Abstract Background Cervical cancer, one of the lethal cancers among women, is a challenging disease to treat. The current therapies often come with severe side effects and the risk of resistance development. Traditional herbal medicine, with its potential to offer effective and less toxic options, is a promising avenue. This study was undertaken to investigate the potential of Rhamnus prinoides (R. prinoides) root bark extracts in selectively inhibiting the proliferation of cervical cancer cells, using the HeLa cell line as an in vitro model. Methods R. prinoides plant extracts were first screened at a fixed concentration of 200 μg/ml to determine the active extract. The selective anti-proliferative activity of the active extract was evaluated in a concentration dilution assay using the (3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide) MTT assay on cancerous (HeLa) cells and non-cancerous (Vero) cells to determine the half-maximal inhibitory (IC[50]) and half-cytotoxic concentrations (CC[50]), respectively. Functional assays on cell morphology (by microscopy), cell migration (wound healing assay) and cell cycle (by flow cytometry) were also conducted. The active extract was analyzed using Gas Chromatography/Mass Spectrometry (GC/MS) to determine any compounds it contained. Following identification of possible gene targets by network pharmacology, the genes were validated by molecular docking and Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR). Results The ethyl acetate extract of R. prinoides (EARP), the most active extract, selectively inhibited the growth of cervical cancer cells, their migration and induced cell cycle arrest at the S phase. In silico analysis revealed that squalene, 3,3a,6,6-tetramethyl-4,5,5a,7,8,9-hexahydro-1H-cyclopenta[i]indene and Olean-12-en-3.beta.-ol, acetate showed acceptable drug-like characteristics and may be partly attributed to the bioactivity demonstrated and the deregulation of the mRNA expression of AKT1, NF-κB, p53, Bax, Bcl-2, and Er-b-B2. Conclusion This study, for the first time, demonstrates the anti-proliferation effects of EARP and forms a firm foundation for further drug development studies. Keywords: Cervical cancer, Cytotoxicity, Rhamnus prinoides, Network pharmacology, Anti-proliferative activity 1. Introduction Worldwide, the most frequent cause of fatalities is cancer, which has even been hailed recently as a “major public health challenge”. After lung, colorectal, and breast cancers, respectively, cervical cancer comes fourth in terms of prevalence. The bulk of new instances and fatalities from cervical cancer occurs in low-to-middle-income nations, which bear a disproportionate amount of the burden when compared to nations with higher income; actually, 84 % of all cervical cancer cases worldwide are found in Sub-Saharan Africa [[29]1]. Cervical cancer accounts for 12.4 % of all malignancies in Kenya, coming second in terms of prevalence, nevertheless, being the primary cause of cancer-related loss of life in the nation, accounting for 11.9 % of all cancer-related deaths, with breast cancer, which is first in terms of prevalence, accounting for 11.5 % of all cancer-related deaths. Factors such as inadequate health facilities, personnel and late diagnosis are responsible for the increased prevalence and high mortality rates [[30][1], [31][2], [32][3], [33][4]]. Cytoreductive surgery, radiation therapy, and chemotherapy are the most widely used treatment options for cervical cancer in Kenya. The high cost of treatment along with high instances of recurrence, chemo-resistance and severe side effects, are some of the problems associated with the chemotherapy [[34]5,[35]6]. Thus, searching for less harmful and more effective alternatives is both necessary and essential. Traditionally, use of therapeutic plants, dates back a long way in treating and mitigating illnesses, and they hold a vast potential as reservoirs of potent anti-cancer agents [[36]7,[37]8]. Rhamnus prinoides L'Herit (R. prinoides), which belongs to the genus Rhamnaceae, is a plant exotic to Kenya and native to many parts of Eastern and Central Africa. The tree is small, attaining an average height of just 4 m. It is known locally as “Mukarakinga” among the Kikuyu community and “Olkonyil” among the Maasai community in Kenya. Traditionally, a variety of plant parts of R. prinoides are used, these include, the leaves, stembark, and root bark, to address chest issues, tonsils, pneumonia, as an appetizer, to manage malaria, sexually transmitted diseases, and to cleanse blood, depending on the region [[38][9], [39][10], [40][11], [41][12]]. R. prinoides extracts were shown to have anti-microbial qualities [[42]13], anti-oxidant properties [[43]14,[44]15], anti-malarial effects [[45]16], and anti-inflammatory action [[46]15]. To the best of our knowledge, R. prinoides anti-proliferation and anti-cancer activities have not been assessed experimentally. We sought to explore the potential of anti-proliferation of crude-, hexane-, water- and ethyl acetate extracts of R. prinoides against human cervical cancer cells (HeLa cells) and non-cancerous Cercopithecus aethiops epithelial line from African green monkey kidney cells (Vero cells). The results demonstrated that the ethyl acetate extract fraction (EARP) exhibited the most effective selective action towards cervical cancer. We identified the compounds in the extracts that can be partly attributed to the recorded bioactivity using GC/MS analysis. We then conducted in silico studies which suggested that certain compounds had promising putative binding affinities with various genes essential for the development and progression of cervical cancer. We further validated these findings through in silico docking studies, cellular functional assays (ability to limit cell migration and induce cell cycle arrest at the S phase) and by gene expression analysis using RT-qPCR. Notably, we found that EARP down-regulated the mRNA expression of AKT1, Bcl-2 NF-κB and Er-b-B2 genes while up-regulating the m-RNA expression of tumor suppressor protein 53 and Bax. Therefore, we showed the potential of R. prinoides ethyl acetate extract as an anti-cancer agent and laid a solid foundation for further studies to isolate the compounds responsible for activity and validate their activity in higher organisms. 2. Materials and methods 2.1. Plant collection and extraction 2.1.1. Plant collection The root barks of R. prinoides were fetched from the region of Mount Kenya at the coordinates 0° 07′ 15.60″ N, 37° 20′ 7.20″ E. A taxonomist assisted in the plant's identification, collection and a voucher specimen was archived in the National Museums of Kenya's East Africa Herbarium (Mwitari/Gathiuru/RM/001/2022). The root barks were sent to the Kenya Medical Research Institute's Center for Traditional Medicine and Drug Research for processing and extraction. 2.1.2. Plant preparation After the root barks were cleaned to get rid of dust and other particles, they were chopped into smaller bits and allowed to air dry at room temperature then crushed into a powder with a laboratory mill [[47]17]. 2.1.3. Solvent extraction The cold solvent extraction method followed by solvent partitioning was used as described previously with minor adjustments [[48]18,[49]19]. 800g of the ground root bark of R. prinoides was weighed using a laboratory electric balance and macerated in dichloromethane: methanol mixture in a 1:1 ratio until submerged for 72 h with occasional shaking to get a crude extract. The solvent mixture was then decanted into a conical flask with a Whatman No.1 filter (Schleicher and Schuell Microscience Gmbh, Germany) and a vacuum rotary evaporator (Buchi, Switzerland) used to condense the extract at 55 °C. A portion of the crude extract was allowed to air dry. The remaining crude extract was re-soaked in 400 ml hexane to extract the non-polar compounds and concentrated in a rotary evaporator at 59 °C and allowed to dry at room temperature. Furthermore, the remaining extract was soaked in a combination of water and ethyl acetate in a 1:1 ratio in a separating funnel to extract the mid-polar and the polar compounds. The mixture was left to separate overnight forming two distinct layers. The two layers were then separated using density. The ethyl acetate partition was concentrated in a vacuum rotary evaporator at 67 °C while the water partition was lyophilized in a freeze drier (Modulyo Edwards high vacuum, Crawley England, Britain, Serial No. 2261). The extracts were left to air dry and kept at −20 °C until use. The percentage yield of the crude-, hexane-, water- and ethyl acetate extracts was calculated as; % yield = [(weight of dried extract) / (weight of dried plant sample)] x 100 With the use of an electric balance, 100 mg each plant extract fraction was measured and dissolved in 1 ml of laboratory-grade 100 % Dimethyl Sulphoxide (DMSO, FINAR, India) and kept at −20 °C until use as the stock solution. 2.2. Cell culture and anti-proliferation assay 2.2.1. Cell culture The American Type Culture Collection (ATCC) supplied the non-cancerous Vero cells (ATCC Cat# CCL-81, RRID: CVCL_0059) and cervical cancer HeLa-229 cells (ATCC Cat# CCL-2.1, RRID: CVCL_1276) that were used for the experiments. The MycoStrip™ Mycoplasma Detection Kit (Invitrogen, USA) was used to ascertain the purity of the cell lines. 10 % Fetal Bovine Serum (FBS, Sigma Aldrich, USA) was added to the Minimum Essential Medium (MEM, GIBCO, USA) along with 7.5 % Sodium Hydrogen Carbonate (LOBA Chemie, India), 1 % L-Glutamine (Sigma Aldrich, USA), 1 % Penicillin/Streptomycin (Sigma Aldrich, USA) and 1 % HEPES buffer (Gold Biotechnology, USA). They were maintained at 37 °C with 5 % CO[2] inside an incubator with humidity (Thermo Fisher Scientific, USA). 2.2.2. Viability and proliferation assay Proliferation and viability of HeLa and Vero cells was determined using the MTT assay as described previously [[50]20]. In brief, HeLa and Vero cells were seeded at a density of 10,000 cells/well in 96 well plates (Thermo Fisher Scientific, USA) and left overnight to grow at 37 °C with 5 % CO[2] in an incubator with humidity. After 24 h, the used media was dispensed with and replenished with 100 μl of fresh media. The plant extract concentrations were made by diluting the initial stock, reconstituted with DMSO, using growth media with the final DMSO concentration being 0.4 %. The first screening was conducted on water-, crude-, hexane- and ethyl acetate extracts and also doxorubicin hydrochloride as a positive control at a fixed concentration of 200 μg/ml for 48 h in HeLa cells. 0.4 % DMSO was used as a negative control. The most active extract was selected and the cells were treated to a range of concentrations of the active extract (starting from 240 to 0 μg/ml in HeLa and 800-0 μg/ml in Vero cells) for 48 h. The positive control utilized was doxorubicin hydrochloride at concentrations starting from 10 to 0.5 μg/ml for HeLa cells and 100–1.5625 μg/ml for Vero cells. The media with the extract was then discarded and the cells washed lightly using phosphate-buffered saline (PBS, Sigma Aldrich, USA). Thereafter, fresh media containing MTT dye (Solar Bio, China) at a concentration of 5 mg/ml in PBS was put in and the plates incubated for 4 h at 5 % CO[2] and 37 °C. After 4 h the formation of formazan crystals was confirmed through the change in color from yellow to purple. The media was dispensed with and 100 μl of DMSO put into the wells and the absorbance read at 570 nm and 720 nm was used reference wavelength to capture non-specific absorbance in a microtiter reader (Thermo Fisher Scientific, USA). The experiment was run in triplicate at least three times. The anti-proliferation effect of the extract was expressed as a percentage of the ratio of the treated cells to those treated with the negative control and the IC[50] (Inhibitory concentration that kills 50 % of cells), IC[25] and IC[12.5]values were estimated with the Graph pad Prism 8.0 software (La Jola, California, USA). % Viability = [Absorbance of Sample/Absorbance of Control] *100 2.3. Cell phenotypic analysis To delve into how the EARP extract affects the morphological phenotype of HeLa cells, for 48 h the cells were treated with EARP at IC[50], IC[25] and IC[12.5] concentrations and the positive control, doxorubicin hydrochloride, at its IC[50] concentration. The resulting phenotypic changes were recorded using an EVOS™ XL Core Imaging system (Thermo Fisher Scientific, USA). 2.4. Wound healing assay The effect of EARP on the limitation of HeLa cells migration was estimated according to the previously described method [[51]21]. HeLa cells were seeded in 24-well plates and left to attach overnight in a humidified incubator at 37 °C with 5 % CO[2]. An artificial wound was made by use of a sterile p200 tip once a confluent monolayer had been formed. After scraping the cells and washing away loose cells with PBS, new media containing the EARP extract at its IC[50], IC[25] and IC[12.5]concentrations was added to the wells. Using an ultra-fine tip marker pen (Sharpie, USA), a reference line was made perpendicular to the artificial wound. The scratch areas were captured at 0 h, 24 h and 48 h using an EVOS™ XL Core Imaging system (Thermo Fisher Scientific, USA) and analyzed using Image J software ([52]imagej.nih.gov). The percentage wound closure was calculated using the formula: %Wound Closure = [(A (0) - A (t))/A (0)] × 100 Whereby, A (0) is the area at time zero (0) and A (t) is the area after incubation time (t). 2.5. Cell cycle analysis In order to examine how EARP affects the cell cycle, HeLa cells at 100,000 cells/ml were propagated in T-25 culture flasks using MEM culture media and incubated overnight at 37 °C with 5 % CO[2]. The cells were then exposed to EARP at IC[50], IC[25] and negative control and incubated for 48 h at 37 °C with CO[2] at 5 %. Thereafter, trypsin was used to harvest the cells after they had been incubated and rinsed twice with ice-cold PBS. Then cells were suspended in 70 % ethanol for 24 h. After discarding the ethanol, the cells were centrifuged for 2 min at 2000 rcf and re-suspended in PBS with 0.25 % Triton X-100 for 15 min on ice. After centrifugation of the cells for 2 min at 2000 rcf, the supernatant was discarded. The cells were kept at 24 °C for 30 min in a dark place after being re-suspended in PBS with 10 μg/ml of RNase A and 25 μg/ml of propidium iodide. A flow cytometer (BD Canto II, BD Sciences) was used for recording and analysis done using FlowJo software (version 10.7.0), to determine the distribution of the cell cycle at G1, S and G2/M phases [[53]21]. 2.6. Phytochemical screening of EARP The color based phytochemical screening tests of EARP were done according to standard methods [[54]14,[55]22]. 2.6.1. Test for alkaloids Half a gram of plant extract was weighed and stirred in 2 ml of 1 % aqueous hydrochloric acid and heated in a boiling water bath for 10 min. The mixture was filtered while hot and treated with Dragendorff's reagent. Turbidity or precipitation was used to indicate the presence of alkaloids. 2.6.2. Test for terpenoids Half a gram of the extract was defatted with hexane. The residue was then extracted in dichloromethane and the solution dehydrated with magnesium sulfate anhydride. The mixture was treated with 0.5 ml acetic anhydride followed by addition of 2 drops of concentrated sulphuric acid. A gradual appearance of green blue color was used to indicate presence of sterols while color change from pink to purple indicated the presence of triterpenes. 2.6.3. Test for saponins Half a gram of the plant extract was dissolved in 5 ml of distilled water and shaken for at least 5 min. Persistent frothing for at least half an hour indicated the presence of saponins. The quantity of froth was used to estimate quantity. 2.6.4. Test for flavonoids Two hundred milligrams of the extract was dissolved in 4 ml of 50 % methanol. The solution was warmed and then metal magnesium added. Five drops of concentrated sulphuric acid was added. Development of a red color indicated the presence of flavonoids while orange color showed presence of flavones. 2.6.5. Test for tannins The ferric chloride test was used. Half a gram of the extract was dissolved in 2 ml of distilled water and filtered. Two drops of ferric chloride was added to the filtrate. Development of a blue-black precipitate indicated the presence of tannins. 2.6.6. Test for phenols (ferric chloride test) 2 ml of distilled water was added to 1 mg of plant extract followed by a few drops of 10 % aqueous ferric chloride solution. Formation of blue or green color indicated the presence of phenols. 2.6.7. Test for glycosides 2 mg of plant extract was dissolved in 1 ml distilled water and then aqueous sodium hydroxide was added. Formation of a yellow color indicated the presence of glycosides. 2.7. Gas Chromatography/Mass Spectrometry (GC/MS) of EARP The compounds contained in EARP were identified through GC/MS QP-2010SE instrument (Shimadzu, Kyoto, Japan) with a BPX5 capillary column with dimensions 30m *0.25 mm * 0.25 μm film in width with a low polarity. In order to reach the isothermal temperature of 280 °C and maintain it for 15 min and 30 s, the oven was configured to start at 50 °C and stay that way for 1 min before progressively raising the temperature by 10 °C per minute. The injector's temperature was retained at 200 °C and its flow rate at 1.08 mL per minute. Helium was used as the carrier gas. As AS30000 auto-sampler connected to a GC in split mode with a split ratio of 10:1 automatically injected 1 μl of the diluted sample in solvent after a 4 min delay. The source of the ions and interface were adjusted to 250 °C and 200 °C, respectively. In full scan mode, the EI mass spectra above the range of m/z 35–550 and at 70 eV were then fetched. The chemical components in the extract were identified using the mass spectrum database of the National Institute of Standards and Technology (NIST) [[56]23]. 2.8. In silico studies 2.8.1. Drug screening PubChem ([57]https://pubchem.ncbi.nlm.nih.gov/) was utilized to generate canonical SMILES of chemical compounds identified through GC/MS. Their ADMET (Absorption, Distribution, Metabolism, Excretion and Toxicity) attributes were gotten using the Swiss ADME online database ([58]http://www.swissadme.ch/index.php) and the pkCSM tool ([59]http://structure.bioc.cam.ac.uk/pkcsm). The parameters that were described in these databases were used to project the drug-likeness of the compounds on the basis of the following criteria: Lipinski's rule of five (molecular weight in Daltons <500, log P o/W, hydrogen bond acceptors ≤10 and hydrogen bond donors ≤5), oral bioavailability; interaction with cytochromes; crossing the blood-brain barrier (BBB) as well as the topological surface area (TPSA) [[60]24,[61]25]. 2.8.2. Identification of therapeutic targets Genes associated with cervical cancer were assembled using Gene Cards ([62]https://www.genecards.org/), DISGeNET ([63]https://www.disgenet.org/), OMIM ([64]https://www.omim.org/) and NCBI ([65]https://www.ncbi.nlm.nih.gov/gene) databases with the phrase “cervical cancer” being the key word. All the results were merged and the duplicates were removed. Targets from EARP were gotten from Swiss Target Prediction ([66]http://www.swisstargetprediction.ch/), Similarity Ensemble Approach (SEA) ([67]https://sea.bkslab.org/) and Super-PRED ([68]https://prediction.charite.de/subpages/target_prediction.php) databases. The gene ids were acquired from the Universal Protein Resource (Uniprot), ([69]https://www.uniprot.org/). The outputs from the databases was put together and duplicates were deleted. Common genes between the targets and the cancer genes were gotten from an online Venn diagram generator ([70]https://bioinformatics.psb.ugent.be/webtools/Venn/) [[71]26]. 2.8.3. Generation of the protein-protein interaction (PPI) network The similar genes were loaded into the STRING 12.0 database ([72]https://string-db.org/), where the minimum threshold was set at 0.4. Cytoscape software (version 3.10.2) was utilized to analyze the topology based on the degree of centrality with the Cytohubba plug-in being used to generate the top 30 genes based on five centralities, MCC (Maximal Clique Centrality), ECC (Eccentricity), BN(Betweenness), DG (Degree) and CN (Closeness). 2.8.4. Gene ontology (GO) and Kyoto Encyclopedia of genes and Genomes (KEGG) enrichment analysis The online enrichment tool Shiny GO version 0.80 ([73]http://bioinformatics.sdstate.edu/go/) was utilized to conduct these studies [[74]27]. The parameters set were; species was “Human”, false discovery rate (FDR) cut-off was 0.05 and the number of pathways displayed were up to 20 [[75]22]. 2.9. Molecular docking 3D Structured Data File (SDF) format of the compounds selected after ADMET parameters were analyzed were gotten from PubChem database. The RCSB protein database ([76]https://www.rcsb.org/) was used to get the structures of the proteins of the top 30 genes. The filters applied were; the species was Human, the experimental method was X-ray Diffraction, the resolution was between 0.5 and 3.5 Å and the structure determination methodology was experimental. Using Discovery Studio 2021, the targeted proteins were curated whereby the Gasteiger charges were added, water molecules were removed, all co-crystallized ligands removed and polar hydrogen were added. The 3D SDF compounds (ligands) were edited and transformed into the “pdbqt” format with the aid of the inbuilt Open Babel software in Pyrx program (([77]https://sourceforge.net/projects/pyrx/). The embedded Autodock Vina software in Pyrx program was then used to dock the prepared ligands to the prepared proteins. The size of the protein was used to centralize the three-dimensional grid box. Discovery studio 2021 ([78]https://discover.3ds.com/discovery-studio-visualizer-download) was used to generate the 3D protein-ligand complexes with the lowest binding energies and their 2D interaction diagrams [[79]28]. 2.10. Survival analysis Some of the genes with the lowest binding affinities were assessed with the aid of the GEPIA database ([80]http://gepia.cancer-pku.cn) to ascertain the correlation between their expression and overall patient survival and overall disease free survival [[81]29]. 2.11. RNA isolation and quantification using RT-qPCR To validate the docked proteins as well as assess the effect of the EARP extract on key oncogenic genes, RT-qPCR was used. HeLa cells were seeded in T-75 culture flasks and upon attaining a confluence of >70 %, they were treated with the EARP extract at IC[50] concentration for 48 h. After treatment, the flasks were rinsed with PBS thrice and RNA was obtained using the Pure Link™ RNA Mini Kit (Thermo Fisher Scientific, USA) extraction kit. A NanoDrop, ND-2000 spectrophotometer (Thermo Scientific, USA), was employed in the quantification of the extracted RNA. The RNA was preserved at −80 °C until use. 1 μg of RNA was converted to cDNA using the Firescript® RT cDNA synthesis kit (Solis Biodyne, Estonia). The cDNA was stored at −20 °C until use. The genes used were purchased from Macrogen (Macrogen, South Korea), ([82]Table 1). The HOT Firepol® Evagreen® qPCR Mix plus (ROX) (Solis Biodyne, Estonia) was utilized for quantitative RT-qPCR analysis using the Quant Studio™ 5 RT-qPCR system (Applied Biosystems, USA). A blank containing the master mix and the primers without the cDNA was used. Analysis of the melt curve verified that there was no non-specific binding. The targeted genes' mRNA levels were quantified using the 2 ^−ΔΔ Ct method. GAPDH and β-actin controls were the internal references