1. Introduction
The whole number of plant secondary metabolites (PSMs) or phytochemicals, largely consisting of polyphenols, have been shown to possess anticancer activity against chemically induced animal tumors of various types [
1,
2,
3,
4]. They reduce the incidence and multiplicity of benign and malignant tumors induced in rodents in colon by 1,2-dimethylhydrazine or azoxymethane, in breast and ovary by 7,12-dimethylbenz(a)anthracene and in breast by N-methyl-N-nitrosourea. Mechanistic data, obtained on human cancer cells growing ex vivo and cultured in vitro, confirmed antiproliferative, proapoptotic, anti-inflammatory and immunomodulatory effects of PSMs, which demonstrated anticancer activity against chemically induced tumors [
5,
6,
7,
8]. Chemopreventive effects of genistein, resveratrol, berberine and some other PSMs were shown in a number of clinical trials [
2,
9,
10,
11].
Anticarcinogenic PSMs, characterized by the presence of aromatic rings with hydroxyl and other substituents, can bind various molecular targets in cells, such as receptors, enzymes of xenobiotic metabolism and epigenetic regulation of transcription, components of signaling pathways, enzymes of DNA repair and metabolism [
12,
13,
14,
15]. A wide range of PSM targets makes it extremely difficult to analyze consequences of different interactions, which impact the integral result of PSM action. Having unique structure, every PSM is characterized by its own spectrum of targets, however, most anticarcinogenic PSMs possess affinity to DNA. PSMs interact with DNA via van der Waals, ionic, and hydrogen bonds without formation of covalent bonds, that explains why they are not genotoxic. Using various sophisticated technics intercalation into DNA helix was shown for apigenin, delphinidin, fisetin, epigallocatechin-3-gallate (EGCG), genistein, naringenin, quercetin, resveratrol and sanguinarine [
16,
17,
18,
19,
20,
21]. Curcumin and sanguinarine interact with DNA as minor groove binders [
16,
22]. G-quadruplex binding and stabilization were shown for berberine, curcumin, EGCG, fisetin, kaempferol, quercetin, sanguinarine [
23,
24,
25,
26,
27,
28,
29]. Formation DNA-PSM complexes can affect the geometric characteristics and thermodynamic stability of DNA duplex, its flexibility and physicochemical properties, as well as its ability to form various alternative DNA structures [
19,
23,
24,
25,
26,
27,
28]. PSMs can cover up DNA sites, which are recognized by enzymes of DNA packaging, epigenetic regulation, repair, transcription and replication.
In our previous study we demonstrated that most of DNA-binding PSMs (11 compounds out of analyzed 15 PSMs with anticarcinogenic activity and ability to bind DNA) cause depletions of linker histones H1.2 and H1.4 from chromatin fraction that should be reflected on three-dimensional organization (3D structure) of chromatin and DNA packaging processes [
30]. Noteworthy, that in multiH1 knocked-down cells, chromatin opening promotes the upregulation of repetitive elements, ultimately triggering an interferon (IFN) response [
31]. In particular, Izquierdo-Bouldstridge et al. demonstrated that histones H1.2 and/or H1.4 are involved in the expression control of transposable elements (TEs) that influences innate immunity regulation. In general, H1 linker histones are enriched in the constitutive heterochromatin with silent repetitive elements LINEs, SINEs, and repeats containing endogenous retroviruses [
32,
33]. Moreover, in our previous study devoted to chromatin destabilization by a new anticancer drug Curaxin CBL0137 we revealed activation of type I IFN signaling accompanied by repetitive DNA transcription [
34]. Chromatin remodeling, caused by ATRX protein, was also demonstrated to activate type I IFN signaling [
35].
IFNs represent key modulators of the immune response. These cytokines with potent antiviral and growth-inhibitory effects play critical roles in the first line of defense against infections and homeostatic disorders during cancer pathogenesis [
36,
37]. IFN signaling activation was described in several studies devoted to effects of some PSMs. In particular, IFN activation was observed when cells were treated with resveratrol [
38,
39], berberine [
37,
40], fisetin [
42], naringenin [
43,
44], sanguinarine [
45], quercetin [
46]. All these studies were performed using separate PSMs and different cancer cells, which makes it difficult to compare their effects, and they do not show possible mechanisms of IFN activation. However, these data and our previously obtained results concerning PSM influence on linker histone location in cell nuclei provide a good basis for clarifying the question of whether PSM-induced chromatin remodeling is accompanied by IFN activation. This clarification should both expand our understanding of molecular effects induced by anticarcinogenic PSM and reveal cell response on chromatin rearrangements caused by different chemicals. The latter may be a strategic basis for the development of new non-genotoxic chemopreventive and anticancer drugs targeting chromatin structure and function.
Thus, the aims of our study include the analysis of the influence of 15 anticarcinogenic DNA-binding on INF-signaling activity, on the patterns of INF-responsive genes and on transcription of repetitive non-coding DNA. At last, the main goal of our study was to compare the data obtained with the abilities of PSMs to cause linker histones H1.2 and H1.4 depletions, which we described in our previous publication [
30].
Previously linker histones H1.2 and H1.4 depletions under PSM treatment were observed on HeLa cells and confirmed on T47D cells, that determined the choice of cell lines for the study presented now. We applied flow cytometry of genetically modified HeLa cells with the reporter mCherry gene under control of IFN sensitive responsive element (ISRE) and using immunofluorescent staining of ORF1 LINE1 and H2Ax-gamma. We also used quantitative RT-PCR for analysis of several IFN-responsive gene activation and the pattern of IFN-responsive genes was determined using Human Signal Transduction Pathway Finder RT2Profiler PCR Array (HSTPF, Qiagen, PARN-014Z).
3. Discussion
Over the last thirty years there has been great progress in understanding innate and adaptive immunity thanks to discovering different pattern recognition receptors (PRRs), which were shown to be associated with pathogens [
49].
Four major sub-families of PRRs include more than 4 hundreds of receptors, in particular, toll-like receptors (TLR), nucleotide-binding oligomerization domain—Leucin Rich Repeats-containing receptors (NLR), the retinoic acid-inducible gene 1 (RIG-1)-like receptors, and the C-type lectin receptors [
50]. PRRs discovery PRR discoveries allowed to explore the genius idea of «chemical binding of exogenous substances to cell», which dominated Paul Erlich’s life [
49,
51]. Based on the information about immune system functioning and the role of PRR activation in immune response, P.Matzinger elaborated “the Danger theory” that immune system responses are less concerned with the self/unself origin of the antigens than with the context of its influence on tissue homeostasis [
52,
53,
54]. When PRRs interact with their ligands, presented as the exogenous molecules (pathogen and environmental chemicals) as well as some internal structures associated with homeostasis disorders, it induces cell response. This response is in turn manifested as significant changes in cell signaling, including type I IFN signaling activation, which is considered to focus on identifying viral nucleic acids in the midst of exceedingly hostderived RNA and DNA [
55].
From this point of view, the agents, which change DNA-protein interactions and influence chromatin structure should also be recognized at the molecular level as the danger signals, as it could influence the pattern of the transcribed sequences including both coding and non-coding DNA. Recently we have found that a number of PSMs could cause linker histone depletion from chromatin [
30]. It may be proposed that PRRs recognize these DNA-binding PSMs or some internal structures appearing after PSM-DNA complex formation.
We separated the studied PSMs into groups depending on their ability to cause depletion of linker histones: compounds 1 to 11 in our previous experiments have this ability while compounds 12 to 15 do not (
Figure 6) [
30]. A more comprehensive analysis of type I IFN signaling activation pattern where we used Human Signal Transduction Pathway Finder RT2Profiler PCR Array showed results that correspond perfectly to our hypothesis: we revealed IFN signaling activation under the treatment of compounds 1-11. However, it should be emphasized that while compounds 9-11 caused more intensive depletion of H1.4 compared to H1.2, their effects were lower than the effects of compounds inducing significant depletion of both H1.2 and H1.4.
These data mainly correspond to the results of our ISRE-mCherry reporter analysis, and we explain some small discrepancies of the PSM effects by the fact that in this part of the study we only used ISRE to assess IFN signaling activation. The result showing that PSMs 12-15, which did not cause linker histone depletion, are not able to induce type I IFN signaling activation was observed in both experiments, when two alternative techniques of IFN signaling analysis were applied.
In regards to previously published data of type I IFN signaling activation by some PSMs, it was not studied very intensively. However, our results correspond to the published data. In particular, it was shown that quercetin and fisetin activate IFN-α in RAW 264.7 cells [
56]. In the study of Lin et al. resveratrol was shown to induce TLR9 activating IFN-β signaling [
57]. Activation of IFN-β signaling was also revealed in RAW264.7 and HEK293T cells after the treatment with berberine [
58]. Naringenin induced IFN-α activation in U2OS cells that was demonstrated both by luciferase reporter assay and RT-PCR [
59]. Sanguinarine was shown to enhance type I INF signaling in cultured monocyte-derived macrophages [
60]. In the study of Ullah et al., contradictory results were published regarding genestein ability to influence type I IFN signaling: using STING competent mouse L929 cells demonstrated genistein positive effect, stably expressing an ISRE-luciferase, while the same cells cocultured with STING-deficient cGAS-overexpressing human HEK-cGASlow cells showed the opposite effect, caused by STING blocking [
61]. At the same time, another study observed antiviral activity of genistein, which is considered to be the result of type I IFN signaling activation [
62].
Concerning PSMs 12-15 that do not cause linker histone depletion, the following data were published: thymoquinone was shown to actually decrease type I IFN signaling activity in RAW 264.7 and MCF-7 cells [
63]; for genosinoside RB1 in CRFK 157 cells after 48h treatment, no activation of type I IFN signaling happened [
64]; apigenin was shown to influence the inhibitory effect of IFN-α on cancer cell viability, wherein said viability is mediated by inhibition of 26S proteasome, however the effect of apigenin itself on type I IFN signaling was not analyzed [
65].
Our research on the activation of IFN signaling by PSMs also revealed that PSMs 3-8, which causes significant depletion of the linker histone H1.2 and H1.4, activate LINE1 expression. This observation concurs with the results of Izquierdo-Bouldstridge et al. who demonstrated that histones H1.2 and/or H1.4 participate in repression of repeats [
31]. It also perfectly corresponds to a well-known fact that LINE1 expression along with other TEs stimulate type I IFN signaling [
32,
66]. PSMs 9-11, which caused more intensive depletion of linker histone H1.4 than H1.2, induce IFN signaling less actively. It may be proposed that it is the consequences of the differential presence of H1 variants within transposable element classes and families described by Salinas-Pena et al. [
32]. For instance, in T47D cells and to some extent also in HeLa cells H1.2 and H1.4 are enriched in different TEs, being H1.4 enriched in evolutionary recent SVA, Alu, L1 and LTR, while H1.2 is enriched in older TEs. Noteworthy, fisetin and quercetin (PSMs 1 and 2 in our study) did not induce LINE1 expression, although they do cause significant depletion of the histones H1.2 and H1.4. It also stands to mention that their chemical structures are very similar (
Figure S1). Thus, we revealed that influence of PSMs on chromatin structure via linker histone depletion may be accompanied by LINE1 expression enhancement, which in turn impacts type I IFN signaling activation and, consequently, impacts anticancer activity of the corresponding PSMs group.
Additionally, literature search showed that for all the PSMs we studied their peculiar direct or indirect influence on PRR-induced signaling had already been described. In particular, it has already been shown that fisetin binds to TLR4, inhibits the binding of lipopolysaccharide (LPS) to the TLR4/MD2 complex and attenuates inflammatory reaction via TLR4/NLRP3 inflammasome pathway [
67,
68,
69,
70], while quercetin and resveratrol inhibit TLR4 and inflammasome activation [
71,
72,
73,
74]. Genistein [
75] and berberine [
40] were also demonstrated to have anti-inflammatory effects via suppression of the toll-like receptor 4-mediated signaling pathway. Naringenin suppresses inflammatory responses by regulation of cell surface TLR2 functioning [
43]. Delphinidin inhibits LPS-induced TLR4, MUC8, and MUC5B expression [
76]. Curcumin was described to inhibit extracellular TLR 2 and 4 and intracellular TLR9 [
77]. Kaempferol attenuates TLR4/NF-κB pathway activation in LPS-activated BV2 cells [
78]. Sanguinarine inhibits TLR4/NF-κB pathway in H9c2 cardiomyocytes and thus attenuates LPS-induced inflammation [
79], while it up-regulates expressions of endosomal TLRs [
80]. EGCG was also revealed to suppress LPS-induced TLR4 activity [
81]. Apigenin inhibits the LPS-mediated inflammatory mediator production in keratinocytes by reducing the TLR4-dependent activation of Akt, mTOR, and NF-κB pathways [
82]. Thymoquinone was shown to block TLR4/NF-κB signaling pathway in microglia cells [
83]. Ginesinoside Rb1 reduces TLR4 dimerization followed by inhibiting the TLR4-MyD88-NF-κB/MAPK pathways [
84]. Coumarines were shown to attenuate inflammation also via TLRs [
85]. Thus, anti-inflammatory effects were described for all the PSMs considered in our study, which should impact anticancer activity along with type I IFN signaling accompanying the depletion of linker histones.
4. Materials and Methods
4.1. Cell Culture
The HeLa cell line was obtained from the Blokhin CRC cell collection. HeLa-TI-ISRE-mCherry cells were kindly provided by Dr. Gurova, the Department of Cell Stress Biology at Roswell Park (Buffalo, NY, USA). Preparation and maintenance of HeLa-TI-ISRE-mCherry cells, containing integrated red fluorescent protein (mCherry) gene, driven by a consensus IFN-sensitive response element (ISRE), were described previously [
34]. Human breast cancer cells T47D were kindly provided by Dr. Jordan, Department of Molecular Genomics, Molecular Biology Institute of Barcelona IBMB-CSIC, Scientific Park of Barcelona, 08028 Barcelona, Catalonia, Spain. Cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM, С420p, PanEco, Moscow, Russia) supplemented with L-glutamine (0.584 mg/mL) (F033Е, PanEco, Moscow, Russia), penicillin (50 U/mL), and streptomycin (50 µg/mL) (А063p, PanEco, Moscow, Russia) and 10% fetal bovine serum (Biowest, S1810-500, Nuaillé, France). Cell lines were incubated at 37 °C and 5% CO2. All cell lines were validated by STR profiling and tested negative for mycoplasma.
4.2. Plant Secondary Metabolites
All of the studied compounds were obtained from Chemlight, Moscow, Russia (1). We studied apigenin (CAS 520- 36-5), (1); berberine (CAS 633-65- 8), (1); coumarin (CAS 91-64-5), (1); curcumin (CAS 458-37-7), (1); delphinidin (CAS 13270- 61-6), (1); EGCG (CAS 989-51-5), (1); fisetin (CAS 528-48-3), (1); genistein (CAS 446-72-0), (1); ginsenoside Rb1 (CAS 41753-43-9), (1); kaempferol (CAS 520-18-3), (1); naringenin (CAS 480-41-1), (1); quercetin (CAS 117-39- 5), (1); resveratrol (CAS 501-36-0), (1); sanguinarine chloride hydrate (CAS 5578-73- 4), (1); thymoquinone (CAS 490-91-5), (1).
4.3. Other Chemicals and Reagents
Curaxin CBL0137 was provided by Incuron, Inc., Russia. TRIzol™ Reagent (15596026), Moloney Murine Leukemia Virus Reverse Transcriptase (M-MLV RT) (18057018) and Random(dN)10 (SB002) were purchased from Evrogen, Moscow, Russia. dNTP mix; deionized water, nuclease-free; Taq DNA polymerase; 10X Taq Turbo Buffer; SYBR® Green dye and primers were purchased from Evrogen, Moscow, Russia. Triton X-100 (CAS 9002-93-1) was purchased from BioInnlabs, Rostov-on-Don, Russia. Dimethyl sulfoxide (DMSO, 67-68-5 | 102952), cOmplete™, Mini Protease Inhibitor Cocktail (cat. 11836153001), Phosphate-buffered saline (PBS, P4417), bovine serum albumin (CAS 9048-46-8), IFN-α A Protein, Recombinant human (P01563) were purchased from Sigma Aldrich (Merck), Bengaluru, Karnataka, India. Versene Solution (Р080p), Trypsin-EDTA 0.25% solution with Hanks salts (P043p) and Phosphate-buffered saline (PBS, P4417) were purchased from PanEco, Moscow, Russia. DC™ Protein Assay Kit I (5000111EDU) was purchased from Bio-Rad (Moscow, Russia). Clarity Max™ Western ECL Substrate for Chemiluminescent Detection of Horseradish Peroxidase (HPR) Conjugates (cat. 1705062) was purchased from Helicon, Moscow, Russia. 2.5x Reaction mixture for qRT-PCR in the presence of SYBR Green I dye (M-427) was purchased from Syntol (Moscow, Russia). Antibodies LINE1-ORF1 (cat# MABC1152, 1:500) were purchased from Sigma Aldrich (Merck), Bengaluru, Karnataka, India; γ-H2AX (cat# ab26350, 1:700) and and Donkey Anti-Mouse IgG H&L (Alexa Fluor® 488, cat# ab150105; 1:1000)—from Abcam, Cambridge, UK).
4.4. Quantitative Reverse Transcriptase-Polymerase Chain Reaction for Analysis of Expression of LINE1 and PSM-Induced Interferon Signaling
For the assay, tumor cells (HeLa) were seeded in 6-well plates (105 cells per well in 2 ml DMEM) and incubated with various concentrations of compounds for 24 h and IFN-α (103UI/ml) used as a positive control for the IFN Signaling analysis. Total RNA was then extracted using TRIzol™ Reagent according to the manufacturer’s protocol. cDNA was synthesized using a reverse transcription reaction. Total RNA (1 μg, from both control and treated cells) was reverse transcribed using M-MLV RT reverse transcriptase and random Random(dN)10 in a reaction volume of 20 μl according to the manufacturer’s protocol (Evrogen, Russia). RNA quantification was performed using NanoDrop Lite (ThermoScientific, Waltham, MA, USA).
For analysis of expression of amplicones LINE1 qRT-PCR was carried out in a reaction mixture containing Master Mix (0.3 mM dNTP mix (10 mM each), 3 mM MgCl2, deionized water, nuclease-free, SYBR® Green dye, 10X Taq Turbo Buffer, 0.2 U/ µl Taq DNA polymerase), 0.2 µM forward and reverse primers, 5 ng of DNA template, in accordance with the manufacturer’s protocol (Evrogen, Russia). Thermal cycling conditions were as follows: initial denaturation step by heating at 95°C for 5 min, followed by 40 cycles of 15 s initial denaturation (at 95°C), 20 s at the appropriate melting temperature according to the primers, and 25 s extension at 72°C. Expression of the gene of interest was normalized to the constitutively expressed housekeeping genes RPL0 and HAPDH. The relative expression level was calculated for each sample using the 2−ΔΔCt method. All experiments were performed at least in triplicate biological replicates.
The sequences of the gene-specific primers used for qRT-PCR were as follows (Primer design from [
47]):
LINE1_amplA_F: 5′GCCAAGATGGCCGAATAGGA 3′
LINE1_amplA_R: 5′AAATCACCCGTCTTCTGCGT 3′
LINE1_amplB_F: 5′CGAGATCAAACTGCAAGGCG 3′
LINE1_amplB_R: 5′CCGGCCGCTTTGTTTACCTA 3′
LINE1_amplC_F: 5′ TAAACAAAGCGGCCGGGAA 3′
LINE1_amplC_R: 5′ AGAGGTGGAGCCTACAGAGG 3′
LINE1_ORF1_F: 5′ ACCTGAAAGTGACGGGGAGA 3′
LINE1_ORF1_R: 5′CCTGCCTTGCTAGATTGGGG 3′
RPL0 F: 5′CCTTCTCCTTTGGGCTGGTCATCC A 3′
RPL0 R: 5′CAGACACTGGCAACATTGCGGACAC 3′
HAPDH F: 5′GTCTCCTCTGACTTCAACAGCG 3′
HAPDH R: 5′ACCACCCTGTTGCTGTAGCCAA 3′
The sequences of the gene-specific primers used for type I IFN signaling qRT-PCR were as follows (Primer design from [
31]):
IFI27_F: 5′ TGCTCTCACCTCATCAGCAGT 3′
IFI27_R: 5′ CACAACTCCTCCAATCACAACT 3′
OASL_F: 5′ GGGACAGAGATGGCACTGAT 3′
OASL_R: 5′ AAATGCTCCTGCCTCAGAAA 3′
IRF1_F: 5′ TTTGTATCGGCCTGTGTGAATG 3′
IRF1_R: 5′ AAGCATGGCTGGGACATCA 3′
For analysis of gene expression of type I IFN signaling qRT-PCR was performed in 96-well Human Signal Transduction PathwayFinder™ RT 2 Profiler™ PCR Array plates (
https://geneglobe.qiagen.com/us/product-groups/rt2-profiler-pcr-arrays/PAHS-064Z, Qiagen, PAHS-064Z, Hilden, Germany) according to the manufacturer’s protocol: 95°C for 10 min, then 40 cycles of 95°C for 15 s and 60°C for 1 min. Each RT2 Profiler PCR array contains gene-specific primers for qRT-PCR assays for a carefully screened set of 84 genes, consisting of IFNs, IFN receptors, IFN regulatory factors, and IFN-responsive genes (
Table 1).
Expression of genes of interest was normalized to constitutively expressed housekeeping genes (ACTB, B2M, GAPDH, HPRT1, RPLP0). Relative expression levels were calculated for each sample using the 2−ΔΔCt method using the manufacturer’s software. All experiments were performed at least in triplicate biological replicates.
4.5. Analysis of ISRE-mCherry Reporter Activation in HeLa-TI-ISRE-mCherry Cells by Flow Cytometry
IFN response in HeLa-TI-ISRE-mCherry cells treated with PSMs was assessed by the proportion of the cells expressing mCherry driven by ISRE as well as by mCherry mean fluorescence intensity (MFI) using a BD FACSCanto™ II flow cytometer (BD Biosciences, San Jose, CA, USA). Cells were seeded in 6-well plates (10
5 cells per well in 2 ml DMEM) and incubated with PSM at non-toxic concentrations for 24 h. For PSMs inducing IFN response after 24 h treatment we studied the dynamics of their effects at 1, 6 and 24 h. After the treatment with PSMs cells were removed from the culture plates using Versene Solution and 0.25% trypsin-EDTA and washed with PBS. To maintain high cell viability, a PBS solution with 2% fetal bovine serum was used as a cell storage buffer. The concentration of dimethyl sulfoxide (DMSO) in the medium for all compounds did not exceed 0.01%. All experiments were performed in triplicate biological replicates. The obtained data were analyzed using WinList™ 3D software (Version 9.0.1, Verity Software House,
https://www.vsh.com/products/winlist/index.asp, Topsham, ME, USA).).
4.6. Analysis of PSM Induced LINE1 Activation by Immunofluorescent Antibody Staining and Flow Cytometry
To analyze PSM induced LINE1 activation, HeLa cells were seeded in 6-well plates (10
5 cells per well in 2 ml DMEM). After 24 h, cells were treated with compounds of interest at IC20 or non-toxic concentrations and incubated for 24/72 h. Then, the cells were removed from the substrate with trypsin, washed three times with PBS and fixed in cold 4% paraformaldehyde for 15 min. After next three washes with PBS, they were permeabilized with cold 0.3% Triton-X100 for 7 minutes and blocked with bovine serum albumin for 1 h. Cells were immunofluorescently stained with antibodies to LINE1-ORF1, γ-H2AX, and subsequent binding with secondary antibodies AlexaFluor488 was carried out in the dark. Cells were washed with PBS and analyzed on a BD FACSCanto™ II flow cytometer (BD Biosciences, San Jose, CA, USA). Proportions of the cells positive for the fluorescent signal and the average intensity of the cell fluorescence normalized to the control were assessed. The obtained data were analyzed using WinList™ 3D software (Version 9.0.1, Verity Software House,
https://www.vsh.com/products/winlist/index.asp, Topsham, ME, USA).
4.7. Annexin-FITC/Propidium Iodide Double Staining
Cells were stained with annexin V-FITC and PI to evaluate apoptosis by flow cytometry according to the manufacturer’s instructions to the FITC Anexin V Apoptosis Detection Kit I (Sigma-Aldrich, St. Louis, MI, USA). Cells were treated with maximum non-toxic concentrations of PSM for 24 h. After treatment, cells were collected, washed twice with ice-cold PBS, and resuspended in 0.5 mL of annexin/V-FITC/PI solution for 30 min in the dark according to manufacturer protocol. After staining at room temperature, cells were analyzed by the BD FACSCanto™ II flow cytometer (BD Biosciences, San Jose, CA, USA). For each sample, 10,000 events were acquired and positive FITC and/or PI cells were quantified using WinList™ 3D software (Version 9.0.1, Verity Software House,
https://www.vsh.com/products/winlist/index.asp, Topsham, ME, USA).
4.8. Statistical Analysis
We compared the data from the experimental and control groups using one-way analysis of variance (ANOVA) and Dunnett’s post hoc test. Differences between groups were considered significant at a p-value <0.05. Statistical analyses were performed using GraphPad Prism 8.3.0 (GraphPad Software Inc., San Diego, CA, USA).
Figure 1.
Flow cytometry data for the expression of mCherry driven by ISRE in HeLa TI ISRE-mCherry cells after PSM treatment for 24 h. A. Color-numeric designation of PSMs in non-toxic concentrations. Ctr- control; IFN- IFN-α, 103 U/ml; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. This color-number legend is used in all figures. B. Proportions of the cells expressing mCherry. C. Mean fluorescence intensity of mCherry per cell. The data are presented as an average value ± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 1.
Flow cytometry data for the expression of mCherry driven by ISRE in HeLa TI ISRE-mCherry cells after PSM treatment for 24 h. A. Color-numeric designation of PSMs in non-toxic concentrations. Ctr- control; IFN- IFN-α, 103 U/ml; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. This color-number legend is used in all figures. B. Proportions of the cells expressing mCherry. C. Mean fluorescence intensity of mCherry per cell. The data are presented as an average value ± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 2.
Flow cytometry data for the expression of mCherry driven by ISRE in HeLa TI ISRE-mCherry cells after PSMs treatment for 1, 6 and 24 h. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 10- sanguinarine, 0.8µM. A. Proportions of the cells expressing mCherry. B. Mean fluorescence intensity of mCherry per cell. The data are presented as m± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 2.
Flow cytometry data for the expression of mCherry driven by ISRE in HeLa TI ISRE-mCherry cells after PSMs treatment for 1, 6 and 24 h. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 10- sanguinarine, 0.8µM. A. Proportions of the cells expressing mCherry. B. Mean fluorescence intensity of mCherry per cell. The data are presented as m± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 3.
Influence of PSMs on gene expression of type I IFN signaling pathway. A. Pattern of gene expression of type I IFN signaling. Data presented as Log2 (fold change) for each of 84 genes (HSTPF, Qiagen, PARN-014Z). IFN- IFN-α, 103 U/ml; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. B-D. mRNA expression of genes IFN-signaling normalized to RPL0 and HAPDH in T-47D and HeLa cell lines. Ctr- control; IFN- IFN-α, 103 U/ml; 3- resveratrol, 50µM; 5- genistein, 60µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. The data are presented as m± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. B. IFN-responsive gene IFI27. C. IFN-responsive gene OASL. D. IFN regulatory factor IRF1.
Figure 3.
Influence of PSMs on gene expression of type I IFN signaling pathway. A. Pattern of gene expression of type I IFN signaling. Data presented as Log2 (fold change) for each of 84 genes (HSTPF, Qiagen, PARN-014Z). IFN- IFN-α, 103 U/ml; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. B-D. mRNA expression of genes IFN-signaling normalized to RPL0 and HAPDH in T-47D and HeLa cell lines. Ctr- control; IFN- IFN-α, 103 U/ml; 3- resveratrol, 50µM; 5- genistein, 60µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. The data are presented as m± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. B. IFN-responsive gene IFI27. C. IFN-responsive gene OASL. D. IFN regulatory factor IRF1.
Figure 4.
Expression of three LINE1 amplicons (A, B, C) and ORF1 LINE1 gene in HeLa cells treated with maximal non-toxic concentrations of PSMs for 24 h. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. The data are presented as an average value ± SD. Significance of the differences between PSM treated cells and control untreated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 4.
Expression of three LINE1 amplicons (A, B, C) and ORF1 LINE1 gene in HeLa cells treated with maximal non-toxic concentrations of PSMs for 24 h. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 9- kaempferol, 2µM; 10- sanguinarine, 0.8µM; 11- EGCG, 65µM; 12- coumarin, 260µM; 13- ginsenoside Rb1, 30µM; 14- thymoquinone, 3µM; 15- apigenin, 5µM. The data are presented as an average value ± SD. Significance of the differences between PSM treated cells and control untreated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001.
Figure 5.
Flow cytometry analysis of Hela cells treated with PSMs in non-toxic concentrations and immunofluorescently stained ORF1 LINE1 or γ-H2AX. B. Example of the analysis of Hela cells with the immunofluorescently stained γ-H2AX treated with delphinidin for 24 h. A.B. The effect of PSM on the proportion of stained cells. C. 24-h PSM treatment. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 10- sanguinarine, 0.8µM. B. 72 h PSM treatment. Ctr- control; 1- fisetin, 13.5µM; 2- quercetin, 5µM; 3- resveratrol, 25µM; 4- berberine, 5µM; 5- genistein, 30µM; 6-naringenin, 26µM; 7-delphinidin, 50µM; 8- curcumin, 3,7µM; 10- sanguinarine, 0.4µM. The data are presented as an average value ± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. С. Measurement of proportion of apoptotic cells in the analyzed populations of fixed cells. D. Example of the analysis of Hela cells with the immunofluorescently stained γ-H2AX treated with delphinidin for 24 h.
Figure 5.
Flow cytometry analysis of Hela cells treated with PSMs in non-toxic concentrations and immunofluorescently stained ORF1 LINE1 or γ-H2AX. B. Example of the analysis of Hela cells with the immunofluorescently stained γ-H2AX treated with delphinidin for 24 h. A.B. The effect of PSM on the proportion of stained cells. C. 24-h PSM treatment. Ctr- control; 1- fisetin, 27µM; 2- quercetin, 10µM; 3- resveratrol, 50µM; 4- berberine, 10µM; 5- genistein, 60µM; 6-naringenin, 52µM; 7-delphinidin, 100µM; 8- curcumin, 7.5µM; 10- sanguinarine, 0.8µM. B. 72 h PSM treatment. Ctr- control; 1- fisetin, 13.5µM; 2- quercetin, 5µM; 3- resveratrol, 25µM; 4- berberine, 5µM; 5- genistein, 30µM; 6-naringenin, 26µM; 7-delphinidin, 50µM; 8- curcumin, 3,7µM; 10- sanguinarine, 0.4µM. The data are presented as an average value ± SD. Significance of the differences between control untreated cells and PSM treated cells was determined using ANOVA test and Dunnett’s post hoc test: significant difference, *— p < 0.05, **— p < 0.01, ***— p < 0.001, ****— p < 0.0001. С. Measurement of proportion of apoptotic cells in the analyzed populations of fixed cells. D. Example of the analysis of Hela cells with the immunofluorescently stained γ-H2AX treated with delphinidin for 24 h.
Figure 6.
Activation of IFN signaling type I by PSMs (number of genes with significantly increased expression, with increased expression by two times and more and with decreased expression). PMS separated into groups depending on their ability to cause depletion of linker histones and LINE1 transcription.
Figure 6.
Activation of IFN signaling type I by PSMs (number of genes with significantly increased expression, with increased expression by two times and more and with decreased expression). PMS separated into groups depending on their ability to cause depletion of linker histones and LINE1 transcription.
Type of gene products |
Gene products |
Number of genes |
IFNs(21)
|
IFN-α; IFN-β; receptor ligands |
5 genes |
IFN-γ; receptor ligands |
1 genes |
Hematopoietin & IFN class (D200-domain) cytokine receptor ligands |
10 genes |
Other IFN related genes |
5 |
IFN receptors(37)
|
IFN-α and IFN-β receptors |
2 |
IFN-γ receptors |
2 |
Hematopoietin, IFN class (D200-domain) receptors |
28 |
IFN regulatory factors (9) |
|
9 |
IFN-responsive genes(23)
|
Response to virus |
13* |
Transcriptional regulation |
2* |
Other IFN responsive genes |
8 |