- Research
- Open access
- Published:
Evaluation of multidrug resistance in the Gram-negative microbiome of cancer patients and the adverse effects of their metabolites on albino rats and epithelial or fibroblasts cell lines
Infectious Agents and Cancer volume 20, Article number: 2 (2025)
Abstract
Background
Cancer is a significant global health issue due to its high incidence and mortality rates. In recent years, the relationship between the human microbiota and cancer has garnered attention across various medical fields. This includes research into the microbial communities that influence cancer development, tumor-associated microorganisms, and the interactions between the microbiome and tumor, collectively referred to as the oncobiome.
Methods
The negative effects of secondary metabolites extracted from selected multidrug-resistant Gram-negative bacteria within the cancer microbiota were evaluated. These effects included carcinogenicity, mutagenicity, hepatotoxicity, nephrotoxicity, and sperm deformities observed in albino rats after one month of oral ingestion of these microbial extracts.
Results
Our findings in the present investigation revealed that among the bacterial community derived from the microbiota, Gram-negative bacteria accounted for 74.87% the total microbiota (146 out of 195) and their spectrum including Escherichia sp. (n = 36, 24.66%) followed by Acinetobacter sp. (n = 34, 23.29%), Stenotrophomonas sp. (n = 29, 19.86%), Pseudomonas sp. (n = 26, 17.81%) and Serratia sp. (n = 21, 14.38%), as the most prevalent pathogens. All isolates derived from the cancer microbiome exhibited multidrug resistance to a large number of conventional therapies. Out of them Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4 and Pseudomonas sp. Esraa 5 strains showed multidrug resistant profile against all antibiotic classes under study including penicillins, cephalosporins, carbapenems, fluoroquinolones, β-lactamase inhibitors combinations, folate synthesis pathway inhibitors, phosphonic, aminoglycosides, polymyxins, tetracyclines, macrolides, and chloramphenicol antibiotics. The adverse effects of oral ingestion of their metabolites were evaluated in albino rats. They induced pronounced carcinogenesis along with severe raise in the inflammatory cytokines, hepatotoxicity, nephrotoxicity, mutagenicity along with sperm deformities in treated animals. Moreover, all metabolites showed marked cytotoxicity against human normal cell lines; human mammary epithelial (MCF10A), human lung fibroblasts (WI38) and human dermal fibroblasts (HDFs).
Conclusion
These bacterial strains isolated from the cancer microbiome may play significant roles in inducing cancer, inflammation, mutagenesis, hepatotoxicity, nephrotoxicity, and sperm abnormalities, along with histopathological changes in the treated animal groups by orally administrated metabolites in compared to the untreated group.
Graphical Abstract

Introduction
Cancer is a major health concern globally because of its high incidence and mortality rates. In recent decades, the microbial association with human body (microbiota) has attracted attention in various medical fields, containing cancer biology and the microbial groups that have any kind of influence on cancer development, tumor-associated microorganisms, and the contact between microbiome and tumor were defined as the oncobiome [1, 2]. The eubiome or oncobiome excretes bioactive metabolites containing short-chain fatty acids, amino acid, hormone-like effects, and other secondary metabolites that trigger cancer initiation or can encourage metastasis creation in tumor like was showed in breast tumor or pancreas adenocarcinoma [3]. Current investigation points out the etiological role of the human microbiome and bacterial infections of the body as risk factors correlated with the development of ovarian, prostate, colorectal and breast cancers by altering the metabolism of steroid hormones and disrupting estrogen metabolism, which play a key role in most cancers [3,4,5].
Recent research has indicated that the development of pancreatic, esophageal, pancreatic and gynecological cancer is among the most lethal malignancies that are closely related to the microbiome and influence cancer development through a different mechanism including inflammation and immunomodulation [6,7,8,9]. In the past era, the role of host-microbial connections in inducing human tumor plasticity and malignant progress has been documented [10]. Development and alterations in the microbiota negatively impacts the structure and function of different organs as well as the host’s behavior [11]. Presently, different countries mentioned that the scale of bacteria in tumor patients has moved from prevalent G-positive to G-negative that seem to take the residence of G-positive, that can be caused by the relatively lower employ of lodging medical devices and lower prescribing of prophylactic antimicrobial usages in tumor patients [12].
Hence in order to effectively inhibit, recognize as well as handle infections, information of the altering epidemiology of infections is critical [13]. It has been displayed that bacterial inflammatory microenvironments mediate the tumor formation due to the contact with certain microorganisms and the immune system resultant in chronic inflammation leading to tumor [14]. Inflammations prompted tumor through various tools containing stimulation of cell proliferation, yield of great levels of metabolically activated free radicals, reactive oxygen species, N-nitrosamines that cause oxidative harm to DNA, regulatory proteins as well as other macromolecules; mutations or even gene deletions and then prompt and impulse different categories of tumor [15]. Amongst the infection of bacterial that prompted tumor, Salmonella typhi induce gallbladder cancer through changing the genomic sequence protein cancer p53 and increase of protooncogene c-MYC, Chlamydia pneumonia induce lung cancer by variation in apoptosis and cell programming signaling; overexpression of miRNA-328; exciting lung-resident γδ T cells; increase of Myd88-dependent IL-1b and IL-23; increase secretion of cytokines, IL-8, IL-10, and TNF but Helicobacter pylori was the cause of distal stomach adenocarcinoma through its capability to cause critical inflammations [16]. Lenický et al., reported that the bacterial Proteus mirabilis, Citrobacter braakii, Staphylococcus lentus and Escherichia coli as the most dominant strains had a negative influence on the sperm quality parameters including sperm motility, acrosome integrity and they stated the positive associations was detected concerning the bacterial load and fragmentation of DNA, ROS over-generation, oxidative mutilation to the proteins, and lipids in semen [17].
Antibiotics remain the mainstay of pathogens treatments and the select of empirical antibiotics therapy should be based on the prevalent pathogens and the sensitivity of those microorganisms to antibiotics that vary greatly from region to another then continuous monitoring of the type of drug-resistant local infection is critical [18, 19]. Cancer patients are at risk of multidrug-resistant nosocomial infections because of unsuitable usage of antibiotics [20]. It is expected that there will be more deaths due to antimicrobial resistance than cancer by 2050. The occurrence of drug resistance in cancer patients is a catastrophic problem because it combines the two biggest problems of humanity, multidrug resistance and tumorigenesis [14, 21,22,23]. Furthermore, Garg et al., reported that in cancer patients among 644 positive bacterial cultures, G-negative isolates were most commonly encountered (84.14%) with the highest level of resistance to various antibiotics [20]. Acinetobacter baumannii, Serratia marcescens, Klebsiella pneumoniae, Pseudomonas aeruginosa, Enterobacter sp., Stenotrophomonas maltophilia and Escherichia coli have gained notoriety worldwide as the most prevalent multidrug resistant pathogens that characterized as the most common carcinogens in tumor patients with moveable rate relying on the recording country and continent [24,25,26,27].
The present work was designed to measure multidrug-resistant G-negative bacteria (MDRGN) in the microbiome of cancer patients against 37 antibiotics of 12 chemical classes. Moreover, the adverse effects of secondary metabolites extracted from the selected strains that characterized as being XDR strains with the highest MICs values among the cancer microbiota including carcinogenicity, mutagenicity, hepatotoxicity, nephrotoxicity and sperm deformities that occurred in albino rats after oral ingestion of these microbial extracts, individually for one month were assessed. Furthermore, anti-proliferative activities of these extracts against different human normal cell lines including mammary epithelial (MCF10A), lung fibroblasts (WI38) and dermal fibroblasts (HDFs) cell lines were described.
Materials and methods
Antimicrobial susceptibility test
All bacterial isolates susceptible to antimicrobial agents performed according to the CLSI reference broth microdilution technique, EUCAST, or FDA categorical interpretations were used [28,29,30,31]. The antibiotics tested were penicillins (ampicillin, ticarcillin, oxacillin, piperacillin and carbenicillin), cephalosporins (cephalothin, cefazolin, cefoxitin, ceftazidime, cefepime, ceftriaxone and cefotaxime), carbapenems (ertapenem, imipenem, meropenem and doripenem), antibiotics with β-lactamase inhibitors (ceftazidime-avibactam, meropenem-vaborbactam as well as imipenem-cilastatin), folate synthesis pathway inhibitors (trimethoprim/sulfamethoxazole), fluoroquinolones (ofloxacin, ciprofloxacin, levofloxacin and moxifloxacin), phosphonic (fosfomycin), aminoglycosides (gentamicin, tobramycin, netilmicin, amikacin, nitrofurantoin and vancomycin), polymyxins (colistin), tetracyclines (doxycycline, minocycline and tigecycline), macrolides (clarithromycin and azithromycin) and chloramphenicol antibiotics (Oxoid, UK).
Clinical strains and extraction of secondary metabolites
The resistant strains Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4 and Pseudomonas sp. Esraa 5 were isolated from cancer patient, cultured and extracted like previously mentioned previously [14]. Briefly, each isolate was inoculated from blood agar plates into tryptic soy broth medium with 5% blood in 500 mL Erlenmeyer flasks and incubated at 37 ºC, 150 rpm for two days followed by extraction with ethyl acetate (EtOAc) at pH 4.0. Each ethyl acetate (EtOAc) extractable metabolite was dried, assigned the same code as its producing strain and stored at – 20 °C.
Animals and experimental design
Adult male Sprague–Dawley albino rats (weighting 150–180 g) were obtained from the animal colony (National Research Centre, Egypt), the rats were allowed free access to tap water and food, for a week before starting the trial for acclimatization. The animals were cared for in compliance with the fundamental standards for the handling and use of experimental rats, as approved by the ethical committee of the National Research Center (FWA 00014747), which had previously authorized this investigation. After acclimation, the animals were randomly arranged into 6 groups (7 rats per group) as group 1 healthy rats orally ingested with physiological saline (1.25 mL/kg body weight twice/week) for four weeks (control), while groups 2, 3, 4, 5, and 6 comprised healthy animals those were subjected to oral administration with Esraa 1, Esraa 2, Esraa 3, Esraa 4 and Esraa 5 extracts, respectively at dosage of 1.25 mL/kg twice/week (11.6 g of each extract was dissolved in 8 mL physiological saline) for a similar period.
Blood and tissue sampling
One day after the last administration, the rats were fasted overnight. Then, under diethyl ether anesthesia, blood samples were collected from retro orbital venous plexus, and centrifuged at 3000 rpm under cooling for 15 min; sera were separated, and stored at − 80 °C until the biochemical studies could be performed as soon as possible.
Biochemical measurements
Serum amino transferases (ALAT and ASAT), gamma glutamyl transferase (GGT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH) activities as well as serum urea and creatinine, alpha fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor necrosis factor alpha (TNFα) and interleukin-1beta (IL-1β) levels were determined [14].
Histopathological studies
Sections of 5 μm thick from rats’ groups treated with extract of Esraa 1, Esraa 2, Esraa 3, Esraa 4 and Esraa 5, individually as well as animals control were cut and stained with hematoxylin and eosin (H & E) and Masson’s trichrome for connective tissue, collagen, assessment [32]. The collagen fibers percentage was measured by image analyzer system.
Micronucleus test and sperm morphology deformity
In all animal groups, after the last ingestion of each microbial extract, the epiphyses were cut and the bone marrow was flushed out by gentle flushing and aspiration with fetal calf serum [33]. Counting the polychromatic erythrocytes and the ratio of micro-nucleated polychromatic erythrocytes (MNPCEs) were established by observing the MN cells numbers from 1000 PCEs per rat [34]. The epididymides were excised and minced in 2.2% isotonic sodium citrate solution. Smears were equipped and sperms were stained with Eosin Y [35, 36]. Thousand sperm/animal were evaluated for morphological deformities.
Preparation of cell lines and The MTT cell proliferation assay and IC50 determination
To study the cytotoxic effects of these microbial extracts, their anti-proliferation activities against three different types of human non-tumorigenic cells including mammary epithelial (MCF10A), lung fibroblasts (WI38) and dermal fibroblasts (HDFs) cell lines (American Type Culture Collection; ATCC) were evaluated. MCF10A were preserved in MEBM basal medium added with the MEGM SingleQuot Kit (Lonza Corporation, USA) containing 100 ng/mL cholera toxin (Sigma-Aldrich), WI38 were cultured in Eagle's minimum essential medium ((EMEM, ATCC) complemented with 2 mM glutamine and 1% non-essential amino acids (NEAA) and HDFs were cultured in Dulbecco’s modified Eagle’s medium (DMEM). All media were accompanied with 10% fetal bovine serum (FBS), and 1% penicillin/streptomycin antibiotics (Gibco, Grand Island, NY, USA), and cultured at 37 °C in a humidified atmosphere with 5% CO2. At confluence 85%, the cells were trypsinized by trypsin–EDTA (Sigma-Aldrich), then MCF10A, HDFs and WI38 cells were seeded in 96-well plates (at a density of 2 × 104 cells/mL) and exposed to various concentrations (0, 10, 20, 30, 40, 50, 60, 70, 80, 90, and 100 µg/mL) of each bacterial extract individually and trial control with cells only was also involved. After incubation for 48 h, the viability of cells and the half maximal inhibitory concentration (IC50) values were assessed. To measure the rate of cell proliferation, the MTT (3-(4, 5-dimethylthiazolyl-2)−2, 5-diphenyltetrazolium bromide) assay based on the yellow compound MTT-dependent was applied. After the handling time, cells were allowed to react with MTT for 3–4 h in the dark at 37 °C and then dark purple formazan crystals were dissolved and the absorbance was measured spectrophotometry at 595 nm. The cell viability % = [(Optical density {OD} of treated cell − OD of blank) / (OD of vehicle control − OD of blank) × 100] was calculated [37].
Statistical analysis and data reconstruction
The achieved results were statistically analyzed using one-way analysis of variance (ANOVA) followed by post hoc (Duncan) test at p ≤ 0.05. T-test was used for cytogenetic analysis.
Results
Microbial spectrum of G-negative bacteria isolated from cancer microbiome
In the current study, G-negative bacteria were the most prevalent bacteria in the obtained microbiome with the rate of 74.87% (146/195 isolates, Table 1). Furthermore, data in Table 1 showed that the general ranking of the most common pathogens among G-negative microbiota of oncology patients was Esherichia sp. (n = 36, 24.66%) > Acinetobacter sp. (n = 34, 23.29%) > Stenotrophomonas sp. (n = 29, 19.86%) > Pseudomonas sp. (n = 26, 17.81%) > Serratia sp. (n = 21, 14.38%).
The prevalence of antibiotics resistance to G-negative bacteria obtained from cancer microbiome
The antibiotic resistance prevalence analysis in Table 1 showed that the obtained G-negative bacteria derived from the cancer microbiome exhibited entirely multidrug resistance to penicillin antibiotics including ampicillin, ticarcillin, oxacillin, piperacillin and carbenicillin (MIC; 128—> 1024, 560—> 1024, 256—> 1024, 512—> 1024 and 440—> 1024 μg/mL), respectively as well as the first-generation cephalosporins including cephalothin, cefazolin and cefoxitin (MIC; 256—> 1024, 360—> 1024 and 480—1024 μg/mL), respectively. Moreover, resistance prevalence to other cephams including ceftazidime, cefepime, ceftriaxone and cefotaxime reached 100% in Stenotrophomonas and Acinetobacter isolates with MIC ranged from 128 to ˃ 1024 μg/mL (Table 1). Additionally, resistance rate equal to 96.15, 76.92, 65.39 and 57.69% with MIC 400–664, 256–512, 128–512 and 64–512 μg/mL, respectively was recorded in Pseudomonas isolates (n = 26) compared to 95.24, 76.19, 66.67 and 61.91% with MIC 512–720, 256–640, 128–560 and 64–512 μg/mL, respectively in Serratia sp. isolates (n = 21 isolates) while 88.89, 80.56, 63.89 and 63.89% of Esherichia isolates (n = 36 isolates) were resist to ceftazidime, cefepime, ceftriaxone and cefotaxime with MIC 256–512, 128–800, 64–480, and 128–256 μg/mL, respectively (Table 1). Then the cancer microbiota under study can be considered as extended-spectrum β-lactamase producers (ESBL).
The highest resistance rates toward imipenem (73.08% with MIC; 128–512 μg/mL) and doripenem (76.92% with MIC; 264–472 μg/mL) was recorded among Pseudomonas species but the highest resistant against meropenem (75.86% with MIC; 256–600 μg/mL) and ertapenem (79.41% with MIC; 400–648 μg/mL) were occurred among Stenotrophomonas and Acinetobacter species, respectively (Table 1). On the other hand, when ceftazidime, imipenem and meropenem were used together with the β-lactamase inhibitors avibactam, cilastatin and vaborbactam, respectively the rates of resistance in the total number of G-negative bacteria decreased significantly from 95.89, 62.33 and 68.49% to 39.73, 25.34 and 29.45%, respectively (Table 1).
Moreover, data in Table 1 showed that the rates of resistance toward fluoroquinolones antibiotics include ofloxacin, ciprofloxacin levofloxacin and moxifloxacin as well as trimethoprim/sulfamethoxazole (folate synthesis pathway inhibitors) were ≥ 61.91% ≤ 80.95%; ≥ 41.67% ≤ 75.00%; ≥ 69.23% ≤ 80.77%; ≥ 75.86% ≤ 93.10%, and ≥ 73.53% to 100.00% with Serratia sp., Esherichia sp., Pseudomonas sp., Stenotrophomonas sp. and Acinetobacter sp., respectively. Because aminoglycosides remain, a significant class of agents when microorganisms of interest are resistant to other classes, especially β-lactams and fluoroquinolones as in the current study, resistance to aminoglycosides among G-negative microbiome under study were evaluated (Table 1). High resistance potency was recorded against nitrofurantoin (n = 93, 63.70%) followed by tobramycin (n = 90, 61.64%), netilmicin (n = 85, 58.22%), gentamicin (n = 82, 56.16%), amikacin (n = 69, 47.26%) and vancomycin (n = 57, 39.04%) (Table 1). Therefore, the current spread of resistance to aminoglycosides amongst already MDR microorganisms is an unwelcome event. The data in Table 1 indicated that the cancer microbiome resistance rates under study were estimated to be 63.70, 42.47 and 34.93% with MIC; 240–1000, 128–500, and 32–280 μg/mL against the tetracycline antibiotics (doxycycline, minocycline and tigecycline), respectively but 79.45, 74.66 and 65.07% of G-negative isolates were resistant to clarithromycin, zithromycin (macrolides) and chloramphenicol, respectively with MIC ranged between 128 and > 1024 μg/mL. Of the results presented in Table 1, the highest sensitivity profile was recorded against colistin followed by imipenem-cilastatin, meropenem-vaborbactam, tigecycline, vancomycin and ceftazidime-avibactam (78.77, 74.66, 70.55, 65.07, 60.96 and 60.27%), respectively (Table 1). The isolates Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4 and Pseudomonas sp. Esraa 5, which showed resistance against all the antibiotics under study and then characterized extensive drug-resistant (XDR) strains with the highest MIC values, were selected for the further studies.
Carcinogenic activities of bacterial extracts
After oral ingestion of microbial extracts, under study by the five animals’ groups, the values of serum cancer markers (AFP, CEA, and LDH) increased significantly in all treated animals’ groups comprised to the healthy animals’ group (Table 2). Serum AFP levels was significantly increased from 0.52 ± 0.019 ng/mL (healthy group) to 2.08 ± 0.07 ng/mL (300%) after treating with Acinetobacter sp. Esraa 3 followed by Pseudomonas sp. Esraa 5, Stenotrophomonas sp. Esraa 2, Escherichia sp. Esraa 4, and Serratia sp. Esraa 1 extracts (1.92 ± 0.41; 269.2%, 0.85 ± 0.04; 63.5%, 0.73 ± 0.04; 40.4%, and 0.57 ± 0.02 ng/mL; 9.6%), respectively (Table 2). Furthermore, serum CEA level increased in descending order in animals’ groups handled by extracts of Pseudomonas sp. Esraa 5, Stenotrophomonas sp. Esraa 2, Escherichia sp. Esraa 4, Acinetobacter sp. Esraa 3, and Serratia sp. Esraa 1 (07.69 ± 0.71; 311.2%, 6.08 ± 0.13; 225.1%, 4.41 ± 0.79; 135.8%, 3.53 ± 0.07; 88.8%, and 1.99 ± 0.14 ng/mL; 6.4%), respectively compared with the control group (1.87 ± 0.04 ng/mL), indicating the carcinogenic activity of the studied G-negative bacteria (Table 2). Moreover, activity of serum lactate dehydrogenase (LDH) significantly increased to 2248 ± 81.1, 1851 ± 73.4, 789 ± 7.49, 623 ± 18.5, and 402 ± 11.6 IU/L (464.8, 365.1, 98.2, 56.5, and 1.1%) in rats after oral ingestion of the extracts of strains Esraa 3, Esraa 5, Esraa 2, Esraa 4, and Esraa 1, respectively comprised with 398 ± 3.74 IU/L in healthy animals’ group (Table 2). For instant, the higher carcinogenic activity with Escherichia strain was achieved by subcutaneous-injection while Serratia sp. Esraa 1 or Pseudomonas sp. Esraa 5 extracts markedly increased serum level of CEA in the case of oral ingestion. However, there is no significant difference in the effect of Stenotrophomonas sp. Esraa 2 extract whether administered orally or by injection while Acinetobacter sp. Esraa 3 extract increased serum LDH activity by 171.5 and 98.2% post subcutaneous-injection and oral administration, respectively.
Hepatotoxic effect of bacterial extracts
Oral ingestion of the microbial extracts in treated animals stimulated liver dysfunction that was monitored through increased activities of liver enzymes. ALAT activity markedly increased from 23.1 ± 1.8 U/L (control group) to 27.1 ± 1.98, 85.3 ± 6.56, 27.7 ± 2.06, and 120.6 ± 3.2 U/L (17.3, 269.3, 19.9, and 422.1%) in the animal groups ingested with the extracts of strains Esraa 2, Esraa 3, Esraa 4, and Esraa 5, respectively; while it decreased by 7.8% post-handling with the extract of strain Esraa 1 (Table 3). Similarly, serum ASAT activity increased from 22.3 ± 0.3.6 U/L (control group) to 25.2 ± 4.1, 29.0 ± 3.8, 78.6 ± 9.6, 25.4 ± 4.12, and 101.7 ± 3.13 U/L (13.0, 30.0, 252.5, 13.9, and 356.1%); GGT increased from 45.6 ± 2.83 U/L (control group) to 52.1 ± 1.44, 64.8 ± 2.84, 142.1 ± 3.8, 48.7 ± 1.38, and 102.7 ± 13.19 U/L (14.3, 42.1, 211.6, 6.8, and 125.2%); as well as serum ALAP activity increased from 80.7 ± 4.38 (control group) to 93.9 ± 9.3, 138.3 ± 6.9, 375.4 ± 10.5, 89.7 ± 1.83, and 256.4 ± 7.43 U/L (16.4, 71.4, 365.2, 11.2, and 217.7%) after the oral ingestion of the extracts of Esraa 1, Esraa 2, Esraa 3, Esraa 4, and Esraa 5, respectively (Table 3). These results referring to sever hepatotoxic potential of these extracts; the highest hepatotoxic effect was exhibited by Esraa 3, and Esraa 5 (Table 3).
Nephrotoxic effect of bacterial extracts
The extracts of cancer microbiota exhibited nephrotoxic effects on the kidneys, which were achieved through a respectable increase in the concentricity of urea and creatinine in the blood compared to normal rats. Esraa 3 extract performed the most nephrotoxic properties as it increased the serum levels of urea and creatinine to 110.4 ± 3.95 (253.9%), and 1.78 ± 0.42 mg/dl (114.5%), respectively; while the extracts of Esraa 1, and Esraa 2 exhibited the smallest effect as they performed the minimal increase in serum urea (34.7 ± 3.25 mg/dl, 11.2%) and creatinine (0.84 ± 0.07 mg/dl, 1.2%), respectively (Table 4). This study revealed that these tumors-associated microbial communities have some sort of influence on tumor development. Therefore, this correlation can be considered as demonstrated that it can be defined as an oncobiome.
Effect of the bacterial extracts on the inflammatory cytokines
Inflammatory cytokines (IL-1β and TNF-α) levels were markedly increased post-oral ingestion of each bacterial extracts under study. Pseudomonas sp. Esraa 5 extract induced the highest production of IL-1β followed by Esraa 2, Esraa 3, Esraa 4, and Esraa 1 extracts (30.2 ± 2.41; 297.4%, 24.5 ± 2.18; 222.4%, 18.6 ± 1.89; 144.7%, 15.3 ± 1.08; 101.3%, and 7.8 ± 1.44 ng/L; 2.6%), respectively compared to 7.6 ± 1.65 ng/L of control group (Table 4). Moreover, TNF-α level was increased by (11.08 ± 0.94; 6.5%, 29.8 ± 1.71; 186.5%, 27.5 ± 0.65; 164.4%, 19.1 ± 2.44; 83.7%, and 41.9 ± 5.32 ng/mL; 302.9%) after oral administration of Esraa 1, Esraa 2, Esraa 3, Esraa 4, and Esraa 5 extracts, respectively compared to 10.4 ± 0.24 ng/mL of control group (Table 4).
Histopathological studies
The histopathological studies of the liver sections of untreated animals (group 1), and oral ingestion treated groups with Esraa 1 (group 2), Esraa 2 (group 3), Esraa 3 (group 4), Esraa 4 (group 5) and Esraa 5 (group 6) extracts, stained by Masson trichrom to examine the alterations in collagen and fibrous tissues are illustrated in Figs. 1 and 2. Liver section of the control group in Figs. 1 and 2 (section 1) showed branching cords of hepatocytes with vesiculated nuclei and intervening blood sinusoids lined with endothelial cells, hepatocytes as polygonal cells with acidophilic granular cytoplasm around the portal tract. The control group has the minimum amount of collagen or fibrous tissues compared to the collagen and fibrous tissues in all other orally treated groups. The hepatic sections of animals group 2 treated with the extract of Esraa 1 exhibited disrupted arrangement of hepatocytes around the portal tract (red arrow) together with severe cytoplasmic vacuolation and pyknotic nuclei (black arrow) but hepatocytes around the central vein are organized and nearly normal (blue arrow, Fig. 1; section 1P and 2; section 2). Treated animals by extract of Esraa 2 (group 3) exhibited disrupted arrangement of hepatocytes around the portal tract (pt) with vacuolization and pyknotic nuclei (arrow) but hepatocytes around the central vein are organized and nearly normal (inset) along with marked increase in connective tissue in liver capsule (right inset) were observed (Fig. 1; section 2P and 2; section 3). Radiating hepatocytes from the central vein, arranged, nearly normal and detached by blood sinusoid in addition to some inflammatory cells around the portal tract along with marked increase in connective tissue in liver capsule were observed in the liver sections of animals group 4 orally treated with Esraa 3 extract, (inset, Fig. 1; section 3P and 2; section 4). Data in Fig. 1 (section 4P) and 2 (section 5) illustrated the liver of animals group 5 with vacuolar and pyknosis in hepatocytes around the dilated and congested portal tract area with marked increase in normal arrangement of hepatic cells around the central vein (right inset) in connective tissue of liver capsule (left inset) after oral ingestion of Escherichia sp. Esraa 4 extract. Furthermore, the liver sections of the treated group with Esraa 5 extract (group 6) exhibited many hepatocytes with eosinophilic cytoplasm, pyknotic nuclei, massive increase in inflammatory cells around the dilated portal tract and central vein (left inset) and marked increase in connective tissue in liver capsule (right inset, Figs. 1; section 5P and 2; section 6).
A section in liver of control and oral treated animals; (section 1; control) group 1 (Hx. & E. × 400), (section 1P) group 2 treated with Serratia sp. Esraa 1 extract (Hx. & E. × 400), (section 2P) group 3 oral treated with Stenotrophomonas sp. Esraa 2 extract (Inset × 400) (Hx. & E. × 400), (section 3P) group 4 treated with Acinetobacter sp. Esraa 3 extract (Hx. & E. × 300), (section 4P) group 5 treated with extract of Escherichia sp. Esraa 4 (Hx. & E. × 300), and (section 5P) group 6 oral treated with extract of Pseudomonas sp. Esraa 5 (Hx. & E. × 200)
Photomicrographs of liver sections of 1; untreated and 2 −6; oral treated animal groups with extracts of Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4 and Pseudomonas sp. Esraa 5 stained with Masson trichrom (Masson's trichrom reaction × 200), respectively
Frequency of micronuclei (MN) induced by the selected bacterial extracts in male rats
Results in Table 5 and Fig. 3a and b indicated that the ratio of micronuclei in poly chromatic erythrocytes from rat bone-marrow cells was statistically significant (p < 0.01) in all treated animals’ groups by oral ingestion compared to negative control. The numbers of MN cells in animals group that took orally the extract of Escherihia sp. Esraa 4 were markedly increased 10.32-fold (n = 392) followed by the animals’ groups that were administered orally extracts of Esraa 2, Esraa 5, Esraa 1, and Esraa 3 (8.45-fold; n = 321, 7.82-fold; n = 297, 7.79-fold; n = 296, and 5.97-fold; n = 227), respectively when compared to n = 38 in the untreated group (Table 5). Moreover, compared to untreated group (0.76 ± 0.48) the ratio of micro-nucleated polychromatic erythrocytes (MNPCEs) that specified by analyzing the number of MN cells from 1000 PCEs per animal was described to be 5.92 ± 0.58, 6.42 ± 0.50, 4.54 ± 0.63, 7.84 ± 0.68, and 5.94 ± 0.69 after oral ingestion by Esraa 1, Esraa 2, Esraa 3, Esraa 4, and Esraa 5 extracts in the current study (Table 5).
Sperm-shape abnormalities
Various forms of head abnormalities as straight (n = , 34, 47, 40, 52, and 57), banana (n = 153, 161, 148, 157, and 191), amorphous (n = 41, 45, 37, 63, and 47) and without hook (n = 53, 39, 48, 61, and 31) in addition to the coiled tail (n = 20, 22, 18, 35, and 25) were discovered in groups treated by Esraa 1, Esraa 2, Esraa 3, Esraa 4, and Esraa 5 extracts, respectively compared to 27, 37, 3, 18, and 0, respectively in the untreated animals (Table 5 and Fig. 4a - d). The highest sperm abnormalities number in all groups was recorded for banana shape but the lowest was detected for coiled tail, that accompanied by male sterility (Table 5).
Assessment of anti-proliferative activities of the selected bacterial extracts against human normal cell lines
Anti-proliferative activities of Esraa 1, Esraa 2, Esraa 3, Esraa 4, and Esraa 5 extracts individually against three different types of human normal cell lines including mammary epithelial (MCF10A), lung fibroblasts (WI38), and dermal fibroblasts (HDFs) are presented in Fig. 5. Total inhibition in the proliferation of WI38, MCF10A, and HDFs, when they were treated by the Serratia sp. Esraa 1 extract at concentrations of 60, 70, and 70 µg/mL and IC50 equal to 22.50, 26.80, and 32.28 µg/mL, respectively (Fig. 5). Stimulation of 100% death in WI38, MCF10A, and HDFs cells was noticed when they treated with Esraa 2 extract at doses of 50, 40, and 50 µg/mL with IC50 equal to 20.1, 12.80, and 11.20 µg/mL. Moreover, Escherichia sp. Esraa 4 extract at concentrations ranged between 70, and 80 µg/mL entirely inhibited the proliferation of WI38, MCF10A, and HDFs with IC50 ranged from 30 to 40 µg/mL, while their growth was totally inhibited at 40 µg/mL of Pseudomonas sp. Esraa 5 extract with 50% inhibition in their proliferation (IC50) achieved at doses of 20.0, 15.38, and 10.0 µg/mL, respectively. Interestingly, the extracted metabolites of Acinetobacter sp. Esraa 3 showed the highest cytotoxic effects against all human normal cells under study. Esraa 3 extract at a concentration of 30 µg/mL caused total inhibition in the growth of WI38, MCF10A, and HDFs cell lines with IC50 estimated to be 9.5, 14.12, 8.90 µg/mL, respectively (Fig. 5).
Discussion
Continuous monitoring of multidrug-resistant microorganisms as well as drug resistance patterns is needed to avoid treatment failure and decrease selective pressure [38]. Gram negative bacteria dominated the microbiome in this study, representing 74.87% of isolates. The most common pathogens among oncology patients were Escherichia sp. (24.66%) > Acinetobacter sp. (23.29%) > Stenotrophomonas sp. (19.86%) > Pseudomonas sp. (17.81%) > Serratia sp. (14.38%). Previous investigations in developed countries indicated a shift in the causative microorganisms to G-positive bacteria, but G-negative bacteria remain the dominant causative pathogen amongst developing countries [39]. Montazeri et al., described that G-negative bacteria were the most prevalent bacteria isolated from cancer patients (74%) and all of them were multidrug resistant strains [27] as well as Bhat et al., reported that G-negative bacteria constituted 66.96% of the strains, which is related to our findings of 74.87%, then the current study focused on the G-negative bacteria [13]. In the study of Siddaiahgari et al., in Mumbai the most common pathogens among microbiota of oncology patients were Pseudomonas sp. (26.2%) > Enterococcus sp. (11.66%) > E. coli (11.34%) > Klebsiella sp. (10.59%) > Acinetobacter sp. (9.95%) > Enterobacter sp. (3.1%) > Burkholderia sp. (2.35%) [40]. The rank order in the study of Bhat et al., was Klebsiella sp. (18.30%) > Pseudomonas sp. (17.65%) > Escherichia coli (14.71%) > Acinetobacter sp. (6.21%) > Enterococcus sp. (3.92%) > Proteus sp. (2.61%) > Haemophilus sp. (1.96%) [13]. Furthermore, Jungrungrueng et al., described the pattern of causative microorganisms in oncology patients and they found that of 192 isolates; the most common bacteria were E. coli (38/154, 24.7%), K. pneumoniae (19/154, 12.3%), and A. baumannii (14/154, 9.1%) [18] as well as Kabanangi et al., reported that the most obtained G-negative bacterium was P. aeruginosa (39.0%), followed by Acinetobacter sp. (28.7%) and Klebsiella sp. (16.2%) as well as among them 80.1% were MDRGN strains [41]. These data indicated that the structure of cancer microbiome varies greatly from region to region, and then continuous monitoring of bacterial pattern in the cancer microbiome is critical.
It is complicated to treat the G-negative microbiome in cancer patients due to the uncontrolled and continuous use of a large number of antibiotic therapies for a prolonged period which has resulted in selective pressure on bacteria, thus increasing the prevalence of antibiotics resistance [14]. In line with our data, Kabanangi et al., reported that 78.6% of Enterobacteriaceae are extended spectrum producers of β-lactamase and amongst them 100% of Klebsiella sp. and E. coli were MDR [41]. Interestingly, resistance against carbapenems including ertapenem, imipenem, meropenem and doripenem was detected in 66.44, 62.33, 68.49, and 59.59% of G-negative bacteria, respectively. These findings raise the alarm because when G-negative microbiome isolated from cancer patients are resistant to third-generation cephalosporines or/and carbapenems treatment options become limited or even result in treatment failure [42]. Smith et al., mentioned that combine β-lactamase inhibitors with antibiotics as ceftazidime-avibactam, imipenem-cilastatin and meropenem-vaborbactam have high potency against isolates carried carbapenem, multidrug, colistin and tigecycline; resistance mechanisms and inhibited activity of Enterobacterales, MDR and XDR isolates by 99.1, 96.5, and 82% of respectively, which are much higher than the current study [43]. Doi et al., reported that aminoglycoside resistance may occur via enzymatic modification mediated by aminoglycoside acetyltransferases, nucleotidyltransferases or phosphotransferases and appears to converge with the carbapenemase epidemic, facilitating the emergence of XDR and in some cases, pan-drug resistant microorganisms [44]. Colistin has regained worldwide interest due to the high prevalence of multidrug resistance [38]. Nevertheless, the incidence of G-negative MDR-resistant microorganisms as P. aeruginosa, A. baumannii, Serratia sp., Escherichia coli, Salmonella sp., and Klebsiella sp. that considered a serious problem due to the lack of alternative antibiotics in cancer patient is mutable depending on the stating country and continent coupled with inadequate empirical/therapeutic antibiotics therapy exposes these patients to increased risk of adverse outcome [12, 45, 46] as well as necessitates more research and treatment development as previously were reported [14, 21, 45,46,47,48,49,50].
Numerous trials have been prepared on the etiology of tumor and most of them have been assessed genetically or environmentally; however, the prospective role of bacterial strains in expanding of adenocarcinoma has not been discussed [51]. These findings are consistent with Al-Hilu and Al-Shujairi, who reported that certain bacteria are associated with stimulation of a specific kind of tumor through various molecular and biochemical mechanisms; among them H. pylori caused gastric cancer through its capability to cause intense inflammations, S. typhi associated with gallbladder carcinoma, C. pneumoniae is one of the ethological reasons of lung cancer but Klebsiella sp., as well as Proteus mirabilis caused bladder cancer [16]. New studies show that the spread of tumor may be exceedingly linked to the microbiome [52]. In harmony with current findings, EL-Gendy et al., indicated that the subcutaneous-injection of Pseudomonas sp. extract significantly increased CEA and AFP in hepatic tissue and LDH in rats’ serum; however, MRSA strains significantly increased various tumor markers including CEA, AFP and LDH [21, 22]. Our previous results dealing with the risks of subcutaneous injection of bacterial extracts into animals [14] and the current work results representative the harmful effects of their oral administration suggested that the risk of each bacterial extract on animals’ groups is affected by the route of exposure.
Some microbial metabolites are carcinogens that undergo metabolic activation to form metabolites that react with cellular macromolecules and initiate carcinogenesis via DNA damage [8, 9]. Our data supported the previous results of El-Gendy et al., they described that the extracts of different Pseudomonas and S. aureus strains prompted a considerable increase in the serum activities of ASAT, ALAT and ALAP because of hepatic harm and cell necrosis of several tissues [21, 22]. EL-Gendy et al., reported that the levels of urea and creatinine were increased considerably post repeated administration of Pseudomonas sp. extract; this effect was attributed to the deterioration of renal function [22]. Thus, with a best comprehension of how bacterial microbiota contribute to carcinogenesis, new strategies for tumor prevention and treatment by targeting the cancer microbiome can be developed [6, 7]. There is a potential association between the serious clinical syndromes including progression of specific cancers and antibiotic resistant G-negative bacteria in patients with malignancy [14, 21, 53]. Furthermore, current investigations have displayed a causal relationship between bacterial infection and emergence of tumor in organs as kidney, cervix, liver, lungs and colon, which are constantly exposed to bacteria [10]. Microbiota colonization has been associated to G-negative bacteria with high inflammatory prospective and then local inflammation possibly participates in the initiation and continuation of carcinogenesis as seen in ovarian cancer that characterized by oncobiosis in neoplastic diseases [3]. Furthermore, Lenický et al., revealed that the bacterial strains E. coli, P. mirabilis, S. lentus, and Citrobacter braakii in turkey induced inflammatory developments, oxidative stress, structural deterioration increased pro-inflammatory markers levels, such as IL-1, IL-6 and C-reactive protein [17].
El-Gendy et al., stated that Masson’s trichrome stained tissues of non-treated group exhibited few collagen or fibers around the portal tract while the collagen fiber content in portal zone is apparently increased while subcutaneous injected animals with Acinetobacter, Serratia, Escherichia, Stenotrophomonas and Pseudomonas species extracts exhibited disorganization of hepatocytes with vacuolization, ballooning and apoptotic nuclei, inset the pyknotic nuclei appear near from the central vein (abnormal hepatocytes) and fibrous tissues are amplified around the portal abroad tract zone [14]. In a previous study animals treated by pseudomonas extract exhibited intense histopathological changes as derangement of hepatic cords with granular alterations in the cytoplasm, multifocal swelling of hepatocytes with congestion of central and portal blood vessels, focal degenerative, necrotic alterations along with mononuclear cell infiltration, moderate to intense nephropathic alterations, distinct cellular changes, marked degenerative, necrobiotic alterations with inflammatory reaction and tubular changes as diffuse tubular swelling and loss of tubular epithelium [22]. Petridou et al., stated that microscopic examination of buffalo liver sections infected by S. maltophilia showed edema, loss of hepatocytes, fibrosis, capillary thrombosis, thrombi comprised of neutrophils, plasma cells and fibrinous material as well as hemorrhages and foci of coagulative necrosis with infiltrates of neutrophils, macrophages and plasma cells present around the centrilobular vein [54].
These data clearly indicated that oral administration of Esraa 1, Esraa 2, Esraa 3, and Esraa 4 extracts caused higher propagation in the number of micronuclei than their subcutaneous injection which previously reported to be 233, 308, 214 and 341 respectively, while subcutaneous injection of Esraa 5 exhibited higher mutagenicity (MN = 375) in albino rats [14] than its oral ingestion (MN = 297) in the current study. These data supported the severe mutagenicity and genotoxicity of these microbial extracts might be through prompt the construction of DNA cross-links, single- or double-strand breaks that can lead genomic instability and mutations through oncogenes and tumor suppressor genes [6, 7]. It has been found that carcinogenic bacteria gave abroad variation of metabolites, which vary in their chemical configuration but possess a common capability to form high levels of free radicals, inflammations, and chemical bonds with DNA, resulting in the construction of DNA adducts which is known as the first stage in carcinogenesis [16].
Bacterial infection of semen is present recognized global as an important cause contributing to sperm sterility as well as prompted sperm deformities suggesting point mutations in germ cells, presumably causing structural variations in cell organelles, which leads to the occurrence of sperm abnormalities [55]. In parallel with our data, Eini et al., suggested that genital inflammation caused by bacteria can affect the male reproductive system in different ways and bacteria remarkably connected with leukocytospermia could weaken male fertility prospective through lowering sperm concentration, motility, morphology and DNA integrity [56]. Hamazah and Al-Dahmoshi, found that G-negative bacterial strains combine 40/70 (57.1%) were E. coli 30/70 (42.9%) Followed by E. aerogenes 8/70 (11.4%) and Proteus sp. 2/70 (2.9%) as well as bacteria mirobiome among bacteriospermic-pyospermic showed high resistant rates [57]. Alcántar-Curiel et al., reported that up to 58% of carbapenem resistant bacteria including A. baumannii outbreak in HRGIZ strains were capable to adhere to A549 epithelial cells and 14.5% of them caused cytotoxicity of over 50% [58]. Chen et al., revealed the function of gastrointestinal microbiome in occurrence of breast tumor as well as how the intestinal bacterial microbiome, in particular, enteric bacterial genes able of metabolizing estrogens (estroboloma) could influence the risk appearance of “estrogen receptor positive” breast cancer after menopause [59]. Furthermore, Banerjee et al., described that dysbiosis of the microbiota have been linked with pathology containing tumor and suggest a robust and specific microbiota associated with ovarian cancer [60].
Conclusion
In conclusion, regular monitoring of susceptibility pattern of G-negative of cancer microbiome in cancer patients to various antibiotics commonly used in routine chemotherapies is critical for antibiotic policy development to manage multidrug-resistant (MDR) pathogens in cancer microbiota and reduce morbidity and mortality. Therefore, in the present work, we highlight and describe the monitoring of antimicrobial resistance among clinical G-negative bacteria obtained from tumor patients. There was high ratio of drug-resistant G-negative bacterial in the oncology microbiome under study. Among them the selected isolates Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4, and Pseudomonas sp. Esraa 5 showed resistant against all antibiotics classes under study and then they were characterized as extensively drug resistance strains (XDR). The oncobiome and the most likely bacterial metabolites play vital roles in mediating the initiation and propagation of cancer as well as the effectiveness of chemotherapy. To advance our understanding of the potential primary or co-factorial possible roles of this bacterial microbiome derived from cancer patients; the trials based on oral ingestion of extracts derived from the G-negative bacteria Serratia sp. Esraa 1, Stenotrophomonas sp. Esraa 2, Acinetobacter sp. Esraa 3, Escherichia sp. Esraa 4, and Pseudomonas sp. Esraa 5 were performed by albino rats individually. The current study proved that these bacterial strains isolated from the cancer microbiome have potential roles in the induction of cancer, inflammation, mutagenesis, hepatotoxicity, nephrotoxicity and sperm abnormalities along with histopathological changes in the treated animal groups by orally administrated extracts in compared to the untreated group. Moreover, we assessed the cytotoxicity of their extracts on three different human cell lines WI38, MCF10A, and HDFs and they exhibited marked anti-proliferative activities against these human cell lines.
Data availability
No datasets were generated or analysed during the current study.
References
Cojocaru M. Breast cancer and the microbiome. J Clini Sexol. 2021;4(1):11–8.
Pourali G, Kazemi D, Chadeganipour AS, Arastonejad M, Kashani SN, Pourali R, Maftooh M, Akbarzade H, Fiuji H, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Khazaei M, Avan A. Microbiome as a biomarker and therapeutic target in pancreatic cancer. BMC Microbiol. 2024;24:16. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12866-023-03166-4.
Sipos A, Ujlaki G, Mikó E, Maka E, Szabo J, Uray K, Krasznai Z, Bai P. The role of the microbiome in ovarian cancer: mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med. 2021;27:33. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-021-00295-2.
Ge Y, Wang X, Guo Y, Yan J, Abuduwaili A, Aximujiang K, Yan J, Wu M. Gut microbiota influence tumor development and alter interactions with the human immune system. J Exp Clin Cancer Res. 2021;40:42.
Yuan Y, Chen Y, Yao F, Zeng M, Xie Q, Shafiq M, Noman SM, Jiao X. Microbiomes and resistomes in biopsy tissue and intestinal lavage fluid of colorectal cancer. Front Cell Dev Biol. 2021;9:736994. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2021.736994.
Baba Y, Iwatsuki M, Naoya Y, Masayuki W, Baba H. Review of the gut microbiome and esophageal cancer: pathogenesis and potential clinical implications. Ann Gastroenterol Surg. 2017;5:1–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ags3.12014.
Borella F, Carosso AR, Cosma S, Preti M, Collemi G, Cassoni P, Bertero L, Benedetto C. Gut microbiota and gynecological cancers: a summary of pathogenetic mechanisms and future directions. ACS Infect Dis. 2021;7(5):987–1009. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsinfecdis.0c00839.
Wang Y, Yang G, You L, Yang J, Feng M, Qiu J, Zhao F, Liu Y, Cao Z, Zheng L, Zhang T, Zhao Y. Role of the microbiome in occurrence, development and treatment of pancreatic cancer. Mol Cancer. 2019;18:173. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-019-1103-2.
Zhou H, Liu J, Shen J, Fang W, Zhang L. Gut microbiota and lung cancer: a Mendelian randomization study. JTO Clin Res Rep. 2020;1(3):100042. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jtocrr.2020.100042.
Vergara D, Simeone P, Damato M, Maffia M, Lanuti P, Trerotola M. The cancer microbiota: EMT and inflammation as shared molecular mechanisms associated with plasticity and progression. J Oncol. 2019;2:16. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2019/1253727.
Murciano-Brea J, Geuna S, Herrera-Rincon C. Gut microbiota and neuroplasticity. Cells. 2021;10(8):2084.
Perdikouri EIA, Arvaniti K, Lathyris D, Apostolidou Kiouti F, Siskou E, Haidich AB, Papandreou C. Infections due to multidrug-resistant bacteria in oncological patients: insights from a five-year epidemiological and clinical analysis. Microorganisms. 2019;7(9):277. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/microorganisms7090277.
Bhat S, Muthunatarajan S, Mulki SS, Bhat KA, Kotian KH. Bacterial infection among cancer patients: analysis of isolates and antibiotic sensitivity pattern. Int J Microbiol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/8883700.
El-Gendy MMAA, Abdel-Wahhab KG, Hassan NS, El-Bondkly EA, Farghaly AA, Ali HF, Ali SA, El-Bondkly AMA. Evaluation of carcinogenic activities and sperm abnormalities of Gram-negative bacterial metabolites isolated from cancer patients after subcutaneous injection in albino rats. Antonie Van Leeuwenhoek. 2021;114:287–302. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10482-021-01522-w.
Banerjee SP, Wei Z, Tian T, Bose D, Shih NNC, Feldman MD, Khoury T, De Michele A, Robertson ES. Prognostic correlations with the microbiome of breast cancer subtypes. Cell Death Dis. 2021;12:831. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-04092-x.
Al-Hilu SA, Al-Shujairi WH. Dual role of bacteria in carcinoma: stimulation and inhibition. Int J Microbiol. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2020/4639761.
Lenický M, Slanina T, Kačániová M, Galovičová L, Petrovičová M, Ďuračka M, Benko F, Kováč J, Tvrdá E. Identification of bacterial profiles and their interactions with selected quality, oxidative, and immunological parameters of Turkey Semen. Animals. 2021;11(6):1771. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ani11061771.
Jungrungrueng T, Anugulruengkitt S, Lauhasurayotin S, Chiengthong K, Poparn H, Sosothikul D, Techavichit P. The pattern of microorganisms and drug susceptibility in pediatric oncologic patients with febrile neutropenia. J Pathogens. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/6692827.
Mukherjee S, De MS, Goel G, Bhattacharyya A, Mallick I, Dabkara D, Bhaumik J, Roy MK, Majumdar PB, Chatterji S, Mukherjee S, Bhattacharya S, Chandy M. Multi-drug resistant (MDR) and extensively drug-resistant (XDR) bacteraemia rates among cancer patients in an oncology hospital in eastern India: an 11-year retrospective observational study. Infect Prev Pract. 2023;5(2):100275. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.infpip.2023.100275.
Garg VK, Seema M, Nishkarsh G, Garg R, Sachidanand B, Vinod K, Gautam H, Kapil A, Bhatnagar S. Microbial and antibiotic susceptibility profile among isolates of clinical samples of cancer patients admitted in the intensive care unit at regional tertiary care cancer center: a retrospective observational study. Indian J Crit Care Med. 2019;23(2):67–72. https://doiorg.publicaciones.saludcastillayleon.es/10.5005/jp-journals-10071-23119.
El-Gendy MMAA, Abdel-Wahhab KG, Mannaa FA, Farghaly AA, El-Bondkly AM. Carcinogenic activities and sperm abnormalities of methicillin resistance Staphylococcus aureus and inhibition of their virulence potentials by Ayamycin. Appl Biochem Biotechnol. 2017;183:833–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12010-017-2467-7.
El-Gendy MMA, Al-Zahrani HAA, Abozinadah NY, EL-Bondkly AMA. In vivo, evaluation of the toxic effect of ethyl acetate extracts of marine antibiotic resistance Pseudomonas species derived from the Red Sea. Appl Biochem Biotechnol. 2018;184(1):323–49. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12010-017-2553-x.
El-Gendy MMA, El-Bondkly AMA, Keera AA, Ali AM. Incidence of methicillin-resistant Staphylococcus aureus (MRSA) in microbial community of cancer patients and evaluation of their resistant pattern. Arab J Sci Eng. 2018;43(1):83–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13369-017-2670-4.
Araoka H, Babaa M, Okadaa C, Abea M, Kimuraa M, Yoneyamaa A. Evaluation of trimethoprim sulfamethoxazole based against Stenotrophomonas maltophilia: in vitro effects and clinical efficacy in cancer patients. Int J Infect Dis. 2017;58:18–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijid.2017.02.020.
Batra U, Goyal P, Jain P, Upadhyay A, Sachdeva N, Agarwal M, Bhurani D, Talwar V, Gupta SK, Doval DC. Epidemiology and resistance pattern of bacterial isolates among cancer patients in a tertiary care oncology center in North India. Indian J Cancer. 2016;53(3):448–51. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/0019-509x.200647.
Fan L, Wang Z, Wang Q, Xiong Z, Xu Y, Li D, Shiwen Z. Increasing rates of Acinetobacter baumannii infection and resistance in an oncology department. J Can Res Ther. 2018;14:68–71. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/jcrt.jcrt_737_17.
Montazeri EA, Khosravi AD, Saki M, Sirous M, Keikhaei B, Mohammadi SS. Prevalence of extended-spectrum beta-lactamase-producing Enterobacteriaceae causing bloodstream infections in cancer patients from Southwest of Iran. Infect Drug Resist. 2020;6(13):1319–26. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/IDR.S254357.
CLSI. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically; approved Standard. 11th ed., 2018; Clinical and Laboratory Standards Institute, Wayne, PA
CLSI. Performance standards for antimicrobial susceptibility testing. 29th informational supplement, 2019; Clinical and Laboratory Standards Institute, Wayne, PA.
EUCAST. Breakpoint tables for interpretation of MICs and zone diameters version 9.0. 2019. http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_9.0_Breakpoint_Tab les.pdf.
FDA. Antibacterial susceptibility test interpretive criteria. 2019. https://www.fda.gov/drugs/development- resources/antibacterial-susceptibility-test-interpretive-criteria.
Drury RA, Wallington EA. Carleton’s histological technique. 5th Edition, 1980; Oxford University Press, New York.
Valette H, Dolle F, Bottlaender M, Hinnen F, Marzin D. Fluro-A-85380 demonstrated no mutagenic properties in vivo rat micronucleus and Ames tests. Nucl Med Boil. 2002;9:849–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0969-8051(02)00331-1.
D’souza UJA, Zain A, Raju S. Genotoxic and cytotoxic effects bone marrow of rats exposed to low dose of paquat via the dermal route. Mutat Res. 2002;581:187–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mrgentox.2004.10.019.
Wyrobek AJ, Bruce WR. The induction of sperm-shape abnormalities in mice and humans. In: Hollaender A, de Serres FJ, (Eds.), Principles and Methods for their Detection, 1978; volume 5, Plenum Press, New York, pp: 257–285.
Wyrobek AJ, Gordon LA, Burkhart JG, Francis MW, Kapp RW, Letz G, Malling HV, Tapham JC, Whorton MD. An evaluation of the mouse sperm morphology test and other sperm tests in non-human mammals: a report of the gene. Mutat Res. 1983;115:1–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0165-1110(83)90014-3.
Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1–2):55–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0022-1759(83)90303-.
Gogry FA, Siddiqui MT, Sultan I, Haq QMR. Current update on intrinsic and acquired colistin resistance mechanisms in bacteria. Front Med. 2021;8:677720. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmed.2021.677720.
Lee JH, Kim SK, Kim SK, Han SB, Lee JW, Lee DG, Chung NG, Cho B, Jeong DC, Kang JH, Kim HK. Increase in antibiotic-resistant gram-negative bacterial infections in febrile neutropenic children. Infect Chemother. 2016;48(3):181–9.
Siddaiahgari S, Manikyam A, Kumar KA, Rauthan A, Ayyar R. Spectrum of systemic bacterial infections during febrile neutropenia in pediatric oncology patients in tertiary care pediatric center. Indian J Cancer. 2014;51(4):403–5. https://doiorg.publicaciones.saludcastillayleon.es/10.4103/0019-509x.175367.
Kabanangi F, Joachim A, Nkuwi EJ, Manyahi J. High level of multidrug-resistant Gram-negative pathogens causing burn wound infections in hospitalized children in Dar es Salaam, Tanzania. Int J Microbiol. 2021;2021(1):1–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/6644185.
Yusuf E, Bax HI, Verkaik NJ, van Westreenen M. An update on eight “new” antibiotics against multidrug-resistant Gram-negative bacteria. J Clin Med. 2021;10(5):1068. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm10051068.
Smith JR, Rybak JM, Claeys KC. Imipenem-cilastatin-relebactam: a novel β-lactam-β-lactamase inhibitor combination for the treatment of multidrug-resistant Gram-negative infections. Pharmacotherapy. 2020;40(4):343–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/phar.2378.
Doi Y, Wachino JI, Arakawa Y. Aminoglycoside resistance: the emergence of acquired 16S ribosomal RNA methyltransferases. Infect Dis Clin North Am. 2016;30(2):523–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.idc.2016.02.011.
El-Gendy MMAA, Mohamed ZK, Hekal NZ, Ali FM, Yousef AEM. Production of bioactive metabolites from different marine endophytic Streptomyces species and testing them against methicillin-resistant Staphylococcus aureus (MRSA) and cancer cell lines. Biotechnologia. 2018;99(1):13–35. https://doiorg.publicaciones.saludcastillayleon.es/10.5114/bta.2018.73559.
El-Gendy MMAA, Yahya SMM, Hamed AR, El-Bondkly AMA. Assessment of the phylogenetic analysis and antimicrobial, antiviral, and anticancer activities of marine endophytic Streptomyces species of the soft coral Sarcophyton convolutum. Int Microbiol. 2022;25:133–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10123-021-00204-x.
Alzahrani NH, El-Bondkly AAM, El-Gendy MMAA, El-Bondkly AM. Enhancement of undecylprodigiosin production from marine endophytic recombinant strain Streptomyces sp. ALAA-R20 through low-cost induction strategy. J Appl Genet. 2021;62(1):165–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13353-020-00597-x.
Awad MF, El-Shenawy FS, El-Gendy MMAA, El-Bondkly EAM. Purification, characterization, and anticancer and antioxidant activities of L-glutaminase from Aspergillus versicolor Faesay4. Int Microbiol. 2021;24(2):169–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10123-020-00156-8.
El-Bondkly AAM, El-Bondkly AAM, El-Gendy MMA, El-Bondkly EAM, Ahmed AM. Biodiversity and biological activity of the fungal microbiota derived from the medicinal plants Salvia aegyptiaca L. and Balanties aegyptiaca L. Biocatal Agri Biotechnol. 2020;28:101720.
El-Gendy MMAA, Yahya SMM, Hamed AR, Soltan MM, El-Bondkly AMA. Phylogenetic analysis and biological evaluation of marine endophytic fungi derived from Red Sea sponge Hyrtios erectus. Appl Biochem Biotechnol. 2018;185(3):755–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12010-017-2679-x.
Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol. 2015;45:17–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eji.201444972.
Kovaleva OV, Romashin D, Zborovskaya IB, Davydov MM, Shogenov MS, Gratchev A. Human lung microbiome on the way to cancer. A review article. J Immunol Res. 2019;2:1394191. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2019/1394191.
Wasfi R, Rasslan F, Hassan SS, Ashour HM, Abd El-Rahman OA. Co-existence of carbapenemase-encoding genes in Acinetobacter baumannii from cancer patients. Infect Dis Ther. 2021;10(1):291–305. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40121-020-00369-4.
Petridou E, Filioussis G, Karavanis E, Kritas SK. Stenotrophomonas maltophilia as a causal agent of pyogranulomatous hepatitis in a buffalo (Bubalus bubalis). J Vet Diagn Invest. 2010;22:772–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/104063871002200522.
Prabha V, Negi S, Chauhan A, Vander H, Rana K, Thaper D. Antifertility effect of sperm agglutinating factor isolated from Serratia marcescens: an in vivo study. J Reproductive Endocrinol & Infert. 2018;3(1):5.
Eini F, Kutenaei MA, Zareei F, Dastjerdi ZS, Shirzeyli MH, Salehi E. Effect of bacterial infection on sperm quality and DNA fragmentation in subfertile men with leukocytospermia. BMC Mol and Cell Biol. 2021;22:42. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12860-021-00380-8.
Hamazah LM, Al-Dahmoshi HOM. Bacterial profile and resistance patterns of bacteriospermia among pyospermic patients in Hilla City, Iraq. Annal RSCB. 2021;25(4):332–42.
Alcántar-Curiel MD, Rosales-Reyes R, Jarillo-Quijada MD, Gayosso-Vázquez C, Fernández-Vázquez JL, Toledano-Tableros JE, Giono-Cerezo S, Garza-Villafuerte P, López-Huerta A, Vences-Vences D, Morfín-Otero R, Rodríguez-Noriega E, López-Álvarez MdR, Espinosa-Sotero MdC, Santos-Preciado JI. Carbapenem-resistant Acinetobacter baumannii in three tertiary care hospitals in Mexico: virulence profiles, innate immune response and clonal dissemination. Front Microbiol. 2019; 10:2116. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2019.02116.
Chen J, Douglass J, Prasath V, Neace M, Atrchian S, Manjili MH, Shokouhi S, Habibi M. The microbiome and breast cancer: a review. Breast Cancer Res Treat. 2019;178(3):493–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10549-019-05407-5.
Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, Alwine JC, Coukos G, Robertson ES. The ovarian cancer oncobiome. Oncotarget. 2017;8(22):36225–45.
Acknowledgements
The authors extend their appreciation to the Deanship of Scientific Research at King Khalid University for funding this work through large group research project under grant number RGP.2/49/45.
Funding
This research was funded by Deanship of Scientific Research at King Khalid University through large group research project under grant number RGP.2/489/45.
Author information
Authors and Affiliations
Contributions
M.M. A. A.El-G., H.A.A., K.G. A.-W., N.S.H., A.A.M.El-B., M.A.-W., A.A.F. and A.M.A.El-B., designed and carried out the work, accomplished literature searches, analyzed data and wrote the manuscript. All the authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
This study was approved by the Ethical Committee of National Research Centre (FWA 00014747), Egypt.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
El-Gendy, M.M.A.A., Alghamdi, H.A., Abdel-Wahhab, K.G. et al. Evaluation of multidrug resistance in the Gram-negative microbiome of cancer patients and the adverse effects of their metabolites on albino rats and epithelial or fibroblasts cell lines. Infect Agents Cancer 20, 2 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13027-024-00634-y
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13027-024-00634-y