Next Article in Journal
Current Methods for Reliable Identification of Species in the Acinetobacter calcoaceticusAcinetobacter baumannii Complex
Previous Article in Journal
The Susceptibility Profiles of Human Peripheral Blood Cells to Staphylococcus aureus Cytotoxins
Previous Article in Special Issue
Effects of Natural Ingredient Xanthohumol on the Intestinal Microbiota, Metabolic Profiles and Disease Resistance to Streptococcus agalactiae in Tilapia Oreochromis niloticus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

From Hue to Health: Exploring the Therapeutic Potential of Plant-Pigment-Enriched Extracts

1
Laboratory of Biotechnology, Department of Microbiology, Agricultural Research Centre (ARC), Animal Health Research Institute (AHRI), Zagazig 44516, Egypt
2
Department of Botany and Microbiology, Faculty of Science, Benha University, Benha 13518, Egypt
3
Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
4
Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Microorganisms 2025, 13(8), 1818; https://doi.org/10.3390/microorganisms13081818
Submission received: 23 June 2025 / Revised: 24 July 2025 / Accepted: 29 July 2025 / Published: 4 August 2025
(This article belongs to the Special Issue Advanced Research on Antimicrobial Activity of Natural Products)

Abstract

The escalating global challenges of antimicrobial resistance (AMR) and cancer necessitate innovative therapeutic solutions from natural sources. This study investigated the multifaceted therapeutic potential of pigment-enriched plant extracts. We screened diverse plant extracts for antimicrobial and antibiofilm activity against multidrug-resistant bacteria and fungi. Hibiscus sabdariffa emerged as the most promising, demonstrating potent broad-spectrum antimicrobial and significant antibiofilm activity. Sub-inhibitory concentrations of H. sabdariffa robustly downregulated essential bacterial virulence genes and suppressed aflatoxin gene expression. Comprehensive chemical profiling via HPLC identified major anthocyanin glucosides, while GC-MS revealed diverse non-pigment bioactive compounds, including fatty acids and alcohols. Molecular docking suggested favorable interactions of key identified compounds (Cyanidin-3-O-glucoside and 1-Deoxy-d-arabitol) with E. coli outer membrane protein A (OmpA), indicating potential antiadhesive and antimicrobial mechanisms. Furthermore, H. sabdariffa exhibited selective cytotoxicity against MCF-7 breast cancer cells. These findings establish H. sabdariffa pigment-enriched extract as a highly promising, multi-functional source of novel therapeutics, highlighting its potential for simultaneously addressing drug resistance and cancer challenges through an integrated chemical, biological, and computational approach.

1. Introduction

The global health landscape is at a critical juncture, facing an escalating demand for natural alternatives across industries ranging from food to pharmaceuticals. This shift is largely driven by growing concerns over adverse health effects linked to synthetic compounds, including allergies, neurological issues, and toxicity [1,2]. Beyond their role as safe colorants, natural compounds, particularly those from plants, offer a vast, untapped arsenal of beneficial pharmacological activities—such as anticancer, anti-inflammatory, and antidiabetic effects—holding immense promise for human health [3].
Compounding this, humanity confronts an existential threat stemming from the alarming rise of antibiotic resistance, dramatically worsening the global burden of infectious diseases. The relentless selective pressure from widespread and often inappropriate antibiotic use has fueled the rapid evolution and dissemination of multidrug-resistant (MDR) pathogens. Foremost among these are the notorious “ESKAPE” organisms (Enterococcus spp., Staphylococcus aureus, Klebsiella spp., Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.), which pose a critical public health crisis by resisting standard treatments, prolonging infections, and rendering once-effective therapies obsolete [4]. This dire situation is further compounded by the critically dwindling pipeline for new antibiotics, underscoring the urgent global priority of discovering novel, effective antibacterial agents [5,6].
Amidst this urgent quest, plants emerge as an extraordinarily rich and promising reservoir of new therapeutic agents. Their unparalleled chemical diversity, natural abundance, affordability, and generally lower incidence of severe side effects compared to synthetic drugs position them as an attractive and sustainable avenue for innovative therapies [7]. Within the vast array of bioactive plant compounds, pigments constitute a particularly significant class with demonstrated therapeutic potential. Distinct pigment classes, including carotenoids, chlorophylls, and anthocyanins, not only fulfill vital roles in plant biology but also exhibit diverse biological activities pertinent to human health [8].
Anthocyanins, a prominent group of water-soluble polyphenolic pigments, captivate with the vibrant pink, red, purple, and blue hues they impart to countless fruits, vegetables, and flowers. As key flavonoid constituents, these compounds are not only essential for plant propagation and ecophysiology but also serve as crucial defenders against environmental stressors [9]. Critically, numerous studies have underscored the broad-spectrum antimicrobial activity of anthocyanins, with mechanisms spanning membrane disruption, enzyme inhibition, and metabolic interference [10,11]. However, the therapeutic efficacy of complex plant extracts, which contain various bioactive components, demands a deeper, more integrated understanding of their active constituents and mechanisms to unlock their full potential.
While the direct antimicrobial activity of many plant pigments and extracts is well-documented, their precise impact on virulence factors—pivotal determinants of infection establishment and severity—remains a critical and largely underexplored frontier. This study aimed to address this gap, providing a deeper understanding of the therapeutic potential of pigment-enriched plant extracts by investigating their effects beyond direct microbial killing. We specifically examined the ability of sub-inhibitory extract concentrations to modulate the expression of key microbial virulence and aflatoxin genes, representing a novel angle in their antimicrobial assessment. Acknowledging the inherent complexity of these extracts, this study additionally assessed their direct antimicrobial and cytotoxic potential against cancer cells. To achieve a comprehensive understanding of their therapeutic potential and to explore the underlying molecular interactions, we employed a multifaceted approach. This included an initial screening for broad-spectrum antimicrobial and antibiofilm activities. We then performed comprehensive chemical profiling, utilizing HPLC for precise pigment identification and quantification, and GC-MS for detailed identification of non-pigment secondary metabolites. Subsequently, we conducted gene expression analyses to quantify the downregulation of critical microbial virulence and aflatoxin genes. Furthermore, molecular docking simulations were meticulously performed to explore the potential direct molecular interactions of key identified compounds—encompassing both characteristic pigments and other influential constituents—with relevant bacterial targets, thereby suggesting plausible mechanistic bases for their observed bioactivities. Finally, we rigorously evaluated the selective cytotoxicity of these extracts against MCF-7 breast cancer cells. This integrated investigation aimed to illuminate the profound therapeutic potential of pigment-enriched extracts as novel, multi-functional agents uniquely capable of simultaneously combating the pressing global challenges of drug resistance and cancer.

2. Materials and Methods

2.1. Extraction of Plant Pigments

2.1.1. Chlorophyll Extraction

Chlorophyll pigments were extracted from fresh leaves of Moringa oleifera, Nerium oleander, Zingiber officinale, Ocimum basilicum, and Citrus limon following the method described by Porra [12] and Alraof et al. [13]. The concentrations of chlorophyll a, chlorophyll b, and total chlorophyll were calculated using the following equations:
Total Chlorophyll (mg/L) = 20.2(A645) + 8.02(A663)
Chlorophyll a (mg/L) = 12.7(A663) – 2.69(A645)
Chlorophyll b (mg/L) = 22.9(A645) – 4.68(A663)
where A645 and A663 represent the absorbance at 645 nm and 663 nm, respectively.

2.1.2. Carotenoid Extraction

Carotenoids were extracted from Curcuma longa, Citrus reticulata, Spinacia oleracea, Daucus carota, Lycopersicon esculentum, and Capsicum annuum following the method described by Jeyanthi Rebecca et al. [14]. The extracts were collected, and the absorbance was measured at 450 nm using a colorimeter. The carotenoid content was then calculated and expressed as mg/100 g fresh weight.

2.1.3. Anthocyanin Extraction

Anthocyanin pigments were extracted from Beta vulgaris roots, Hibiscus sabdariffa petals, Solanum melongena peels, Brassica oleracea (red cabbage), and Prunus domestica following the method described by Shehata et al. [15]. Two hundred grams of each plant material was homogenized into a smooth puree using a standard kitchen blender. The puree was extracted using a solution of 0.1 M HCl in 85:15 (v/v) ethanol. The resulting slurry was stirred for 1 h with a mechanical stirrer. The mixture was then filtered through a Buchner funnel using a vacuum filtration system (MilliporeSigma, Burlington, MA, USA). The filtrate was collected and stored at 4 °C. The total anthocyanin content was determined spectrophotometrically and expressed as mg cyanidin 3-glucoside equivalents per 100 g of plant material using a standard curve of cyanidin 3-glucoside, following the method reported by Lohachoompol et al. [16].

2.2. Assessment of Antimicrobial Activity

2.2.1. Collection of Microbial Pathogens

The following multidrug-resistant bacterial strains were obtained from the Animal Health Research Institute (AHRI), Serology Department, Dokki Branch, and Microbiology Department, Zagazig Branch, Egypt: Klebsiella pneumoniae, Klebsiella oxytoca, Pseudomonas fluorescens, Pseudomonas aeruginosa, Morganella morganii, Escherichia coli, Salmonella typhimurium, Pasteurella haemolytica, Shigella flexneri, and Staphylococcus aureus. The fungal strains Candida albicans, Aspergillus niger, and Aspergillus flavus were also obtained from the same sources.
Upon receipt, all the bacterial isolates were re-cultivated on nutrient agar (NA) plates and incubated at 37 °C for 24 h to ensure viability and purity. Similarly, the fungal strains were re-cultivated on potato dextrose agar (PDA) plates and incubated at 25 °C for 72 h. Prior to experimental testing, all the isolates were activated by sub-culturing twice consecutively in appropriate liquid broth media (nutrient broth for bacteria, potato dextrose broth for fungi) and incubated under their respective optimal conditions (37 °C for 24 h for bacteria; 25 °C for 48–72 h for fungi) to ensure active growth and consistent physiological states. Stock cultures were maintained at −80 °C in cryoprotective media for long-term storage.

2.2.2. Agar Diffusion Assay

The agar well diffusion method was used for initial screening of the antimicrobial properties of the pigment extracts against the selected bacterial and fungal strains [17]. Twenty-four-hour-old bacterial broth cultures and forty-eight-hour-old fungal cultures were adjusted to a 0.5 McFarland standard, resulting in an inoculum of approximately 1.5 × 108 CFU/mL. The inoculum was uniformly spread onto Mueller–Hinton agar (Oxoid Ltd., Basingstoke, UK) for bacteria and Sabouraud dextrose agar (Hardy Diagnostics, Santa Maria, CA, USA) for fungi using sterile swabs.
Immediately after inoculation, 6 mm diameter wells were aseptically punched into the agar using a sterile cork borer. Each well was then loaded with 100 µL of the respective pigment extract. For controls, negative control wells received 100 µL of the extraction solvent (1% DMSO), while positive control wells received 100 µL of a standard antimicrobial agent (100 µL chloramphenicol for bacteria and nystatin for fungi). The plates were then incubated at 37 °C for 24 h (bacteria) or 28 °C for 48 h (fungi). The diameters of the inhibition zones were measured in millimeters, and the results were recorded.

