1, Systems pharmacology uncovers that licorice targets cell cycle progression and immune process
As a comprehensive system, the systems pharmacology approach was used to investigate the complex molecular mechanisms of licorice as a treatment for NSCLC in this study (as shown in Fig. 1).
Altogether, 89 ingredients were identified in licorice with the searching literatures and using Traditional Chinese Medicine Systems Pharmacology Database (TCMSP), and a total of 23 active ingredients (shown in Table 1) were screened out by in silico ADME (absorption, distribution, metabolism, and excretion) system, with the criteria of oral bioavailability (OB) ≥ 50% and drug-likeness (DL) ≥ 0.40. Then, predicted by the weighted ensemble similarity method (WES) [17] and systematic drug targeting tool (SysDT)[18], we found that these 23 ingredients in licorice were investigated interacted with 109 targets (shown in Table 2 and table S1). And we constructed the compound-target (C-T) network graph to greatly illustrate the relationships between compounds and targets. In terms of the targets interacted with licorice, we observed that most of which were related to cell cycle, immune, inflammation, cancer and neoplasm metastasis with higher scores. Specifically, including CDK2, ESR1, PPARG, ESRRA, PRKACA, CXCL8, PLAA, RXRB, MAPK14 and so on (shown in Fig. 2a).
To detect the potential role of these targets, we performed Gene Ontology (GO) biological processes enrichment analysis, and found that most of biological processes were involved in immune progress. Including “regulation of myeloid cell differentiation”, “neutrophil mediated immunity”, and “regulation of cytokine production involved in inflammatory response” (shown in Fig. 2b and 2c). Then, to further understand the relationship between licorice and diseases, using the Database for Annotation Visualization, and Integrated Discovery (DAVID), we performed the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. And the results showed that the most targets of licorice mainly enriched in signaling pathways related to the cancer process. Including “non-small cell lung cancer”, “small cell lung cancer” “pathways in cancer”, “prostate cancer”, “T cell receptor signaling pathway” and so on (Fig. 2d).
Therefore, the systems pharmacology analysis uncovers that licorice mainly targets cell cycle and immune progress to exert its anti-cancer effect, and paves the way for in-depth understanding of the multi-target molecular mechanism of licorice treating for NSCLC.
Table 1. Chemical information and pharmacokinetics parameters of 23 active compounds of licorice.
Table 2
The targets information of licorice.
UniProt-ID
|
Protein names
|
Gene names
|
Degree
|
Species
|
P0DP23
|
Calmodulin-1
|
CALM1
|
19
|
homo sapiens
|
P35368
|
Alpha-1B adrenergic receptor
|
ADRA1B
|
5
|
homo sapiens
|
P00918
|
Carbonic anhydrase 2
|
CA2
|
17
|
homo sapiens
|
P18031
|
Tyrosine-protein phosphatase non-receptor type 1
|
PTPN1
|
17
|
homo sapiens
|
P46098
|
5-hydroxytryptamine receptor 3A
|
HTR3A
|
1
|
homo sapiens
|
P20309
|
Muscarinic acetylcholine receptor M3
|
CHRM3
|
3
|
homo sapiens
|
P23219
|
Prostaglandin G/H synthase 1
|
PTGS1
|
8
|
homo sapiens
|
Q14524
|
Sodium channel protein type 5 subunit alpha
|
SCN5A
|
11
|
homo sapiens
|
P07477
|
Trypsin-1
|
PRSS1
|
18
|
homo sapiens
|
P17612
|
cAMP-dependent protein kinase catalytic subunit alpha
