hirsuteine showed high permeability, with apparent permeability coefficient values for bidirectional transport at the 10−5 cm/s level [27]. Corynoxine, a natural neuroprotective autophagy enhancer [28], has the greatest number of therapeutic targets for AD (degree = 48). Corynoxine can regulate several kinases, including RPS6KB1, MAP2K2, and PLK1, contributing to the clearance of AD-associated β-amyloid precursor protein (APP) and Parkinson disease-associated α-synuclein [29]. The molecular docking results also showed that corynoxine strongly binds CCR5, MMP3, CCND1, and NTRK2. Treatment with isorhynchophylline (20 or 40 mg/kg/day) could ameliorate Aβ25-35- or D-galactose-induced cognitive impairment in animal models [21,23]. Furthermore, in vitro experiments demonstrated that isorhynchophylline and rhynchophylline (pretreating 100 μM for 2 h) significantly decreased Aβ-induced cell death, intracellular calcium overload, and tau protein hyperphosphorylation in PC12 cells [30]. Notably, there have been few studies on the treatment of AD with corynantheine and dihydrocorynatheine until now. For future research, more attention might be paid to these alkaloids in the treatment of AD.
Senile plaques (SPs), neurofibrillary tangles (NFTs), neuronal synaptic dysfunction and neuronal loss are characteristic pathologic changes in AD [7,31]. KEGG pathway enrichment analysis revealed that the Alzheimer disease pathway (hsa05010) was the most significantly enriched pathway in UR alkaloids against AD. Among the associated targets, ADAM17 (an α-secretase), BACE1 (a β-secretase), APH1B, NCSTN, PSEN1, and PSENEN (a γ-secretase) are involved in APP cleavage. In AD brains, ADAM17-positive neurons often colocalize with amyloid plaques and are considered potential therapeutic targets for AD [32]. Moreover, ADAM17 is also involved in the cleavage of many other membrane-bound proteins, especially some inflammatory factors related to microglial activation [33]. BACE1 is a rate-limiting enzyme for Aβ production. The inhibition of BACE1 activity could block one of the earliest pathologic events in AD. Several BACE1 inhibitor drug candidates have advanced to phase 3 clinical trials [34]. γ-Secretase activity is mediated by a multiprotein complex consisting of Presenilin, APH1, PEN2 and Nicastrin (NCT) [35]. PSEN1 mutations are responsible for the majority of familial AD cases, and more than 300 mutations in PSEN1 have been reported [36]. Thus, UR alkaloids could decrease Aβ generation and deposition in the brain through several important drug targets. In addition, targets for regulating tau phosphorylation, such as CAPN1, CDK5 and GSK3B, are involved in the anti-AD effects of the UR alkaloids. CDK5 and GSK3B belong to the nonreceptor serine/threonine protein kinase family. GSK3B is a major tau kinase involved in the development of AD tau pathology [37]. A previous study showed that Aβ accumulation in the AD brain can activate kinases that promote tau phosphorylation, including GSK3B [38]. Interestingly, apart from the targets related to the Alzheimer’s disease pathway, up to 28 out of 90 targets were significantly correlated with tau, Aβ, or Aβ and tau. Among these targets, the response to amyloid-beta (GO:1904645) was the most significantly enriched, and the synaptic signaling term (GO:0099536) exhibited the highest number of target connections. CCR5, a cytokine belonging to the β chemokine receptor family of integral membrane proteins, is significantly positively associated with both Aβ and tau pathology. CCR5 expression is strongly related to microglia and inflammation, which accelerate the development of AD [39]. Of the 28 targets related to Aβ and tau pathology, CCR5 had the highest degree in the PPI network and was significantly increased in the hippocampus of AD patients. We should pay more attention to CCR5 in future studies. In addition to affecting Aβ and tau pathology, a series of synaptic-related KEGG pathways, such as serotonergic synapses (hsa04726), dopaminergic synapses (hsa04728), and cholinergic synapses (hsa04725), were also enriched. Cholinergic synapses are ubiquitous in the human central nervous system. The cholinergic hypothesis of AD centers on the progressive loss of limbic and neocortical cholinergic innervation [40]. Cholinesterase inhibitors increase the availability of acetylcholine at synapses in the brain and are one of the few drug therapies that have been proven clinically useful in the treatment of AD, thus validating the cholinergic system as an important therapeutic target in this disease. Overall, our findings indicate that UR alkaloids can directly treat AD by acting on multiple pathological processes in AD.
Akt is a serine/threonine protein kinase and an important kinase downstream of PI3K. There are three AKT isoforms (AKT1-3), among which AKT1 is the most important subtype. In this study, the PI3K-Akt signaling pathway (hsa04151) was found by KEGG pathway enrichment analysis to be an important pathway by which UR alkaloids counteract AD, and 17 targets are involved (including the core targets AKT1, CCND1, and MTOR). Among signal transduction pathways, this pathway is a key mediator that regulates cell growth, inflammation, metabolism, and cell survival in response to growth factors. Previous studies have found that Aβ oligomers inhibit the PI3K-Akt pathway, which leads to neuronal death [41]. In the AD brain, normalized pT308 AKT1 was positively correlated with both the amyloid burden and tau tangle density [42]. The PI3K-Akt signaling pathway plays an important role in GSK3B activity regulation, as AKT promotes the phosphorylation of GSK3B, resulting in GSK3B inactivation [43,44]. Furthermore, the core target CASPS (degree = 38) was also involved in AD treatment. CASP3 is a pivotal executioner of apoptosis that plays a major role in neuronal death during nervous system development and under certain pathological conditions. Neuronal cell injury and loss are major neuropathological features of AD. The level of activated CASP3 was elevated in the brains of patients with severe definitive AD [45]. The positive regulation of cell death (GO:0010942) and neuron death (GO:0070997) were also significantly enriched in the present study.
A total of 10 alkaloids in UR and 90 common targets against AD were screened by network pharmacology analysis. GO enrichment and KEGG pathway enrichment analyses suggested that UR alkaloids can directly treat AD by acting on multiple AD pathological processes, such as processes involving Aβ and tau, neuronal synaptic function, and neuronal loss. However, clinical studies are necessary to verify these theoretical observations in the future. The bioavailability of UR alkaloids after oral administration was not reported in the clinic. The oral bioavailability of rhynchophylline, isorhynchophylline, hirsutine, and angustidine was 41.82%, 12.71%, 34.44%, and 51.85%, respectively, based on the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP, https://tcmspw.com/tcmsp.php (accessed on 30 March 2021)). It is necessary to further optimize the structure or improve the dosage form to increase the oral bioavailability for its clinical applicability. Taken together, our findings might provide a theoretical basis for the use of UR alkaloids as a therapeutic for AD.