2.2.3. Minimum Inhibitory Concentration (MIC) and Minimum Microbiocidal Concentration (MMC) Determination

The minimum inhibitory concentration (MIC) and minimum microbiocidal concentration (MMC) of the pigment extracts were determined using the broth microdilution method [18,19]. Serial two-fold dilutions of the extracts were prepared in Luria–Bertani (LB) broth (Acumedia, MI, USA) in 96-well plates (TPP, Switzerland). Each well was inoculated with 1 × 105 CFU of the respective bacterial strain and incubated at 37 °C for 24 h. The MIC was defined as the lowest extract concentration that visibly inhibited bacterial growth. The MMC was determined by sub-culturing aliquots from the wells showing no visible growth onto agar plates and incubating them. The MMC was defined as the lowest extract concentration that resulted in no viable microbial growth after sub-culturing. The tested extract concentrations ranged from 0.062 to 64 μg/mL.

2.3. Biofilm Formation and Antibiofilm Activity Assay

A standard microtiter plate method was employed [20,21] for the detection and assessment of the biofilm formation and the antibiofilm activity of the tested pigments. Initially, a fresh overnight culture of the target microorganism, grown in an appropriate liquid medium such as tryptic soy broth (TSB) or potato dextrose broth (PDB), was prepared for biofilm formation. This culture was then diluted to a standardized optical density (0.5 McFarland standard) to ensure consistent inoculation.
For the biofilm formation assay, sterile 96-well flat-bottom microtiter plates were utilized. Each well was filled with the appropriate growth medium. For the antibiofilm assay, the tested pigment extracts were prepared at various sub-inhibitory concentrations and added directly to the wells alongside the microbial inoculum. Control wells were included, comprising an untreated microbial culture (positive control for biofilm formation), wells with only medium and pigment extract (to check for pigment interference with optical density readings), and sterile medium only (negative control for biofilm). The inoculated plates were then incubated under static conditions at a specific temperature (37 °C for bacteria, 28 °C for fungi) for a predetermined period, commonly 24 to 48 h, to allow for robust biofilm development.
Following the incubation period, non-adherent planktonic cells (freely suspended, individual microbial cells in liquid culture) were carefully removed by gently aspirating the liquid from each well. The wells were then washed meticulously, typically three times with sterile phosphate-buffered saline (PBS), to ensure that only the adherent biofilm remained. After washing, the plates were allowed to air dry completely.
To quantify the adherent biofilm, a staining solution, such as 0.1% (w/v) Crystal Violet solution, was added to each well and allowed to stain the biomass for a specific duration, usually 15 to 20 min. After staining, the wells were again thoroughly washed multiple times with sterile distilled water to remove any unbound or excess stain. The plates were then air-dried once more.
Finally, the Crystal Violet stain bound to the biofilm was solubilized by adding a known volume of a solvent, such as 95% ethanol or 33% (v/v) glacial acetic acid, to each well. The plates were gently agitated for a few minutes to ensure complete destaining. The optical density of the solubilized stain in each well was then measured using a microplate reader at an appropriate wavelength (570 nm). A reduction in the optical density of the treated wells compared to the untreated biofilm control wells indicated the antibiofilm efficacy of the tested pigments.
Biofilm formation assays were performed in triplicate for each isolate, and the entire experiment was repeated independently three times. For both the tested isolates and the negative controls, the average optical density (OD) values and standard deviations (SDs) were determined.
The cut-off value for biofilm formation (ODc) was computed as follows: ODc = average OD of negative control + (3 × SD of negative control)
The isolates were then categorized based on their OD values relative to the ODc as:
  • Non-biofilm producer: OD ≤ ODc
  • Weak biofilm producer: ODc < OD ≤ (2 × ODc)
  • Moderate biofilm producer: (2 × ODc) < OD ≤ (4 × ODc)
  • Strong biofilm producer: OD > (4 × ODc)

2.4. Quantitative Assessment of Pigment Extract Effects on Virulence Gene Expression Using qRT-PCR

The effect of sub-inhibitory concentrations of the pigment extracts on microbial virulence gene expression was analyzed using quantitative reverse transcriptase real-time PCR (qRT-PCR) in the Biotechnology Unit, Animal Health Research Institute, Zagazig Branch, Egypt. Specific primers for the target genes were designed using Primer3 and FastPCR software (version number 6.9.21, http://primerdigital.com/fastpcr.html, accessed on 28 July 2025), optimized for specificity and sensitivity using touchdown PCR, and validated experimentally prior to use. The qRT-PCR reactions (25 μL) contained 12.5 μL of 2x QuantiTect SYBR Green PCR Master Mix (Qiagen, Germany, GmbH), 0.25 μL of RevertAid Reverse Transcriptase (200 U/μL) (Thermo Fisher Scientific, Waltham, MA, USA), 0.5 μL of each primer (20 pmol concentration), 8.25 μL of water, and 5 μL of RNA template. Amplification was performed using a StepOne real-time PCR system (Applied Biosystems, Foster City, CA, USA) with the specific cycling conditions listed in Table 1. The amplification curves and Ct values were determined using the StepOne software version 2.3. The relative gene expression was calculated using the ΔΔCt method [22,23], comparing the Ct values of each sample to the positive control group.

2.5. HPLC Analysis

High-performance liquid chromatography (HPLC) analysis of the Hibiscus sabdariffa pigment extract was performed using a Shimadzu LC-1620A HPLC system (Kyoto, Japan). This system was equipped with a binary pump and a UV-Vis detector (SPD-20A), allowing for specific detection at a fixed wavelength of 521 nm, characteristic of anthocyanins. For the unequivocal identification of specific anthocyanin glucosides, their retention times and UV-Vis absorption spectra were rigorously compared against those of commercially available authentic standards. The standards used were as follows: Delphinidin-3-O-glucoside (Sigma-Aldrich, St. Louis, MO, USA; Catalog No. 6906-38-3), Pelargonidin-3-O-glucoside (Sigma-Aldrich, St. Louis, MO, USA; Catalog No. 18466-51-8), and Cyanidin-3-O-glucoside (Sigma-Aldrich, St. Louis, MO, USA; Catalog No. 7084-24-4). These authentic standards were prepared in HPLC-grade methanol (or an appropriate solvent) at known concentrations and run under identical chromatographic conditions to the sample extracts to ensure accurate comparison. Chromatographic separation was achieved on a Phenomenex Luna C18 column (125 mm × 4.60 mm, 5 µm particle size). Data acquisition and analysis were handled by Shimadzu LabSolutions software (version 5.110). The overall methodology adhered to principles recommended by Shehata et al. [15] for anthocyanin analysis. Isocratic elution utilized a mobile phase composed of 27.5% (v/v) aqueous 0.01% (v/v) formic acid (Component A), 22.5% (v/v) HPLC-grade methanol (Component B), and 50% (v/v) HPLC-grade acetonitrile (Component C). Prior to use, the mobile phase was filtered through a 0.22 µm membrane filter and degassed by sonication for 15 min. Samples underwent similar preparation, being filtered through a 0.22 µm syringe filter and sonicated before injection. The flow rate was maintained at 1.0 mL/min. The column temperature remained at ambient temperature, and the injection volume was 20 µL. Analytes were detected at the aforementioned fixed wavelength of 521 nm.

2.6. Gas Chromatography–Mass Spectrometry (GC-MS) Analysis

The chemical composition of the plant extract was determined using a Thermo Scientific Trace 1310 GC coupled to an ISQ mass spectrometer (Waltham, MA, USA). A TG-5MS capillary column (30 m × 0.25 mm × 0.25 μm film thickness) was used for separation. The oven temperature program was as follows: initial temperature 50 °C, increased at a rate of 5 °C/min to 230 °C and held for 2 min, then increased at 30 °C/min to a final temperature of 290 °C and held for 2 min. The injector and MS transfer line temperatures were maintained at 250 °C and 260 °C, respectively. Helium was used as the carrier gas at a constant flow rate of 1 mL/min. A solvent delay of 3 min was implemented. One microliter of the diluted sample was injected automatically in split mode using an AS 1300 autosampler (Thermo Fisher Scientific S.p.A., Milan, Italy) coupled with the GC. The electron ionization (EI) mass spectra were acquired at 70 eV ionization voltage over a mass-to-charge (m/z) range of 40–1000 in full scan mode. The ion source temperature was set at 200 °C. Component identification was performed by comparing their retention times and mass spectra with those in the WILEY 09 and NIST 11 mass spectral databases.

2.7. Molecular Docking

The three-dimensional crystal structure of the protein (PDB ID: 1QJP) was downloaded from the RCSB protein data bank (https://www.rcsb.org/, accessed on 28 July 2025). The retrieved files were opened in the Discovery studio visualizer [31], and it was found that protein 1QJP was missing the amino acid residues. The missing residues were added using the SwissModel web tool (https://swissmodel.expasy.org/, accessed on 28 July 2025) using the original PDB file as a template. Furthermore, the protein files were further prepared by removing the water molecules and other additional ligands occupying the binding site of the protein.
Molecular docking of ligands against proteins is recognized as a crucial strategy for accelerating the development and delivery of novel drug candidates, significantly reducing both research times and financial costs. In this study, molecular docking studies were performed using AutoDock Vina (https://github.com/ccsb-scripps/AutoDock-Vina, accessed on 28 July 2025) [32] on the virtual screening tool PyRx 0.8 software [33]. The PDB files of the protein were loaded on the 3D screen of the PyRx tool and were converted into PDBQT format. An open docking was performed. For protein 1QJP, the Vina search space was centered at X = 24.13, Y = 19.32, and Z = 35.57 with XYZ dimensions of 54.07 Å × 45.06 Å × 34.40 Å to cover the entire protein.

2.8. Cytotoxicity Assessment

2.8.1. Cell Culture

The assay was performed in accordance with procedures approved by the Research Ethics Committee at Institutional Review Board, King Abdullah Bin Abdulaziz University Hospital, Riyadh, Saudi Arabia (IRB number: 25-0036). The non-tumorigenic MCF-10A cell line and the breast cancer MCF-7 cell line were obtained from the Natural and Health Sciences Research Centre in Riyadh, Saudi Arabia. Cells were cultured in DMEM supplemented with 10% FBS, 100 U/mL penicillin, and 100 μg/mL streptomycin and maintained in a humidified incubator at 37 °C with 5% CO2. Cells were trypsinized and passaged according to standard procedures, maintaining approximately 100,000–150,000 cells per passage at 80–90% confluence.

2.8.2. Anticancer Assay

The MTT assay was used to evaluate the anticancer activity of the pigment extracts. MCF-7 (breast cancer) and MCF-10A (non-tumorigenic breast) cells were seeded at a density of 5000 cells per well in 96-well plates and allowed to adhere for 24 h. Cells were then treated with serial dilutions of the pigment extracts for 72 h. Following treatment, the medium was replaced with fresh medium, and 20 μL of 20 mM MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) solution in PBS was added to each well. The plates were incubated for 3 h at 37 °C in a 5% CO2 atmosphere. After incubation, the MTT solution was carefully removed, and 100 μL of DMSO (dimethyl sulfoxide) was added to each well to solubilize the resulting formazan crystals. The absorbance was measured at 560 nm, and the results were expressed as the cell viability relative to untreated control cells.

2.8.3. Selectivity Index (SI) Calculation

The selectivity index (SI) was calculated to assess the pigment’s selective toxicity toward cancer cells compared to normal cells. The SI was determined using the following formula:
SI = IC50 (MCF-10A)/IC50 (MCF-7)
where IC50 represents the concentration of the pigment extract that inhibits 50% of cell growth.