|
PRKACA
|
6
|
homo sapiens
|
O14757
|
Serine/threonine-protein kinase
|
CHEK1
|
18
|
homo sapiens
|
P11309
|
Serine/threonine-protein kinase pim-1
|
PIM1
|
20
|
homo sapiens
|
P35354
|
Prostaglandin G/H synthase 2
|
PTGS2
|
20
|
homo sapiens
|
P27487
|
Dipeptidyl peptidase 4
|
DPP4
|
13
|
homo sapiens
|
Q16539
|
Mitogen-activated protein kinase 14
|
MAPK14
|
13
|
homo sapiens
|
P48736
|
Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform
|
PIK3CG
|
3
|
homo sapiens
|
P21730
|
C5a anaphylatoxin chemotactic receptor 1
|
AR
|
22
|
homo sapiens
|
P49841
|
Glycogen synthase kinase-3 beta
|
GSK3B
|
17
|
homo sapiens
|
P24941
|
Cyclin-dependent kinase 2
|
CDK2
|
17
|
homo sapiens
|
Q92731
|
Estrogen receptor beta
|
ESR2
|
16
|
homo sapiens
|
P07900
|
Heat shock protein HSP 90-alpha
|
HSP90AA1
|
12
|
homo sapiens
|
P20248
|
Cyclin-A2
|
CCNA2
|
20
|
homo sapiens
|
B2RXH2
|
Lysine-specific demethylase 4E
|
KDM4E
|
1
|
homo sapiens
|
O00767
|
Stearoyl-CoA desaturase
|
SCD
|
10
|
homo sapiens
|
O95622
|
Adenylate cyclase type 5
|
ADCY5
|
7
|
homo sapiens
|
P08842
|
Steryl-sulfatase
|
STS
|
13
|
homo sapiens
|
P11474
|
Steroid hormone receptor ERR1
|
ESRRA
|
12
|
homo sapiens
|
P12644
|
Bone morphogenetic protein 4
|
BMP4
|
1
|
homo sapiens
|
P16152
|
Carbonyl reductase [NADPH] 1
|
CBR1
|
7
|
homo sapiens
|
P28223
|
5-hydroxytryptamine receptor 2A
|
HTR2A
|
18
|
homo sapiens
|
P51843
|
Nuclear receptor subfamily 0 group B member 1
|
NR0B1
|
7
|
homo sapiens
|
Q99814
|
Endothelial PAS domain-containing protein 1
|
EPAS1
|
3
|
homo sapiens
|
Q9Y263
|
Phospholipase A-2-activating protein
|
PLAA
|
3
|
homo sapiens
|
O60218
|
Aldo-keto reductase family 1 member B10
|
AKR1B10
|
1
|
homo sapiens
|
P05093
|
Steroid 17-alpha-hydroxylase/17,20 lyase
|
CYP17A1
|
1
|
homo sapiens
|
P10276
|
Retinoic acid receptor alpha
|
RARA
|
1
|
homo sapiens
|
P11413
|
Glucose-6-phosphate 1-dehydrogenase
|
G6PD
|
1
|
homo sapiens
|
P11473
|
Vitamin D3 receptor
|
VDR
|
1
|
homo sapiens
|
P16662
|
UDP-glucuronosyltransferase 2B7
|
UGT2B7
|
1
|
homo sapiens
|
P18405
|
3-oxo-5-alpha-steroid 4-dehydrogenase 1
|
SRD5A1
|
1
|
homo sapiens
|
P19793
|
Retinoic acid receptor RXR-alpha
|
RXRA
|
7
|
homo sapiens
|
P36873
|
Serine/threonine-protein phosphatase PP1-gamma catalytic subunit
|
PPP1CC
|
1
|
homo sapiens
|
P80365
|
Corticosteroid 11-beta-dehydrogenase isozyme 2
|
HSD11B2
|
2
|
homo sapiens
|
Q08828
|
Adenylate cyclase type 1
|
ADCY1
|
1
|
homo sapiens
|
Q12908
|
Ileal sodium/bile acid cotransporter
|
SLC10A2
|
1
|
homo sapiens
|
Q9NRD8
|
Dual oxidase 2
|
DUOX2
|
1
|
homo sapiens
|
Q9UBM7
|
7-dehydrocholesterol reductase
|
DHCR7
|
1
|
homo sapiens
|
P03372
|
Estrogen receptor
|
ESR1
|
13
|
homo sapiens
|
P03420
|
Fusion glycoprotein F2
|
F2
|
18
|
homo sapiens
|
P37231
|
Peroxisome proliferator-activated receptor gamma
|
PPARG
|
19
|
homo sapiens
|
P30291
|
Wee1-like protein kinase
|
WEE1
|
3
|
homo sapiens
|
P23141
|
Liver carboxylesterase 1
|
CES2
|
7
|
homo sapiens
|
P05067
|
Amyloid-beta precursor protein
|
APP
|
7
|
homo sapiens
|
P09960
|
Leukotriene A-4 hydrolase
|
LTA4H
|
10
|
homo sapiens
|
P10636
|
Microtubule-associated protein tau
|
MAPT
|
9
|
homo sapiens
|
Q04206
|
Transcription factor p65
|
RELA
|
6
|
homo sapiens
|
P22303
|
Acetylcholinesterase
|
ACHE
|
11
|
homo sapiens
|
Q15596
|
Nuclear receptor coactivator 2
|
NCOA2
|
10
|
homo sapiens
|
P11388
|
DNA topoisomerase 2-alpha
|
TOP2A
|
11
|
homo sapiens
|
P35968
|
Vascular endothelial growth factor receptor 2
|
KDR
|
8
|
homo sapiens
|
P00742
|
Coagulation factor X
|
F10
|
16
|
homo sapiens
|
P08709
|
Coagulation factor VII, EC 3.4.21.21
|
F7
|
7
|
homo sapiens
|
P11926
|
Ornithine decarboxylase
|
ODC1
|
10
|
homo sapiens
|
P14061
|
17-beta-hydroxysteroid dehydrogenase type 1
|
HSD17B1
|
5
|
homo sapiens
|
P18054
|
olyunsaturated fatty acid lipoxygenase ALOX12
|
ALOX12
|
7
|
homo sapiens
|
Q9UHC3
|
Acid-sensing ion channel 3
|
ASIC3
|
12
|
homo sapiens
|
P05091
|
Aldehyde dehydrogenase
|
ALDH2
|
4
|
homo sapiens
|
P37058
|
Testosterone 17-beta-dehydrogenase 3
|
HSD17B3
|
3
|
homo sapiens
|
Q13887
|
Krueppel-like factor 5
|
KLF5
|
2
|
homo sapiens
|
Q15788
|
Nuclear receptor coactivator 1
|
NCOA1
|
6
|
homo sapiens
|
Q12809
|
Potassium voltage-gated channel subfamily H member 2
|
KCNH2
|
5
|
homo sapiens
|
Q9H4B7
|
Tubulin beta-1 chain
|
TUBB1
|
5
|
homo sapiens
|
P12268
|
Inosine-5'-monophosphate dehydrogenase 2
|
IMPDH2
|
1
|
homo sapiens
|
P11308
|
Transcriptional regulator ERG
|
ERG
|
1
|
homo sapiens
|
P45985
|
Dual specificity mitogen-activated protein kinase kinase 4
|
MAP2K4
|
1
|
homo sapiens
|
P25100
|
Alpha-1D adrenergic receptor
|
ADRA1D
|
2
|
homo sapiens
|
P36544
|
Neuronal acetylcholine receptor subunit alpha-7
|
CHRNA7
|
1
|
homo sapiens
|
P28702
|
Retinoic acid receptor RXR-beta
|
RXRB
|
2
|
homo sapiens
|
P08912
|
Muscarinic acetylcholine receptor M5
|
CHRM5
|
1
|
homo sapiens
|
P11229
|
Muscarinic acetylcholine receptor M1
|
CHRM1
|
2
|
homo sapiens
|
P07550
|
Beta-2 adrenergic receptor
|
ADRB2
|
4
|
homo sapiens
|
P35372
|
Mu-type opioid receptor
|
OPRM1
|
1
|
homo sapiens
|
P41143
|
Delta-type opioid receptor
|
OPRD1
|
1
|
homo sapiens
|
O60502
|
Protein O-GlcNAcase
|
OGA
|
1
|
homo sapiens
|
P08514
|
Integrin alpha-IIb
|
ITGA2B
|
1
|
homo sapiens
|
P16278
|
Beta-galactosidase
|
GLB1
|
1
|
homo sapiens
|
P28838
|
Cytosol aminopeptidase
|
LAP3
|
1
|
homo sapiens
|
P31639
|
Sodium/glucose cotransporter 2
|
SLC5A2
|
1
|
homo sapiens
|
P53396
|
ATP-citrate synthase
|
ACLY
|
1
|
homo sapiens
|
P54577
|
Tyrosine–tRNA ligase, cytoplasmic
|
YARS
|
1
|
homo sapiens
|
O75907
|
Diacylglycerol O-acyltransferase 1
|
DGAT1
|
3
|
homo sapiens
|
P14222
|
Perforin-1
|
PRF1
|
1
|
homo sapiens
|
P51684
|
C-C chemokine receptor type 6
|
CCR6
|
2
|
homo sapiens
|
P05177
|
Cytochrome P450 1A2
|
CYP1A2
|
1
|
homo sapiens
|
Q16678
|
Cytochrome P450 1B1
|
CYP1B1
|
1
|
homo sapiens
|
Q92959
|
Solute carrier organic anion transporter family member 2A1
|
SLCO2A1
|
1
|
homo sapiens
|
P29474
|
Nitric oxide synthase
|
NOS3
|
2
|
homo sapiens
|
P08684
|
Cytochrome P450 3A4
|
CYP3A4
|
1
|
homo sapiens
|
P09211
|
Glutathione S-transferase P
|
GSTP1
|
2
|
homo sapiens
|
Q99835
|
Smoothened homolog
|
SMO
|
1
|
homo sapiens
|
Q9NYA1
|
Sphingosine kinase 1
|
SPHK1
|
1
|
homo sapiens
|
P48039
|
Melatonin receptor type 1A
|
MTNR1A
|
1
|
homo sapiens
|
Q03181
|
Peroxisome proliferator-activated receptor delta
|
PPARD
|
1
|
homo sapiens
|
P10145
|
Interleukin-8
|
CXCL8
|
1
|
homo sapiens
|
P62993
|
Growth factor receptor-bound protein 2
|
GRB2
|
1
|
homo sapiens
|
P01857
|
Immunoglobulin heavy constant gamma 1
|
IGHG1
|
2
|
homo sapiens
|
P35228
|
Nitric oxide synthase
|
NOS2
|
20
|
homo sapiens
|
P04798
|
Cytochrome P450 1A1
|
CYP1A1
|
4
|
homo sapiens
|
Q12791
|
Calcium-activated potassium channel subunit alpha-1
|
KCNMA1
|
1
|
homo sapiens
|
2, Licorice induced tumor cells cycle arrest mainly by down-regulating Cyclin D1-CDK4
To further study the anti-cancer effect of licorice on NSCLC, we first tested the effects of licorice on the growth of tumor cells. According to the CCK8 assay results shown in Fig. 3a, we could recognize that licorice induced a concentration-dependent inhibition of H1975 cell proliferation. Treating licorice two days with concentrations of 3200, 5600 and 7200µg/mL, we found that compared with DMSO treatment, the H1975 cell growth decreased by 25, 48 and 87%, respectively. Moreover, the IC50 value on it were 5400µg/mL.
Next, given the analysis of systems pharmacology for licorice, and a number of studies have shown that the negative effects of licorice or its relatives on cell cycle progression[15, 16, 19, 20], we reasoned that licorice might influenced cell cycle to exert the anti-tumor effect on NSCLC to some extent. To test the hypothesis, we treated H1975 cells with different concentrations of licorice followed by flow cytometry analysis of cell cycle profile. Strikingly, H1975 cells subjected to licorice led to a significant increase in the number of cells arrested at G0/G1 growth phase, in a dose-dependent manner, compared with vehicle control containing media (shown in Fig. 3b and Fig. 3c). At the same time, the number of cells at both S growth phase and G2/M growth phase slightly decreased (Fig. 3c). This finding consistent with previous study that licorice induced G1 cell cycle arrest in MCF-7 human breast cancer cells[16].
It has been known that cyclin-dependent kinase (CDK)/cyclin complexes, such as CDK2/Cyclin E, CDK4, CDK6/CyclinD1, and P21 play crucial roles in cell cycle progression[21]. Therefore, to elucidate the underlying molecular mechanism with which licorice induced cell cycle arrest at G0/G1 growth phase, immunoblot analysis were performed to evaluate cell cycle-related protein abundance in vitro experiment. Notably, we found that the levels of CDK4, cyclin D1 were reduced, and the effect was concentration dependent, while the expression of CyclinB1 and CyclinA2 was relatively maintained at the level of the control group following licorice treatment (Fig. 3d and Fig. 3e). Interestingly, the expression of p21, a CDK inhibitor, was slightly decreased in response to licorice exposure vs control group (shown in Fig. 2d).
In addition, previous work uncovered that cyclin D1 degradation occurs mainly at the G1/S phase boundary[21, 22]. Collectively, these results indicated that licorice is likely to induce tumor cells arrested at G0/G1 growth phase by down-regulating CDK4-Cyclin D1 complex.
3, Licorice positively regulates PD-L1 protein abundance
It has been shown that PD-L1 expression can be modulated at both transcriptional and post-translational levels, however, it is not yet clear whether PD-L1 expression is regulated under physiological conditions for example during cell cycle progression[23–26]. In this setting, to further understand the connection between PD-L1 and cell cycle, we used cell synchronization by nocodazole arrest and immunoblot analysis to explore variation of PD-L1 during cell cycle. As shown in Fig. 4a and Fig. 4b, we found that PD-L1 protein expression increased in M/early G1 phases, followed by a great decrease in late G1/S phases.
As our results showed that licorice down-regulated Cyclin D1-CDK4 expression to arrest cell cycle progression, we probed whether licorice participated in variation of PD-L1. To do this, we treated H1975 cells with different concentration of licorice, followed by immunoblotting analysis. Strikingly, licorice administration results in a significant increase in the expression of PD-L1 protein (Fig. 4c and Fig. 4d), in a dose-dependent manner. Furthermore, recent finding had shown that Cyclin D-CDK4 kinase destabilized PD-L1, inhibition of CDK4/6 in vivo increased PD-L1 protein levels[27]. Together, these findings indicating that increased levels of PD-L1 expression by licorice correlated with down-regulation of Cyclin D1-CDK4 expression.
4、Licorice induce tumor regression by affecting Cyclin D1-CDK4-PD-L1 axis
Based on previous studies that various natural compounds in licorice possess effective antitumor activity[14, 16, 28, 29], we wanted to know whether licorice can function in vivo to suppress tumor progression for NSCLC. To do so, we utilized C57/BL6 female mice bearing LLC tumor to assess the anti-tumor impact of licorice. And size-matched tumor-bearing mice (TBMs) were divided into 4 groups randomly and received the administrations (as depicted in Fig. 5a).
By day 20 of treatment, as expected, all control mice encountered humane endpoints. Then mouse from each group were killed and dissected tumor, mouse serum was taken out and stored for subsequent experiment.
It is critical to note that licorice treatment result in a 64.9% tumor volume regression, and we found that there was slightly inhibitory effect on tumor volume of mice treated with anti-PD-L1 antibody alone vs control mice. Interestingly, we also observed a 54.7% tumor volume reduction in licorice + anti-PD-L1 mice compared with control mice over time. (Fig. 5a).
In keeping with our finding of tumor volume, treatment of licorice led to a significant induction of tumor weight, this also occurred in licorice + anti-PD-L1 group compared with untreated group. However, slight reduction of tumor weight was observed in anti-PD-L1 alone group (Fig. 5b).
Having pinpointed the critical role for licorice in affecting Cyclin D1-CDK4 expression in vitro, we next examined whether licorice had similar influence in vivo. Therefore, we assayed cell cycle-related protein for tumor tissue using the immunoblot method. Consistent with earlier observations in vitro (Fig. 3d), licorice treatment markedly reduced the abundance of CDK4、Cyclin D1, and importantly led to a dramatic PD-L1 accumulation compared with control group (Fig. 5c and 5d).
Therefore, these results coherently indicated that licorice might mainly function through down-regulating CDK4-Cyclin D1 to stabilize PD-L1 and subsequently suppress tumor progression.
5, Licorice increased antigen presentation and infiltration of CD8+ T cell
Furthermore, the results of systems pharmacology analysis indicated that kinds of compounds of licorice correlated with CD8+ T-cell (Fig. 6a, figure S2a, figure S2b), then intratumoral CD8+ T-cell infiltration in tumor tissue lysates were measured by flow cytometry analysis. Importantly, CD8+ T cell infiltration of licorice-treated mice we detected increased by 6% of that in untreated mice (Fig. 6c, Fig. 6d and figure S2c). In further support of a physiological role for licorice in promoting CD8+T cell infiltration, we used the mice serum to perform ELISA-based assays and found a remarkable increase of IFN-γin licorice-treated mice (Fig. 6e). These results were in line with a previous study that CDK4/6 inhibitors induce breast cancer cell cytostasis and enhance their capacity to present antigen and stimulate cytotoxic T cells[30].
Next, to gain insights into the physiological role of licorice in modulating tumor regression at gene level, RT-qPCR analysis was performed. Specifically, we sought to determine relative mRNA levels of antigen presentation genes by RT-qPCR analysis, and observed that transporter–MHC interactions (Tap-bp) had at least a 15x fold increase in licorice-treated tumor tissues compared to control tumor samples, and peptide transporters (Tap1 and Tap2) were also markedly up-regulated in licorice-treated tumors, although directing peptide cleavage (Erap1) hardly change to some extent. (Fig. 6b).
Altogether, these studies indicated that licorice increased expression of antigen presentation genes and promoted CD8+ T cell infiltration for tumor tissue.