4. Materials and Methods

4.1. Collection of the Main Alkaloids from UR and ADME Evaluation

The main alkaloids in UR were obtained from a recent study based on HPLC [17]. Lipinski’s rule of five was used to assess the in vivo absorption abilities of the designed compounds [46,47]. Compounds that meet the requirements of Lipinski’s rule of five seem to be more likely to become a drug. Specifically, Lipinski’s rule of five includes the following: a molecule weight (MW) < 500, number of hydrogen bond donors (Hdon) ≤ 5, number of hydrogen bond acceptors (Hacc) ≤ 10, lipid–water partition coefficient (LogP) ≤ 5 and number of rotatable bonds (Rbon) ≤ 10. The SwissADME web tool (http://www.swissadme.ch (accessed on 30 March 2021)) [48,49] was used to evaluate the ADME of the compounds. Moreover, the TPSA, LogS, and skin permeation (Log Kp) were also measured.

4.2. Target Fishing

4.2.1. Collection of the Main Alkaloid-Related Targets

All chemical structures and canonical SMILES strings for the main alkaloids were obtained from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 30 March 2021)) [50]. To obtain the targets of the main alkaloids in UR, SwissTargetPrediction (http://www.swisstargetprediction.ch/ (accessed on 30 March 2021)) [51] was employed.

4.2.2. Screening of Targets of the Main Alkaloids against AD

Disease targets were collected from the GeneCards database (https://www.genecards.org/ (accessed on 30 March 2021)), DrugBank database (https://go.drugbank.com/ (accessed on 30 March 2021)) [52], TTD (http://bidd.nus.edu.sg/group/cjttd/ (accessed on 30 March 2021)) [53], and Chemogenomics Database for Alzheimer’s Disease (https://www.cbligand.org/AD/mainpage.php (accessed on 30 March 2021)) using “Alzheimer’s disease” as a key phrase, and duplicate targets were removed using Microsoft Excel software. The intersection of UR alkaloid-related targets and AD-related targets was assessed by Venny 2.1 (https://bioinfogp.cnb.csic.es/tools/venny/index.html (accessed on 30 March 2021)). Common targets represent the target of UR alkaloids against AD. In addition, protein classification was performed using the Panther classification system (http://pantherdb.org/ (accessed on 30 March 2021)) [54].

4.3. PPI Network Construction and Clustering Analysis

A PPI network was constructed by using the STRING database version 11.0 (https://string-db.org/ (accessed on 30 March 2021)) [25]. The organism was set to Homo sapiens, and only the minimum required interaction score > 0.4 was chosen as significant. PPI networks consist of nodes, which represent a target protein, and edges, which represent protein–protein interactions. The thickness of an edge represents the combined score. Degree refers to the number of other nodes directly connected to a node. The higher the degree is, the more important the node is. Core targets were identified through network analysis using Cytoscape software (v.3.7.1) [55] and its plugin (Network Analysis). In the present study, the top 10 proteins ranked by degree were selected and identified as core targets.
The Cytoscape plugin MCODE [56] was applied to analyze clustering modules in the PPI network. The MCODE criteria for selection were as follows: degree cutoff = 2, node score cutoff = 0.2, k-core = 2, and max depth = 100. A node with the highest weighted vertex was defined as a seed node, the key target of this cluster, by MCODE. Moreover, a potential alkaloid target-AD target network was constructed using Cytoscape software.

4.4. GO and KEGG Pathway Enrichment Analyses

Metascape (https://metascape.org/gp (accessed on 30 March 2021)) [57] is a comprehensive tool for gene annotation and enrichment analysis. GO biological process and KEGG pathway enrichment analyses were performed using Metascape. The enriched terms with p < 0.01, a minimum count of 3, and an enrichment factor > 1.5 were considered significant. The top 20 enriched terms were visualized using an online tool (www.bioinformatics.com.cn (accessed on 30 March 2021)). A target-enriched KEGG pathway network for the main alkaloids against AD was also constructed by Cytoscape software. Red nodes represent enriched KEGG pathways, and brown nodes represent target proteins.

4.5. Analysis of Alkaloid Targets Related to AD Pathology

AlzData (http://www.alzdata.org/ (accessed on 30 March 2021)) [26] is an AD database that collects current high-throughput omics data. Gene symbols for the target proteins of UR alkaloids against AD were given as input to the AlzData database for correlation analysis of AD pathology (Aβ and tau). Microsoft Excel was then used to collate the obtained results. The targets of the UR alkaloids related to AD pathology were used to conduct further GO and KEGG pathway enrichment analyses. Normalized expression targets of alkaloids against AD in the control and AD groups of the GEO dataset were analyzed by the “Differential expression” module of AlzData. GraphPad Prism software was used for graphical visualization. Values are presented as the mean ± SD.

4.6. Molecular Docking

To validate the binding of UR alkaloids on predicted core targets, the 3D molecular structure of UR alkaloids was retrieved from the PubChem database and the structure files of target proteins were acquired from the RCSB Protein Data Bank (PDB database, http://www.rcsb.org/ (accessed on 30 March 2021)) [58]. Molecular docking calculations were performed using the SwissDock web service (http://www.swissdock.ch/docking (accessed on 30 March 2021)) [59].

5. Conclusions

In this study, a network-based computational strategy and molecular docking were used to uncover the pharmacological mechanism of the UR alkaloids on AD. A total of 10 alkaloids in UR and 90 common targets against AD were screened by network pharmacology. GO enrichment analysis and KEGG pathway enrichment analysis suggested that UR alkaloids exerted the effect of treating AD directly act on multiple AD pathological processes such as Aβ overproduction, tau hyperphosphorylation, synaptic dysfunction, and neuronal loss. Taken together, our findings might provide a theoretical basis for the use of UR alkaloids as a therapeutic for AD.

Author Contributions

Conceptualization, P.Z. and Q.T.; investigation, P.Z.; methodology, X.-M.W., C.-Y.Y., and H.-F.S.; supervision, X.-M.W. and C.-Y.Y.; visualization, P.Z. and H.-F.S.; writing—original draft, P.Z.; writing—review and editing, Q.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (91539112) and the Health Committee of Hubei Province (ZY2021M041).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Processed data is contained within the article. Raw data is available from the corresponding author upon request.

Conflicts of Interest

The authors declare that there are no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

ADAlzheimer’s disease
APH1B Aph-1 homolog b
APPβ-amyloid precursor protein
amyloid beta
BCHEButyrylcholinesterase
BPBiological process
CYP2D6Cytochrome p450 family 2 subfamily d member 6
GEOGene Expression Omnibus
GOGene Ontology
HaccHydrogen bond acceptors
HdonHydrogen bond donors
HPLCHigh-performance liquid chromatography
KEGGKyoto Encyclopedia of Genes and Genomes
MCODEMolecular complex detection
METMet proto-oncogene receptor tyrosine kinase
MWMolecule weight
NCTNicastrin
NFTNeurofibrillary tangles
N-methyl D-aspartateNmda
NTRK1Neurotrophic receptor tyrosine kinase 1
PPIProtein–protein interaction
RbonRotatable bonds
SPSenile plaques
SSTR3Somatostatin receptor 3
TCMSPTraditional Chinese Medicine Systems Pharmacology Database and Analysis Platform
TPSATopological polar surface area
TTDTherapeutic target database
URUncaria rhynchophylla

References

  1. McKhann, G.; Drachman, D.; Folstein, M.; Katzman, R.; Price, D.; Stadlan, E.M. Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984, 34, 939–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zhang, F.; Zhong, R.J.; Cheng, C.; Li, S.; Le, W.D. New therapeutics beyond amyloid-beta and tau for the treatment of Alzheimer’s disease. Acta Pharmacol. Sin. 2020, 1–8. [Google Scholar] [CrossRef]
  3. Guo, Y.; Yang, H.; Huang, Z.; Tian, S.; Li, Q.; Du, C.; Chen, T.; Liu, Y.; Sun, H.; Liu, Z. Design, Synthesis, and Evaluation of Acetylcholinesterase and Butyrylcholinesterase Dual-Target Inhibitors against Alzheimer’s Diseases. Molecules 2020, 25, 489. [Google Scholar] [CrossRef] [Green Version]
  4. Wallace, R.A.; Dalton, A.J. What can we learn from study of Alzheimer’s disease in patients with Down syndrome for early-onset Alzheimer’s disease in the general population? Alzheimers Res. Ther. 2011, 3, 13. [Google Scholar] [CrossRef] [Green Version]
  5. Asaad, M.; Lee, J.H. A guide to using functional magnetic resonance imaging to study Alzheimer’s disease in animal models. Dis. Model. Mech. 2018, 11, dmm031724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sheng, J.G.; Zhou, X.Q.; Mrak, R.E.; Griffin, W.S. Progressive neuronal injury associated with amyloid plaque formation in Alzheimer disease. J. Neuropathol. Exp. Neurol. 1998, 57, 714–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Scheltens, P.; Blennow, K.; Breteler, M.M.; de Strooper, B.; Frisoni, G.B.; Salloway, S.; Van der Flier, W.M. Alzheimer’s disease. Lancet 2016, 388, 505–517. [Google Scholar] [CrossRef]
  8. Mehta, D.; Jackson, R.; Paul, G.; Shi, J.; Sabbagh, M. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin. Investig. Drugs 2017, 26, 735–739. [Google Scholar] [CrossRef]
  9. Melnikova, I. Therapies for Alzheimer’s disease. Nat. Rev. Drug Discov. 2007, 6, 341–342. [Google Scholar] [CrossRef]
  10. Inglis, F. The tolerability and safety of cholinesterase inhibitors in the treatment of dementia. Int. J. Clin. Pract. Suppl. 2002, 127, 45–63. [Google Scholar]
  11. Lee, J.; Jin, C.; Cho, S.Y.; Park, S.U.; Jung, W.S.; Moon, S.K.; Park, J.M.; Ko, C.N.; Cho, K.H.; Kwon, S. Herbal medicine treatment for Alzheimer disease: A protocol for a systematic review and meta-analysis. Medicine 2020, 99, e21745. [Google Scholar] [CrossRef]
  12. Zhou, J.; Zhou, S. Antihypertensive and neuroprotective activities of rhynchophylline: The role of rhynchophylline in neurotransmission and ion channel activity. J. Ethnopharmacol. 2010, 132, 15–27. [Google Scholar] [CrossRef]
  13. Hsieh, C.L.; Chen, M.F.; Li, T.C.; Li, S.C.; Tang, N.Y.; Hsieh, C.T.; Pon, C.Z.; Lin, J.G. Anticonvulsant effect of Uncaria rhynchophylla (Miq) Jack. in rats with kainic acid-induced epileptic seizure. Am. J. Chin. Med. 1999, 27, 257–264. [Google Scholar] [CrossRef] [PubMed]
  14. Hsieh, C.L.; Tang, N.Y.; Chiang, S.Y.; Hsieh, C.T.; Lin, J.G. Anticonvulsive and free radical scavenging actions of two herbs, Uncaria rhynchophylla (MIQ) Jack and Gastrodia elata Bl., in kainic acid-treated rats. Life Sci. 1999, 65, 2071–2082. [Google Scholar] [CrossRef]
  15. Liu, L.; Zhao, Y.H.; Zeng, C.Q.; Zeng, Y. Research progress in pharmacological effects of Uncaria Hook on Alzheimer disease models. Acta Pharm. Sin. 2016, 51, 536–542. [Google Scholar]
  16. Yuan, D.; Ma, B.; Yang, J.Y.; Xie, Y.Y.; Wang, L.; Zhang, L.J.; Kano, Y.; Wu, C.F. Anti-inflammatory effects of rhynchophylline and isorhynchophylline in mouse N9 microglial cells and the molecular mechanism. Int. Immunopharmacol. 2009, 9, 1549–1554. [Google Scholar] [CrossRef]
  17. Zheng, M.; Chen, M.; Liu, C.; Fan, Y.; Shi, D. Alkaloids extracted from Uncaria rhynchophylla demonstrate neuroprotective effects in MPTP-induced experimental parkinsonism by regulating the PI3K/Akt/mTOR signaling pathway. J. Ethnopharmacol. 2021, 266, 113451. [Google Scholar] [CrossRef]
  18. Shi, J.S.; Yu, J.X.; Chen, X.P.; Xu, R.X. Pharmacological actions of Uncaria alkaloids, rhynchophylline and isorhynchophylline. Acta Pharmacol. Sin. 2003, 24, 97–101. [Google Scholar]
  19. Shin, S.J.; Jeong, Y.; Jeon, S.G.; Kim, S.; Lee, S.K.; Choi, H.S.; Im, C.S.; Kim, S.H.; Kim, S.H.; Park, J.H.; et al. Uncaria rhynchophylla ameliorates amyloid beta deposition and amyloid beta-mediated pathology in 5XFAD mice. Neurochem. Int. 2018, 121, 114–124. [Google Scholar] [CrossRef]
  20. Yang, Y.; Ji, W.G.; Zhu, Z.R.; Wu, Y.L.; Zhang, Z.Y.; Qu, S.C. Rhynchophylline suppresses soluble Abeta1-42-induced impairment of spatial cognition function via inhibiting excessive activation of extrasynaptic NR2B-containing NMDA receptors. Neuropharmacology 2018, 135, 100–112. [Google Scholar] [CrossRef]
  21. Xian, Y.F.; Mao, Q.Q.; Wu, J.C.; Su, Z.R.; Chen, J.N.; Lai, X.P.; Ip, S.P.; Lin, Z.X. Isorhynchophylline treatment improves the amyloid-beta-induced cognitive impairment in rats via inhibition of neuronal apoptosis and tau protein hyperphosphorylation. J. Alzheimers Dis. 2014, 39, 331–346. [Google Scholar] [CrossRef]
  22. Li, H.Q.; Ip, S.P.; Yuan, Q.J.; Zheng, G.Q.; Tsim, K.; Dong, T.; Lin, G.; Han, Y.; Liu, Y.; Xian, Y.F.; et al. Isorhynchophylline ameliorates cognitive impairment via modulating amyloid pathology, tau hyperphosphorylation and neuroinflammation: Studies in a transgenic mouse model of Alzheimer’s disease. Brain Behav. Immun. 2019, 82, 264–278. [Google Scholar] [CrossRef]
  23. Xian, Y.F.; Su, Z.R.; Chen, J.N.; Lai, X.P.; Mao, Q.Q.; Cheng, C.H.; Ip, S.P.; Lin, Z.X. Isorhynchophylline improves learning and memory impairments induced by D-galactose in mice. Neurochem. Int. 2014, 76, 42–49. [Google Scholar] [CrossRef]
  24. Hopkins, A.L. Network pharmacology: The next paradigm in drug discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef] [PubMed]