2.9. Statistical Analysis

Data were compiled and managed using Microsoft Excel (Microsoft Corporation). Prior to statistical analysis, the normality of the data distribution was assessed using the Shapiro–Wilk test, and the homogeneity of variance was determined using Levene’s test [34]. For comparisons involving more than two groups, the effect of different pigment extracts on the inhibition zones of the various microbial isolates was analyzed using the Kruskal–Wallis H test, with Dunn’s test used for post hoc comparisons when significant differences were observed. Gene transcription data, as well as the antibiofilm efficacy of hibiscus and prunes against various pathogens, were evaluated using one-way analysis of variance (ANOVA), followed by Tukey’s honestly significant difference (HSD) post hoc test for pairwise comparisons. The results are presented as the mean ± standard error of the mean (SEM), and statistical significance was defined as a p-value less than 0.05 (p < 0.05). All the statistical analyses were primarily performed using IBM SPSS Statistics version 26 (IBM Corp, Armonk, NY, USA) and SAS software version 9.4 TS1M9 (SAS Institute, Cary, NC, USA). Figures and graphical representations were generated using GraphPad Prism version 8 (GraphPad Software, San Diego, CA, USA) and R-software version 4.4.3 (https://www.r-project.org/, accessed on 28 July 2025).

3. Results and Discussion

3.1. Pigment Content of Different Plants

Table S1 presents the pigment content of various plants. The highest total chlorophyll content (28.37 μg/g) was measured in Moringa oleifera, closely followed by Nerium oleander (28.31 μg/g). Ocimum basilicum exhibited the lowest total chlorophyll content (11.13 μg/g). Curcuma longa had the highest carotenoid content (180.8 µg/100 g), while Daucus carota had the lowest (120.5 µg/100 g). Among the tested plants, Beta vulgaris showed the highest anthocyanin content (30.761 mg/100 g), followed by Brassica oleracea (14.913 mg/100 g). Prunes contained the lowest amount of anthocyanin (1.14 mg/100 g). The variations in the pigment content across the tested plant species reflect diverse physiological adaptations and potential health benefits. Moringa oleifera’s and Nerium oleander’s high chlorophyll concentrations suggest efficient photosynthetic capabilities, while Ocimum basilicum’s lower levels may indicate different light adaptation strategies. Curcuma longa’s exceptional carotenoid content, far exceeding that of Daucus carota, highlights its potent antioxidant potential and vibrant coloration. Similarly, Beta vulgaris’s and Brassica oleracea’s substantial anthocyanin levels, in stark contrast to the low content in prunes, underscore their roles as rich sources of these antioxidant compounds. These findings emphasize the importance of considering the pigment profiles when evaluating the nutritional and functional properties of plants.

3.2. Antimicrobial Efficacy

The chlorophyll pigment extracts exhibited significant antimicrobial activity against the tested pathogenic organisms (p < 0.0001). While both the Moringa oleifera and Citrus limon extracts showed antimicrobial effects, no statistically significant differences were observed between them across all the tested organisms. Similarly, the Nerium oleander and Ocimum basilicum extracts displayed antimicrobial activity, although O. basilicum showed significantly greater inhibition against Aspergillus flavus and Aspergillus niger compared to N. oleander (p < 0.05), with increases in the inhibition zone diameters of 17.77% and 26.87%, respectively. Olive extract demonstrated no inhibitory activity against Pseudomonas fluorescens and Candida albicans. The inhibition zones for the remaining pathogenic organisms ranged from 1.3 cm to 2.00 cm (Table S2).
The carotenoid pigment extracts tested in this study demonstrated significant antimicrobial activity against most pathogenic organisms (p < 0.001). However, Aspergillus niger was not significantly affected (p = 0.0649). Furthermore, the carotenoid extracts did not exhibit inhibitory activity against Klebsiella oxytoca, Pseudomonas aeruginosa, Escherichia coli, Salmonella typhimurium, or Pasteurella haemolytica (Table S3).
Aboody and Mickymaray [35] reported the antimicrobial activity of the anthocyanin and carotenoid extracts against the tested fungi, yeast, and pathogenic bacteria. Plants produce flavonoids in response to microbial infection, and the antimicrobial effects of these compounds are likely due to their ability to interact with bacterial cell walls. Carotenoid pigments extracted from flowers have also been shown to exhibit strong antimicrobial activity, particularly against Staphylococcus aureus [36].
The anthocyanin pigment extracts from the various plants tested in this study exhibited significant antimicrobial activity against most pathogenic microorganisms (p < 0.0001), with the exception of the extract from eggplant peels (Table 2, Figure 1). The prune and hibiscus extracts were the most effective, inhibiting microbial growth within ranges of 1.3–2.5 cm and 2.5–3.7 cm, respectively.
Anthocyanins are known for their diverse biological and pharmacological activities, including antioxidant, antimicrobial, antipyretic, hepatoprotective, and nephroprotective effects [37]. Abdel-Shafi et al. [38] reported that pigments extracted from Hibiscus sabdariffa inhibited all the tested bacteria and fungi, with anthocyanins demonstrating relatively higher antibacterial activity compared to antifungal activity. Previous studies have also shown that H. sabdariffa inhibits Staphylococcus aureus, Bacillus cereus, Escherichia coli, Clostridium sp., Klebsiella pneumoniae, and Pseudomonas fluorescens [39,40]. More recently, Chowdhury et al. [41] observed antibacterial activity of anthocyanins from fresh roselle against Shigella boydii and Shigella dysenteriae.
The microdilution assay revealed that both the hibiscus and prune pigment extracts exhibited antimicrobial activity, as demonstrated by their minimum inhibitory concentrations (MICs) and minimum microbiocidal concentrations (MMCs) (Table 3).
Against bacterial pathogens, the hibiscus extract had MIC values ranging from 0.25 to 1 μg/mL and MMC values ranging from 0.5 to 2 μg/mL. However, Klebsiella pneumoniae was less susceptible to the hibiscus extract, exhibiting a higher MIC value. The prune extract had MIC values ranging from 1 to 4 μg/mL and MMC values ranging from 2 to 8 μg/mL against bacterial pathogens.
Against fungal pathogens, the hibiscus extract had MIC values ranging from 4 to 16 μg/mL and MMC values ranging from 8 to 32 μg/mL. The prune extract had MIC values ranging from 4 to 8 μg/mL and MMC values ranging from 8 to 16 μg/mL against fungal pathogens.

3.3. Biofilm Formation and Inhibition Profile of Tested Pathogens

The inherent ability of various bacterial and fungal pathogens to form robust biofilms was initially assessed using the microtiter plate method. As presented in Figure 2, all the tested isolates, encompassing a diverse range of clinically relevant microorganisms, demonstrated substantial biofilm formation in the untreated control group. Untreated cultures consistently exhibited strong biofilm formation (SBF), with representative OD570 values ranging from 0.298 ± 0.006 for Pseudomonas aeruginosa to 0.992 ± 0.022 for Klebsiella oxytoca. These consistently high optical density values across the isolates confirm their strong biofilm-producing capabilities, underscoring the widespread nature of this virulence mechanism among the studied pathogens. This finding is consistent with numerous reports highlighting the significant role of biofilm formation in enhancing microbial resistance to antimicrobials and host immune responses, thereby contributing to persistent infections and treatment failures in both clinical and environmental settings [42,43].
Figure 2 further display the antibiofilm activities of the hibiscus and prune extracts against these diverse pathogens. Statistical analysis (ANOVA, p < 0.05) revealed highly significant differences (p < 0.0001 for most pathogens, and p = 0.003 for Morganella morganii) in biofilm formation among the untreated control, hibiscus-treated, and prune-treated groups for nearly all the examined isolates. Both pigment extracts significantly reduced the biofilm formation compared to their respective untreated controls, as evidenced by the lower OD values and different superscript letters (b or c) compared to the untreated ‘a’ group.
Detailed analysis of the treated groups indicates that the hibiscus extract generally exhibited superior antibiofilm activity compared to the prune extract for the majority of tested isolates where a statistical difference between the two treatments was observed (indicated by different superscript letters ‘b’ and ‘c’). For instance, the hibiscus extract notably reduced the biofilm formation of E. coli to an OD570 of 0.078 ± 0.004, classifying it as weak biofilm formation (WBF), significantly outperforming the prune extract (OD570 0.247 ± 0.008, SBF). Similarly, K. oxytoca and P. haemolytica exhibited WBF after hibiscus treatment (0.088 ± 0.018 and 0.095 ± 0.007, respectively), while prune treatment resulted in significantly higher ODs (0.334 ± 0.003 and 0.35 ± 0.01, respectively), classifying them as SBF. This potent antibiofilm effect of hibiscus is often attributed to its rich content of phytochemicals, such as anthocyanins, phenolic acids, and flavonoids, which are known to interfere with bacterial adhesion, extracellular polymeric substance (EPS) production, and quorum sensing pathways, all critical for biofilm development [44,45].
For some pathogens, however, the antibiofilm efficacy between the two extracts was statistically comparable (indicated by both treatments having the same superscript letter, ‘b’). For example, against S. aureus (hibiscus: 0.22 ± 0.015, prunes: 0.308 ± 0.026) and M. morganii (hibiscus: 0.146 ± 0.028, prunes: 0.19 ± 0.011), both extracts demonstrated a significant reduction in biofilm formation compared to the untreated control, with no statistically significant difference between hibiscus and prunes, although hibiscus consistently showed numerically lower ODs (better inhibition). Conversely, for Salmonella typhimurium, the prune extract (0.218 ± 0.004) numerically led to slightly lower biofilm formation (MBF) compared to hibiscus (0.238 ± 0.014, SBF), though this difference was also not statistically significant. The antibiofilm properties of the prunes extract are linked to their high concentrations of polyphenols (chlorogenic acid and neochlorogenic acid), tannins, and dietary fiber, which have been documented to possess antimicrobial and antiadhesion properties, thus inhibiting biofilm formation in various microbial species [46].
The observed pathogen-specific variations in the efficacy of the hibiscus and prune extracts highlight the diverse mechanisms by which these natural products interact with different microbial species and their biofilm matrices. The significant antibiofilm activities demonstrated, particularly by the hibiscus extract, underscore their promising potential as natural agents for developing novel therapeutic or preventive strategies against biofilm-associated infections, offering a complementary approach to conventional antimicrobial treatments.

3.4. Effect of Prune and Hibiscus Extracts on Virulence Gene Expression

In this study, we investigated the effects of sub-inhibitory concentrations of prune and hibiscus pigment extracts on the expression of key virulence genes in a panel of pathogenic microorganisms. The relative expression of all the target virulence genes in the untreated control samples was normalized to one, serving as the baseline for comparison. Overall, both extracts exhibited a tendency to downregulate gene expression, suggesting a potential for anti-virulence activity (Figure 3). However, significant variations were observed, indicating the differential effects of the extracts on specific genes and pathogens. The accurate measurement of gene expression is dependent on the specificity of the primers used. Primers were designed to specifically target the genes of interest, to avoid off-target binding, and to ensure the validity of the data obtained. The present study investigated the effects of hibiscus and prune extracts on the expression of key virulence genes across a diverse panel of pathogenic microorganisms. Consistent with our hypothesis, both extracts generally induced significant downregulation of these genes, suggesting a broad-spectrum anti-virulence potential. This downregulation, observed across genes involved in adhesion, invasion, toxin production, motility, and capsule regulation, signifies a multifaceted approach to attenuating pathogen virulence.
In Staphylococcus aureus, the observed downregulation of icaA (intercellular adhesin A) and hlg (gamma-hemolysin) by both extracts suggests a reduction in biofilm formation and hemolytic activity, respectively. IcaA is essential for biofilm development, a critical factor in S. aureus pathogenesis, enabling persistent infections and antibiotic resistance [47]. The reduced expression of icaA indicates a potential disruption of this process, weakening the bacteria’s ability to form robust biofilms. Similarly, the downregulation of hlg, a pore-forming toxin, implies a decrease in the bacteria’s capacity to damage host cells [48], potentially mitigating tissue damage and inflammation.
For Klebsiella pneumoniae and Klebsiella oxytoca, the significant downregulation of mrkD (mannose-resistant hemagglutinin) and rmpA (regulator of mucoid phenotype A) by both extracts is noteworthy. MrkD mediates bacterial adherence to host cells, while rmpA regulates capsule production, a crucial virulence factor [49]. The reduced expression of mrkD suggests a diminished ability of Klebsiella spp. to colonize host tissues, while the downregulation of rmpA indicates a potential decrease in capsule production, rendering the bacteria more susceptible to host immune defenses [50].
In Pseudomonas fluorescens and Pseudomonas aeruginosa, the downregulation of pslA (polysaccharide synthesis locus A) and fliC (flagellin) by both extracts suggests a reduction in biofilm formation and motility. PslA is critical for exopolysaccharide synthesis, essential for biofilm formation [51,52], and fliC is the major component of flagella, required for bacterial motility [53]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to colonize and spread.
The observed downregulation of ompA (outer membrane protein A) and neuC (N-acetylneuraminic acid synthase) in Escherichia coli by both extracts suggests a reduction in adhesion, invasion, biofilm formation, and capsule production. OmpA is involved in multiple virulence functions, and neuC is involved in capsule biosynthesis. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to cause infections [54].
Furthermore, in Salmonella typhimurium, the downregulation of sopB (Salmonella outer protein B) and mgtC (magnesium transporter C) by both extracts indicates a reduction in invasion and intracellular survival. SopB is an effector protein that manipulates host cell signaling [55], and mgtC is essential for intracellular survival [56]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to cause systemic infections.
The downregulation of Gcp (glycopeptide-containing protein) and lktC (leukotoxin C) in Pasteurella haemolytica by both extracts suggests a reduction in adhesion, colonization, and leukotoxin production. Gcp is involved in adhesion and colonization, and lktC is a toxin that damages bovine leukocytes [57]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to cause respiratory infections.
In Morganella morganii, the downregulation of ureC (urease C) and hdc (histidine decarboxylase) indicates a reduction in urease activity and histamine production, respectively. UreC contributes to urinary tract infections, and hdc contributes to inflammation [58]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to cause infections.
The downregulation of paH (plasmid antigen H) and ial (invasion-associated locus) in Shigella flexneri indicates a reduction in adherence, invasion, and intracellular spread. PaH contributes to adherence and invasion, and ial is essential for invasion and intracellular spread [59]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the bacteria’s ability to cause shigellosis.
In Candida albicans, the downregulation of ALS3 (adhesin-like sequence 3) and PLD1 (phospholipase D1) indicates a reduction in adhesion, biofilm formation, and tissue damage. ALS3 is an adhesin, and PLD1 is involved in cell signaling and virulence [60]. The reduced expression of these genes indicates a potential disruption of these processes, weakening the fungus’s ability to cause candidiasis.
Finally, in Aspergillus flavus and Aspergillus niger, the downregulation of the aflatoxin gene indicates a reduction in aflatoxin production, a direct virulence factor and a mycotoxin harmful to animal and human health [61]. Furthermore, the downregulation of pgxB (polygalacturonase xylanase B), predominantly associated with plant cell wall degradation [62], suggests a broader impact on fungal metabolism and overall pathogenicity. The reduced expression of these key genes collectively indicates a potential disruption of virulence-associated processes, thereby weakening the fungi’s ability to cause disease.
Overall, the consistent downregulation of virulence genes across diverse pathogens by pigment-rich extracts suggests a promising anti-virulence strategy. The observed reductions in gene expression, indicative of decreased adhesion, invasion, toxin production, motility, and capsule formation, collectively contribute to a weakened pathogen phenotype. This highlights the potential of these pigment-containing botanical extracts as novel therapeutic agents. Further research is warranted to elucidate the specific bioactive compounds responsible for these effects and to evaluate the efficacy of these extracts in in vivo models.

3.5. Chemical Characterization of Hibiscus sabdariffa Extract

The chemical composition of the Hibiscus sabdariffa extract was comprehensively evaluated using a complementary approach of high-performance liquid chromatography (HPLC) and gas chromatography–mass spectrometry (GC-MS) analyses, targeting different classes of phytochemicals.

3.5.1. HPLC Analysis of Hibiscus Profile and Molecular Docking Insights

High-performance liquid chromatography (HPLC) analysis was conducted to identify and quantify the major anthocyanin glucosides within the Hibiscus sabdariffa pigment-enriched extract. By direct comparison of the retention times and UV-Vis absorption spectra to authentic standards run under identical conditions, three primary anthocyanin glucosides were confidently identified and quantified (Figure S1).
Specifically, the following compounds were detected and quantified:
  • Delphinidin-3-O-glucoside was observed at a retention time (RT) of 5.5 min with a concentration of 5.22 µg/mL, representing 17.57% of the total quantified anthocyanins.
  • Pelargonidin-3-O-glucoside eluted at 7.0 min, present at a concentration of 10.13 µg/mL, representing 34.09% of the total quantified anthocyanins.
  • Cyanidin-3-O-glucoside was identified at an RT of 9.0 min, showing the highest concentration among the three at 14.36 µg/mL, representing 48.33% of the total quantified anthocyanins.
Collectively, these three quantified anthocyanins total 29.71 µg/mL in the extract. These findings indicate that the hibiscus extract is rich in these specific anthocyanin pigments, with Cyanidin-3-O-glucoside being the most abundant, followed by Pelargonidin-3-O-glucoside and Delphinidin-3-O-glucoside. This profile is consistent with the previous literature on Hibiscus sabdariffa (roselle), a species well-known for its abundant anthocyanin content, particularly these specific glucosides [63,64]. The detected anthocyanins are widely recognized for their robust antioxidant and anti-inflammatory properties [65].
Molecular Docking of Identified Anthocyanins Against OmpA
To explore the potential molecular mechanisms of antimicrobial activity, the three major identified anthocyanins (Cyanidin-3-O-glucoside, Delphinidin-3-O-glucoside, and Pelargonidin-3-O-glucoside) were selected for molecular docking studies due to their confirmed presence and high concentration in the active hibiscus extract. The E. coli outer membrane protein A (OmpA; PDB ID: 1QJP) was chosen as a target due to its critical role in bacterial membrane stability, structural integrity, and interactions with environmental factors, making it a key focus for antimicrobial studies [66,67]. Molecular docking of the selected anthocyanins against the 1QJP protein was performed using AutoDock Vina software version 1.2.7. The grid boxes were maximized to cover the entire protein. The docking scores ranged from –4.012 to –3.524 kcal/mol, indicating moderate binding affinities across all the compounds. Analysis of the interactions revealed that Cyanidin-3-O-glucoside exhibited the highest affinity (–4.012 kcal/mol). Graphical investigation of the complex (Figure 4 and Figure 5) depicted hydrogen bond interactions with PHE51 and MET53. Additionally, the ligand established Pi–alkyl interactions with LEU79 and PHE51, which further assisted in maintaining the ligand within the hydrophobic pocket of the receptor. These combined interactions suggest a robust binding mode for Cyanidin-3-O-glucoside within the active site. Delphinidin-3-O-glucoside showed moderate binding affinity, forming hydrogen bonds with ASN46, ASP44, and ASP42. Additional hydrophobic interactions with VAL45 (Pi–alkyl) and ASN5 (van der Waals) contributed to binding stability. Pelargonidin-3-O-glucoside showed the weakest binding among the three ligands, forming a single hydrogen bond with TYR43, supported by Pi–Pi stacking and Pi–cation interactions with PHE51 (Table 4, Figure 4 and Figure 5).

3.5.2. GC-MS Identification of Non-Pigment Bioactive Compounds in Hibiscus sabdariffa Extract and Related Molecular Docking

Gas chromatography–mass spectrometry (GC-MS) is a powerful analytical technique employed to identify and quantify volatile and semi-volatile compounds within complex mixtures, including diverse classes such as fatty acids, esters, alcohols, and hydrocarbons [50]. Crucially, GC-MS is suited for these specific compound types due to their volatility and thermal stability, whereas pigmented compounds are generally non-volatile and thermally labile, making them unsuitable for direct GC analysis [68,69]. A total of 25 distinct peaks were detected in the GC-MS chromatogram of the Hibiscus sabdariffa extract, collectively accounting for 99.92% of the total volatile and semi-volatile components detected by GC-MS.
This indicates a thorough quantification of this fraction of the extract’s chemical profile. From this comprehensive analysis, Table 5 presents 14 key bioactive compounds that were unequivocally identified and are known to possess antimicrobial properties. These 14 compounds collectively constitute approximately 53.09% of the total detected volatile and semi-volatile fraction of the extract. The selection of these compounds for detailed reporting in the main text is based on their established antimicrobial relevance and/or their relatively higher abundance within the extract.
The identified components, along with their retention times (RTs), relative concentrations (peak areas %), molecular formulas, and molecular weights (MWs), are summarized in Table 5. Among the prominent bioactive compounds identified were 1-Deoxy-d-arabitol, 2,5-Methylene-d,l-rhamnitol, 1,3,5-Trimethylbenzene, and several fatty acids and their derivatives including 1-Nonanol, 1-Dodecene, 1-Tetradecanol, 2-Myristynoyl pantetheine, Octadecanoic acid, 9-Octadecenoic acid (oleic acid), 11-Octadecenoic acid methyl ester, Trans-13-octadecenoic acid, Cis-13-Eicosenoic acid, and 9-Hexadecenoic acid. The presence of this diverse array of bioactive constituents strongly suggests the potential for various medicinal activities. Notably, a literature review concerning the identified compounds indicated that these 14 bioactive compounds within the Hibiscus sabdariffa pigment-enriched extract possess known antimicrobial properties.
While Table 5 focuses on these 14 highly relevant antimicrobial compounds, the remaining detected peaks (representing approximately 46.91% of the volatile/semi-volatile fraction) include other minor or less characterized components, some of which could not be definitively identified by spectral database matching. A complete list of all the detected peaks, including their retention times, area percentages, and tentative identifications (where available), is provided in the Supplementary Data (Supplementary Table S4), ensuring the full transparency of our GC-MS findings.
Molecular Docking of Non-Pigment Compounds Against OmpA
1-Deoxy-d-arabitol and oleic acid were selected for docking against the 1QJP protein due to their relative abundance in the extract and/or their known biological relevance from the literature. In both docking studies, the grid box size was maximized to encompass the entire protein. Analysis of the 1-Deoxy-d-arabitol docking results revealed that the lowest energy pose (−4.4 kcal/mol) interacted with the beta-sheet and a modeled loop region of the protein. Visualization of the docked complex showed that all but one hydroxyl group of 1-Deoxy-d-arabitol participated in hydrogen bond interactions with the protein’s side chain residues. Specifically, Asn25 and Asn26 acted as hydrogen bond acceptors (HBAs), Ala104 acted as a hydrogen bond donor (HBD), and Thr106 acted as both an HBA and an HBD. Hydrophobic interactions were also observed with Asp105 (van der Waals) and Ile24 (alkyl–alkyl) (Figure 6). The lowest energy (–5.6 kcal/mol) pose of oleic acid attained in the docking against 1QJP showed that the polar end of oleic acid performed three hydrogen bond interactions with the side chain residues, i.e., Trp15 (HBA, 2.52 Å), Gln17 (HBD, 2.90 Å), and Gly160 (HBA, 2.28 Å), respectively. The hydrophobic tail of oleic acid performed hydrophobic interactions (both alkyl–alkyl and p–alkyl) with the side chain residues of both the beta sheets and the modeled part of the protein, including Leu139, Tyr141, Pro157, and Leu162, as shown in Figure 7.

3.5.3. Overall Phytochemical Profile and Bioactivity Nexus of Hibiscus sabdariffa Extract

Collectively, the quantification of major pigments through HPLC and the detailed and comprehensive identification of various non-pigment secondary metabolites through GC-MS provide a robust initial characterization of our Hibiscus sabdariffa pigment-enriched extract. While pigments distinctly contribute to the vibrant coloration and exhibit significant antioxidant and anti-inflammatory capacities, the co-existence of numerous other bioactive compounds (as identified by GC-MS) highlights the complex phytochemical richness of this extract. The initial insights from the molecular docking studies further suggest that both anthocyanins and non-pigment compounds, such as 1-Deoxy-d-arabitol and oleic acid, are capable of favorable interactions with relevant microbial targets. This integrated approach of chemical characterization and computational investigation against a specific bacterial target (OmpA) provides novel insights into the potential molecular basis of hibiscus bioactivity. This intricate blend of compounds suggests that the observed broad-spectrum antimicrobial, antibiofilm, and anticancer biological activities are likely the result of synergistic or additive effects among these diverse constituents, rather than being attributable to a single class of compounds. The presence of these intensely colored anthocyanins also underscores the potential of this hibiscus extract not only as a natural food colorant, offering a viable alternative to synthetic dyes, but also as a valuable functional ingredient. Their demonstrated bioactivities, supported by our phytochemical characterization, position the extract for applications in the food, nutraceutical, or even pharmaceutical sector, contributing to both aesthetic appeal and significant health promotion.

3.6. Cytotoxicity

Based on its robust and broad-spectrum antimicrobial and antibiofilm activities observed in the initial screenings among the 14 tested plant extracts, the Hibiscus sabdariffa pigment extract was selected for further investigation into its cytotoxicity against cancer cells. This extract exhibited moderate to high cytotoxicity against MCF-7 breast cancer cells, with a determined IC50 value of 62.075 ± 15.161 μM. Notably, the extract demonstrated significantly lower toxicity toward the non-tumorigenic MCF-10A cells, with an IC50 value exceeding 1000 μM. Due to this extremely high IC50 value in MCF-10A cells, it was not possible to calculate a selectivity index (SI) for the pigment extract. While the lower toxicity observed in MCF-10A cells suggests potential for selective anticancer activity, the SI remains undetermined (Table 6 and Figure 8). These results can be compared to the known cytotoxicity of the standard chemotherapy agent cisplatin, which has been reported to have a much lower IC50 value of 5.35 ± 1.54 μM. Anthocyanins, in general, have been reported to exhibit anticarcinogenic activity against various cancer cell lines in both in vitro and in vivo tumor models [70].

4. Conclusions

In an era defined by escalating antimicrobial resistance and the persistent global burden of cancer, the urgent need for novel, sustainable therapeutic alternatives is undeniable. Our study, focusing on plant-pigment-enriched extracts from Hibiscus sabdariffa, provides compelling evidence of a multifaceted therapeutic potential, offering a promising answer to these critical health challenges.
This work uniquely advances the understanding of H. sabdariffa (and Prunus domestica) by demonstrating its integrated efficacy across multiple biological fronts. Our findings reveal significant broad-spectrum antimicrobial and potent antibiofilm capabilities against diverse, clinically relevant pathogens, including crucial multidrug-resistant strains. Beyond direct inhibition, we innovatively show that H. sabdariffa extracts can strategically modulate microbial virulence gene expression even at sub-inhibitory concentrations, effectively disrupting fundamental pathogenic mechanisms and mitigating toxin production.
Crucially, this investigation also unravels the molecular underpinnings of these effects. Through detailed pigment characterization via HPLC and complementary chemical profiling via GC-MS, we identified a rich and diverse array of bioactive compounds. These include key anthocyanin glycosides (representative pigments) and a wide range of non-pigment secondary metabolites, such as fatty acids, alcohols, and other unique phytochemicals. Molecular docking provided critical mechanistic insights into their favorable interactions with microbial targets, suggesting potential modes of action for both pigment and non-pigment constituents. Furthermore, the observed selective cytotoxicity of H. sabdariffa against MCF-7 breast cancer cells broadens its therapeutic spectrum beyond infectious diseases.
Collectively, this comprehensive investigation firmly establishes plant-pigment-enriched extracts from H. sabdariffa as a highly promising and sustainable source of next-generation therapeutics. This study not only offers a novel and readily available approach to address the global challenge of drug-resistant infections but also positions H. sabdariffa as a significant candidate in the burgeoning field of natural-product-derived anticancer drug development. Moving forward, comprehensive metabolomic analysis is essential to fully characterize the complete spectrum of non-pigment compounds, followed by precise quantitative profiling. This will enable a thorough assessment of the synergistic contributions of all the bioactive constituents within these complex extracts, paving the way for targeted therapeutic applications, and ultimately, clinical translation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms13081818/s1, Figure S1. HPLC Chromatogram and Quantification of Major Anthocyanin Glycosides Identified in Hibiscus Pigment; Table S1. Quantification of Pigment Concentrations (Chlorophyll, Carotenoids, Anthocyanins) in Various Plants; Table S2. Effect of chlorophyll pigment from different plants on the growth of pathogenic organisms; Table S3. Effect of carotenoid pigment from different plants on the growth of pathogenic organisms; Table S4. Comprehensive List of Compounds Identified in Hibiscus sabdariffa Extract by GC-MS Analysis. References [14,15,71,72,73] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, A.S.E.-D., A.E.S. and G.E.D.; methodology, A.S.E.-D., A.E.S., A.A., H.H., Y.M. and G.E.D.; software, A.S.E.-D.; validation, A.S.E.-D.; formal analysis, A.S.E.-D., A.A., Y.M. and G.E.D.; investigation, A.S.E.-D., A.E.S., Y.M. and G.E.D.; resources, A.S.E.-D., A.E.S., A.A., H.H., Y.M. and G.E.D.; data curation, A.S.E.-D., A.E.S., A.A., H.H. and Y.M.; writing—original draft, A.S.E.-D., A.E.S. and G.E.D.; writing—review and editing, A.S.E.-D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Princess Nourah bint Abdulrahman University Researchers Supporting Project Number (PNURSP2025R305), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Materials. Further inquiries can be directed to the corresponding author(s).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Karimi-Maleh, H.; Darabi, R.; Shabani-Nooshabadi, M.; Baghayeri, M.; Karimi, F.; Rouhi, J.; Alizadeh, M.; Karaman, O.; Vasseghian, Y.; Karaman, C. Determination of D&C Red 33 and Patent Blue V Azo Dyes Using an Impressive Electrochemical Sensor Based on Carbon Paste Electrode Modified with ZIF-8/g-C3N4/Co and Ionic Liquid in Mouthwash and Toothpaste as Real Samples. Food Chem. Toxicol. 2022, 162, 112907. [Google Scholar] [CrossRef] [PubMed]
  2. Ebrahem, A.F.; El-Demerdash, A.S.; Orady, R.M.; Nabil, N.M. Modulatory Effect of Competitive Exclusion on the Transmission of ESBL E. coli in Chickens. Probiotics Antimicrob. Proteins 2023, 16, 1087–1098. [Google Scholar] [CrossRef]
  3. Buledi, J.A.; Mahar, N.; Mallah, A.; Solangi, A.R.; Palabiyik, I.M.; Qambrani, N.; Karimi, F.; Vasseghian, Y.; Karimi-Maleh, H. Electrochemical Quantification of Mancozeb through Tungsten Oxide/Reduced Graphene Oxide Nanocomposite: A Potential Method for Environmental Remediation. Food Chem. Toxicol. 2022, 161, 112843. [Google Scholar] [CrossRef]
  4. Gorlenko, C.L.; Kiselev, H.Y.; Budanova, E.V.; Zamyatnin, A.A.; Ikryannikova, L.N. Plant Secondary Metabolites in the Battle of Drugs and Drug-Resistant Bacteria: New Heroes or Worse Clones of Antibiotics? Antibiotics 2020, 9, 170. [Google Scholar] [CrossRef]
  5. El-Demerdash, A.S.; Mohamady, S.N.; Megahed, H.M.; Ali, N.M. Evaluation of Gene Expression Related to Immunity, Apoptosis, and Gut Integrity That Underlies Artemisia’s Therapeutic Effects in Necrotic Enteritis-Challenged Broilers. 3 Biotech 2023, 13, 181. [Google Scholar] [CrossRef]
  6. Hashem, N.M.; Essawi, W.M.; El-Demerdash, A.S.; El-Raghi, A.A. Biomolecule-Producing Probiotic Bacterium Lactococcus lactis in Free or Nanoencapsulated Form for Endometritis Treatment and Fertility Improvement in Buffaloes. J. Funct. Biomater. 2024, 15, 138. [Google Scholar] [CrossRef] [PubMed]
  7. El-Demerdash, A.S.; Kamel, S.A.; Elariny, E.Y.T.; Henidi, H.; Mahran, Y.; Alahdal, H.; Saleh, A.M.; Ibrahim, R.A. Natural Inhibitors of Salmonella MDR Efflux Pumps AcrAB and AcrD: An Integrated In Silico, Molecular, and In Vitro Investigation. Int. J. Mol. Sci. 2024, 25, 12949. [Google Scholar] [CrossRef]
  8. Iqbal, Z.; Iqbal, M.S.; Hashem, A.; Abd_Allah, E.F.; Ansari, M.I. Plant Defense Responses to Biotic Stress and Its Interplay with Fluctuating Dark/Light Conditions. Front. Plant Sci. 2021, 12, 631810. [Google Scholar] [CrossRef]
  9. Singh, S.; Bhatt, P.; Kumar, V.; Singh, N.P. Phytonutrients, Anthocyanidins, and Anthocyanins: Dietary and Medicinal Pigments with Possible Health Benefits. In Advances in Flavonoids for Human Health and Prevention of Diseases; Apple Academic Press: Palm Bay, FL, USA, 2024. [Google Scholar]
  10. Ma, Y.; Ding, S.; Fei, Y.; Liu, G.; Jang, H.; Fang, J. Antimicrobial Activity of Anthocyanins and Catechins against Foodborne Pathogens Escherichia coli and Salmonella. Food Control 2019, 106, 106712. [Google Scholar] [CrossRef]
  11. Dong, Y.; Yang, C.; Zhong, W.; Shu, Y.; Zhang, Y.; Yang, D. Antibacterial Effect and Mechanism of Anthocyanin from Lycium ruthenicum Murr. Front. Microbiol. 2022, 13, 974602. [Google Scholar] [CrossRef]
  12. Porra, R.J. The Chequered History of the Development and Use of Simultaneous Equations for the Accurate Determination of Chlorophylls a and b. Photosynth. Res. 2002, 73, 149–156. [Google Scholar] [CrossRef]
  13. Alraof, A.M.A.; Ismaeil, F.H.M.; Elnabarway, A.A.E.; Eldesouky, H.S.A. Improvement of Squash Plants Growth Growing under Cold Stress Conditions by Using Calcium and Phosphorus Forms. Benha J. Appl. Sci. 2024, 9, 43–53. [Google Scholar] [CrossRef]
  14. Jeyanthi Rebecca, L.; Sharmila, S.; Das, M.P.; Seshiah, C. Extraction and Purification of Carotenoids from Vegetables. J. Chem. Pharm. Res. 2014, 6, 594–598. [Google Scholar]
  15. Shehata, W.A.; Akhtar, S.; Alam, T. Extraction and Estimation of Anthocyanin Content and Antioxidant Activity of Some Common Fruits. Trends Appl. Sci. Res. 2020, 15, 179–186. [Google Scholar] [CrossRef]
  16. Lohachoompol, V.; Mulholland, M.; Srzednicki, G.; Craske, J. Determination of Anthocyanins in Various Cultivars of Highbush and Rabbiteye Blueberries. Food Chem. 2008, 111, 249–254. [Google Scholar] [CrossRef]
  17. Essawi, W.M.; El-Demerdash, A.S.; El-Mesalamy, M.M.; Abonorag, M.A. Validation of Camel’s Fetal Fluids as Antimicrobial Agents. Curr. Microbiol. 2020, 77, 1399–1404. [Google Scholar] [CrossRef]
  18. El-Demerdash, A.S.; Orady, R.M.; Matter, A.A.; Ebrahem, A.F. An Alternative Approach Using Nano-Garlic Emulsion and Its Synergy with Antibiotics for Controlling Biofilm-Producing Multidrug-Resistant Salmonella in Chicken. Indian J. Microbiol. 2023, 63, 632–644. [Google Scholar] [CrossRef]
  19. Al Siraj, S.S.; Abd El-Tawab, A.A.; El-Hofy, F.L.; Elmasry, D.M.A. Comparison between Antimicrobial Activity of Thymus and Cumin Extracts and Their Nanoparticle on Salmonella Enteritidis. Benha Vet. Med. J. 2022, 43, 51–59. [Google Scholar]
  20. Stepanović, S.; Vuković, D.; Dakić, I.; Savić, B.; Švabić-Vlahović, M. A Modified Microtiter-Plate Test for Quantification of Staphylococcal Biofilm Formation. J. Microbiol. Methods 2000, 40, 175–179. [Google Scholar] [CrossRef]
  21. Sewid, A.H.; Sharaf, M.; El-Demerdash, A.S.; Ragab, S.M.; Al-Otibi, F.O.; Taha Yassin, M.; Liu, C.G. Hexagonal Zinc Oxide Nanoparticles: A Novel Approach to Combat Multidrug-Resistant Enterococcus faecalis Biofilms in Feline Urinary Tract Infections. Front. Cell Infect. Microbiol. 2024, 14, 1505469. [Google Scholar] [CrossRef]
  22. Yuan, J.S.; Reed, A.; Chen, F.; Stewart, C.N. Statistical Analysis of Real-Time PCR Data. BMC Bioinform. 2006, 7, 85. [Google Scholar] [CrossRef]
  23. Ali, N.M.; Mohamed, G.A.E.; El-Demerdash, A.S. Impact of Oral Administration of Chitosan—Nanoparticles on Oxidative Stress Index and Gut Microbiota of Heat Stressed Broilers. J. Adv. Vet. Res. 2023, 13, 997–1003. [Google Scholar]
  24. Ghadaksaz, A.; Fooladi, A.A.I.; Hosseini, H.M.; Amin, M. The Prevalence of Some Pseudomonas Virulence Genes Related to Biofilm Formation and Alginate Production among Clinical Isolates. J. Appl. Biomed. 2015, 13, 61–68. [Google Scholar] [CrossRef]
  25. Talukder, K.A.; Mondol, A.S.; Islam, M.A.; Islam, Z.; Dutta, D.K.; Khajanchi, B.K.; Azmi, I.J.; Hossain, M.A.; Rahman, M.; Cheasty, T.; et al. A Novel Serovar of Shigella Dysenteriae from Patients with Diarrhoea in Bangladesh. J. Med. Microbiol. 2007, 56, 654–658. [Google Scholar] [CrossRef]
  26. Klima, C.L.; Alexander, T.W.; Hendrick, S.; McAllister, T.A. Characterization of Mannheimia Haemolytica Isolated from Feedlot Cattle That Were Healthy or Treated for Bovine Respiratory Disease. Can. J. Vet. Res. 2014, 78, 38–45. [Google Scholar]
  27. Mason, W.J.; Blevins, J.S.; Beenken, K.; Wibowo, N.; Ojha, N.; Smeltzer, M.S. Multiplex PCR Protocol for the Diagnosis of Staphylococcal Infection. J. Clin. Microbiol. 2001, 39, 3332–3338. [Google Scholar] [CrossRef]
  28. Bugarel, M.; Granier, S.A.; Weill, F.X.; Fach, P.; Brisabois, A. A Multiplex Real-Time PCR Assay Targeting Virulence and Resistance Genes in Salmonella Enterica Serotype Typhimurium. BMC Microbiol. 2011, 11, 151. [Google Scholar] [CrossRef]
  29. Morse, D.J.; Wilson, M.J.; Wei, X.; Lewis, M.A.O.; Bradshaw, D.J.; Murdoch, C.; Williams, D.W. Denture-Associated Biofilm Infection in Three-Dimensional Oral Mucosal Tissue Models. J. Med. Microbiol. 2018, 67, 364. [Google Scholar] [CrossRef]
  30. Alameri, K.; A Al-Ameri, K.H.; Kadhim Al-Shibly, M. Studying the gene expression of virulence factors in aspergillus (flavus and niger) associated with the diabetic foot. Biochem. Cell. Arch. 2019, 19, 849–852. [Google Scholar]
  31. Dassault Systèmes BIOVIA. Discovery Studio Visualizer, V21.1.0.20298; Dassault Systèmes BIOVIA: San Diego, CA, USA.
  32. Trott, O.; Olson, A.J. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef]
  33. Dallakyan, S.; Olson, A.J. Small-Molecule Library Screening by Docking with PyRx. Methods Mol. Biol. 2015, 1263, 243–250. [Google Scholar] [CrossRef]
  34. Razali, N.M.; Wah, Y.B. Power Comparisons of Shapiro-Wilk, Kolmogorov-Smirnov, Lilliefors and Anderson-Darling Tests. J. Stat. Model. Anal. 2011, 2, 21–33. [Google Scholar]
  35. Al Aboody, M.S.; Mickymaray, S. Anti-Fungal Efficacy and Mechanisms of Flavonoids. Antibiotics 2020, 9, 45. [Google Scholar] [CrossRef]
  36. Shakeri, A.; Soheili, V.; Karimi, M.; Hosseininia, S.A.; Fazly Bazzaz, B.S. Biological Activities of Three Natural Plant Pigments and Their Health Benefits. J. Food Meas. Charact. 2018, 12, 356–361. [Google Scholar] [CrossRef]
  37. Riaz, G.; Chopra, R. A Review on Phytochemistry and Therapeutic Uses of Hibiscus sabdariffa L. Biomed. Pharmacother. 2018, 102, 575–586. [Google Scholar] [CrossRef] [PubMed]
  38. Abdel-Shafi, S.; Al-Mohammadi, A.R.; Sitohy, M.; Mosa, B.; Ismaiel, A.; Enan, G.; Osman, A. Antimicrobial Activity and Chemical Constitution of the Crude, Phenolic-Rich Extracts of Hibiscus sabdariffa, Brassica Oleracea and Beta Vulgaris. Molecules 2019, 24, 4280. [Google Scholar] [CrossRef] [PubMed]
  39. Al-Hashimi, A. Antioxidant and Antibacterial Activities of Hibiscus sabdariffa L. Extracts. Afr. J. Food Sci. 2012, 6, 506–511. [Google Scholar]
  40. Da-Costa-Rocha, I.; Bonnlaender, B.; Sievers, H.; Pischel, I.; Heinrich, M. Hibiscus sabdariffa L.—A Phytochemical and Pharmacological Review. Food Chem. 2014, 165, 424–443. [Google Scholar] [CrossRef]
  41. Chowdhury, K.; Chowdhury, S.; Bari, L.; Yeasmin, S. Improved extraction of anthocyanins with antibacterial activity from fresh roselle (Hibiscus sabdariffa L.) calyces. Dhaka Univ. J. Biol. Sci. 2024, 33, 139–147. [Google Scholar] [CrossRef]
  42. Elashkar, E.; Alfaraj, R.; El-Borady, O.M.; Amer, M.M.; Algammal, A.M.; El-Demerdash, A.S. Novel Silver Nanoparticle-Based Biomaterials for Combating Klebsiella Pneumoniae Biofilms. Front. Microbiol. 2024, 15, 1507274. [Google Scholar] [CrossRef]
  43. Almatroudi, A. Biofilm Resilience: Molecular Mechanisms Driving Antibiotic Resistance in Clinical Contexts. Biology 2025, 14, 165. [Google Scholar] [CrossRef]
  44. Husnah, M.; Suhartono, S.; Ismail, Y.S. A Current Perspective on Antibacterial and Antibiofilm Properties of Waru (Hibiscus tiliaceus L.). IOP Conf. Ser. Earth Environ. Sci. 2021, 711, 012019. [Google Scholar] [CrossRef]
  45. Lahiri, D.; Dash, S.; Dutta, R.; Nag, M. Elucidating the Effect of Anti-Biofilm Activity of Bioactive Compounds Extracted from Plants. J. Biosci. 2019, 44, 52. [Google Scholar] [CrossRef]
  46. De Rossi, L.; Rocchetti, G.; Lucini, L.; Rebecchi, A. Antimicrobial Potential of Polyphenols: Mechanisms of Action and Microbial Responses—A Narrative Review. Antioxidants 2025, 14, 200. [Google Scholar] [CrossRef] [PubMed]
  47. Abdel-Shafi, S.; El-Serwy, H.; El-Zawahry, Y.; Zaki, M.; Sitohy, B.; Sitohy, M. The Association between IcaA and IcaB Genes, Antibiotic Resistance and Biofilm Formation in Clinical Isolates of Staphylococci spp. Antibiotics 2022, 11, 389. [Google Scholar] [CrossRef] [PubMed]
  48. Los, F.C.O.; Randis, T.M.; Aroian, R.V.; Ratner, A.J. Role of Pore-Forming Toxins in Bacterial Infectious Diseases. Microbiol. Mol. Biol. Rev. 2013, 77, 173–207. [Google Scholar] [CrossRef]
  49. Li, Y.; Ni, M. Regulation of Biofilm Formation in Klebsiella Pneumoniae. Front. Microbiol. 2023, 14, 1238482. [Google Scholar] [CrossRef]
  50. El-Demerdash, A.S.; Alfaraj, R.; Fared, F.; Saleh, A.; Dawwam, G.E. Essential oils as Capsule Disruptors: Enhancing Antibiotic Efficacy Against Multidrug-Resistant Klebsiella Pneumoniae. Front. Microbiol. 2024, 15, 1467460. [Google Scholar] [CrossRef]
  51. Ghafoor, A.; Hay, I.D.; Rehm, B.H.A. Role of Exopolysaccharides in Pseudomonas aeruginosa Biofilm Formation and Architecture. Appl. Environ. Microbiol. 2011, 77, 5238–5246. [Google Scholar] [CrossRef]
  52. Saad, M.F.; Elsayed, M.M.; Khder, M.; Abdelaziz, A.S.; El-Demerdash, A.S. Biocontrol of Multidrug Resistant Pathogens Isolated from Fish Farms Using Silver Nanoparticles Combined with Hydrogen Peroxide Insight to Its Modulatory Effect. Sci. Rep. 2024, 14, 7971. [Google Scholar] [CrossRef]
  53. Bouteiller, M.; Dupont, C.; Bourigault, Y.; Latour, X.; Barbey, C.; Konto-ghiorghi, Y.; Merieau, A. Pseudomonas Flagella: Generalities and Specificities. Int. J. Mol. Sci. 2021, 22, 3337. [Google Scholar] [CrossRef]
  54. Maciel, J.F.; Matter, L.B.; Trindade, M.M.; Camillo, G.; Lovato, M.; de Ávila Botton, S.; Castagna de Vargas, A. Virulence Factors and Antimicrobial Susceptibility Profile of Extraintestinal Escherichia Coli Isolated from an Avian Colisepticemia Outbreak. Microb. Pathog. 2017, 103, 119–122. [Google Scholar] [CrossRef] [PubMed]
  55. Ruan, H.; Zhang, Z.; Tian, L.; Wang, S.; Hu, S.; Qiao, J.J. The Salmonella Effector SopB Prevents ROS-Induced Apoptosis of Epithelial Cells by Retarding TRAF6 Recruitment to Mitochondria. Biochem. Biophys. Res. Commun. 2016, 478, 618–623. [Google Scholar] [CrossRef] [PubMed]
  56. Retamal, P.; Castillo-Ruiz, M.; Mora, G.C. Characterization of MgtC, a Virulence Factor of Salmonella Enterica Serovar Typhi. PLoS ONE 2009, 4, e5551. [Google Scholar] [CrossRef] [PubMed]
  57. Confer, A.W.; Ayalew, S. Mannheimia Haemolytica in Bovine Respiratory Disease: Immunogens, Potential Immunogens, and Vaccines. Anim. Health Res. Rev. 2018, 19, 79–99. [Google Scholar] [CrossRef]
  58. Liu, H.; Zhu, J.; Hu, Q.; Rao, X. Morganella Morganii, a Non-Negligent Opportunistic Pathogen. Int. J. Infect. Dis. 2016, 50, 10–17. [Google Scholar] [CrossRef]
  59. Al-Maamory, E.H.; Al-Khafaji, J.K.; Al-Masoudi, H.K. Detection the Virulence-Associated Genes in Shigella Species Isolated from Diarrheal Samples in Babylon Province. J. Pharm. Sci. Res. 2018, 10, 3201–3206. [Google Scholar]
  60. Nikou, S.A.; Kichik, N.; Brown, R.; Ponde, N.O.; Ho, J.; Naglik, J.R.; Richardson, J.P. Candida Albicans Interactions with Mucosal Surfaces during Health and Disease. Pathogens 2019, 8, 53. [Google Scholar] [CrossRef]
  61. Liu, C.-Q.; Hu, K.-D.; Li, T.-T.; Yang, Y.; Yang, F.; Li, Y.-H.; Liu, H.-P.; Chen, X.-Y.; Zhang, H. Polygalacturonase Gene PgxB in Aspergillus Niger Is a Virulence Factor in Apple Fruit. PLoS ONE 2017, 12, e0173277. [Google Scholar] [CrossRef]
  62. Raruang, Y.; Omolehin, O.; Hu, D.; Wei, Q.; Promyou, S.; Parekattil, L.J.; Rajasekaran, K.; Cary, J.W.; Wang, K.; Chen, Z.Y. Targeting the Aspergillus Flavus P2c Gene through Host-Induced Gene Silencing Reduces A. Flavus Infection and Aflatoxin Contamination in Transgenic Maize. Front. Plant Sci. 2023, 14, 1150086. [Google Scholar] [CrossRef]
  63. Borrás-Linares, I.; Fernández-Arroyo, S.; Arráez-Roman, D.; Palmeros-Suárez, P.A.; Del Val-Díaz, R.; Andrade-Gonzáles, I.; Fernández-Gutiérrez, A.; Gómez-Leyva, J.F.; Segura-Carretero, A. Characterization of Phenolic Compounds, Anthocyanidin, Antioxidant and Antimicrobial Activity of 25 Varieties of Mexican Roselle (Hibiscus sabdariffa). Ind. Crops Prod. 2015, 69, 385–394. [Google Scholar] [CrossRef]
  64. Rasheed, D.M.; Porzel, A.; Frolov, A.; El Seedi, H.R.; Wessjohann, L.A.; Farag, M.A. Comparative Analysis of Hibiscus sabdariffa (Roselle) Hot and Cold Extracts in Respect to Their Potential for α-Glucosidase Inhibition. Food Chem. 2018, 250, 236–244. [Google Scholar] [CrossRef] [PubMed]
  65. Shahrajabian, M.H.; Cheng, Q.; Sun, W. The Importance of Neglected and Underutilized Medicinal Plants from South America in Modern Pharmaceutical Sciences. Lett. Drug Des. Discov. 2022, 20, 1688–1706. [Google Scholar] [CrossRef]
  66. Reusch, R.N. Insights into the Structure and Assembly of Escherichia Coli Outer Membrane Protein A. FEBS J. 2012, 279, 894–909. [Google Scholar] [CrossRef]
  67. Pautsch, A.; Schulz, G.E. High-Resolution Structure of the OmpA Membrane Domain. J. Mol. Biol. 2000, 298, 273–282. [Google Scholar] [CrossRef]
  68. McNair, H.M.; Miller, J.M.; Snow, N.H. Basic Gas Chromatography; John Wiley & Sons: New York, NY, USA, 2019. [Google Scholar]
  69. Orata, F. Derivatization Reactions and Reagents for Gas Chromatography Analysis, Advanced Gas Chromatography—Progress in Agricultural, Biomedical and Industrial Applications; InTech: Rijeka, Croatia, 2012; Volume 11, pp. 83–156. [Google Scholar]
  70. Wang, L.S.; Stoner, G.D. Anthocyanins and Their Role in Cancer Prevention. Cancer Lett. 2008, 269, 281–290. [Google Scholar] [CrossRef]
  71. Soni, D.K.; Shahi, S.K.; Khandel, P.; Mahobiya, D.; Singh, R.; Yadaw, R.K.; Kanwar, L. Extraction and Estimation of Chlorophylls from Epiphytic Orchids and Their Antioxidants Scavenging Activity analysis. Plant Arch. 2018, 18, 2448–2452. [Google Scholar]
  72. Rahman, M.M.; Rahaman, M.S.; Islam, M.R.; Hossain, M.E.; Mithi, F.M.; Ahmed, M.; Saldías, M.; Akkol, E.K.; Sobarzo-Sánchez, E. Multifunctional Therapeutic Potential of Phytocomplexes and Natural Extracts for Antimicrobial Properties. Antibiotics 2021, 10, 1076. [Google Scholar] [CrossRef]
  73. Ahmadi, A.; Shahidi, S.A.; Safari, R.; Motamedzadegan, A.; Ghorbani-HasanSaraei, A. Evaluation of Stability and Antibacterial Properties of Extracted Chlorophyll from Alfalfa (Medicago sativa L.). Food Chem. Toxicol. 2022, 163, 112980. [Google Scholar] [CrossRef]
Figure 1. Agar well diffusion assay results of both fluids and a blank (control negative 1% DMSO) on a lawn of S. typhimurium.
Figure 1. Agar well diffusion assay results of both fluids and a blank (control negative 1% DMSO) on a lawn of S. typhimurium.
Microorganisms 13 01818 g001
Figure 2. Antibiofilm efficacy of hibiscus and prune pigment extracts against diverse pathogens. (A) Visual confirmation by microtiter plate biofilm assay. (B) Heatmap illustrating antibiofilm activities by optical density (OD) and (C) quantitative inhibition of biofilm formation by extracts. a–c Bars with different superscript letters imply a statistical difference (p < 0.05).
Figure 2. Antibiofilm efficacy of hibiscus and prune pigment extracts against diverse pathogens. (A) Visual confirmation by microtiter plate biofilm assay. (B) Heatmap illustrating antibiofilm activities by optical density (OD) and (C) quantitative inhibition of biofilm formation by extracts. a–c Bars with different superscript letters imply a statistical difference (p < 0.05).
Microorganisms 13 01818 g002
Figure 3. Effect of hibiscus and prune extracts on the transcription of different genes in different studied microorganisms relative to the untreated control. *, p < 0.05; ns, non-significant.
Figure 3. Effect of hibiscus and prune extracts on the transcription of different genes in different studied microorganisms relative to the untreated control. *, p < 0.05; ns, non-significant.
Microorganisms 13 01818 g003aMicroorganisms 13 01818 g003b
Figure 4. Three-dimensional interactions between the protein and ligand: (A) Delphinidin 3-O-glucoside, (B) Pelargonidin 3-glucoside, and (C) Cyanidin 3-glucoside.
Figure 4. Three-dimensional interactions between the protein and ligand: (A) Delphinidin 3-O-glucoside, (B) Pelargonidin 3-glucoside, and (C) Cyanidin 3-glucoside.
Microorganisms 13 01818 g004
Figure 5. Two-dimensional interactions between the ligand and the protein with the distances in Angstrom: (A) Pelargonidin 3-glucoside, (B) Cyanidin 3-glucoside, and (C) Delphinidin 3-O-glucoside.
Figure 5. Two-dimensional interactions between the ligand and the protein with the distances in Angstrom: (A) Pelargonidin 3-glucoside, (B) Cyanidin 3-glucoside, and (C) Delphinidin 3-O-glucoside.
Microorganisms 13 01818 g005
Figure 6. (A) Structure of 1QJP protein with docked the 1-Deoxy-D-arabitol ligand, (B) three-dimensional interactions between the protein and the ligand, and (C) two-dimensional interactions between the ligand and the protein with the distances in Angstrom.
Figure 6. (A) Structure of 1QJP protein with docked the 1-Deoxy-D-arabitol ligand, (B) three-dimensional interactions between the protein and the ligand, and (C) two-dimensional interactions between the ligand and the protein with the distances in Angstrom.
Microorganisms 13 01818 g006
Figure 7. (A) Structure of 1QJP protein with the docked oleic acid ligand, (B) three-dimensional interactions between the protein and the ligand, and (C) two-dimensional interactions between the ligand and the protein with the distances in Angstrom.
Figure 7. (A) Structure of 1QJP protein with the docked oleic acid ligand, (B) three-dimensional interactions between the protein and the ligand, and (C) two-dimensional interactions between the ligand and the protein with the distances in Angstrom.
Microorganisms 13 01818 g007
Figure 8. Concentration-dependent cytotoxicity of pigment extract in MCF-7 and MCF-10A cells, demonstrating selective toxicity.
Figure 8. Concentration-dependent cytotoxicity of pigment extract in MCF-7 and MCF-10A cells, demonstrating selective toxicity.
Microorganisms 13 01818 g008
Table 1. Primer sequences, target genes and cycling conditions for SYBR green RT-PCR.
Table 1. Primer sequences, target genes and cycling conditions for SYBR green RT-PCR.
PathogenTarget GenePrimers SequencesAnnealing (T °C)Reference
S. aureus16S rRNAGAGGCAGCAGTAGGGAATCT60This study (Accession: LN794238.1)
GATAACGCTTGCCACCTACG
icaAAGCACAATGAAAACGAAAAGGT60This study (Accession: KU670830.1)
GCGACTATCAATAAAGAGTGCGA
hlgGGTAATTTCCAATCAGCCCCA60This study (Accession: KM116014.1)
TGACCTGATTCAGTGGCGAA
Klebsiella species16S rRNATCCAGGTGTAGCGGTGAAAT60This study (Accession: PP094549.1)
TGAGTTTTAACCTTGCGGCC
mrkDCTCGAAACCTATCTGAGCGC58This study (Accession: KF777787.1)
ATTAAAATCCTTCCCGCCGG
rmpAAGAGTATTGGTTGACTGCAGGATTT
AAACATCAAGCCATATCCATTGG
60This study (Accession: KF801503.1)
Pseudomonas species16S rRNAGGGAGGAAGGGCAGTAAGTT60This study (Accession: FJ972538.1)
ACCACCCTCTACCGTACTCT
pslATACCGGGCCTGGATGAAAC60This study (Accession: OM567545.1)
CGAGTTGTAGTTCTCCGGGA
fliCTGAACGTGGCTACCAAGAACG60[24]
TCTGCAGTTGCTTCACTTCGC
E. coli16S rRNATGAGTTTTAACCTTGCGGCC58This study (Accession: LC848137.1)
TCCAGGTGTAGCGGTGAAAT
ompACGGAAGTACAGACCAAGCAC58This study (Accession: MK941176.1)
CCCAGAACAACTACGGAACC
neuCTCCCTCTGACGATTGCATTT58This study (Accession: OL778841.1)
GTGGGCTAATTGGGAACTCC
Shigella flexneri16S rRNATGGCGCATACAAAGAGAAGC58This study (Accession: LC521305.1)
TTTTGCAACCCACTCCCATG
IpaHTGCCCGGGATAAAGTCAGAA58This study (Accession: OR804338.1)
CGGAGGTCATTTGCTGTCAC
IalGCTATAGCAGTGACATGG58[25]
ACGAGTTCGAAGCACTC
Pasteurella haemolytica
(Mannheimia)
16S rRNAACCAAGCCGTCGATCTCTAG58This study (Accession: U57072.1)
AAAACAACCACCTTCCTCGC
GcpCGCCCCTTTTGGTTTTCTAA58[26]
GTAAATGCCCTTCCATATGG
lktCGGAAACATTACTTGGCTATGG58[26]
TGTTGCCAGCTCTTCTTGATA
Morganella morganii16S rRNAGGCGTATACAAAGGGAAGCG60This study (Accession: AB680150.1)
TTTTGCAACCCACTCCCATG
ureCCAACCTGAACCCGAATGTCC58This study (Accession: U69175.1)
GTTAGCGGTCTGGATCACAC
hdcTACCTGGGCCGTGAAATCTT60This study (Accession: FJ469558.1)
TGCGCTTCTTTATCGTCAGC
Salmonella typhimurium16S rRNACAGAAGAAGCACCGGCTAACTC60[27]
GCGCTTTACGCCCAGTAATT
sopBTTTTCGGCAAAGAGGGAACG60This study (Accession: JQ067617.1)
GCCAGCTCATTAACACCCAC
mgtCAAGAGGCCGCGATCTGTTTA60[28]
CGAATTTCTTTATAGCCCTGTTCCT
Candida albicansACT1TGCTGAACGTATGCAAAAGG60[29]
TGAACAATGGATGGACCAGA
ALS3CTGGACCACCAGGAAACACT60[29]
GGTGGAGCGGTGACAGTAGT
PLD1GCCAAGAGAGCAAGGGTTAGCA60[29]
CGGATTCGTCATCCATTTCTCC
A. flavus18S rRNACGGAGACACCACGAACTCTG58[30]
CCCTACCTGATCCGAGGTCA
Afla toxin GeneTTGCTGCTTTTCGCTAGCAC58
TCATCAGGTTGCACGAACTG
A. niger18S rRNATTGTACCCTGTTGCTTCGGC58[30]
TTCAGCGGGTATCCCTACCT
pgxBTTGCGGCCGCTTTTGCGTCTTGATTGTGAG58
CGACAGACCCAAGCTTTGATGTGGGTAGATGCGTAG
Table 2. Effect of anthocyanin pigment from different plants on the growth of pathogenic organisms.
Table 2. Effect of anthocyanin pigment from different plants on the growth of pathogenic organisms.
Microbial IsolatesAnthocyanin Pigment
Diameter of the Inhibition Zone (cm)
p-Value
Beet Roots (Beta vulgaris)Eggplant Peels (Solanum melongena)Prunes (Prunus domestica)Red Cabbage (Brassica oleracea)Roselle Petals (Hibiscus sabdariffa)
S. aureus2.63 ± 0.110.02.2 ± 0.02.5 ± 0.03.5 ± 0.0<0.0001
Klebsiella pneumoniae0.00.01.3 ± 0.00.02.9 ± 0.0<0.0001
Klebsiella oxytoca2.43 ± 0.20.02.4 ± 0.02.5 ± 0.02.6 ± 0.0<0.0001
Pseudomonas fluorescens2.4 ± 0.050.01.9 ± 0.02.5 ± 0.03.7 ± 0.0<0.0001
Pseudomonas aeruginosa2.7 ± 0.20.02.1 ± 0.102.6 ± 0.053.6 ± 0.0<0.0001
E. coli3.2 ± 0.20.02.06 ± 0.052.4 ± 0.03.1 ± 0.0<0.0001
Salmonella typhimurium2.9 ± 0.170.02.2 ± 0.202.5 ± 0.02.9 ± 0.0<0.0001
Pasteurella haemolytica2 ± 0.00.02.2 ± 0.00.03 ± 0.0<0.0001
Morganella morganii3 ± 0.00.02.2 ± 0.02.7 ± 0.02.7 ± 0.0<0.0001
Shigella flexneri2.9 ± 0.00.02.5 ± 0.102.5 ± 0.02.5 ± 0.0<0.0001
Candida albicans2.8 ± 0.00.01.8 ± 0.101.4 ± 0. 02.5 ± 0.0<0.0001
Aspergillus flavus2.4 ± 0.00.02.1 ± 0.02.1 ± 0.02.5 ± 0.0<0.0001
Aspergillus niger2.4 ± 0.00.02 ± 0.01.7 ± 0.02.5 ± 0.0<0.0001
Table 3. SIC, MIC, and MMC of hibiscus and prune extracts against tested isolates.
Table 3. SIC, MIC, and MMC of hibiscus and prune extracts against tested isolates.
MicroorganismConcentration of SIC, MIC, MMC (µg/mL)
Hibiscus ExtractsPrunes Extracts
SICMICMMCSICMICMMC
S. aureus0.250.51248
Klebsiella pneumoniae3264128124
P. aeruginosa0.1250.250.5124
FP. Fluorescent0.1250.250.5124
Morganella morganii0.1250.250.50.512
E. coli0.1250.250.5124
S. typhimurium0.5120.512
Pasteurella haemolytica0.512124
Klebsiella oxytoca0.5120.512
Shigella flexneri0.250.51124
Candida albicans248248
Aspergillus flavus816322816
Aspergillus niger4816248
SIC: subinhibitory concentration (1/2 × MIC), MIC: minimum inhibitory concentration. MMC: minimum microbiocidal concentration.
Table 4. Predicted binding interactions and energies of hibiscus anthocyanin glycosides via molecular docking.
Table 4. Predicted binding interactions and energies of hibiscus anthocyanin glycosides via molecular docking.
LigandH-Bond Interaction—ResidueH-Bond Interaction—Distance (Å)Hydrophobic InteractionBinding Energy (kcal/mol)
Cyanidin 3-glucosidePHE51, MET532.25LEU79 (Pi–alkyl), PHE51 (Pi–alkyl)−4.012
Delphinidin 3-O-glucosideASN46, ASP44, ASP424.11, 4.15, 3.59VAL45 (Pi–alkyl), ASN5 (van der Waals)−3.967
Pelargonidin 3-glucosideTYR435.81PHE51 (Pi–Pi Stacked, Pi–Cation)−3.524
Table 5. Key non-pigment bioactive compounds identified in Hibiscus sabdariffa extract by GC-MS analysis.
Table 5. Key non-pigment bioactive compounds identified in Hibiscus sabdariffa extract by GC-MS analysis.
RT (Min)Compound NameArea %Molecular FormulaMolecular WeightKnown Bioactivities/Relevance
5.751-Deoxy-d-arabitol12.45C5H12O4136Sugar alcohol, potential antioxidant, and osmotic agent
6.492,5-Methylene-d,l-rhamnitol13.52C7H12O6192Sugar alcohol derivative; potential role in carbohydrate metabolism
6.60BENZENE, 1,3,5-TRIMETHYL3.18C9H12120Aromatic hydrocarbon; industrial solvent
7.711-NONANOL2.92C9H20O144Long-chain primary alcohol; used in flavor/fragrance industries; mild antimicrobial potential
8.301-DODECENE0.76C12H24168Alkene hydrocarbon; limited direct bioactivity; used in surfactants or as an intermediate
8.461-TETRADECANOL1.54C14H30O214Long-chain fatty alcohol; emollient and surfactant with possible antimicrobial effects
9.322-Myristynoyl pantetheine1.77C25H44N2O5S484Pantetheine derivative; involved in coenzyme A biosynthesis; metabolic cofactor; also noted for anti-inflammatory activity
11.03OCTADECANOIC ACID2.69C18H36O2284Saturated fatty acid; commonly used in pharmaceuticals and cosmetics; mild antibacterial effect
29.589-Octadecenoic acid1.35C18H34O2282Oleic acid isomer; monounsaturated fatty acid with anti-inflammatory activity
32.85Oleic acid3.92C18H34O2282Well-known monounsaturated fatty acid; supports cardiovascular and skin health; exhibits antibacterial activity
32.8511-octadecenoic acid, methyl ester3.92C19H36O2296Fatty acid ester; potential in antimicrobial or lipid metabolism studies
34.67Trans-13-octadecenoic acid1.39C18H34O2282Unsaturated fatty acid; investigated for lipid modulation and bioactivity
34.67Cis-13-Eicosenoic acid1.39C20H38O2310Monounsaturated fatty acid; limited data but structural analogs suggest anti-inflammatory roles
34.749-Hexadecenoic acid2.29C16H30O2254Palmitoleic acid; bioactive lipid with antimicrobial and anti-inflammatory effects
RT: retention time.
Table 6. IC50 values in μM reflect the cytotoxicity activity of the pigment in MCF-7 and non-tumorigenic MCF10A cells, along with the selectivity index (SI) for the pigment.
Table 6. IC50 values in μM reflect the cytotoxicity activity of the pigment in MCF-7 and non-tumorigenic MCF10A cells, along with the selectivity index (SI) for the pigment.
CompoundIC50 * (µM)
IC50 MCF-7 IC50 Mcf10ASelectivity Index (SI)
Pigment62.075 ± 15.161>1000ND a
* IC50 values are expressed as mean ± SD for three independent observations; a ND: not determined.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

El-Demerdash, A.S.; Sehim, A.E.; Altamimi, A.; Henidi, H.; Mahran, Y.; Dawwam, G.E. From Hue to Health: Exploring the Therapeutic Potential of Plant-Pigment-Enriched Extracts. Microorganisms 2025, 13, 1818. https://doi.org/10.3390/microorganisms13081818

AMA Style

El-Demerdash AS, Sehim AE, Altamimi A, Henidi H, Mahran Y, Dawwam GE. From Hue to Health: Exploring the Therapeutic Potential of Plant-Pigment-Enriched Extracts. Microorganisms. 2025; 13(8):1818. https://doi.org/10.3390/microorganisms13081818

Chicago/Turabian Style

El-Demerdash, Azza SalahEldin, Amira E. Sehim, Abeer Altamimi, Hanan Henidi, Yasmin Mahran, and Ghada E. Dawwam. 2025. "From Hue to Health: Exploring the Therapeutic Potential of Plant-Pigment-Enriched Extracts" Microorganisms 13, no. 8: 1818. https://doi.org/10.3390/microorganisms13081818

APA Style

El-Demerdash, A. S., Sehim, A. E., Altamimi, A., Henidi, H., Mahran, Y., & Dawwam, G. E. (2025). From Hue to Health: Exploring the Therapeutic Potential of Plant-Pigment-Enriched Extracts. Microorganisms, 13(8), 1818. https://doi.org/10.3390/microorganisms13081818

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop