1. Introduction
Cancer is one of the major cause of death globally, and a significant contributor to decreased life expectancy [
1]. Cancer of the prostate is an adenocarcinoma that often forms in the glandular prostate and can be identified by its characteristic glandular patterns under the microscope [
2]. This cancer can advance by spreading outside the prostate or to remain contained in the prostate for a very long time. The lymph nodes and bone are common sites where prostate cancer metastasizes; it is hypothesized that the prostatic venous plexus's connection to the spinal veins contributes to the latter [
3]. According to the World Health Organisation (WHO), there are 1,414,259 new cases of prostate cancer (PCa) each year, making it the second most common cancer in men and the fifth greatest cause of cancer-related mortality globally [
4]. In Africa, the rate of prostate cancer is 26.6% per 100,000 people, and in sub-Saharan Africa, there are an estimated 70,000 new cases per year [
5]. Countries with low socio-demographic indexes such as Nigeria and Kenya are affected the most by prostate cancer, and this has been attributed to the lack of effective preventive and treatment strategies in the countries [
6].
Prostatectomy and local radiation are used to treat localised PCa, which accounts for 90 % of PCa cases. However, PCa is diagnosed in an advanced stage in around 90 % of men with the disease, when androgen deprivation therapy (ADT) and chemotherapy are common treatment options. [
5,
7]. ADT treatment leads to recurrent androgen-independent prostate cancer within 2–3 years, with frequent metastases to regional lymph nodes or the pelvis, making the disease advanced [
8]. Although there are several prostate cancer chemotherapy treatments, there is still a gap in the therapeutic options for advanced prostate cancer. Drugs that target rapidly proliferating cancer cells such as docetaxel and paclitaxel can also damage healthy cells, causing fatigue, hypertension, hot flushes, arthralgia, fractures, peripheral oedema and rash [
9]. Therefore, there is a serious need to prospect better therapeutic interventions against PCa and herbal plant sources provide the greatest promise, and the “lowest hanging fruits”.
The use of herbal plants in cancer has gained substantial attention, and recently, research is ongoing, with the US National Cancer Institute (NCI) playing a pivotal role in the research of traditional medicine to treat cancer [
10]. Herbal plants have various advantages over chemical products due to their lower chances of inducing adverse effects, cost-effectiveness, their tolerability, and their reduced chances of developing resistance [
11]. Findings have shown that herbal plants exercise their anticancer properties owing to the existence of phytochemicals which act by creating antioxidant effects, repairing damaged DNA, boosting the immune system, inducing apoptosis and suppressing cell cycle [
12,
13].
Euphorbia ingens E.Mey. ex Boiss belongs to the plant family of
Euphorbiaceae [
14], it is known to contain latex, which is applied in traditional medicine for the treatment of cancer and other abnormalities including swellings, fistula, lesions, wounds, abscesses and burns [
15,
16]. In East Africa,
E. ingens is also used for the treatment of snakebites suggesting that the plant may possess an anti-venom for snakes[
17]. Additionally, there are scientific data demonstrating ichthyoidal, antitubercular, antimicrobial and antifungal activities of
E. ingens [
18,
19,
20]. We have previously shown that
E. ingens extract contain phytochemicals including phenols, tannins, terpenoids, flavonoids and saponins, which are generally associated with anticancer activity [
21]. However, despite the aforementioned roles of
E. ingens as a viable target for the management and treatment of diseases, scientific validation of its ethnobotanical use in cancer management and treatment is lacking. Therefore, we hypothesised that
E. ingens has the ability to selectively stop the proliferation of prostate cancer cells while not harming normal cells.
To test this hypothesis, we first applied network pharmacology approaches to determine whether compounds in E. ingens extract have molecular targets associated with PCa and further investigated the antiproliferative activity of the extract so as to validate the putative activity and targets. We showed that E. ingens has selective antiproliferative effect against prostate cancer cell line, with no toxicity towards the non-cancerous Vero cell line.
3. Discussion
Active surveillance, surgery, radiotherapy, hormonal therapy, immunotherapy and chemotherapy are treatment options currently available for prostate cancer. These options have major limitations, particularly the accompanying adverse side effects; hence, prostate cancer remains to be incurable. Nevertheless, natural products provide an outlet for identification of promising new anticancer agents that are highly efficient with low toxicity [
23]. Cancer cells exhibit sustained proliferation and resistance to cell death; and thus, compounds that can stop or slow down cell proliferation in rapidly dividing cells hold great promise as anticancer therapeutics [
24]. Based on the cell-based MTT assay, our findings demonstrated a significant selective antiproliferative effects of extracts from
E. ingens roots towards the DU-145 prostate cancer cell lines without affecting the non-cancerous ones (Vero E6). An IC
50 < 30 μg/ml is one of the established criteria by the US National Cancer Institute (NCI) to consider crude extracts as being cytotoxic. Interestingly, we observed that the ethyl acetate fractions of
E. ingens caused cytotoxicity in DU-145 with IC
50 value that was less than 10 µg/mL (
Table 1). Therefore,
E. ingens has significant potential for further studies as a potential chemotherapeutic agent. The inhibitory effects of
E. ingens ethyl acetate on DU-145 is better when compared to that reported for longifolene isolated from
Chrysopogon zizanioides (IC
50 of 78.64 µg/mL) on the same DU-145 cells [
25]. Meanwhile, El-Hawary et al.[
26] found
E. ingens to be inactive when tested against human colon adenocarcinoma (CACO2), human hepatoma (HepG2) and human breast adenocarcinoma (MCF-7) cell lines. This might be related to the difference in the solvents extraction, hence a likely different set of chemical compounds; while they used the methanolic extract of the plant, we test the ethyl acetate fraction of crude (dichloromethane:methanol) extract. A future study to investigate the cytotoxic effect of ethyl acetate fraction of
E. ingens on the CACO2 and HepG2 cell lines is encouraged. Meanwhile, a drug with a measure of the safety margin (selectivity index) of ≥2 is considered highly selective [
27]. The ethyl acetate fraction of
E. ingens had an SI greater than 2, an indicator of high selectivity for the cancer cells. Previous studies have also shown that
E. ingens bioactivities are not a result of general toxicity [
19].
The findings of this study depict a high abundance of terpenoids which act on different stages of tumour development and exhibit their anticancer properties by inducing autophagy in cancer cells
via a complex signalling pathway [
28,
29]. In recent study, sesquiterpenoids have been shown to restrict cell cycle in prostate cancer through induction of apoptosis [
30]. Flavonoids are known antioxidants under normal conditions and potent pro-oxidants in cancer cells, triggering the apoptotic pathways and downregulating pro-inflammatory signalling pathways [
31,
32]. Other present phytoconstituents, phenols, tannins, saponins, and sterols, were previously shown to possess anticancer properties. Phenols and tannins have been demonstrated to participate in cell cycle arrest, induce apoptosis, and suppress cancer cell proliferation and invasiveness [
32,
33]. Similar mechanisms as well as regulation of angiogenesis have been attributed to saponins and sterols [
13,
34]. Some of the GC-MS-identified compounds in this study have been isolated and reported to elicit anticancer activity while others have been identified to synergistically act with other compounds in many medicinal plants. For example, andrographolide, diterpenoid, that was isolated from
Andrographis paniculata Nees induced cell cycle arrest and apoptosis in HT-29 human colon cancer cells [
35]. By increasing intracellular reactive oxygen species, squalene induced anti-proliferative activity against ovarian, breast, lung, and colon cancers [
36]. Similarly, diterpene phenol, ferruginol has pharmacological properties such as inhibition of the growth rate of cancer cells [
37]. Other compounds which have also been reported to have anticancer activity include 1-octadecene, 1-heneicosanol, and 2,4-di-tert-butylphenol [
38,
39,
40]. Given that these compounds are found in the ethyl acetate fraction of
E. ingens, we suggest that they may be responsible for the demonstrated antiproliferative effects of the plant on DU-145 cells. However, further research is required to determine the antiproliferative activity of these isolated compounds, particularly on prostate cell lines.
Network pharmacology is currently used in cancer therapy to develop new drugs [
41]. The multi-target pathways application of network pharmacology is widely adopted to study the mechanism of action of traditional medicine; it identifies the active ingredients of plants, predict their targets, and subsequently combine them with disease targets to generate a presentable drug-target-disease relationship [
42]. We selected 7 GC-MS-identified compounds with good absorption, distribution, metabolism, and excretion (ADME) activity using the RO5, as well as blood–brain barrier, total polar surface area, CYP2D6 and CYP3A4. Drug screening and development relies heavily on pharmacokinetic characteristics. Without appropriate pharmacokinetic qualities, drugs will fail to attain requisite concentration in the target organs where they are needed to produce therapeutic effects [
43]. The candidate targets of the selected
E. ingens bioactive compounds for prostate cancer treatment were obtained; of which ESR1, IL6, MMP9, CDK2, MAP2K1, AR, PRKCD, CDK1, CDC25B and JAK2 were at the core position in the PPI network. These targets are considered the possible molecular targets of the drug-like compounds in
E. ingens ethyl acetate fraction against prostate cancer cells.
The MMPs family is known to have proteolytic effect on the cell membrane; member proteins such as MMP9 release proangiogenic factors which acts on endothelial cells to induce cell migration and proliferation [
44,
45]. An increased level of MMP9 was reported to cause metastasis in androgen-independent prostate cancer [
46]. There have been reports on the role of ESR1, PRKCD, and IL6 in the proliferation and migration of PCa cells, and inhibitors of JAK2 have be suggested to be important in the treatment of advanced PCa [
47,
48,
49,
50]. Targeted inhibition of MAP2K1 expression has been shown to elicit cell apoptosis and weakening of cell proliferation in DU-145 and PC-3 prostate cancer cells [
51]. The cell cycle protein-dependent kinase (CDK) and cell cycle proteins, as well as CDK inhibitors are essential in the regulation of cell cycle, hence, impairment in the activities of these cell cycle mediators is observed in many types of cancer. CDK2 is a core regulator of cell cycle through late G1-phase and S-phase. CDK2 is thought to be strongly linked to development of cancer, and accumulating evidence shows that inhibition of CDK2 induces cancer cell apoptosis without cellular damage [
52]. CDC25B is a cell cycle transitions regulatory enzyme; it is an important target of the checkpoint machinery in maintaining genome stability during DNA damage [
53]. Overexpression of CDC25B has been reported in many types of human cancers, and targeted cellular depletion of the enzyme in DU-145 facilitated rapamycin anticancer effects [
54]. Given the clear evidence that these genes are involved in the development and progression of prostate cancer, the ability to modulate them would have contributed to the observed antiproliferative property of the ethyl acetate fraction of
E. ingens.
The KEGG enrichment results showed that many disease pathways that were not relevant to this study were enriched, probably because the same molecular targets exist in the development of different diseases. The prostate cancer pathway was selected for analysis, the pathway showed a sets of multiple pathways, through which the therapeutic effect of
E. ingens on prostate cancer may be produced; these include the PI3K/Akt, MAPK and p53 signalling pathways. The phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) signalling pathway is an important tumour cell pathway which participates in the occurrence, invasion, and distant metastasis of prostate cancer [
55]. Long-term ADT can abnormally activate the PI3K/Akt pathway thereby enhancing the antiapoptotic ability of tumour [
56]. Therefore, the PI3K/Akt pathway is an important potential target of non-AR pathway in the treatment of PCa. Mitogen-activated protein kinases (MAPKs) are serine-threonine kinases that could link extracellular signals to fundamental cellular processes such as cell growth, proliferation, differentiation, migration, and apoptosis. The p53 pathway is one of the crucial signalling pathways for cancer cell apoptosis [
57], as a cancer suppressor gene, p53 regulate the downstream genes and take a part in DNA repair and regulation of the cell cycle and apoptosis. Hence, reactivation and restoration of p53 function holds great potential for the treatment of PCa.
Multiple lines of evidence in the field of cancer research have demonstrated the role of secondary metabolites in natural products is to affect gene expression by influencing significant transcription factors involved in carcinogenesis [
58]. We used RT-qPCR to validate the predicted molecular targets of
E. ingens in prostate cancer. Overexpression of CDK1 promotes progression of PCa, and when this kinase is hyperactivated, it mediates the phosphorylation that activates AR when there are no ligands [
59,
60]. AR signalling plays a crucial role in the growth of normal prostate tissue and PCa pathogenesis and progression. In advanced PCa stages, AR mutations and overexpression contribute to sustained proliferation [
61]. The treatment of DU-145 prostate cancer cells with
E. ingens resulted into a significant downregulation in AR when compared with the untreated control. Similar results were reported on the modulation of AR expression by the flavonoid genistein. [
62] However,
E. ingens showed upregulation in the expression of CDK1. This unusual finding was also observed by Mustafa et al.[
63]. CDK1 has been said to be capable of avoiding an accumulation of oncogenic mutations during cell division, that might explain the result [
64]. In cancer cells, the evasion of cell death is often achieved by either upregulating anti-apoptotic proteins like BCL-2 or by impairing the function of pro-apoptotic proteins such as caspase-3. BCL-2 controls the mitochondrial apoptotic pathway by binding to pro-apoptotic proteins and preventing pore formation and cytochrome c release. Apoptosis is executed by caspases and various upstream regulatory factors, including p53. We observed an upregulation in the expression of caspase-3 and p53 with concomitant downregulation of BCL-2 when
E. ingens-treated prostate cancer cells were compared with the untreated prostate cancer cells; this pattern was also reported by Eltamany et al.[
65]. Moreover, recent computational study revealed that flavonoids, found to be abundant in our study, have strong interaction with caspase-3, BCL-2 and p53 [
66]. The difference in the expression level of AR, p53, BCL-2 and caspase-3 between the
E. ingens-treated human prostate cancer cells and the untreated cells justifies the network pharmacology predictions; as these genes are distributed in the PI3K/Akt, MAPK and p53 signalling pathways, the results further suggest that
E. ingens’ antiprofelirative activity is likely by modulating the pathways to cause induction of apoptosis as well as suppression of cell cycle.
Figure 1.
Screening for cellular proliferation inhibition of E. ingens fractions: Inhibition of cellular proliferation following 48 h treatment at 200 µg/ml concentration of the water and ethyl acetate fractions of E. ingens on DU-145. Doxorubicin at 200 µg/ml was used as positive control, and 0.2% DMSO as negative control. Values are expressed as Mean ± SEM. All treatments were done in triplicates (n = 3).
Figure 1.
Screening for cellular proliferation inhibition of E. ingens fractions: Inhibition of cellular proliferation following 48 h treatment at 200 µg/ml concentration of the water and ethyl acetate fractions of E. ingens on DU-145. Doxorubicin at 200 µg/ml was used as positive control, and 0.2% DMSO as negative control. Values are expressed as Mean ± SEM. All treatments were done in triplicates (n = 3).
Figure 2.
Inhibition of cellular proliferation by E. ingens ethyl acetate fraction: Inhibition of cellular proliferation following 48 hours treatment with serial concentrations of E. ingens ethyl acetate fraction on DU-145 so as to determine IC50. 0.2% DMSO acted as negative control. Values are expressed as Mean ± SEM. All treatments were done in triplicates (n = 3).
Figure 2.
Inhibition of cellular proliferation by E. ingens ethyl acetate fraction: Inhibition of cellular proliferation following 48 hours treatment with serial concentrations of E. ingens ethyl acetate fraction on DU-145 so as to determine IC50. 0.2% DMSO acted as negative control. Values are expressed as Mean ± SEM. All treatments were done in triplicates (n = 3).
Figure 3.
Cellular safety: The potential cellular safety of E. ingens ethyl acetate was measured using noncancerous Vero E6. Cells were treated with serial concentrations of the E. ingens extract fraction for 48 hours. Values were expressed as Mean ± SEM, all treatments were done in triplicates (n = 3).
Figure 3.
Cellular safety: The potential cellular safety of E. ingens ethyl acetate was measured using noncancerous Vero E6. Cells were treated with serial concentrations of the E. ingens extract fraction for 48 hours. Values were expressed as Mean ± SEM, all treatments were done in triplicates (n = 3).
Figure 4.
GC-MS chromatogram of the chemical constituents present in E. ingens ethyl acetate fraction.
Figure 4.
GC-MS chromatogram of the chemical constituents present in E. ingens ethyl acetate fraction.
Figure 5.
Compound-disease targets Venny intersection diagram, with the compound targets on the left, the disease (PCa) targets on the right, and the two intersection targets in the middle.
Figure 5.
Compound-disease targets Venny intersection diagram, with the compound targets on the left, the disease (PCa) targets on the right, and the two intersection targets in the middle.
Figure 6.
PPI network of the key targets in E. ingens’ action against human prostate cancer.
Figure 6.
PPI network of the key targets in E. ingens’ action against human prostate cancer.
Figure 7.
Obtained ten hub genes. The larger the node is, the more important the target in the network.
Figure 7.
Obtained ten hub genes. The larger the node is, the more important the target in the network.
Figure 8.
Euphorbia ingens GO enrichment analysis (A) BP terms (B) CC terms (C) MF terms.
Figure 8.
Euphorbia ingens GO enrichment analysis (A) BP terms (B) CC terms (C) MF terms.
Figure 9.
KEGG signaling pathway analysis and prostate cancer pathway of E. ingens-PCa. (A) KEGG signaling pathway analysis. Each bubble represents a KEGG pathway on the vertical axis. The - logP values are shown on the horizontal axis. The size of each bubble indicates the number of genes enriched in each KEGG pathway. Larger bubbles indicate more genes involved in the pathway. The colour of each bubble represents the adjusted P-value of each KEGG pathway, with redder color indicating smaller adjusted P-value (B) Prostate cancer pathway (red marks represent potential targets for E. ingens intervention).
Figure 9.
KEGG signaling pathway analysis and prostate cancer pathway of E. ingens-PCa. (A) KEGG signaling pathway analysis. Each bubble represents a KEGG pathway on the vertical axis. The - logP values are shown on the horizontal axis. The size of each bubble indicates the number of genes enriched in each KEGG pathway. Larger bubbles indicate more genes involved in the pathway. The colour of each bubble represents the adjusted P-value of each KEGG pathway, with redder color indicating smaller adjusted P-value (B) Prostate cancer pathway (red marks represent potential targets for E. ingens intervention).
Figure 10.
Relative gene expression analysis of E. ingens treated and untreated DU-145 cells (A) AR, (B) BCL-2, (C) CDK1, (D) caspase-3 and (E) p53. ns p ˃ 0.05, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001 as compared to untreated control.
Figure 10.
Relative gene expression analysis of E. ingens treated and untreated DU-145 cells (A) AR, (B) BCL-2, (C) CDK1, (D) caspase-3 and (E) p53. ns p ˃ 0.05, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001 as compared to untreated control.
Table 1.
Summary of IC50, CC50 and selectivity index values.
Table 1.
Summary of IC50, CC50 and selectivity index values.
Extract |
IC50 (µg/ml) |
CC50 (µg/ml) |
SI |
E. ingens ethyl acetate |
9.71 ± 0.40a
|
80.19 ± 6.12a
|
8.26 |
Doxorubicin |
5.30 ± 0.11b
|
176.10 ± 8.09b
|
33.23 |
Table 2.
Phytochemical profile of E. ingens ethyl acetate fraction.
Table 2.
Phytochemical profile of E. ingens ethyl acetate fraction.
Phytoconstituent |
E. ingens ethyl acetate |
Alkaloids |
- |
Phenols |
++ |
Tannins |
+++ |
Terpenoids |
+++ |
Flavonoids |
+++ |
Saponins |
+++ |
Quinones |
- |
Sterols |
+++ |
Table 3.
The GC-MS identified compounds from E. ingens ethyl acetate fraction.
Table 3.
The GC-MS identified compounds from E. ingens ethyl acetate fraction.
Peak$Nr. |
Rt (min) |
Compound identified |
Peak$Area % |
MW$(g/mol) |
MF |
Structure Type |
|
5.302 |
1-decene |
0.29 |
140 |
C10H20
|
Alkene |
|
7.952 |
Bicyclo[3.1.1]heptan-3-ol |
0.23 |
152 |
C10H16O |
Terpenoid |
|
8.381 |
1-dodecene |
2.11 |
168 |
C12H24
|
Alkene |
|
9.034 |
Bicyclo[3.1.1]hept-3-en-2-one |
0.25 |
150 |
C10H14O |
Terpenoid |
|
11.339 |
1-tridecene |
3.64 |
182 |
C13H26
|
Alkene |
|
13.123 |
2,4-di-tert-butylphenol |
2.03 |
206 |
C14H22O |
Phenol |
|
14.003 |
1-octadecene |
4.05 |
252 |
C18H36
|
Alkene |
|
14.003 |
1-octadecene |
4.05 |
252 |
C18H36
|
Alkene |
|
18.543 |
1-heneicosanol |
1.86 |
312 |
C21H44O |
Fatty alcohol |
|
18.543 |
1-heneicosanol |
1.86 |
312 |
C21H44O |
Fatty alcohol |
|
18.543 |
1-heneicosanol |
1.86 |
312 |
C21H44O |
Fatty alcohol |
|
24.128 |
Octadecyl trifluoroacetate |
0.37 |
366 |
C20H37F3O2
|
Fatty Acid |
|
24.639 |
Prasterone |
0.14 |
288 |
C19H28O2
|
Sterol |
|
25.177 |
Andrographolide |
1.22 |
350 |
C20H30O5
|
Diterpenoid |
|
25.695 |
Ferruginol |
0.13 |
286 |
C20H30O |
Diterpenoid |
|
25.834 |
(1R,7S,E)-7-isopropyl-4,10-dimethylenecyclodec-5-enol |
0.63 |
220 |
C15H24O |
Sesquiterpenoid |
|
26.079 |
2-bornanol |
16.75 |
348 |
C16H20N4O5
|
Terpenoid |
|
26.435 |
11-oxoandrosterone |
1.53 |
376 |
C22H36O3Si |
Sterol |
|
26.933 |
Squalene |
0.20 |
410 |
C30H50
|
Triterpenoid |
|
28.535 |
6-pentylidene-4,5-secoandrostane-4,17.beta.-diol |
55.39 |
362 |
C24H42O2
|
Sterol |
|
28.827 |
17.beta.-hydroxy-6.alpha.-pentyl-4-nor-3,5-secoandrostan-3-oic acid, methyl ester |
1.78 |
378 |
C24H42O3
|
Fatty acid methyl ester |
|
28.827 |
17.beta.-hydroxy-6.alpha.-pentyl-4-nor-3,5-secoandrostan-3-oic acid, methyl ester |
1.78 |
378 |
C24H42O3
|
|
Table 4.
E. ingens ethyl acetate compounds with ideal drug candidate qualities.
Table 4.
E. ingens ethyl acetate compounds with ideal drug candidate qualities.
S/N |
Compounds |
MW $(g/mol) |
HD |
HA |
Mol$LogP |
Lipinski’s $Rule $(Violation) |
BBB |
CYP2D6 |
CYP3A4 |
TPSA |
|
1-dodecene |
168.32 |
0 |
0 |
5.25 |
Yes; 1 |
No |
No |
No |
0 |
|
1-heneicosanol |
312.6 |
1 |
1 |
5.62 |
Yes; 1 |
No |
No |
No |
20.23 |
|
1-octadecene |
252.5 |
0 |
0 |
6.77 |
Yes; 1 |
No |
No |
No |
0 |
|
1-tridecene |
182.35 |
0 |
0 |
5.52 |
Yes; 1 |
No |
No |
No |
0 |
|
Octadecyl trifluoroacetate |
366.5 |
0 |
5 |
5.47 |
Yes; 1 |
No |
No |
No |
26.30 |
|
Andrographolide |
350.4 |
3 |
5 |
1.98 |
Yes; 0 |
No |
No |
No |
86.99 |
|
Squalene |
410.7 |
0 |
0 |
3.59 |
Yes; 0 |
No |
No |
No |
0 |
Table 5.
Primers sequences.
Table 5.
Primers sequences.
Genes |
Primers |
AR |
Forward- GCTTTATCAGGGAGAACAGCCT$Reverse- TGCAGCTCTCTCGCAATCTG |
BCL2 |
Forward- GGCCTCAGGGAACAGAATGAT$Reverse- TCCTGTTGCTTTCGTTTCTTTC |
CDK1 |
Forward- GAACACCACTTGTCCCTCTAAGAT$Reverse- CTGCTTAGTTCAGAGAAAAGTGC |
Caspase-3 |
Forward- CAAAGAGGAAGCACCAGAACCC$Reverse- GGACTTGGGAAGCATAAGCGA |
P53 |
Forward- CTTCGAGATGTTCCGAGAGC$Reverse- GACCATGAAGGCAGGATGAG |
β –Actin |
Forward- GCCAACTTGTCCTTACCCAGA$Reverse- AGGAACAGAGACCTGACCCC |