  25. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Xu, M.; Zhang, D.F.; Luo, R.; Wu, Y.; Zhou, H.; Kong, L.L.; Bi, R.; Yao, Y.G. A systematic integrated analysis of brain expression profiles reveals YAP1 and other prioritized hub genes as important upstream regulators in Alzheimer’s disease. Alzheimers Dement. 2018, 14, 215–229. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, Y.N.; Yang, Y.F.; Xu, W.; Yang, X.W. The Blood-Brain Barrier Permeability of Six Indole Alkaloids from Uncariae Ramulus Cum Uncis in the MDCK-pHaMDR Cell Monolayer Model. Molecules 2017, 22, 1944. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, L.L.; Song, J.X.; Lu, J.H.; Yuan, Z.W.; Liu, L.F.; Durairajan, S.S.; Li, M. Corynoxine, a natural autophagy enhancer, promotes the clearance of alpha-synuclein via Akt/mTOR pathway. J. Neuroimmune Pharmacol. 2014, 9, 380–387. [Google Scholar] [CrossRef]
  29. Chen, L.L.; Wang, Y.B.; Song, J.X.; Deng, W.K.; Lu, J.H.; Ma, L.L.; Yang, C.B.; Li, M.; Xue, Y. Phosphoproteome-based kinase activity profiling reveals the critical role of MAP2K2 and PLK1 in neuronal autophagy. Autophagy 2017, 13, 1969–1980. [Google Scholar] [CrossRef] [Green Version]
  30. Xian, Y.F.; Lin, Z.X.; Mao, Q.Q.; Hu, Z.; Zhao, M.; Che, C.T.; Ip, S.P. Bioassay-Guided Isolation of Neuroprotective Compounds from Uncaria rhynchophylla against Beta-Amyloid-Induced Neurotoxicity. Evid. Based Complement. Alternat. Med. 2012, 2012, 802625. [Google Scholar] [CrossRef]
  31. Frey, H.J.; Mattila, K.M.; Korolainen, M.A.; Pirttila, T. Problems associated with biological markers of Alzheimer’s disease. Neurochem. Res. 2005, 30, 1501–1510. [Google Scholar] [CrossRef]
  32. Skovronsky, D.M.; Fath, S.; Lee, V.M.; Milla, M.E. Neuronal localization of the TNFalpha converting enzyme (TACE) in brain tissue and its correlation to amyloid plaques. J. Neurobiol. 2001, 49, 40–46. [Google Scholar] [CrossRef]
  33. Qian, M.; Shen, X.; Wang, H. The Distinct Role of ADAM17 in APP Proteolysis and Microglial Activation Related to Alzheimer’s Disease. Cell. Mol. Neurobiol. 2016, 36, 471–482. [Google Scholar] [CrossRef]
  34. Koelsch, G. BACE1 Function and Inhibition: Implications of Intervention in the Amyloid Pathway of Alzheimer’s Disease Pathology. Molecules 2017, 22, 1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. De Strooper, B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-Secretase complex. Neuron 2003, 38, 9–12. [Google Scholar] [CrossRef] [Green Version]
  36. Ryan, N.S.; Nicholas, J.M.; Weston, P.; Liang, Y.; Lashley, T.; Guerreiro, R.; Adamson, G.; Kenny, J.; Beck, J.; Chavez-Gutierrez, L.; et al. Clinical phenotype and genetic associations in autosomal dominant familial Alzheimer’s disease: A case series. Lancet Neurol. 2016, 15, 1326–1335. [Google Scholar] [CrossRef] [Green Version]
  37. Ly, P.T.; Wu, Y.; Zou, H.; Wang, R.; Zhou, W.; Kinoshita, A.; Zhang, M.; Yang, Y.; Cai, F.; Woodgett, J.; et al. Inhibition of GSK3beta-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J. Clin. Investig. 2013, 123, 224–235. [Google Scholar] [CrossRef] [Green Version]
  38. Ochalek, A.; Mihalik, B.; Avci, H.X.; Chandrasekaran, A.; Teglasi, A.; Bock, I.; Giudice, M.L.; Tancos, Z.; Molnar, K.; Laszlo, L.; et al. Neurons derived from sporadic Alzheimer’s disease iPSCs reveal elevated TAU hyperphosphorylation, increased amyloid levels, and GSK3B activation. Alzheimers Res. Ther. 2017, 9, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Li, T.; Zhu, J. Entanglement of CCR5 and Alzheimer’s Disease. Front. Aging Neurosci. 2019, 11, 209. [Google Scholar] [CrossRef] [PubMed]
  40. Hampel, H.; Mesulam, M.M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J.; et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018, 141, 1917–1933. [Google Scholar] [CrossRef] [PubMed]
  41. Gabbouj, S.; Ryhanen, S.; Marttinen, M.; Wittrahm, R.; Takalo, M.; Kemppainen, S.; Martiskainen, H.; Tanila, H.; Haapasalo, A.; Hiltunen, M.; et al. Altered Insulin Signaling in Alzheimer’s Disease Brain–Special Emphasis on PI3K-Akt Pathway. Front. Neurosci. 2019, 13, 629. [Google Scholar] [CrossRef] [PubMed]
  42. Arvanitakis, Z.; Wang, H.Y.; Capuano, A.W.; Khan, A.; Taib, B.; Anokye-Danso, F.; Schneider, J.A.; Bennett, D.A.; Ahima, R.S.; Arnold, S.E. Brain Insulin Signaling, Alzheimer Disease Pathology, and Cognitive Function. Ann. Neurol. 2020, 88, 513–525. [Google Scholar] [CrossRef] [PubMed]
  43. Grimes, C.A.; Jope, R.S. The multifaceted roles of glycogen synthase kinase 3beta in cellular signaling. Prog. Neurobiol. 2001, 65, 391–426. [Google Scholar] [CrossRef]
  44. Cross, D.A.; Alessi, D.R.; Cohen, P.; Andjelkovich, M.; Hemmings, B.A. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995, 378, 785–789. [Google Scholar] [CrossRef] [PubMed]
  45. Stadelmann, C.; Deckwerth, T.L.; Srinivasan, A.; Bancher, C.; Bruck, W.; Jellinger, K.; Lassmann, H. Activation of caspase-3 in single neurons and autophagic granules of granulovacuolar degeneration in Alzheimer’s disease. Evidence for apoptotic cell death. Am. J. Pathol. 1999, 155, 1459–1466. [Google Scholar] [CrossRef]
  46. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef]
  47. Lipinski, C.A. Lead- and drug-like compounds: The rule-of-five revolution. Drug Discov. Today Technol. 2004, 1, 337–341. [Google Scholar] [CrossRef]
  48. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [Green Version]
  49. Huang, J.; Liang, Y.; Tian, W.; Ma, J.; Huang, L.; Li, B.; Chen, R.; Li, D. Antitumor Activity and Mechanism of Robustic Acid from Dalbergia benthami Prain via Computational Target Fishing. Molecules 2020, 25, 3919. [Google Scholar] [CrossRef]
  50. Kim, S.; Thiessen, P.A.; Cheng, T.; Yu, B.; Shoemaker, B.A.; Wang, J.; Bolton, E.E.; Wang, Y.; Bryant, S.H. Literature information in PubChem: Associations between PubChem records and scientific articles. J. Cheminform. 2016, 8, 32. [Google Scholar] [CrossRef] [Green Version]
  51. Daina, A.; Michielin, O.; Zoete, V. SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 2019, 47, W357–W364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Wishart, D.S.; Knox, C.; Guo, A.C.; Cheng, D.; Shrivastava, S.; Tzur, D.; Gautam, B.; Hassanali, M. DrugBank: A knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res. 2008, 36, D901–D906. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Y.; Zhang, S.; Li, F.; Zhou, Y.; Zhang, Y.; Wang, Z.; Zhang, R.; Zhu, J.; Ren, Y.; Tan, Y.; et al. Therapeutic target database 2020: Enriched resource for facilitating research and early development of targeted therapeutics. Nucleic Acids Res. 2020, 48, D1031–D1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mi, H.; Guo, N.; Kejariwal, A.; Thomas, P.D. PANTHER version 6: Protein sequence and function evolution data with expanded representation of biological pathways. Nucleic Acids Res. 2007, 35, D247–D252. [Google Scholar] [CrossRef] [Green Version]
  55. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef]
  56. Bader, G.D.; Hogue, C.W. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinform. 2003, 4, 2. [Google Scholar] [CrossRef] [Green Version]
  57. Zhou, Y.; Zhou, B.; Pache, L.; Chang, M.; Khodabakhshi, A.H.; Tanaseichuk, O.; Benner, C.; Chanda, S.K. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 2019, 10, 1523. [Google Scholar] [CrossRef]
  58. Westbrook, J.; Feng, Z.; Jain, S.; Bhat, T.N.; Thanki, N.; Ravichandran, V.; Gilliland, G.L.; Bluhm, W.; Weissig, H.; Greer, D.S.; et al. The Protein Data Bank: Unifying the archive. Nucleic Acids Res. 2002, 30, 245–248. [Google Scholar] [CrossRef] [Green Version]
  59. Grosdidier, A.; Zoete, V.; Michielin, O. SwissDock, a protein-small molecule docking web service based on EADock DSS. Nucleic Acids Res. 2011, 39, W270–W277. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flowchart of the study.
Figure 1. Flowchart of the study.
Ijms 22 03612 g001
Figure 2. Structures of the main alkaloids extracted from Uncaria rhynchophylla (UR).
Figure 2. Structures of the main alkaloids extracted from Uncaria rhynchophylla (UR).
Ijms 22 03612 g002
Figure 3. Protein–protein interaction (PPI) network construction for target proteins of UR alkaloids against Alzheimer’s disease (AD). (A) A Venn diagram was applied to obtain the intersection between the 10 UR alkaloids and AD targets. (B) Panther classification was used categorize common targets of UR alkaloids against AD. (C) Target proteins involved in protein modification as enzyme (PC00260). (D) PPI network of UR alkaloids against AD. Nodes represent target proteins, and edges represent interactions among targets. The numbers below the nodes indicate the degree. The darker the color and the larger the node are, the greater the degree is.
Figure 3. Protein–protein interaction (PPI) network construction for target proteins of UR alkaloids against Alzheimer’s disease (AD). (A) A Venn diagram was applied to obtain the intersection between the 10 UR alkaloids and AD targets. (B) Panther classification was used categorize common targets of UR alkaloids against AD. (C) Target proteins involved in protein modification as enzyme (PC00260). (D) PPI network of UR alkaloids against AD. Nodes represent target proteins, and edges represent interactions among targets. The numbers below the nodes indicate the degree. The darker the color and the larger the node are, the greater the degree is.
Ijms 22 03612 g003
Figure 4. PPI network clusters of common targets of both UR alkaloids and in AD. (AF) Clusters 1 to 6 were found with Molecular Complex Detection (MCODE), which can identify densely connected regions. The seed node of each cluster is indicated by red font. (G) Comparison of the MCODE scores of different clusters.
Figure 4. PPI network clusters of common targets of both UR alkaloids and in AD. (AF) Clusters 1 to 6 were found with Molecular Complex Detection (MCODE), which can identify densely connected regions. The seed node of each cluster is indicated by red font. (G) Comparison of the MCODE scores of different clusters.
Ijms 22 03612 g004
Figure 5. Potential alkaloid target-AD target network. Red nodes represent the main alkaloids extracted from UR, and the numbers beneath each node represent the number of UR targets against AD. Green nodes represent shared targets between potential targets of UR alkaloids and AD targets. UR: Uncaria rhynchophylla; AD: Alzheimer’s disease.
Figure 5. Potential alkaloid target-AD target network. Red nodes represent the main alkaloids extracted from UR, and the numbers beneath each node represent the number of UR targets against AD. Green nodes represent shared targets between potential targets of UR alkaloids and AD targets. UR: Uncaria rhynchophylla; AD: Alzheimer’s disease.
Ijms 22 03612 g005
Figure 6. GO biological process (BP) (A) terms and the results of KEGG (B) pathway enrichment analysis of target proteins of UR alkaloids against AD. The X-axis represents the rich factor, the bubble size represents the number of targets enriched in terms, and the color indicates the p-value. (C) Schematic drawing of the Alzheimer disease pathway (hsa05010). Red font indicates the targets of UR alkaloids involved in the Alzheimer disease pathway.
Figure 6. GO biological process (BP) (A) terms and the results of KEGG (B) pathway enrichment analysis of target proteins of UR alkaloids against AD. The X-axis represents the rich factor, the bubble size represents the number of targets enriched in terms, and the color indicates the p-value. (C) Schematic drawing of the Alzheimer disease pathway (hsa05010). Red font indicates the targets of UR alkaloids involved in the Alzheimer disease pathway.
Ijms 22 03612 g006
Figure 7. Target-enriched KEGG pathway network for UR alkaloids against AD. Red nodes represent enriched KEGG pathways, and brown nodes represent target proteins.
Figure 7. Target-enriched KEGG pathway network for UR alkaloids against AD. Red nodes represent enriched KEGG pathways, and brown nodes represent target proteins.
Ijms 22 03612 g007
Figure 8. Bioinformatics analysis of targets of UR alkaloids related to Aβ and tau pathology. (A) Radial bar plot showing targets of the alkaloids significantly correlated with tau, Aβ, or Aβ and tau. (B) PPI network of proteins associated with the pathology of Aβ and tau. The darker the color and the larger the node are, the higher the degree is. (C) Bubble chart of the top 20 biological process (BP) terms identified by GO enrichment analysis. The X-axis represents the rich factor, the bubble size represents the number of targets enriched in terms, and the color indicates the p-value. (D) KEGG pathway enrichment analysis of targets correlated with tau, Aβ, or Aβ and tau. Numbers next to bar graphs indicate the numbers of targets enriched in the terms.
Figure 8. Bioinformatics analysis of targets of UR alkaloids related to Aβ and tau pathology. (A) Radial bar plot showing targets of the alkaloids significantly correlated with tau, Aβ, or Aβ and tau. (B) PPI network of proteins associated with the pathology of Aβ and tau. The darker the color and the larger the node are, the higher the degree is. (C) Bubble chart of the top 20 biological process (BP) terms identified by GO enrichment analysis. The X-axis represents the rich factor, the bubble size represents the number of targets enriched in terms, and the color indicates the p-value. (D) KEGG pathway enrichment analysis of targets correlated with tau, Aβ, or Aβ and tau. Numbers next to bar graphs indicate the numbers of targets enriched in the terms.
Ijms 22 03612 g008
Figure 9. (AQ) Targets of UR alkaloids against AD in the control and AD groups of the GEO dataset. Entorhinal cortex, n = 39 in each group. Hippocampus, n = 66 in the healthy control group, n = 74 in the AD group. Temporal cortex, n = 39 in the healthy control group, n = 52 in the AD group. Values are presented as the mean ± SD.
Figure 9. (AQ) Targets of UR alkaloids against AD in the control and AD groups of the GEO dataset. Entorhinal cortex, n = 39 in each group. Hippocampus, n = 66 in the healthy control group, n = 74 in the AD group. Temporal cortex, n = 39 in the healthy control group, n = 52 in the AD group. Values are presented as the mean ± SD.
Ijms 22 03612 g009
Table 1. Pharmacological and molecular properties of the main alkaloids in UR.
Table 1. Pharmacological and molecular properties of the main alkaloids in UR.
NameFormulaMW (g/mol)HdonHaccRbonTPSA (Å)LogPLogSLog Kp (cm/s)
RhynchophyllineC22H28N2O4384.4715567.872.43−3.51−7.01
CorynantheineC22H26N2O3366.4514554.563.08−4.03−6.28
CorynoxineC22H28N2O4384.4715567.872.38−3.51−7.01
IsocorynoxeineC22H26N2O4382.4515567.872.26−3.34−7.17
IsorhynchophyllineC22H28N2O4384.4715567.872.4−3.51−7.01
DihydrocorynatheineC22H28N2O3368.4714554.563.22−4.21−6.11
HirsutineC22H28N2O3368.4714554.563.22−4.21−6.11
HirsuteineC22H26N2O3366.4514554.563.1−4.03−6.28
AngustolineC20H17N3O2331.3723170.912.52−3.52−7.05
AngustidineC19H15N3O301.3412050.683.02−4.01−6.24
MW: molecule weight; Hdon: hydrogen bond donors; Hacc: hydrogen bond acceptors; Rbon: rotatable bonds; TPSA: topological polar surface area; LogP: lipid–water partition coefficient; LogS: solubility; Log Kp: skin permeation.
Table 2. Targets of UR alkaloids against AD.
Table 2. Targets of UR alkaloids against AD.
NumberGene IDGene SymbolDescription
125ABL1ABL proto-oncogene 1, non-receptor tyrosine kinase
243ACHEacetylcholinesterase (Cartwright blood group)
36868ADAM17ADAM metallopeptidase domain 17
4152ADRA2Cadrenoceptor alpha 2C
5154ADRB2adrenoceptor beta 2
6207AKT1AKT serine/threonine kinase 1
7240ALOX5arachidonate 5-lipoxygenase
883464APH1Baph-1 homolog B, gamma-secretase subunit
923621BACE1beta-secretase 1
10590BCHEbutyrylcholinesterase
1123476BRD4bromodomain containing 4
12823CAPN1calpain 1
13834CASP1caspase 1
14836CASP3caspase 3
15595CCND1cyclin D1
16729230CCR2C-C motif chemokine receptor 2
171232CCR3C-C motif chemokine receptor 3
181234CCR5C-C motif chemokine receptor 5
19983CDK1cyclin dependent kinase 1
201020CDK5cyclin dependent kinase 5
211129CHRM2cholinergic receptor muscarinic 2
221131CHRM3cholinergic receptor muscarinic 3
231137CHRNA4cholinergic receptor nicotinic alpha 4 subunit
241141CHRNB2cholinergic receptor nicotinic beta 2 subunit
251436CSF1Rcolony stimulating factor 1 receptor
263579CXCR2C-X-C motif chemokine receptor 2
271565CYP2D6cytochrome P450 family 2 subfamily D member 6
281812DRD1dopamine receptor D1
291813DRD2dopamine receptor D2
301814DRD3dopamine receptor D3
311815DRD4dopamine receptor D4
321859DYRK1Adual specificity tyrosine phosphorylation regulated kinase 1A
331889ECE1endothelin converting enzyme 1
349451EIF2AK3eukaryotic translation initiation factor 2 alpha kinase 3
352064ERBB2erb-b2 receptor tyrosine kinase 2
362078ERGETS transcription factor ERG
372260FGFR1fibroblast growth factor receptor 1
382261FGFR3fibroblast growth factor receptor 3
392534FYNFYN proto-oncogene, Src family tyrosine kinase
402558GABRA5gamma-aminobutyric acid type A receptor subunit alpha5
4151738GHRLghrelin and obestatin prepropeptide
422904GRIN2Bglutamate ionotropic receptor NMDA type subunit 2B
432932GSK3Bglycogen synthase kinase 3 beta
443091HIF1Ahypoxia inducible factor 1 subunit alpha
453290HSD11B1hydroxysteroid 11-beta dehydrogenase 1
463350HTR1A5-hydroxytryptamine receptor 1A
473356HTR2A5-hydroxytryptamine receptor 2A
483358HTR2C5-hydroxytryptamine receptor 2C
493360HTR45-hydroxytryptamine receptor 4
503362HTR65-hydroxytryptamine receptor 6
513363HTR75-hydroxytryptamine receptor 7
523480IGF1Rinsulin like growth factor 1 receptor
533643INSRinsulin receptor
543832KIF11kinesin family member 11
553815KITKIT proto-oncogene, receptor tyrosine kinase
563932LCKLCK proto-oncogene, Src family tyrosine kinase
57120892LRRK2leucine rich repeat kinase 2
584128MAOAmonoamine oxidase A
595602MAPK10mitogen-activated protein kinase 10
6010461MERTKMER proto-oncogene, tyrosine kinase
614233METMET proto-oncogene, receptor tyrosine kinase
624311MMEmembrane metalloendopeptidase
634313MMP2matrix metallopeptidase 2
644314MMP3matrix metallopeptidase 3
654318MMP9matrix metallopeptidase 9
662475MTORmechanistic target of rapamycin kinase
6723385NCSTNnicastrin
684842NOS1nitric oxide synthase 1
694843NOS2nitric oxide synthase 2
704846NOS3nitric oxide synthase 3
714835NQO2N-ribosyldihydronicotinamide:quinone reductase 2
724914NTRK1neurotrophic receptor tyrosine kinase 1
734915NTRK2neurotrophic receptor tyrosine kinase 2
744916NTRK3neurotrophic receptor tyrosine kinase 3
755027P2RX7purinergic receptor P2X 7
76142PARP1poly(ADP-ribose) polymerase 1
775144PDE4Dphosphodiesterase 4D
788622PDE8Bphosphodiesterase 8B
795159PDGFRBplatelet derived growth factor receptor beta
807941PLA2G7phospholipase A2 group VII
815663PSEN1presenilin 1
825664PSEN2presenilin 2
8355851PSENENpresenilin enhancer, gamma-secretase subunit
845743PTGS2prostaglandin-endoperoxide synthase 2
856571SLC18A2solute carrier family 18 member A2
866531SLC6A3solute carrier family 6 member 3
876532SLC6A4solute carrier family 6 member 4
886752SSTR2somatostatin receptor 2
896753SSTR3somatostatin receptor 3
906755SSTR5somatostatin receptor 5
Table 3. Top 20 KEGG pathway terms enriched in UR alkaloids against AD.
Table 3. Top 20 KEGG pathway terms enriched in UR alkaloids against AD.
TermPathwayRich Factorp-ValueCountSymbols
hsa05010Alzheimer disease0.082.445 × 10−1816CAPN1,CASP3,CDK5,GRIN2B,GSK3B,MME,NOS1,PSEN1,PSEN2,PTGS2,ADAM17,EIF2AK3,NCSTN,BACE1,PSENEN,APH1B
hsa04726Serotonergic synapse0.119.421 × 10−1713ALOX5,CASP3,CYP2D6,HTR1A,HTR2A,HTR2C,HTR4,HTR6,HTR7,MAOA,PTGS2,SLC6A4,SLC18A2
hsa04020Calcium signaling pathway0.071.693 × 10−1615ADRB2,CHRM2,CHRM3,DRD1,ERBB2,HTR2A,HTR2C,HTR4,HTR6,HTR7,NOS1,NOS2,NOS3,P2RX7,PDGFRB
hsa04151PI3K-Akt signaling pathway0.044.79 × 10−1517AKT1,CCND1,CHRM2,CSF1R,ERBB2,FGFR1,FGFR3,MTOR,GSK3B,IGF1R,INSR,KIT,MET,NOS3,NTRK1,NTRK2,PDGFRB
hsa04728Dopaminergic synapse0.085.086 × 10−1311AKT1,DRD1,DRD2,DRD3,DRD4,GRIN2B,GSK3B,MAOA,MAPK10,SLC6A3,SLC18A2
hsa04722Neurotrophin signaling pathway0.072.555 × 10−109ABL1,AKT1,GSK3B,NTRK1,NTRK2,NTRK3,MAPK10,PSEN1,PSEN2
hsa04330Notch signaling pathway0.121.685 × 10−86PSEN1,PSEN2,ADAM17,NCSTN,PSENEN,APH1B
hsa04725Cholinergic synapse0.069.258 × 10−87ACHE,AKT1,CHRM2,CHRM3,CHRNA4,CHRNB2,FYN
hsa04144Endocytosis0.032.33 × 10−79ADRB2,CCR5,CSF1R,FGFR3,IGF1R,CXCR2,KIT,MET,NTRK1
hsa04520Adherens junction0.072.494 × 10−76ERBB2,FGFR1,FYN,IGF1R,INSR,MET
hsa04926Relaxin signaling pathway0.052.92 × 10−77AKT1,MMP2,MMP9,NOS1,NOS2,NOS3,MAPK10
hsa04540Gap junction0.074.675 × 10−76CDK1,DRD1,DRD2,HTR2A,HTR2C,PDGFRB
hsa04210Apoptosis0.055.935 × 10−77PARP1,AKT1,CAPN1,CASP3,NTRK1,MAPK10,EIF2AK3
hsa04012ErbB signaling pathway0.066.051 × 10−76ABL1,AKT1,ERBB2,MTOR,GSK3B,MAPK10
hsa04657IL-17 signaling pathway0.066.442 × 10−76CASP3,GSK3B,MMP3,MMP9,MAPK10,PTGS2
hsa04371Apelin signaling pathway0.047.882 × 10−66AKT1,CCND1,MTOR,NOS1,NOS2,NOS3
hsa05014Amyotrophic lateral sclerosis0.073.464 × 10−54CASP1,CASP3,GRIN2B,NOS1
hsa05032Morphine addiction0.040.00022494DRD1,GABRA5,PDE4D,PDE8B
hsa04360Axon guidance0.030.00043395ABL1,CDK5,FYN,GSK3B,MET
hsa04115p53 signaling pathway0.040.002133CCND1,CASP3,CDK1
Table 4. Molecular docking of core targets with corynoxine and corynantheine.
Table 4. Molecular docking of core targets with corynoxine and corynantheine.
LigandsTargetPDBdeltaG (kcal/mol)deltaGvdwFullFitness (kcal/mol)Energy (kcal/mol)
CorynoxineCCR56AKY−9.28−83.41−1260.0216.61
CorynoxineMMP32D1O−8.52−70.64−866.9416.06
CorynoxineCCND12W9F−8.17−66.74−1571.1321.40
CorynoxineNTRK24AT4−8.10−56.35−1692.151.58
CorynoxineGRIN2B7KL4−7.95−76.52−1938.7729.92
CorynoxineCHRM24MQT−7.92−51.24−874.5914.59
CorynoxineCDK51UNH−7.90−54.59−3964.0520.41
CorynoxineMMP21QIB−7.67−54.18−1258.8516.61
CorynoxineGSK3B2O5K−7.66−62.37−1986.9817.42
CorynantheineCHRM24MQT−9.38−82.53−860.06−5.88
CorynantheineMMP32D1O−9.30−82.62−857.66−0.88
CorynantheineMMP21QIB−9.24−94.70−1246.61−5.34
CorynantheineGRIN2B7KL4−9.02−87.46−1941.76−5.64
CorynantheineCCR56AKY−8.92−75.97−1250.691.62
CorynantheineCCND12W9F−8.77−83.19−1556.058.76
CorynantheineNTRK24AT4−8.10−53.43−1676.61−3.78
CorynantheineCDK51UNH−7.95−61.20−3952.57−1.90
CorynantheineGSK3B2O5K−7.38−56.22−1952.8724.20
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zeng, P.; Wang, X.-M.; Ye, C.-Y.; Su, H.-F.; Tian, Q. The Main Alkaloids in Uncaria rhynchophylla and Their Anti-Alzheimer’s Disease Mechanism Determined by a Network Pharmacology Approach. Int. J. Mol. Sci. 2021, 22, 3612. https://doi.org/10.3390/ijms22073612

AMA Style

Zeng P, Wang X-M, Ye C-Y, Su H-F, Tian Q. The Main Alkaloids in Uncaria rhynchophylla and Their Anti-Alzheimer’s Disease Mechanism Determined by a Network Pharmacology Approach. International Journal of Molecular Sciences. 2021; 22(7):3612. https://doi.org/10.3390/ijms22073612

Chicago/Turabian Style

Zeng, Peng, Xiao-Ming Wang, Chao-Yuan Ye, Hong-Fei Su, and Qing Tian. 2021. "The Main Alkaloids in Uncaria rhynchophylla and Their Anti-Alzheimer’s Disease Mechanism Determined by a Network Pharmacology Approach" International Journal of Molecular Sciences 22, no. 7: 3612. https://doi.org/10.3390/ijms22073612

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop