Next Article in Journal
Transgenerational Epigenetic DNA Methylation Editing and Human Disease
Previous Article in Journal
The Blocking of Drug Resistance Channels by Selected Hydrophobic Statins in Chemoresistance Human Melanoma
Previous Article in Special Issue
Anthocyanins: Molecular Aspects on Their Neuroprotective Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antimicrobial Potential of Different Isolates of Chaetomium globosum Combined with Liquid Chromatography Tandem Mass Spectrometry Chemical Profiling

1
Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
2
Department of Microbiology, Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Giza 11562, Egypt
3
Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Industries, Sinai University, Al-Arish, North Sinai 45511, Egypt
4
Department of Epidemiology, Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Giza 11562, Egypt
5
Department of Clinical Pathology, Al-Sahel Teaching Hospital, Cairo 11697, Egypt
6
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
7
Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Biomolecules 2023, 13(12), 1683; https://doi.org/10.3390/biom13121683
Submission received: 29 September 2023 / Revised: 28 October 2023 / Accepted: 9 November 2023 / Published: 21 November 2023

Abstract

:
The antimicrobial resistance of pathogenic microorganisms against commercial drugs has become a major problem worldwide. This study is the first of its kind to be carried out in Egypt to produce antimicrobial pharmaceuticals from isolated native taxa of the fungal Chaetomium, followed by a chemical investigation of the existing bioactive metabolites. Here, of the 155 clinical specimens in total, 100 pathogenic microbial isolates were found to be multi-drug resistant (MDR) bacteria. The Chaetomium isolates were recovered from different soil samples, and wild host plants collected from Egypt showed strong inhibitory activity against MDR isolates. Chaetomium isolates displayed broad-spectrum antimicrobial activity against C. albicans, Gram-positive, and Gram-negative bacteria, with inhibition zones of 11.3 to 25.6 mm, 10.4 to 26.0 mm, and 10.5 to 26.5 mm, respectively. As a consecutive result, the minimum inhibitory concentration (MIC) values of Chaetomium isolates ranged from 3.9 to 62.5 µg/mL. Liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) analysis was performed for selected Chaetomium isolates with the most promising antimicrobial potential against MDR bacteria. The LC-MS/MS analysis of Chaetomium species isolated from cultivated soil at Assuit Governate, Upper Egypt (3), and the host plant Zygophyllum album grown in Wadi El-Arbaein, Saint Katherine, South Sinai (5), revealed the presence of alkaloids as the predominant bioactive metabolites. Most detected bioactive metabolites previously displayed antimicrobial activity, confirming the antibacterial potential of selected isolates. Therefore, the Chaetomium isolates recovered from harsh habitats in Egypt are rich sources of antimicrobial metabolites, which will be a possible solution to the multi-drug resistant bacteria tragedy.

1. Introduction

Due to the explosive increase in microorganisms resistant to a number of antibacterial and antifungal compounds over the past decade, managing infections in both community and hospital settings has become more difficult and has grown to be a substantial worldwide worry [1,2]. Clinical routine frequently links infections to resistant microorganisms in hospital settings; however, they can also happen in the community as a result of selective pressure from antibiotic use. Antimicrobial resistance (AMR) is on the rise in part due to the environment’s overuse of antimicrobials [3]. Furthermore, the growth, spread, and persistence of multi-drug-resistant (MDR) bacteria pose an increasing hazard to the health of people, animals, and the environment [4]. MDR bacteria often exhibit resistance to three or more antibiotics. In the end, the discovery of antibiotics has lagged due to the advent and evolution of MDR [4]. Natural products remain a promising source of new efficacious antimicrobials to counter increasing resistance and prevent the emergence of multi-drug resistant bacteria [5]. Novel antimicrobial metabolites from endophytic fungi are now becoming an alternative option to overcome the increasing levels of drug resistance by human pathogens. An outstanding model that is utilized as a biotechnological tool in many disciplines related to bioactive molecules is the genus Chaetomium [6].
Genus Chaetomium is primarily found in soil and organic compost, but some species of Chaetomium have also recently been isolated from coral, soft coral, and marine algae [7]. Chaetomium species are worldwide species that often live on plant debris and are found in soil and the air. Various metabolites were characterized from different taxa of Chaetomium and showed cytotoxicity against a panel of human solid tumour cell lines: NCI-H460 lung cancer, MCF-7 breast cancer, SF-268 brain cancer, and different prostate cancer cell lines (PC-3, LNCaP, and DU-145) as mentioned by [8]. Some members of Chaetomium isolates have been identified to produce a number of antimicrobial compounds with antagonistic mechanisms against other pathogenic fungi [9].
From a chemical perspective, more than 350 species of the genus Chaetomium have been extensively investigated to find new bioactive secondary metabolites with unique structures from mycelium or spores [10,11]. More than 200 secondary metabolites were isolated and identified from Chaetomium globosum, with cytotoxicity, antimicrobial, antimalarial, anticancer, and antiviral actions [12]. Among them, emodins, chrysophanols, chaetoglobosins A–G, isochaetoglobosin, chetomin, azaphilones, chaetoviridins, terpenoids, chaetoglobosins, tetramic acids, steroids, xanthones, diketopiperazines, bis (3-indolyl)-benzoquinones, azaphilones, anthraquinones, pyranones, and orsellides were identified [12,13]. Various studies carried out by various researchers since 1944 on the antibacterial potential of Chaetomium have shown that the Gram-positive and Gram-negative bacteria Escherichia coli and Staphylococcus aureus were particularly susceptible to the antibacterial effects of Chaetomium metabolites [14] Additionally, cochliodones were recovered from C. globosum species, showing antifungal activities in low doses (1–10 g/mL) against various genera of microfungi, including Botrytis allii and Fusarium moniliforme. The polysaccharides produced by Chaetomium globosum CGMCC 6882 recorded anticancer properties against human lung cancer A549 cells [15].
Until now, neither antimicrobial activity nor chemical profiling of Chaetomium species isolated from different ecological habitats in Egypt have been studied. Therefore, this study aimed to assess the antimicrobial potentialities of secondary metabolites of native C. globosum against MDR microbes, followed by the rapid identification of natural products using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) of the most promising taxa.

2. Materials and Methods

2.1. Fungal Isolation and Identification

2.1.1. Collection of Samples

Different samples from cultivated and desert soils were collected from various habitats and locations as described in Table 1.
Samples of the most common medicinal plant species in the Saint Katherine protectorate (SKP), Sinai-Egypt, were collected. The taxa of medicinal plants were Artemisia herba-alba Asso; Achillea fragrantissima (Forssk) Sch.; Ballota undulata (Sieber ex Fresen.) Benth.; Chiliadenus montanus (Vahl) Brullo; Alkanna orientallis (L.) Boiss.; Origanum syriacum L. Peganum harmala L.; Phlomis aurea Decne; Teucrium polium L.; Verbascum sinaiticum Benth.; Thymus decussates Benth.; Tanacetum sinaicum (Fresen.) Delile ex K. Bremer & Humphries; Zygophyllum album; and Adiantum capillus-veneris L. (Table 2). All plant materials were collected for scientific purposes under permission from SKP (#1312-SKP), and no threatened or endangered species were included in this study.
Also, mangal samples of Avicennia marina (Forssk.) Vierh., Ziziphus spinosus Semen, and Adiantum capillus-veneris L. were collected from Safaga, Nuwieba, and Port Fouad, respectively (Table 2).
All collected samples were transported in sterilized polyethylene bags to the lab, where they were later plated out.

2.1.2. Isolation and Preservation of Mycobiota

For the isolation of terricolous taxa of Chaetomium, an alcohol immersion technique was applied according to [16]. Regarding the isolation of endobiotic (endophytic) fungi, the aerial parts (leaf and stem) of each plant were washed in running water, cut into small pieces, and surface-sterilized by dipping in 75% ethanol (EtOH) (v/v) for 1–5 min depending on the plant thickness, and then dipped in 0.05 g/mL sodium hypochlorite (NaOCl) solution (v/v) for 3–5 min, followed by two rinses in sterile distilled water [17].
Different types of isolation media, including malt extract agar (MEA), Czapek’s yeast extract agar (CYA), and potato dextrose agar (PDA) supplemented with rose Bengal (1/15,000) as bacteriostatic and chloramphenicol (50 ppm) as bactericide [18], were used for the isolation of fungi. Plates were plated and then incubated for 10 days at 27 °C.

2.1.3. Phenotypic Identification of Chaetomium Isolates

The following identification keys were utilized to identify the isolated culturable Chaetomium isolates phenotypically down to the species level on standard medium: for Ascomycetes [19] and for Chaetomium [20]. Carl Zeiss amplival microscope (GmbH, Germany) was used to examine the microscopic characteristics. All name corrections, authorities, and taxonomic assignments of recorded species in the present study were checked against the Index Fungorum database (www.indexfungorum.org) (accessed on 4 May 2023), followed by the 10th edition of Ainsworth and Bisby’s Dictionary of the Fungi [21]. Heatmaps were generated using the ggplot2 package in R software Ver. 2023.09.0+463 for MacOS.

2.2. Culture of Chaetomium Isolates and Extraction of Their Active Metabolites

2.2.1. Subcultures of Pure Isolates of C. globosum

A pure culture of Chaetomium isolates was grown on an oatmeal agar medium for 14 days at 27 °C. Forty grams of rice were mixed with 100 mL of distilled water in a 500 mL Erlenmeyer flask (three replicas for each isolate) before being autoclaved [22]. Under sterile conditions, five discs from each isolate were inoculated into each flask by using a cork borer (1 cm in diameter). All flasks were fermented and incubated for 21 days at 27 °C.

2.2.2. Extraction of Active Metabolites

The fermented flasks for each isolate (3 flasks) were homogenized in 300 mL of deionized water (100 mL per flask) using a high-speed blender (Tornado electric blender, 500 Watt, Cairo, Egypt). The resulting mixture was macerated overnight with ethyl acetate in a ratio of 3:1 with 3 repetitions to extract fungal metabolites. The organic layers were then combined and evaporated under reduced pressure at 40 °C to obtain a crude extract.

2.3. Assessment of Antimicrobial Activity of Different Isolates of Chaetomium Isolates

2.3.1. Collection of Clinical Samples and Isolation of Microbes

About 155 clinical specimens of blood, sputum, wound, pus, urine, and cerebrospinal fluid (CSF) were taken from patients suffering from different infections, admitted to El-Sahel Teaching Hospital, the general organization for teaching hospitals and institutes (GOTHI), Cairo, Egypt. Various specimens were taken from patients whose infections were determined to be present based on clinical manifestations. The patients were of both sexes, with ages ranging from 19 to 80 years old. For each patient, clinical and laboratory data were gathered and recorded. All laboratory procedures were carried out in accordance with the Clinical and Laboratory Standards Institute’s (CLSI) guidelines and regulations (Ethics Code: IME00071). The collected specimens were inoculated onto appropriate isolation culture media, McConkey agar, blood agar and nutrient agar (Oxoid Ltd., Co. ®, Nepean, ON, Canada), and then they were incubated at 37 °C. Microbial identification was largely based on colony features and Gram-stain reactions, while final microbial isolate identification was performed using the traditional VITEK 2 compact 15 system (BioMérieux ®, Inc., Hazelwood, MO, USA).

2.3.2. Antibiotics Susceptibility Test for Determination of MDR Isolates

All bacterial isolates were tested for antibiotic susceptibility using the standard Kirby–Bauer disk diffusion technique, as defined by Clinical Laboratory Standard Institute guidelines (2018). The most widely used classes of antibiotics, such as tetracyclines, aminoglycosides, penicillins, cephalosporins, fluoroquinolones, and carbapenems, were employed. These Gram-positive and Gram-negative antibiotics were prepared at the following concentrations: amikacin (30 µg), amoxycillin/clavulanic acid (30 µg), ampicillin/sulbactam (30 µg), cefepime (30 µg), cefotaxime (10 µg), ceftazidime (30 µg), gentamicin (10 µg), imipenem (10 µg), meropenem (10 µg), tigecycline (15 µg), ciprofloxacin (5 µg), tetracycline (30 µg), levofloxacin (5 µg), cefoxitin (30 µg), and doxycycline (5 µg). All antibiotics were purchased from Oxoid ®, Basingstoke, UK. Multi-drug-resistant (MDR) microbes have been described as those that are resistant to at least one antibiotic in at least three antimicrobial classes [23].

2.3.3. Screening of Antimicrobial Activity of Chaetomium Taxa against MDR Isolates

The stock solutions of 10 crude extracts of Chaetomium taxa were reconstituted using 10% dimethyl sulfoxide (DMSO), and the extracts were diluted to a concentration of 50 mg/mL. They were screened for their antibacterial and antifungal activities against 100 pathogenic microbial isolates, including Gram-positive bacterial isolates (Staphylococcus aureus, Enterococcus faecalis, and Streptococcus pyogenes), Gram-negative bacteria isolates (Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniea, Proteus mirabilis, Pseudomonas aeruginosa, and Serratia marcescens), and Candida albicans. About 20 μL/disc solvent extract with concentrations of 50 mg/mL was soaked in each sterile Whatman disc with a diameter of 6 mm and allowed to dry before being placed on the inoculated media. Bacterial strains (108 CFU) were seeded into Mueller–Hinton sterile agar plates, while Candida albicans were seeded into Sabouraud dextrose agar media. As a negative control, dimethyl sulfoxide (DMSO) was used. The zones of growth inhibition surrounding the disks were measured using a calibrated ruler after 48 h for fungi at 28 °C and after 18 to 24 h of incubation at 37 °C for bacteria. The sensitivities of the microorganisms to the fungal extracts were assessed by measuring the sizes of inhibitory zones in millimetres on the agar surface surrounding the disks, and those with no zones were labelled negative. Results are presented as the mean value (±standard deviation). Minimum inhibitory concentrations (MIC) were determined for fungal crude extracts that showed inhibition activity against the test microorganisms.

2.3.4. Determination of Minimum Inhibitory Concentrations (MIC) of Active Isolates

The MIC was obtained with the microdilution technique using a 96-well microtitre plate according to the Clinical Laboratory Standards Institute (CLSI, 2015). A volume of 100 µL of two-fold diluted extracts in Muller–Hinton broth was introduced in the wells of the plate. Thereafter, 100 µL of inoculum standardized at 0.5 Macfarland and extracts were added to each well to a final volume of 200 μL containing the test substances except for the blank column for sterility control. The concentrations of the test substances ranged from 0.5 to 500 µg/mL. For bacterial and fungal pathogens, the plates were incubated at 37 °C for 24 h and 28 °C for 48 h, respectively. Turbidity was considered an indicator of growth, and the MIC was determined as the lowest concentration that inhibits observable bacterial growth.

2.4. Metabolic Profiling of Selected Chaetomium Isolates Using LC-MS/MS

The metabolic profiling and composition were established using (HPLC/triple-TOFMS and MS) [24,25]. The ethyl acetate extracts of selected isolates of C. globosum (3 and 5) were dissolved in water, methanol, and acetonitrile (50:25:25) mixture to afford a solution with a concentration of 0.5 mg/mL. The prepared solution was centrifuged, and then 50 µL was picked up and completed to 1000 µL with water, methanol, and acetonitrile (50:25:25). Ten µL was injected in both positive and negative modes. The LC/Triple-TOF-MS/MS analysis was conducted using an ExionLC system (AB Sciex, Framingham, MA, USA) with an autosampler system, an in-line filter disk precolumn (0.5 µm × 3.0 mm, Phenomenex, Torrance, CA, USA), and an X-select HSS T3 column (2.5 µm, 2.1 × 150 mm, Waters Corporation, Milford, MA, USA) sustained at 40 °C. The mobile phase consisted of 5 mM ammonium format buffer in 1% methanol, with the pH adjusted to 3.0 and 8.0 for positive and negative modes, respectively. The mobile phase was gradually eluted by raising the acetonitrile concentration over 20 min, and then a steady period of 4 min, followed by a reduction in the acetonitrile concentration over 3 min at a constant flow rate of 0.3 mL/min. This compartment was linked to a Triple TOFTM 5600+ system (AB SCIEX, Concord, ON, Canada) to monitor the analytes’ MS/MS transitions. The detected metabolites were recognized by their m/z and MS/MS transitions to those reported in previously documented databases. The mass accuracy was calculated as follows: [measured mass-expected mass/expected mass] × 106 and expressed in parts per million (ppm) error [26,27,28]. Mzmine ID, retention time, adduct formula, and molecular formula were also detected.

3. Results and Discussion

3.1. Fungal Sources and Identification of Chaetomium sp.

As shown in Figure 1, it was possible to encounter as many as 10 isolates of Chaetomium recovered during the entire study from all habitats. All identified taxa were deposited in the Suez Canal University Fungarium at the Botany and Microbiology Department, Faculty of Science (https://ccinfo.wdcm.org/collection/by_id/1180 (accessed on 15 November 2022). Out of the 27 collected samples that were plated out, a total of 10 teleomorphic isolates were morphologically identified as members of Chaetomiaceae (Table 3).
Chaetomium globosum colonies are characterized by a pale or olivaceous aerial mycelium with yellow, greyish green, green, or red exudates. Ascomata mature within 7–9 days (Figure 2A), showing olivaceous, grey-green, or brown colour in reflected light. The colonies are superficial, spherical, ovate, or obovate in shape with a brown intricate ascomatal wall and 2–3.5 μm broad, having ostiolate pores with a 175–280 μm width. The ascomatal hairs are numerous, usually unbranched, flexuous, undulate or coiled, often tapering, septate, brownish, 3–4.5 μm broad at the base, and up to 500 μm long (Figure 2B). Asci are clavate or slightly fusiform, stalked, 30–40 × 11–16 μm, 8-spored, and evanescent. Ascospores are limoniform, usually biapiculate, bilaterally flattened, brownish when mature, thick-walled, and containing numerous droplets (9–12 × 8–10 × 6–8 μm) with an apical germ pore (Figure 2C).

3.2. Antimicrobial Assessment of Chaetomium Isolates

3.2.1. Microbial Isolates and Their Antibiotic Resistance Pattern

Of the 155 clinical specimens in total, 100 pathogenic microbial isolates were obtained and found to be MDR (Table 4). According to [29], Gram-negative isolates were more commonly found than Gram-positive ones: Staphylococcus aureus, Candida albicans, Enterococcus faecalis, and Streptococcus pyogenes.
The antibiotics that are used for repeated empirical treatment might be the reason for the development of high antibiotic resistance. The antibiotic resistance patterns of all isolated and identified strains are shown in Table 5.
S. aureus strains were found to be 100% resistant to amoxycillin/clavulanic acid, ampicillin/sulbactam, cefepime, and cefotaxime; however, about 100% of S. aureus strains were sensitive to cefoxitin. S. aureus is one of the most common multi-drug-resistant bacterial pathogens, causing different infections [30].
Also, E. faecalis strains showed 100% resistance to amikacin, ampicillin/sulbactam, cefepime, cefotaxime, ceftazidime, and cefoxitin, while it showed 100% sensitivity to tigecycline, similar to results found by [31]. The multi-drug resistance of E. faecalis was observed due to its ability to acquire and transfer antibiotic resistance genes [32].
Moreover, 100% of S. pyogenes strains were resistant to amoxycillin/clavulanic acid, ampicillin/sulbactam, cefepime, cefotaxime, ceftazidime, and cefoxitin. On the other hand, tigecycline, imipenem, and meropenem were completely active against 100% of S. pyogenes strains; such results were represented by [33]. In earlier studies, [34] reported that carbapenems are still sensitive to upcoming resistance from both Gram-positive and Gram-negative bacteria.
Similar to the results of the study stated by [35], all A. baumannii strains showed resistance to cefoxitin and doxycycline; 85.7% of the same strain showed resistance to ampicillin/sulbactam, cefepime, ceftazidime, and ciprofloxacin.
E. coli strains were mostly resistant to cefoxitin, amoxycillin/clavulanic acid, ampicillin/sulbactam, ceftazidime, gentamicin, cefotaxime, and ciprofloxacin with a percentage of 100%, 90.9%, 90.9%, 90.9%, 90.9%, 81.8%, and 81.8%, respectively. This was similar to the previous study by [36,37].
K. pneumoniae is one of the WHO’s list of antibiotic-resistant pathogens that necessitate the development of new antibiotics to combat them [38]. It is a well-known nosocomial pathogen that has recently emerged as an MDR and pan-drug-resistant issue [39]. In our study, K. pneumoniea found high antibiotic resistance to amoxycillin/clavulanic acid (100%), ampicillin/sulbactam (100%), levofloxacin (100%), cefotaxime (90%), cefoxitin (90%), ciprofloxacin (80%), tetracycline (80%), amikacin (73.3%), ceftazidime (73.3%), gentamicin (70%), cefepime (70%), doxycycline (60%), and tigecycline (60%), while 70% and 66.7% were susceptible to imipenem and meropenem, respectively, similar results reported by [40].
P. mirabilis shows varying degrees of resistance to antibacterial drugs, with the highest resistance (100%) to cefotaxime and cefoxitin, followed by (80%) resistance to ampicillin/sulbactam, ciprofloxacin, gentamicin, and levofloxacin, while 60% resistance was recorded to amoxycillin/clavulanic acid, cefepime, ceftazidime, doxycycline, and tetracycline; furthermore, 80% of this strain were sensitive to imipenem, meropenem, and tigecycline; these findings are in agreement with a previous study by [41]. P. mirabilis may cause a number of opportunistic and nosocomial infections due to their virulence factors [42].
P. aeruginosa isolates showed susceptibility to meropenem, imipenem, and tigecycline, 66.7%, 88.9%, and 88.9%, respectively. In contrast, the same strain showed 100% resistance to amoxycillin/clavulanic acid, cefepime, cefotaxime and tetracycline, followed by 88.9% resistance to ampicillin/sulbactam, cefoxitin, ciprofloxacin, gentamicin, and levofloxacin. These results are in agreement with those found by [35]. According to [43], P. aeruginosa is basically resistant to many antibiotics and is capable of easily acquiring antibiotic resistance. Furthermore, P. aeruginosa has a high potential to evolve multi-drug resistance phenotypes [44].
Imipenem, meropenem, and tigecycline were completely effective against S. marcescens, while the same strain showed complete resistance to amoxycillin/clavulanic acid, ampicillin/sulbactam, ceftazidime, gentamicin, and levofloxacin. Although S. marcescens was previously considered non-pathogenic, this species has emerged as a prominent opportunistic pathogen found in nosocomial infections [45,46]. S. marcescens, associated with hospital outbreaks or epidemic events, is commonly resistant to several antibiotics.
Here, all the isolated microbes proved their multi-drug resistance against most tested antibiotics.

3.2.2. The Antimicrobial Activity of Chaetomium Isolates against MDR Strains

Because of the emergence and spread of antimicrobial resistance by pathogenic microorganisms to commercial drugs, the current study was carried out to determine the antimicrobial potential of terricolous and endophytic fungi isolated from different habitats. As described in Table 6, all isolates of Chaetomium had significant inhibitory activity against Gram-negative bacteria. At the same time, Chaetomium globosum isolates (3,4,5,6) and Chaetomium madrasense (10) showed a wide spectrum of antimicrobial activity against Gram-positive and Gram-negative pathogenic microorganisms that were isolated from different clinical samples and showed multi-drug resistance ability.
C. globosum isolates demonstrated substantial inhibitory activity against Gram positive bacteria (S. aureus, E. faecalis, and S. pyogenes) with different inhibition zones ranging from 10.4 to 21 mm. Additionally, they offered variable sensitivity against Gram negative bacteria (A. baumannii, E. coli, P. mirabilis, P. aeruginosa, and S. marcescens) with inhibition zones of 11.3 to 26.5 mm.
The maximum ranges of zones of inhibition were provided by C. globosum isolated from cultivated soil in Assiut Governorate (3) and Zygophyllum album from Wadi El-Arbaein (5), as represented in Figure 3 and Figure 4.
In comparison to other taxa, both isolates (3 and 5) displayed the highest inhibition zones against C. albicans (Figure 5).
Obviously, the C. globosum isolated from cultivated soil in Assiut Governorate (3) and from host plant Zygophyllum album (5) showed strong antimicrobial activity against most tested pathogenic microorganisms; our findings were similar to previous studies reported by [15,47]. On the other hand, Chaetomium iranianum (7) recorded no inhibitory activity with S. aureus, E. faecalis and S. pyogenes, A. baumannii, K. pneumonia, P. mirabilis, and C. albicans, but it showed a high inhibitory effect against E. coli, P. aeruginosa, and S. marcescens with inhibition zones of 18.4, 25.2, and 26.0 mm, respectively. C. madrasense (10) was effective against Gram-positive bacteria, and C. albicans, but no inhibition zones were observed with A. baumannii, K. pneumonia and P. mirabilis strains.

3.2.3. Minimum Inhibitory Concentration (MIC) of Fungal Extracts

The minimum inhibitory concentration (MIC) of crude extracts of Chaetomium was found to range from 3.9 to 62.5 µg/mL, depending on the fungal extracts and the tested microorganisms (Table 7).
Crude extracts of Chaetomium showed significant activity against Gram-negative bacteria and Candida albicans, followed by Gram-positive bacteria. Our findings are in agreement with [15,48], but they are lower than those found by [49,50].
Many antimicrobial compounds isolated from endophytic fungi can be used for pharmaceutical, medicinal, and agricultural applications [51]. Examples of these secondary metabolites are chaetocin, ergosterol, and terpenes [52,53]. These variations in susceptibility might be linked to the type of isolates, the nature and concentration of antimicrobial compounds present in their extracts, as well as their mechanism of action on various microorganisms. Regarding the promising antimicrobial activity of the two isolates of Chaetomium (3, 5), they were subjected to further chemical studies.

3.3. LC-MS/MS Metabolic Profiling of Crude Extracts of C. globosum Isolates 3 and 5

For the rapid identification of the bioactive metabolites correlated to the promising antimicrobial activity of isolates of C. globosum (3 and 5), LC-MS/MS analysis was conducted. The physical separation of various metabolites depends on their affinity for the reversed stationary phase. At the same time, their identification is based on the calculation of mass error accuracy and detection of the daughter mass fragments [24,25].

3.3.1. Identification of Natural Metabolites in the Crude Extract of C. globosum Soil Taxon Recovered from Cultivated Soil in Assiut Governorate, Upper Egypt, (3) Using LC-MS/MS Technique

Alkaloids are the major metabolites detected in the C. globosum soil taxon recovered from cultivated soil in Assiut Governorate, as shown in Table 8.
In terms of alkaloids being the major detected metabolites as shown in Figure 6, trigonelline has been shown to have antibacterial, antiviral, hypoglycemic, hypolipidemic, neuroprotective, memory-improving, and anti-tumour activities, in addition to reducing platelet aggregation [73]. Aporphine isoboldine alkaloid was found to have an antifungal effect against Tricophyton rubrum [74]. Coclaurine is an isoquinoline alkaloid that has antiviral properties [75] and cytotoxic effects against human colon cancer (HCT116), human breast cancer (MCF7), and human liver cancer (HEPG-2) cells, with corresponding IC50 values of 8.233, 15.345, and 1.674, respectively [76]. Scoulerine is a benzylisoquinoline alkaloid showing inhibitory activity against acetylcholinesterase (anti-AChE), tumour necrosis factor-alpha (anti-TNF-α), and the bacterial strain Helicobacter pylori [77]. Numerous fungi from the genera Phoma, Helminthosporium, Zygosporium, Metarrhizium, Chaetomium, and Rosellinia produce cytochalasins [78]. Indole alkaloids, known as chaetoglobinol A and B, were detected; they displayed a hypoglycemic effect via the inhibition of α-glycosidase [66]. Chaetoglobinol A has antibacterial properties against Bacillus subtilis [79]. Another azaphilone alkaloid, chaetomugilide A, displayed cytotoxic effects against HepG2 [80] in addition to its antimicrobial property against Pseudomonas putida and Bacillus subtilis at concentrations of less than 20 μM [81]. Penochalasin A showed cytotoxic activity against the human nasopharyngeal epidermoid tumour KB cell line with an IC50 value of 48 μM [82]. Cytochalasin alkaloids can alter cellular shape, prevent cellular activities like cell division, and even trigger apoptosis [83]. Moreover, the antibacterial activity of sclerotiorin against Micrococcus luteus, Bacillus cereus, Bacillus subtilis, Klebisiella pneumoniae, E. coli, Salmonella typhimurium, and L. monocytogenes was reported [84].
Other chemical categories were found. Among them is shikimic acid, which is a source of precursors involved in the biosynthesis of aromatic amino acids and phenylpropanoid metabolites. Its antioxidant, anti-inflammatory, antiviral, neuroprotective, and antibacterial properties have been the subject of several investigations. Zearalenone macrolide has a fourteen-membered lactone ring fused to 1,3-dihydroxybenzene, which displays an estrogen-like action via binding to the estrogenic receptors in the ovaries, mammary glands, uterus, or vagina [85]. Vanillic acid is a phenolic acid that has been employed as a food flavouring agent, preservative, anti-inflammatory, antioxidant, and antihypertensive [86,87]. It also showed antimicrobial, anti-filarial, snake venom antagonist properties in addition to its antibacterial action against foodborne pathogens such as Staphylococcus aureus and E. coli [88,89]. It was previously mentioned that L- proline-rich peptides are prospective medicines to fight multi-drug-resistant microbes [90].
An isocoumarin derivative known as prochaetoviridin A displayed moderate antifungal activity against five pathogenic fungi, including Sclerotinia sclerotiorum, Botrytis cinerea, Fusarium graminearum, Phytophthora capsici, and Fusarium moniliforme, with inhibition rates ranging from 13.7% to 39.0% [61]. Nicotinamide is an effective antimicrobial agent against the human immunodeficiency virus, Mycobacterium tuberculosis, and Plasmodium falciparum [91]. Perylenequinone altertoxin I is a pentacyclic aromatic polyketide and exhibited strong cytotoxicity with LC50 values (concentration causing 50% inhibition) of 6.43 µM [92]. Altertoxin I was found to be a potential inhibitor of the replication of the HIV-1 virus [93]. Due to the potent antiproliferative, antioxidant, antiestrogenic, and/or antiangiogenic properties of secoisolariciresinol lignan, it has been found to prevent various cancers, including breast, lung, and colon cancers [94]. Verrucarin J is one of the trichothecenes or sesquiterpene metabolites containing a tricyclic skeleton and an epoxide group. Without impacting cell viability, macrocyclic trichothecene verrucarin J suppressed the arenavirus Junin, which causes hemorrhagic fever with IC50 values in the range of 1.2–4.9 ng/mL [95]. It is worth noting that several cytotoxic agents have been proven to suppress or delay microorganism growth in vitro [96,97].
Now, it is obvious to deduce that the antibacterial property of the crude extract of C. globosum soil taxon (3) against multi-drug resistant bacteria refers to the antimicrobial activity of the detected metabolites, as indicated above.

3.3.2. Identification of Natural Metabolites in the Crude Extract of C. globosum Endophytic Taxon Recovered from Zygophyllum album (5), Wadi El-Arbaein, Saint Katherine, South Sinai, Using LC-MS/MS Technique

In the same manner, it was found that the majority of identified metabolites were chemically classified as alkaloids in the C. globosum endophytic taxon recovered from Zygophyllum album (5) (Table 9, Figure 7).
In terms of detected alkaloids, trigonelline, coclaurine, N-methyl coclaurine, N,N-dimethyl coclaurine, and demethyl isoboldine are common alkaloids in isolates of C. globosum (3 and 5). Moreover, piperidine has been shown to have antibacterial, antimalarial, anti-inflammatory, analgesic, antioxidant, anti-hypertensive, and antiproliferative actions [109].
Citramalic acid is an analog of malic acid, containing an alpha-hydroxyl di-carboxylic group. It is involved in the biosynthesis of the branched-chain amino acid pathway and also in cosmetics production to reduce skin wrinkles [110]. Quinic acid is an alpha-hydroxyl acid that demonstrated strong antibacterial effects on S. aureus via lysis of the cell membrane and interfering with cellular metabolism [111]. It also acts as a radioprotective, anti-diabetic, anti-neuroinflammatory, neuroprotective, anti-mutagenic, and anti-inflammatory agent [112].
Chaetoviridins have a pyrone–quinone structure and are typically referred to as azaphilones [113]. Chaetoviridin A has antimalarial, antimycobacterial, antifungal, and cytotoxic activities [114]. The growth of S. sclerotiorum, Rhizoctonia solani, Magnaporthe grisea, and Pythium ultimum can be inhibited by chaetoviridin A [61,115].
Several in vitro and in vivo investigations have looked at cinnamaldehyde as a possible substitute for antimicrobial therapy [116]. By inhibiting pathogens such as Candida spp., E. coli, Listeria monocytogenes, Pseudomonas aeruginosa, and Staphylococcus aureus, cinnamaldehyde has demonstrated a broad spectrum of antimicrobial action [117].
In addition to being essential for sustaining human health, recent research revealed that riboflavin can inhibit or inactivate the growth of a variety of microorganisms, including bacteria, viruses, fungi, and parasites. This suggests that riboflavin may have antimicrobial properties [118,119].
Globosumin is an azaphilone, which is a subclass of fungal polyketide metabolites having a highly oxygenated pyranoquinone bicyclic structure. It showed cytotoxic effects against HepG2 and the lung cancer A549 cell line with an IC50 of 6.82 μM, demonstrating its biological activity [103]. At the same time, chaetominedione inhibits p56lck tyrosine kinase with less damage to healthy cells [120].
The dipeptide carnosine possesses functional characteristics that are unique to muscles and excitable tissues. Recent in vivo and in vitro research has demonstrated that carnosine has anti-inflammatory, metal chelating, antioxidant, and free radical scavenging properties [121].
Syringetin is an O-methylated flavonol that has antioxidant and antimicrobial properties. Its broad-spectrum antimicrobial activity was observed against Staphylococcus aureus, Escherichia coli, Proteus vulgaris, Pseudomonas aeruginosa, Candida albicans, and Microsporum canis. It is also able to inhibit the growth of cancer cells via the induction of cell cycle arrest in the G2/M phase and the initiation of apoptosis. Syringetin can inhibit the alpha-glucosidase enzyme, so it can reduce postprandial glycemia [122].
Stigmasterol had strong anti-inflammatory and immunomodulatory effects via a reduction in the production of pro-inflammatory cytokines, nitric oxide, and tumour necrosis factor-α (TNF-α), as well as the suppression of cyclooxygenase-2 (COX-2) [123]. In colitis, stigmasterol has been discovered to up-regulate the intestinal mucosal immune response linked to inflammatory bowel disease by activating the butyrate-PPAR axis [124].
At last, it is important to note that prochaetoviridin A [125], chaetoglobinol A [79], chaetomugilide A [68], chaetoglobinol B [66], globosumin [103], and chaetominedione [120] were previously isolated from Chaetomium. According to [81], 19 out of 25 strains of C. globosum had chaetoviridin A isolated from their filtrates.

4. Conclusions

In conclusion, this study sheds light on the potentiality of native Chaetomium isolates, which recorded high antibacterial activity against multi-drug resistant clinical Gram-positive and Gram-negative bacteria. Our findings proved that endophytic and terricolous mycobiota, especially those isolated from extreme environments such as Upper Egypt and Sinai, are considered sustainable sources of antimicrobial bioactive compounds. Their anti-microbial activity may be referred to by several compounds, such as chaetomugilide A, chaetoviridin A, prochaetoviridin A, and chaetoglobinol A, as proven by LC-MS/MS chemical profiling. More research involving the isolation of bioactive compounds, safety profile, nanoformulation, and clinical trials of native fungi in Egypt should be conducted in order to discover new drugs for medical, industrial, and nanotechnology applications.

Author Contributions

Conceptualization, J.M.B. and S.S.E.; methodology M.S.G., N.E.-K., M.A.A.-A., S.S.E. and E.E.E.; data curation, M.S.G., N.E.-K., M.A.A.-A., S.S.E., A.J.A., M.A. and E.E.E.; resources, K.A.M.A., N.A.N., A.J.A., M.A. and S.S.E.; isolation of fungi, M.A.A.-A., writing—original draft preparation, M.S.G., N.E.-K., M.A.A.-A. and E.E.E.; writing—review and editing, M.S.G., N.E.-K., M.A.A.-A., J.M.B., E.E.E., A.J.A., M.A. and S.S.E.; supervision, J.M.B., E.E.E. and M.S.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was funded by Institutional Fund Projects under grant no. (IFPIP:1135-166-1443). The authors gratefully acknowledge the technical and financial support provided by the Ministry of Education and King Abdulaziz University, DSR, Jeddah, Saudi Arabia.

Institutional Review Board Statement

The study was conducted in accordance with the guidelines and regulations of the Clinical and Laboratory Standards Institute (CLSI) (Ethics Code: IME00071).

Informed Consent Statement

This study was performed in strict accordance with the GOTHI guidelines and ethics regulations issued by the Minister of Health & Population Cairo, Egypt: No. 238/2003, Articles 52–6121, approved by the medical research ethics committee. In addition, the approval for performing this study has been registered under No. IME00071 in 2022.

Data Availability Statement

The data are available within the article.

Acknowledgments

The author M.A.A-A would like to thank Ahmed Mohamed Abdel-Azeem, Department of Botany and Microbiology, Faculty of Science, Suez Canal University, Ismailia Egypt for his guidance support and providing fungal taxa and required data and facilities to finalize this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Badau, E. A One Health Perspective on the Issue of the Antibiotic Resistance. Parasite 2021, 28, 16. [Google Scholar] [CrossRef]
  2. Hernando-Amado, S.; Coque, T.M.; Baquero, F.; Martínez, J.L. Defining and Combating Antibiotic Resistance from One Health and Global Health Perspectives. Nat. Microbiol. 2019, 4, 1432–1442. [Google Scholar] [CrossRef]
  3. Sridhar, S.; Turbett, S.E.; Harris, J.B.; Larocque, R.C. Antimicrobial-Resistant Bacteria in International Travelers. Curr. Opin. Infect. Dis. 2021, 34, 423–431. [Google Scholar] [CrossRef]
  4. Aslam, B.; Wang, W.; Arshad, M.I.; Khurshid, M.; Muzammil, S.; Rasool, M.H.; Nisar, M.A.; Alvi, R.F.; Aslam, M.A.; Qamar, M.U.; et al. Antibiotic Resistance: A Rundown of a Global Crisis. Infect. Drug Resist. 2018, 11, 1645. [Google Scholar] [CrossRef]
  5. Bate, P.N.N.; Orock, A.E.; Nyongbela, K.D.; Babiaka, S.B.; Kukwah, A.; Ngemenya, M.N. In Vitro Activity against Multi-Drug Resistant Bacteria and Cytotoxicity of Lichens Collected from Mount Cameroon. J. King Saud Univ. -Sci. 2020, 32, 614–619. [Google Scholar] [CrossRef]
  6. Elkhateeb, W.A. Highlights on Chaetomium Morphology, Secondary Metabolites and Biological Activates. Pharm. Pharmacol. Res. 2021, 4, 1–5. [Google Scholar] [CrossRef]
  7. Abdel-Azeem, A.M. Taxonomy and Biodiversity of the Genus Chaetomium in Different Habitats-Recent Developments on Genus Chaetomium; Abdel-Azeem, A.M., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 3–77. ISBN 978-3-030-31612-9. [Google Scholar]
  8. Wijeratne, E.M.K.; Turbyville, T.J.; Fritz, A.; Whitesell, L.; Gunatilaka, A.A.L. A New Dihydroxanthenone from a Plant-Associated Strain of the Fungus Chaetomium globosum Demonstrates Anticancer Activity. Bioorg. Med. Chem. 2006, 14, 7917–7923. [Google Scholar] [CrossRef]
  9. El-Kassem, L.A.; Hawas, U.W.; El-Souda, S.; Ahmed, E.F.; El-Khateeb, W.; Fayad, W. Anti-HCV Protease Potential of Endophytic Fungi and Cytotoxic Activity. Biocatal. Agric. Biotechnol. 2019, 19, 101170. [Google Scholar] [CrossRef]
  10. Demain, A.L.; Fang, A. The Natural Functions of Secondary Metabolites. Adv. Biochem. Eng. Biotechnol. 2000, 69, 1–39. [Google Scholar] [CrossRef]
  11. Zhang, J.; Ge, H.M.; Jiao, R.H.; Li, J.; Peng, H.; Wang, Y.R.; Wu, J.H.; Song, Y.C.; Tan, R.X. Cytotoxic Chaetoglobosins from the Endophyte Chaetomium globosum. Planta Med. 2010, 76, 1910–1914. [Google Scholar] [CrossRef]
  12. Zhang, Q.; Li, H.-Q.; Zong, S.-C.; Gao, J.-M.; Zhang, A.-L. Chemical and Bioactive Diversities of the Genus Chaetomium Secondary Metabolites. Mini-Rev. Med. Chem. 2012, 12, 127–148. [Google Scholar] [CrossRef]
  13. Madbouly, A.K.; Abdel-Wareth, M.T.A. The Use of Chaetomium Taxa as Biocontrol Agents BT-Recent Developments on Genus Chaetomium; Abdel-Azeem, A.M., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 251–266. ISBN 978-3-030-31612-9. [Google Scholar]
  14. Zhao, S.S.; Zhang, Y.Y.; Yan, W.; Cao, L.L.; Xiao, Y.; Ye, Y.H. Chaetomium Globosum CDW7, a Potential Biological Control Strain and Its Antifungal Metabolites. FEMS Microbiol. Lett. 2017, 364, fnw287. [Google Scholar] [CrossRef]
  15. Gao, W.; He, Y.; Li, F.; Chai, C.; Zhang, J.; Guo, J.; Chen, C.; Wang, J.; Zhu, H.; Hu, Z.; et al. Antibacterial Activity against Drug-Resistant Microbial Pathogens of Cytochalasan Alkaloids from the Arthropod-Associated Fungus Chaetomium Globosum TW1-1. Bioorganic Chem. 2019, 83, 98–104. [Google Scholar] [CrossRef]
  16. Warcup, J.H.; Baker, K.F. Occurrence of Dormant Ascospores in Soil. Nature 1963, 197, 1317–1318. [Google Scholar] [CrossRef]
  17. Abdel-Azeem, A.M.; Abdel-Azeem, M.A.; Khalil, W.F. Endophytic Fungi as a New Source of Antirheumatoid Metabolites. In Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases; Academic Press: Cambridge, MA, USA, 2019; pp. 355–384. [Google Scholar] [CrossRef]
  18. Smith, N.R.; Dawson, V.T. The Bacteriostatic Action of Rose Bengal in Media Used for Plate Counts of Soil Fungi. Soil Sci. 1944, 58, 467–472. [Google Scholar] [CrossRef]
  19. Guarro, J. Atlas of Soil Ascomycetes; Centraalbureau voor Schimmelcultures: Utrecht, The Netherlands, 2012; p. 486. [Google Scholar]
  20. Abdel-Azeem, A.M. Recent Developments on Genus Chaetomium; Springer Science and Business Media LLC: Dordrecht, The Netherlands, 2020; p. 453. [Google Scholar] [CrossRef]
  21. Kirk, P.; Cannon, P.; Stalpers, J.; Minter, D.W. Dictionary of the Fungi, 10th ed.; CABI Publishing: Wallingford, UK, 2008. [Google Scholar]
  22. Cheng, M.; Zhao, S.; Liu, H.; Liu, Y.; Lin, C.; Song, J.; Thawai, C.; Charoensettasilp, S.; Yang, Q. Functional Analysis of a Chaetoglobosin A Biosynthetic Regulator in Chaetomium globosum. Fungal Biol. 2021, 125, 201–210. [Google Scholar] [CrossRef]
  23. Magiorakos, A.P.; Srinivasan, A.; Carey, R.B.; Carmeli, Y.; Falagas, M.E.; Giske, C.G.; Harbarth, S.; Hindler, J.F.; Kahlmeter, G.; Olsson-Liljequist, B.; et al. Multidrug-Resistant, Extensively Drug-Resistant and Pandrug-Resistant Bacteria: An International Expert Proposal for Interim Standard Definitions for Acquired Resistance. Clin. Microbiol. Infect. 2012, 18, 268–281. [Google Scholar] [CrossRef]
  24. Abdel-hamed, A.R.; Mehanna, E.T.; Hazem, R.M.; Badr, J.M.; Abo-elmatty, D.M.; Abdel-kader, M.S.; Goda, M.S. Plicosepalus Acacia Extract and Its Major Constituents, Methyl Gallate and Quercetin, Potentiate Therapeutic Angiogenesis in Diabetic Hind Limb Ischemia: HPTLC Quantification and LC-MS/MS Metabolic Profiling. Antioxidants 2021, 10, 1701. [Google Scholar] [CrossRef]
  25. Goda, M.S.; Nafie, M.S.; Awad, B.M.; Abdel-Kader, M.S.; Ibrahim, A.K.; Badr, J.M.; Eltamany, E.E. In Vitro and In Vivo Studies of Anti-Lung Cancer Activity of Artemesia judaica L. Crude Extract Combined with LC-MS/MS Metabolic Profiling, Docking Simulation and HPLC-DAD Quantification. Antioxidants 2021, 11, 17. [Google Scholar] [CrossRef]
  26. Abdelhameed, R.F.A.; Habib, E.S.; Goda, M.S.; Fahim, J.R.; Hassanean, H.A.; Eltamany, E.E.; Ibrahim, A.K.; Aboulmagd, A.M.; Fayez, S.; El-Kader, A.M.A.; et al. Thalassosterol, a New Cytotoxic Aromatase Inhibitor Ergosterol Derivative from the Red Sea Seagrass Thalassodendron Ciliatum. Mar. Drugs 2020, 18, 354. [Google Scholar] [CrossRef]
  27. Eltamany, E.E.; Elhady, S.S.; Goda, M.S.; Aly, O.M.; Habib, E.S.; Ibrahim, A.K.; Hassanean, H.A.; Abdelmohsen, U.R.; Safo, M.K.; Ahmed, S.A. Chemical Composition of the Red Sea Green Algae Ulva Lactuca: Isolation and in Silico Studies of New Anti-COVID-19 Ceramides. Metabolites 2021, 11, 816. [Google Scholar] [CrossRef]
  28. Elhady, S.S.; Habib, E.S.; Abdelhameed, R.F.A.; Goda, M.S.; Hazem, R.M.; Mehanna, E.T.; Helal, M.A.; Hosny, K.M.; Diri, R.M.; Hassanean, H.A.; et al. Anticancer Effects of New Ceramides Isolated from the Red Sea Red Algae Hypnea musciformis in a Model of Ehrlich Ascites Carcinoma: LC-HRMS Analysis Profile and Molecular Modeling. Mar. Drugs 2022, 20, 63. [Google Scholar] [CrossRef]
  29. Mulu, W.; Abera, B.; Yimer, M.; Hailu, T.; Ayele, H.; Abate, D. Bacterial Agents and Antibiotic Resistance Profiles of Infections from Different Sites That Occurred among Patients at Debre Markos Referral Hospital, Ethiopia: A Cross-Sectional Study. BMC Res. Notes 2017, 10, 254. [Google Scholar] [CrossRef]
  30. Mancuso, G.; Midiri, A.; Gerace, E.; Biondo, C. Bacterial Antibiotic Resistance: The Most Critical Pathogens. Pathogens 2021, 10, 1310. [Google Scholar] [CrossRef]
  31. Abdelkareem, M.Z.; Sayed, M.; Hassuna, N.A.; Mahmoud, M.S.; Abdelwahab, S.F. Multi-Drug-Resistant Enterococcus faecalis among Egyptian Patients with Urinary Tract Infection. J. Chemother. 2017, 29, 74–82. [Google Scholar] [CrossRef]
  32. Farman, M.; Yasir, M.; Al-Hindi, R.R.; Farraj, S.A.; Jiman-Fatani, A.A.; Alawi, M.; Azhar, E.I. Genomic Analysis of Multidrug-Resistant Clinical Enterococcus faecalis Isolates for Antimicrobial Resistance Genes and Virulence Factors from the Western Region of Saudi Arabia. Antimicrob. Resist. Infect. Control 2019, 8, 55. [Google Scholar] [CrossRef]
  33. Mukherjee, S.; Mishra, S.; Tiwary, S. Microbial Profile and Antibiogram of Pus Isolate in a Tertiary Care Hospital of Western Odisha. Orig. Res. Artic. J. Evol. Med. Dent. Sci 2020, 9, 1325–1330. [Google Scholar] [CrossRef]
  34. Khanam, R.A.; Islam, M.R.; Sharif, A.; Parveen, R.; Sharmin, I.; Yusuf, M.A. Bacteriological Profiles of Pus with Antimicrobial Sensitivity Pattern at a Teaching Hospital in Dhaka City. Bangladesh J. Infect. Dis. 2018, 5, 10–14. [Google Scholar] [CrossRef]
  35. Bazaid, A.S.; Aldarhami, A.; Gattan, H.; Barnawi, H.; Qanash, H.; Alsaif, G.; Alharbi, B.; Alrashidi, A.; Eldrehmy, E.H. Antibiogram of Urinary Tract Infections and Sepsis among Infants in Neonatal Intensive Care Unit. Children 2022, 9, 629. [Google Scholar] [CrossRef]
  36. Omololu-Aso, J.; Omololu-Aso, O.O.; Adekanye, N.; Alex, T.; Owolabi, T.A.; Shesha, A. Antimicrobial Susceptibility Pattern of Escherichia coli Isolates from Clinical Sources at Tertiary Health Care Setting, Ile Ife, South Western Nigeria. Eur. J. Exp. Biol. 2017, 7, 5. [Google Scholar] [CrossRef]
  37. Naqid, I.A.; Balatay, A.A.; Hussein, N.R.; Saeed, K.A.; Ahmed, H.A.; Yousif, S.H. Antibiotic Susceptibility Pattern of Escherichia coli Isolated from Various Clinical Samples in Duhok City, Kurdistan Region of Iraq. Int. J. Infect. 2020, 7, e103740. [Google Scholar] [CrossRef]
  38. World Health Organization. Prioritization of Pathogens to Guide Discovery, Research and Development of New Antibiotics for Drug-Resistant Bacterial Infections, Including Tuberculosis; World Health Organization: Geneva, Switzerland, 2017. [Google Scholar]
  39. Bhagirath, A.Y.; Li, Y.; Patidar, R.; Yerex, K.; Ma, X.; Kumar, A.; Duan, K. Two Component Regulatory Systems and Antibiotic Resistance in Gram-Negative Pathogens. Int. J. Mol. Sci. 2019, 20, 1781. [Google Scholar] [CrossRef]
  40. Oliveira, R.; Castro, J.; Silva, S.; Oliveira, H.; Saavedra, M.J.; Azevedo, N.F.; Almeida, C. Exploring the Antibiotic Resistance Profile of Clinical Klebsiella pneumoniae Isolates in Portugal. Antibiotics 2022, 11, 1613. [Google Scholar] [CrossRef]
  41. Mirzaei, A.; Habibi, M.; Bouzari, S.; Karam, M.R.A. Characterization of Antibiotic-Susceptibility Patterns, Virulence Factor Profiles and Clonal Relatedness in Proteus mirabilis Isolates from Patients with Urinary Tract Infection in Iran. Infect. Drug Resist. 2019, 12, 3967–3979. [Google Scholar] [CrossRef]
  42. Hasan, T.H.; Alasedi, K.; Aljanaby, A. Proteus Mirabilis Virulence Factors: Review. Int. J. Pharm. Res. 2020, 13, 2145–2149. [Google Scholar] [CrossRef]
  43. Imanah, E.O.; Beshiru, A.; Igbinosa, E.O. Antibiogram Profile of Pseudomonas aeruginosa Isolated from Some Selected Hospital Environmental Drains. Asian Pac. J. Trop. Dis. 2017, 7, 604–609. [Google Scholar] [CrossRef]
  44. Golle, A.; Janezic, S.; Rupnik, M. Low Overlap between Carbapenem Resistant Pseudomonas aeruginosa Genotypes Isolated from Hospitalized Patients and Wastewater Treatment Plants. PLoS ONE 2017, 12, e0186736. [Google Scholar] [CrossRef]
  45. Ghaith, D.M.; Zafer, M.M.; Ismail, D.K.; Al-Agamy, M.H.; Bohol, M.F.F.; Al-Qahtani, A.; Al-Ahdal, M.N.; Elnagdy, S.M.; Mostafa, I.Y. First Reported Nosocomial Outbreak of Serratia Marcescens Harboring Bla IMP-4 and Bla VIM-2 in a Neonatal Intensive Care Unit in Cairo, Egypt. Infect. Drug Resist. 2018, 11, 2211–2217. [Google Scholar] [CrossRef]
  46. Cristina, M.L.; Sartini, M.; Spagnolo, A.M. Serratia Marcescens Infections in Neonatal Intensive Care Units (NICUs). Int. J. Environ. Res. Public Health 2019, 16, 610. [Google Scholar] [CrossRef]
  47. Mohammed, A.E.; Sonbol, H.; Alwakeel, S.S.; Alotaibi, M.O.; Alotaibi, S.; Alothman, N.; Suliman, R.S.; Ahmedah, H.T.; Ali, R. Investigation of Biological Activity of Soil Fungal Extracts and LC/MS-QTOF Based Metabolite Profiling. Sci. Rep. 2021, 11, 4760. [Google Scholar] [CrossRef]
  48. Deshmukh, S.K.; Dufossé, L.; Chhipa, H.; Saxena, S.; Mahajan, G.B.; Gupta, M.K. Fungal Endophytes: A Potential Source of Antibacterial Compounds. J. Fungi 2022, 8, 164. [Google Scholar] [CrossRef]
  49. Du, W.; Yao, Z.; Li, J.; Sun, C.; Xia, J.; Wang, B.; Shi, D.; Ren, L. Diversity and Antimicrobial Activity of Endophytic Fungi Isolated from Securinega suffruticosa in the Yellow River Delta. PLoS ONE 2020, 15, e0229589. [Google Scholar] [CrossRef]
  50. Prima, S.R.; Elfahmi; Julianti, E.; Fidrianny, I. Evaluation of Antibacterial and Antioxidant Activity of Endophytic Fungi Isolated from Capsicum annuum L. and Allium cepa L. Pharmacogn. J. 2022, 14, 329–334. [Google Scholar] [CrossRef]
  51. Dramae, A.; Intaraudom, C.; Bunbamrung, N.; Boonyuen, N.; Auncharoen, P.; Pittayakhajonwut, P. Antimicrobial Tanzawaic Acid Derivatives from the Endophytic Penicillium Citrinum BCC71086. Tetrahedron 2022, 106–107, 132645. [Google Scholar] [CrossRef]
  52. Khan, B.; Yan, W.; Wei, S.; Wang, Z.; Zhao, S.; Cao, L.; Rajput, N.A.; Ye, Y. Nematicidal Metabolites from Endophytic Fungus Chaetomium globosum YSC5. FEMS Microbiol. Lett. 2019, 366, 169. [Google Scholar] [CrossRef]
  53. Manganyi, M.C.; Ateba, C.N. Untapped Potentials of Endophytic Fungi: A Review of Novel Bioactive Compounds with Biological Applications. Microorganisms 2020, 8, 1934. [Google Scholar] [CrossRef]
  54. Johansson, L.; Lindskog, A.; Silfversparre, G.; Cimander, C.; Nielsen, K.F.; Lidén, G. Shikimic Acid Production by a Modified Strain of E. coli (W3110.Shik1) under Phosphate-Limited and Carbon-Limited Conditions. Biotechnol. Bioeng. 2005, 92, 541–552. [Google Scholar] [CrossRef]
  55. Buiarelli, F.; Di Filippo, P.; Riccardi, C.; Pomata, D.; Rumolo, E.; Giannetti, L.; Neri, B. Analytical Method for the Determination of Mycotoxins in Indoor/Outdoor Airborne Particulate Matter by HPLC-MS-MS. Int. J. Environ. Anal. Chem. 2015, 95, 713–729. [Google Scholar] [CrossRef]
  56. García-Salas, P.; Gómez-Caravaca, A.M.; Morales-Soto, A.; Segura-Carretero, A.; Fernández-Gutiérrez, A. Identification and Quantification of Phenolic and Other Polar Compounds in the Edible Part of Annona Cherimola and Its By-Products by HPLC-DAD-ESI-QTOF-MS. Food Res. Int. 2015, 78, 246–257. [Google Scholar] [CrossRef]
  57. Kramberger, K.; Barlič-Maganja, D.; Bandelj, D.; Baruca Arbeiter, A.; Peeters, K.; Miklavčič Višnjevec, A.; Pražnikar, Z.J. HPLC-DAD-ESI-QTOF-MS Determination of Bioactive Compounds and Antioxidant Activity Comparison of the Hydroalcoholic and Water Extracts from Two Helichrysum italicum Species. Metabolites 2020, 10, 403. [Google Scholar] [CrossRef]
  58. Szczesny, D.; Bartosińska, E.; Jacyna, J.; Patejko, M.; Siluk, D.; Kaliszan, R. Quantitative Determination of Trigonelline in Mouse Serum by Means of Hydrophilic Interaction Liquid Chromatography–MS/MS Analysis: Application to a Pharmacokinetic Study. Biomed. Chromatogr. 2018, 32, e4054. [Google Scholar] [CrossRef]
  59. Liang, S.; Sanchez-Espiridion, B.; Xie, H.; Ma, J.; Wu, X.; Liang, D. Determination of Proline in Human Serum by a Robust LC-MS/MS Method: Application to Identification of Human Metabolites as Candidate Biomarkers for Esophageal Cancer Early Detection and Risk Stratification. Biomed. Chromatogr. 2015, 29, 570–577. [Google Scholar] [CrossRef]
  60. Guo, Y.P.; Yang, H.; Wang, Y.L.; Chen, X.X.; Zhang, K.; Wang, Y.L.; Sun, Y.F.; Huang, J.; Yang, L.; Wang, J.H. Determination of Flavonoids Compounds of Three Species and Different Harvesting Periods in Crataegi Folium Based on Lc-Ms/Ms. Molecules 2021, 26, 1602. [Google Scholar] [CrossRef]
  61. Yan, W.; Cao, L.L.; Zhang, Y.Y.; Zhao, R.; Zhao, S.S.; Khan, B.; Ye, Y.H. New Metabolites from Endophytic Fungus Chaetomium globosum CDW7. Molecules 2018, 23, 2873. [Google Scholar] [CrossRef]
  62. Wang, Y.; Song, M.; Hang, T.; Wen, A.; Yang, L. LC-MS-MS Simultaneous Determination of Niacin, Niacinamide and Nicotinuric Acid in Human Plasma LC-MS-MS and Its Application to a Human Pharmacokinetic Study. Chromatographia 2010, 72, 245–253. [Google Scholar] [CrossRef]
  63. Varga, E.; Glauner, T.; Berthiller, F.; Krska, R.; Schuhmacher, R.; Sulyok, M. Development and Validation of a (Semi-)Quantitative UHPLC-MS/MS Method for the Determination of 191 Mycotoxins and Other Fungal Metabolites in Almonds, Hazelnuts, Peanuts and Pistachios. Anal. Bioanal. Chem. 2013, 405, 5087–5104. [Google Scholar] [CrossRef]
  64. Del Mar Contreras, M.; Bribi, N.; Gómez-Caravaca, A.M.; Gálvez, J.; Segura-Carretero, A. Alkaloids Profiling of Fumaria capreolata by Analytical Platforms Based on the Hyphenation of Gas Chromatography and Liquid Chromatography with Quadrupole-Time-of-Flight Mass Spectrometry. Int. J. Anal. Chem. 2017, 2017, 5178729. [Google Scholar] [CrossRef]
  65. Menéndez-Perdomo, I.M.; Hagel, J.M.; Facchini, P.J. Benzylisoquinoline Alkaloid Analysis Using High-Resolution Orbitrap LC-MSn. J. Mass Spectrom. 2021, 56, e4683. [Google Scholar] [CrossRef]
  66. Yao, C.; Bai, H.-H.; Zhang, Q.; Qian, X.-Q.; Zhang, X.; Wu, L.-W.; Yang, T. Secondary Metabolites from the Fungus Chaetomium elatum CIB-412. Chem. Nat. Compd. 2019, 55, 899–902. [Google Scholar] [CrossRef]
  67. Dwibedi, V.; Rath, S.K.; Jain, S.; Martínez-Argueta, N.; Prakash, R.; Saxena, S.; Rios-Solis, L. Key Insights into Secondary Metabolites from Various Chaetomium Species. Appl. Microbiol. Biotechnol. 2023, 107, 1077–1093. [Google Scholar] [CrossRef]
  68. Li, X.; Tian, Y.; Yang, S.X.; Zhang, Y.M.; Qin, J.C. Cytotoxic Azaphilone Alkaloids from Chaetomium globosum TY1. Bioorganic Med. Chem. Lett. 2013, 23, 2945–2947. [Google Scholar] [CrossRef]
  69. Dar, P.M.; Abdel-Azeem, A.M.; Hamed, S.R. Chaetomium’s Alkaloids. In Recent Developments on Genus Chaetomium; Springer: Cham, Switzerland, 2020; pp. 205–225. [Google Scholar] [CrossRef]
  70. Cole, R.J.; Jarvis, B.B.; Schweikert, M.A. Handbook of Secondary Fungal Metabolites; Gulf Professional Publishing: Houston, TX, USA, 2003; pp. 1–672. [Google Scholar]
  71. Hebra, T.; Elie, N.; Poyer, S.; Van Elslande, E.; Touboul, D.; Eparvier, V. Dereplication, Annotation, and Characterization of 74 Potential Antimicrobial Metabolites from Penicillium sclerotiorum Using t-SNE Molecular Networks. Metabolites 2021, 11, 444. [Google Scholar] [CrossRef]
  72. Hoeksma, J.; Misset, T.; Wever, C.; Kemmink, J.; Kruijtzer, J.; Versluis, K.; Liskamp, R.M.J.; Boons, G.J.; Heck, A.J.R.; Boekhout, T.; et al. A New Perspective on Fungal Metabolites: Identification of Bioactive Compounds from Fungi Using Zebrafish Embryogenesis as Read-Out. Sci. Rep. 2019, 9, 17546. [Google Scholar] [CrossRef]
  73. Zhou, J.; Chan, L.; Zhou, S. Trigonelline: A Plant Alkaloid with Therapeutic Potential for Diabetes and Central Nervous System Disease. Curr. Med. Chem. 2012, 19, 3523–3531. [Google Scholar] [CrossRef]
  74. Paulo, D.Q.M.; Barbosa-Filho, J.M.; Lima, E.O.; Maia, R.F.; de Cassia, R.; Barbosa, B.B.C.; Kaplan, M.A.C. Antimicrobial Activity of Benzylisoquinoline Alkaloids from Annona Salzmanii D.C. J. Ethnopharmacol. 1992, 36, 39–41. [Google Scholar] [CrossRef]
  75. Conceição, R.S.; Perez, C.J.; Branco, A.; Botura, M.B.; Ifa, D.R. Identification of Sassafras Albidum Alkaloids by High-Performance Thin-Layer Chromatography Tandem Mass Spectrometry and Mapping by Desorption Electrospray Ionization Mass Spectrometry Imaging. J. Mass Spectrom. 2021, 56, e4674. [Google Scholar] [CrossRef]
  76. Al-Ghazzawi, A.M. Anti-Cancer Activity of New Benzyl Isoquinoline Alkaloid from Saudi Plant Annona Squamosa. BMC Chem. 2019, 13, 13. [Google Scholar] [CrossRef]
  77. Wangchuk, P.; Sastraruji, T.; Taweechotipatr, M.; Keller, P.A.; Pyne, S.G. Anti-Inflammatory, Anti-Bacterial and Anti-Acetylcholinesterase Activities of Two Isoquinoline Alkaloids–Scoulerine and Cheilanthifoline. Nat. Prod. Commun. 2016, 11, 1801–1804. [Google Scholar] [CrossRef]
  78. Siddiquee, S. Recent Advancements on the Role of Biologically Active Secondary Metabolites from Aspergillus. In New and Future Developments in Microbial Biotechnology and Bioengineering; Elsevier: Amsterdam, The Netherlands, 2018; pp. 69–94. [Google Scholar] [CrossRef]
  79. Xu, G.B.; He, G.; Bai, H.H.; Yang, T.; Zhang, G.L.; Wu, L.W.; Li, G.Y. Indole Alkaloids from Chaetomium globosum. J. Nat. Prod. 2015, 78, 1479–1485. [Google Scholar] [CrossRef]
  80. Chen, C.; Tao, H.; Chen, W.; Yang, B.; Zhou, X.; Luo, X.; Liu, Y. Recent Advances in the Chemistry and Biology of Azaphilones. RSC Adv. 2020, 10, 10197. [Google Scholar] [CrossRef]
  81. McMullin, D.R.; Sumarah, M.W.; Miller, J.D. Chaetoglobosins and Azaphilones Produced by Canadian Strains of Chaetomium globosum Isolated from the Indoor Environment. Mycotoxin Res. 2013, 29, 47–54. [Google Scholar] [CrossRef]
  82. Ding, G.; Song, Y.; Chen, J.R.; Xu, C.; Ge, H.; Wang, X.T.; Tan, R. Chaetoglobosin U, a Cytochalasan Alkaloid from Endophytic Chaetomium globosum IFB-E019. J. Nat. Prod. 2006, 69, 302–304. [Google Scholar] [CrossRef]
  83. Cooper, J.A. Effects of Cytochalasin and Phalloidin on Actin. J. Cell Biol. 1987, 105, 1473–1478. [Google Scholar] [CrossRef]
  84. Takahashi, J.A.; de Castro, M.C.M.; Souza, G.G.; Lucas, E.M.F.; Bracarense, A.A.P.; Abreu, L.M.; Marriel, I.E.; Oliveira, M.S.; Floreano, M.B.; Oliveira, T.S. Isolation and Screening of Fungal Species Isolated from Brazilian Cerrado Soil for Antibacterial Activity against Escherichia coli, Staphylococcus aureus, Salmonella typhimurium, Streptococcus pyogenes and Listeria Monocytogenes. J. Mycol. Med. 2008, 18, 198–204. [Google Scholar] [CrossRef]
  85. Zheng, W.; Wang, B.; Li, X.; Wang, T.; Zou, H.; Gu, J.; Yuan, Y.; Liu, X.; Bai, J.; Bian, J.; et al. Zearalenone Promotes Cell Proliferation or Causes Cell Death? Toxins 2018, 10, 184. [Google Scholar] [CrossRef]
  86. Stanely Mainzen Prince, P.; Rajakumar, S.; Dhanasekar, K. Protective Effects of Vanillic Acid on Electrocardiogram, Lipid Peroxidation, Antioxidants, Proinflammatory Markers and Histopathology in Isoproterenol Induced Cardiotoxic Rats. Eur. J. Pharmacol. 2011, 668, 233–240. [Google Scholar] [CrossRef]
  87. Salau, V.F.; Erukainure, O.L.; Ibeji, C.U.; Olasehinde, T.A.; Koorbanally, N.A.; Islam, M.S. Vanillin and Vanillic Acid Modulate Antioxidant Defense System via Amelioration of Metabolic Complications Linked to Fe2+-Induced Brain Tissues Damage. Metab. Brain Dis. 2020, 35, 727–738. [Google Scholar] [CrossRef]
  88. Mourtzinos, I.; Konteles, S.; Kalogeropoulos, N.; Karathanos, V.T. Thermal Oxidation of Vanillin Affects Its Antioxidant and Antimicrobial Properties. Food Chem. 2009, 114, 791–797. [Google Scholar] [CrossRef]
  89. Ul Amin, F.; Shah, S.A.; Kim, M.O. Vanillic Acid Attenuates Aβ1-42-Induced Oxidative Stress and Cognitive Impairment in Mice. Sci. Rep. 2017, 7, 40753. [Google Scholar] [CrossRef]
  90. Mardirossian, M.; Sola, R.; Beckert, B.; Collis, D.W.P.; Di Stasi, A.; Armas, F.; Hilpert, K.; Wilson, D.N.; Scocchi, M. Proline-Rich Peptides with Improved Antimicrobial Activity against E. coli, K. pneumoniae, and A. baumannii. ChemMedChem 2019, 14, 2025. [Google Scholar] [CrossRef]
  91. Xing, X.R.; Liao, Z.; Tan, F.; Zhu, Z.Y.; Jiang, Y.; Cao, Y.Y. Effect of Nicotinamide against Candida albicans. Front. Microbiol. 2019, 10, 595. [Google Scholar] [CrossRef]
  92. Kiefer, A.; Arnholdt, M.; Grimm, V.; Geske, L.; Groß, J.; Vierengel, N.; Opatz, T.; Erkel, G. Structure Elucidation and Biological Activities of Perylenequinones from an Alternaria Species. Mycotoxin Res. 2023, 1, 303–316. [Google Scholar] [CrossRef]
  93. Mahmoud, M.M.; Abdel-Razek, A.S.; Soliman, H.S.M.; Ponomareva, L.V.; Thorson, J.S.; Shaaban, K.A.; Shaaban, M. Diverse Polyketides from the Marine Endophytic Alternaria sp. LV52: Structure Determination and Cytotoxic Activities. Biotechnol. Rep. 2021, 33, e00628. [Google Scholar] [CrossRef]
  94. Kezimana, P.; Dmitriev, A.A.; Kudryavtseva, A.V.; Romanova, E.V.; Melnikova, N.V. Secoisolariciresinol Diglucoside of Flaxseed and Its Metabolites: Biosynthesis and Potential for Nutraceuticals. Front. Genet. 2018, 9, 641. [Google Scholar] [CrossRef]
  95. De Carvalho, M.; Weich, H.; Abraham, W.-R. Macrocyclic Trichothecenes as Antifungal and Anticancer Compounds. Curr. Med. Chem. 2016, 23, 23–35. [Google Scholar] [CrossRef]
  96. Bodet, C.A.; Jorgensen, J.H.; Drutz, D.J. Antibacterial Activities of Antineoplastic Agents. Antimicrob. Agents Chemother. 1985, 28, 437. [Google Scholar] [CrossRef]
  97. Peiris, V.; Oppenheim, B.A. Antimicrobial activity of cytotoxic drugs may influence isolation of bacteria and fungi from blood cultures Influence Isolation. J. Clin. Pathol. 1993, 1124–1125. [Google Scholar] [CrossRef]
  98. Wu, B.; Zhang, B.; Feng, X.; Rubens, J.R.; Huang, R.; Hicks, L.M.; Pakrasi, H.B.; Tang, Y.J. Alternative Isoleucine Synthesis Pathway in Cyanobacterial Species. Microbiology 2010, 156, 596–602. [Google Scholar] [CrossRef]
  99. Peng, J.; Xie, J.; Shi, S.; Luo, L.; Li, K.; Xiong, P.; Cai, W. Diagnostic Fragment-Ion-Based for Rapid Identification of Chlorogenic Acids Derivatives in Inula Cappa Using UHPLC-Q-Exactive Orbitrap Mass Spectrometry. J. Anal. Methods Chem. 2021, 2021, 6393246. [Google Scholar] [CrossRef]
  100. Kumar, R.; Kundu, A.; Dutta, A.; Saha, S.; Das, A.; Bhowmik, A. Chemo-Profiling of Bioactive Metabolites from Chaetomium globosum for Biocontrol of Sclerotinia Rot and Plant Growth Promotion. Fungal Biol. 2021, 125, 167–176. [Google Scholar] [CrossRef]
  101. Duffield, A.M.; Budzikiewicz, H.; Williams, D.H.; Djerassi, C. Mass Spectrometry in Structural and Stereochemical Problems. LXIV. A Study of the Fragmentation Processes of Some Cyclic Amines. J. Am. Chem. Soc. 1965, 87, 810–816. [Google Scholar] [CrossRef]
  102. Mahrous, F.; Mohammed, H.; Sabour, R.; Ismail, L. LC-ESI-QTOF-MS/MS of Holoptelea integrifolia (Roxb.) Planch. Leaves and In Silico Study of Phenolic Compounds’ Antiviral Activity against the HSV1 Virus. Azhar Int. J. Pharm. Med. Sci. 2021, 1, 91–101. [Google Scholar] [CrossRef]
  103. Zhang, X.Y.; Tan, X.M.; Yu, M.; Yang, J.; Sun, B.; Qin, J.C.; Guo, L.P.; Ding, G. Bioactive Metabolites from the Desert Plant-Associated Endophytic Fungus Chaetomium globosum (Chaetomiaceae). Phytochemistry 2021, 185, 112701. [Google Scholar] [CrossRef]
  104. Zhang, M.; Liu, H.J.; Huang, X.L.; Shao, L.J.; Xie, X.L.; Wang, F.; Yang, J.; Pei, P.; Zhang, Z.Q.; Zhai, Y.C.; et al. A Novel LC-MS/MS Assay for Vitamin B 1, B 2 and B 6 Determination in Dried Blood Spots and Its Application in Children. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1112, 33–40. [Google Scholar] [CrossRef]
  105. Kanazawa, H.; Atsumi, R.; Matsushima, Y.; Kizu, J. Determination of Theophylline and Its Metabolites in Biological Samples by Liquid Chromatography-Mass Spectrometry. J. Chromatogr. A 2000, 870, 87–96. [Google Scholar] [CrossRef]
  106. Husain, K.; Abid, M.; Azam, A. Synthesis, Characterization and Antiamoebic Activity of New Indole-3-Carboxaldehyde Thiosemicarbazones and Their Pd(II) Complexes. Eur. J. Med. Chem. 2007, 42, 1300–1308. [Google Scholar] [CrossRef]
  107. Uenoyama, R.; Miyazaki, M.; Miyazaki, T.; Shigeno, Y.; Tokairin, Y.; Konno, H.; Yamashita, T. LC-ESI-MS/MS Quantification of Carnosine, Anserine, and Balenine in Meat Samples. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2019, 1132, 121826. [Google Scholar] [CrossRef]
  108. Gachumi, G.; El-Aneed, A. Mass Spectrometric Approaches for the Analysis of Phytosterols in Biological Samples. J. Agric. Food Chem. 2017, 65, 10141–10156. [Google Scholar] [CrossRef]
  109. Ravindernath, A.; Reddy, M.S. Synthesis and Evaluation of Anti-Inflammatory, Antioxidant and Antimicrobial Activities of Densely Functionalized Novel Benzo [d] Imidazolyl Tetrahydropyridine Carboxylates. Arab. J. Chem. 2017, 10, S1172–S1179. [Google Scholar] [CrossRef]
  110. Wu, Y.; Xu, J.; Shi, M.; Han, X.; Li, W.; Zhang, X.; Wen, X. Pitaya: A Potential Plant Resource of Citramalic Acid. CyTA-J. Food 2020, 18, 249–256. [Google Scholar] [CrossRef]
  111. Bai, J.; Wu, Y.; Bu, Q.; Zhong, K.; Gao, H. Comparative Study on Antibacterial Mechanism of Shikimic Acid and Quinic Acid against Staphylococcus aureus through Transcriptomic and Metabolomic Approaches. LWT 2022, 153, 112441. [Google Scholar] [CrossRef]
  112. Eren, B.; Fen, Ü.; Dergisi, B.; Ercan, L.; Doğru, M. Antioxidant and Antimicrobial Capacity of Quinic Acid. Bitlis Eren Üniversitesi Fen Bilim. Derg. 2022, 11, 1018–1025. [Google Scholar] [CrossRef]
  113. Park, J.H.; Gyung, J.C.; Kyoung, S.J.; He, K.L.; Heung, T.K.; Kwang, Y.C.; Kim, J.C. Antifungal Activity against Plant Pathogenic Fungi of Chaetoviridins Isolated from Chaetomium globosum. FEMS Microbiol. Lett. 2005, 252, 309–313. [Google Scholar] [CrossRef]
  114. Makrerougras, M.; Coffinier, R.; Oger, S.; Chevalier, A.; Sabot, C.; Franck, X. Total Synthesis and Structural Revision of Chaetoviridins A. Org. Lett. 2017, 19, 4146–4149. [Google Scholar] [CrossRef]
  115. Awad, N.E.; Kassem, H.A.; Hamed, M.A.; El-Naggar, M.A.A.; El-Feky, A.M.M. Bioassays Guided Isolation of Compounds from Chaetomium globosum. J. Mycol. Med. 2014, 24, e35–e42. [Google Scholar] [CrossRef]
  116. Wang, Y.; Zhang, Y.; Shi, Y.; Pan, X.; Lu, Y.; Cao, P. Antibacterial Effects of Cinnamon (Cinnamomum zeylanicum) Bark Essential Oil on Porphyromonas gingivalis. Microb. Pathog. 2018, 116, 26–32. [Google Scholar] [CrossRef]
  117. Bakhtiari, S.; Jafari, S.; Taheri, J.B.; Kashi, T.S.J.; Namazi, Z.; Iman, M.; Poorberafeyi, M. The Effects of Cinnamaldehyde (Cinnamon Derivatives) and Nystatin on Candida albicans and Candida glabrata. Open Access Maced. J. Med. Sci. 2019, 7, 1067–1070. [Google Scholar] [CrossRef]
  118. Ahgilan, A.; Sabaratnam, V.; Periasamy, V. Antimicrobial Properties of Vitamin B2. Int. J. Food Prop. 2016, 19, 1173–1181. [Google Scholar] [CrossRef]
  119. Tonnetti, L.; Thorp, A.M.; Reddy, H.L.; Keil, S.D.; Goodrich, R.P.; Leiby, D.A. Riboflavin and Ultraviolet Light Reduce the Infectivity of Babesia Microti in Whole Blood. Transfusion 2013, 53, 860–867. [Google Scholar] [CrossRef]
  120. Abdel-Lateff, A. Chaetominedione, a New Tyrosine Kinase Inhibitor Isolated from the Algicolous Marine Fungus Chaetomium sp. Tetrahedron Lett. 2008, 49, 6398–6400. [Google Scholar] [CrossRef]
  121. Xing, L.; Chee, M.E.; Zhang, H.; Zhang, W.; Mine, Y. Carnosine—A Natural Bioactive Dipeptide: Bioaccessibility, Bioavailability and Health Benefits. J. Food Bioact. 2019, 5, 8–17. [Google Scholar] [CrossRef]
  122. Chmiel, M.; Stompor-Gorący, M. The Spectrum of Pharmacological Actions of Syringetin and Its Natural Derivatives—A Summary Review. Nutrients 2022, 14, 5157. [Google Scholar] [CrossRef]
  123. Villa-De La Torre, F.; Kinscherf, R.; Bonaterra, G.; Arana-Argaez, V.E.; Méndez-González, M.; Cáceres-Farfán, M.; Borges-Argáez, R. Anti-Inflammatory and Immunomodulatory Effects of Critonia Aromatisans Leaves: Downregulation of pro-Inflammatory Cytokines. J. Ethnopharmacol. 2016, 190, 174–182. [Google Scholar] [CrossRef]
  124. Wen, S.; He, L.; Zhong, Z.; Zhao, R.; Weng, S.; Mi, H.; Liu, F. Stigmasterol Restores the Balance of Treg/Th17 Cells by Activating the Butyrate-PPARγ Axis in Colitis. Front. Immunol. 2021, 12, 741934. [Google Scholar] [CrossRef]
  125. Zheng, C.J.; Liao, H.X.; Mei, R.Q.; Huang, G.L.; Yang, L.J.; Zhou, X.M.; Shao, T.M.; Chen, G.Y.; Wang, C.Y. Two New Benzophenones and One New Natural Amide Alkaloid Isolated from a Mangrove-Derived Fungus Penicillium citrinum. Nat. Prod. Res. 2019, 33, 1127–1134. [Google Scholar] [CrossRef]
Figure 1. Heatmap of presence/absence of Chaetomium isolates per source, soil (A); plants (B).
Figure 1. Heatmap of presence/absence of Chaetomium isolates per source, soil (A); plants (B).
Biomolecules 13 01683 g001
Figure 2. Chaetomium globosum: (A) colony; (B) ascomata showing peridial hairs; (C) Limoniform ascospores.
Figure 2. Chaetomium globosum: (A) colony; (B) ascomata showing peridial hairs; (C) Limoniform ascospores.
Biomolecules 13 01683 g002
Figure 3. Antimicrobial activity of crude extracts of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against Gram-positive bacteria: S. aureus (A); S. pyogenes (B); E. faecalis (C), using DMSO as a negative control.
Figure 3. Antimicrobial activity of crude extracts of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against Gram-positive bacteria: S. aureus (A); S. pyogenes (B); E. faecalis (C), using DMSO as a negative control.
Biomolecules 13 01683 g003
Figure 4. Antimicrobial activity of crude extracts of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against Gram-negative bacteria: A. baumannii (A); E. coli (B); K. pneumonia (C); P. mirabilis (D); S. marcescens (E); P. aeruginosa (F), using DMSO as a negative control.
Figure 4. Antimicrobial activity of crude extracts of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against Gram-negative bacteria: A. baumannii (A); E. coli (B); K. pneumonia (C); P. mirabilis (D); S. marcescens (E); P. aeruginosa (F), using DMSO as a negative control.
Biomolecules 13 01683 g004
Figure 5. Antimicrobial activity of crude extract of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against C. albicans, using DMSO as a negative control.
Figure 5. Antimicrobial activity of crude extract of C. globosum isolated from cultivated soil at Assuit (3) and Zygophyllum album (5) against C. albicans, using DMSO as a negative control.
Biomolecules 13 01683 g005
Figure 6. Chemical structures of metabolites detected by LC-MS/MS listed in Table 8.
Figure 6. Chemical structures of metabolites detected by LC-MS/MS listed in Table 8.
Biomolecules 13 01683 g006
Figure 7. Chemical Structures of metabolites detected by LC-MS/MS listed in Table 9.
Figure 7. Chemical Structures of metabolites detected by LC-MS/MS listed in Table 9.
Biomolecules 13 01683 g007
Table 1. Global positioning system (GPS) data of soils’ collection sites.
Table 1. Global positioning system (GPS) data of soils’ collection sites.
Soil TypeSiteGPS
NorthEast
1- Desert SoilSaint Katherine Mountain28°30′23″34°02′00″
Nuwieba28.9°73′44.4″34.6°53′43.3″
Wadi El-Arbaein28°33′00″33°58′11″
El-Arish31°11′10″33°50′25″
El-Kantara30°51′01″32°16′33″
2- Cultivated SoilIsmailia Governorate30°40′00″32°20′21″
El-Sharkia Governorate30°45′00″31°50′12″
Assiut Governorate27.1°80′96″31.18°36′8″
Giza Governorate30°00′20″31°10′37″
Cairo Governorate30°01′11″31°20′34″
° Degrees; ′ minutes; ″ seconds.
Table 2. GPS data of collection sites of host plants.
Table 2. GPS data of collection sites of host plants.
Host plantsSiteGPS
NorthNorth
1- Medicinal plants in SKPGebel Ahmar28°52′83″33°61′83″
Wadi El-Arbaein28°54′54″33°55′36″
Wadi Talaa28°37′22″33°52′48″
2- Mangrove plantsPort Fouad31°14′60.00″32°18′60.00″
Safaga26°43′59.99″33°55′59.99″
Nuwieba28.9°73′44.4″34.6°53′43.3″
° Degrees; ′ minutes; ″ seconds.
Table 3. Origin and codes of Chaetomium taxa.
Table 3. Origin and codes of Chaetomium taxa.
CodeStrainInterpretation
1Chaetomium globosumSoil taxon recovered from the desert in Nuwieba, South Sinai, Egypt.
2Chaetomium globosumSoil taxon isolated from cultivated rice straw soil, Ismailia Governorate (Suez Canal area), Egypt.
3Chaetomium globosumSoil taxon recovered from cultivated soil in Assiut Governorate (Upper Egypt).
4Chaetomium globosumEndophytic taxon recovered from Thymus decussatus, Gebel Ahmer, Saint Katherine, South Sinai, Egypt.
5Chaetomium globosumEndophytic taxon recovered from Zygophyllum album, Wadi El-Arbaein, Saint Katherine, South Sinai, Egypt.
6Chaetomium globosumEndophytic taxon recovered from Chiliadenus montanus, Wadi El-Arbaein, Saint Katherine, South Sinai, Egypt.
7Chaetomium iranianumEndophytic taxon recovered from Origanum syriacum, Wadi El-Arbaein, Saint Katherine, South Sinai, Egypt.
8Chaetomium globosumEndophytic taxon recovered from Adiantum capillus-veneris, Wadi Talaa, Saint Katherine, South Sinai, Egypt.
9Chaetomium globosumEndophytic taxon recovered from Adiantum capillus-veneris, Port Fouad, Port Said Governorate (Mediterranean area), Egypt.
10Chaetomium madrasenseEndophytic taxon recovered from Ziziphus spinosa, Nuwieba, South Sinai, Egypt.
Table 4. Distribution of the microbial isolates from clinical specimens.
Table 4. Distribution of the microbial isolates from clinical specimens.
OrganismsNo of Isolates
Klebsiella pneumoniea30
Staphylococcus aureus21
Escherichia coli11
Pseudomonas aeruginosa9
Candida albicans8
Acinetobacter baumannii7
Enterococcus faecalis5
Proteus mirabilis5
Streptococcus pyogenes2
Serratia marcescens2
Total100
Table 5. Antibiotic resistance pattern (%) of bacterial isolates.
Table 5. Antibiotic resistance pattern (%) of bacterial isolates.
Antibiotics’
Classes
Antibiotic DiscsS.
aureus
E.
faecalis
S.
pyogenes
A.
baumannii
E.
coli
K.
pneumoniea
Pr.
mirabilis
P.
aeruginosa
S.
marcescens
PenicillinAmoxycillin/Clavulanic acid (30 µg)1006010057.190.910060100100
Ampicillin/Sulbactam (30 µg)10010010085.790.91008088.9100
CephalosporinCefepime (30 µg)10010010085.754.5706010050
Cefotaxime (10 µg)10010010071.481.89010010050
Ceftazidime (30 µg)80.910010085.790.973.36066.7100
Cefoxitin (30 µg)01001001001009010088.950
FluoroquinoloneCiprofloxacin (5 µg)71.4805085.781.8808088.950
Levofloxacin (5 µg)76.21005057.154.51008088.9100
AminoglycosidesGentamicin (10 µg)66.710010071.490.9708055.6100
Amikacin (30 µg)76.21005071.454.573.34066.750
CarbapenemImipenem (10 µg)28.6100014.363.6302033.30
Meropenem (10 µg)33.3100028.663.633.32011.10
TetracyclineTetracycline (30 µg)38.06050.057.163.6806010050
Doxycycline (5 µg)76.2805010072.7606010050
Tigecycline (15 µg)19.00014.336.4602033.30
Table 6. Antimicrobial activity of Chaetomium crude extracts (110) against clinical microbial isolates.
Table 6. Antimicrobial activity of Chaetomium crude extracts (110) against clinical microbial isolates.
Crude Extracts12345678910
MicroorganismMean of Zone Inhibition in mm (Mean ± SD)
Staphylococcus aureus0019.5 ± 1.3618.4 ± 1.417.2 ± 1.4816.0 ± 1.28014.8 ± 0.77025.7 ± 1.23
Enterococcus faecalis014.6 ± 0.5500010.4 ± 0.5500011.6 ± 0.55
Streptococcus pyogenes0021.0 ± 0.014.5 ± 0.7015.0 ± 0.000026.0 ± 1.4
Acinetobacter baumannii0000015.4 ± 1.270000
Escherichia coli21.2 ± 2.0919.8 ± 2.0924.0 ± 2.1916.0 ± 1.4825.4 ± 2.4615.4 ± 1.1218.6 ± 2.4615.4 ± 1.0316.3 ± 0.6510.5 ± 0.93
Klebsiella pneumoniea0000000000
Proteus mirabilis000021.6 ± 1.3400019.6 ± 1.340
Pseudomonas aeruginosa26.0 ± 0.8725.4 ± 1.2426.3 ± 0.517.0 ± 1.121.7 ± 2.516.3 ± 1.125.2 ± 1.317.3 ± 0.721.7 ± 2.018.4 ± 1.5
Serratia marcescens18.0 ± 0.016.5 ± 0.722.5 ± 2.114.5 ± 0.724.0 ± 0.0026.0 ± 0.0026.5 ± 0.712.0 ± 0.0
Candida albicans0024.5 ± 0.7618.8 ± 1.4925.6 ± 0.740011.3 ± 0.7015.5 ± 0.76
0, no inhibition zone.
Table 7. Minimum inhibitory concentration (MIC) of Chaetomium crude extracts (110).
Table 7. Minimum inhibitory concentration (MIC) of Chaetomium crude extracts (110).
Crude Extracts12345678910
MicroorganismMIC µg/mL (Mean ± SD)
Staphylococcus aureus--29.8 ± 4.75.0 ± 3.530.5 ± 3.462.5 ± 0.0-15.0 ± 2.6-3.9 ± 0.0
Enterococcus faecalis-14.0 ± 3.5---15.6 ± 0.0---31.2 ± 0.0
Streptococcus pyogenes--31.2 ± 0.015.6 ± 0.0-15.6 ± 0.0---3.9 ± 0.0
Acinetobacter baumannii-----31.2 ± 0.0----
Escherichia coli4.6 ± 1.68.2 ± 3.55.7 ± 2.031.2 ± 0.05.0 ± 1.829.8 ± 4.714.2 ± 3.212.8 ± 3.915.6 ± 0.014.2 ± 3.1
Klebsiella pneumoniea----------
Proteus mirabilis----5.5 ± 2.1---7.0 ± 1.7-
Pseudomonas aeruginosa4.3 ± 1.34.7 ± 1.73.9 ± 0.08.7 ± 2.65.2 ± 1.914.7 ± 2.66.0 ± 3.98.7 ± 2.615.6 ± 0.09.5 ± 3.4
Serratia marcescens15.6 ± 0.015.6 ± 0.03.9 ± 0.015.6 ± 0.03.9 ± 0.0-3.9 ± 0.0-3.9 ± 0.031.2 ± 0.0
Candida albicans--3.9 ± 0.08.8 ± 2.74.9 ± 1.8--31.2 ± 0.0-14.6 ± 2.8
Table 8. LC-MS/MS Metabolic profiling of the crude extract of C. globosum soil taxon recovered from cultivated soil in Assiut Governorate (3).
Table 8. LC-MS/MS Metabolic profiling of the crude extract of C. globosum soil taxon recovered from cultivated soil in Assiut Governorate (3).
NoPolarity ModeMZmine IDRet. Time (min)Observed
m/z
Calculated m/zMass Error (ppm)AdductMolecular FormulaMS/MS SpectrumDeduced
Compound
Ref.
1Negative5221.10173.0448173.0450−1.16[M − H] C7H10O5173, 155, 111Shikimic acid[54]
2Positive2011.11319.1526319.1545−5.95[M + H] +C18H22O5319, 301, 283, 230Zearalenone[55]
3Negative6631.15329.0897329.086210.64[M − H] C14H18O9329, 167Vanillic acid
hexoside
[56,57]
4Positive8851.34138.0549138.0555−4.35[M + H] +C7H7NO2138, 94, 92Trigonelline[58]
5Positive11161.38116.071116.0712−1.72[M + H] +C5H9NO2116, 70L-Proline[59]
6Negative18891.41309.1176309.1200−7.76[M − H] C12H22O9309, 89Hex-2-ulofuranosyl-4,6- dideoxyhexopyranoside[60]
7Positive17301.76297.1083297.1099−5.39[M + Na] +C16H18O4297, 274Prochaetoviridin A[61]
8Positive19241.89123.0544123.0558−11.38[M + H] +C6H6N2O123, 80Nicotinamide (Niacinamide)[62]
9Negative26013.71351.0866351.0869−0.85[M − H] C20H16O6351, 263Altertoxin I[63]
10Positive25294.94314.1368314.1381−4.14[M + H] +C18H19NO4314, 283, 268, 251, 233, 223Demethyl isoboldine[64]
11Positive25415.02286.1434286.1443−3.15[M + H] +C17H19NO3286, 269, 237, 209, 175, 145, 143, 107Coclaurine[64]
12Positive26095.34328.156328.15493.35[M + H] +C19H21NO4328, 178, 151Scoulerine[65]
13Positive26875.66545.2042545.2052−1.83[M + Na] +C32H30N2O5522.60Chaetoglobinol B[66]
14Positive27476.08314.1765314.17630.64[M + H] +C19H24NO3+314, 299, 298, 269, 237, 209, 175, 121, 107N,N-Dimethyl coclaurine[64]
15Positive27776.18500.1878500.18407.60[M + H] +C27H30ClO6N500Chaetomugilide A[67,68]
16Positive28876.50523.219523.2233−8.22[M + H] +C32H30N2O5523Chaetoglobinol A[66,69]
17Positive28836.50300.1592300.1600−2.67[M + H] +C18H21NO3300, 269, 237, 209, 177, 175, 145, 107N-Methyl coclaurine[65]
18Positive29166.67496.2383496.23359.67[M + H] +C28H33NO7496Penochalasin A[69]
19Positive29216.69548.2684548.26486.57[M + H] +C32H37NO7548Cytochalasin L[70]
20Negative28476.84523.2174523.2177−0.57[M − H] C26H36O11523, 361, 346Secoisolariciresinol
-D-hexoside
[56]
21Positive32168.95391.1363391.131213.04[M + H] +C21H23ClO5391, 363, 147Sclerotiorin[71]
22Positive333713.47485.2115485.217512.37[M + H] +C27H32O8485Verrucarin J[72]
Table 9. LC-MS/MS Metabolic profiling of the crude extract of C. globosum endophytic taxon recovered from Zygophyllum album, Wadi El-Arbaein, Saint Katherine, South Sinai (5).
Table 9. LC-MS/MS Metabolic profiling of the crude extract of C. globosum endophytic taxon recovered from Zygophyllum album, Wadi El-Arbaein, Saint Katherine, South Sinai (5).
NoPolarity ModeMZmine IDRet. Time (min)Obseved
m/z
Calculated m/zMass Error (ppm)AdductMolecular FormulaMS/MS SpectrumDeduced
Compound
Ref.
1Negative561.08147.0313147.029313.60[M − H] C5H8O5147, 85, 57Citramalic acid[98]
2Positive3611.12319.1508319.1545−11.59[M + H] +C18H22O5319, 301, 283, 230Zearalenone[55]
3Negative2071.12191.0542191.0556−7.33[M − H] C7H12O6191, 173, 111, 85Quinic acid[99]
4Positive10981.37433.1741433.1762−4.85[M + H] +C23H25ClO6433, 296Chaetoviridin A[100]
5Positive13221.40138.0553138.0555−1.45[M + H] +C7H7NO2138, 94, 92Trigonelline[58]
6Negative25341.77309.1176309.1200−7.76[M − H] C12H22O9309, 179, 119, 89Hex-2-ulofuranosyl-4,6- dideoxyhexopyranoside[60]
7Positive19411.7886.0957286.097014.87[M + H] +C5H11N86, 56Piperidine[101]
8Positive20501.80133.1041133.102313.52[M + H] +C9H8O133, 115, 105, 79Cinnamaldehyde[102]
9Positive22981.90402.1937402.19194.48[M + H] +C22H27NO6402Globosumin[103]
10Positive28113.59286.1442286.1443−0.35[M + H] +C17H19NO3286, 269, 237, 175, 145, 143, 107Coclaurine[64]
11Positive29594.41314.1372314.1381−2.87[M + H] +C18H19NO4314, 283, 268, 251, 233, 223Demethyl isoboldine[64]
12Positive31635.30377.149377.14617.69[M + H] +C17H20N4O6377, 243Riboflavin[104]
13Positive33586.07314.1752314.1763−3.50[M + H] +C19H24NO3+413, 299, 298, 269, 209, 175, 121, 107N,N-Dimethyl coclaurine[64]
14Positive34496.42197.07196.067512.69[M + H] +C7H8N4O3197, 1691,3-Dimethyl urate[105]
15Positive35056.54300.1621300.16006.99[M + H] +C18H21NO3300, 269, 237, 209, 177, 175, 145, 107N-Methyl coclaurine[65]
16Positive40608.10146.0613146.06064.79[M + H] +C9H7NO146, 1173-Formylindole[106]
17Positive460910.15331.0695331.06950[M + Na] +C17H12N2O4331, 308Chaetominedione[69]
18Positive462010.40227.1166227.11449.69[M + H] +C9H14N4O3227, 181, 156,Carnosine[107]
19Positive462810.51509.2021509.2022−0.20[M + H] +C23H24O13509, 347Syringetin 3-O-galactoside[102]
20Positive549722.47395.3634395.3678−11.13[M + H − H2O] +C29H48O395, 147Stigmasterol[108]
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

Goda, M.S.; El-Kattan, N.; Abdel-Azeem, M.A.; Allam, K.A.M.; Badr, J.M.; Nassar, N.A.; Almalki, A.J.; Alharbi, M.; Elhady, S.S.; Eltamany, E.E. Antimicrobial Potential of Different Isolates of Chaetomium globosum Combined with Liquid Chromatography Tandem Mass Spectrometry Chemical Profiling. Biomolecules 2023, 13, 1683. https://doi.org/10.3390/biom13121683

AMA Style

Goda MS, El-Kattan N, Abdel-Azeem MA, Allam KAM, Badr JM, Nassar NA, Almalki AJ, Alharbi M, Elhady SS, Eltamany EE. Antimicrobial Potential of Different Isolates of Chaetomium globosum Combined with Liquid Chromatography Tandem Mass Spectrometry Chemical Profiling. Biomolecules. 2023; 13(12):1683. https://doi.org/10.3390/biom13121683

Chicago/Turabian Style

Goda, Marwa S., Noura El-Kattan, Mohamed A. Abdel-Azeem, Kamilia A. M. Allam, Jihan M. Badr, Nourelhuda Ahmed Nassar, Ahmad J. Almalki, Majed Alharbi, Sameh S. Elhady, and Enas E. Eltamany. 2023. "Antimicrobial Potential of Different Isolates of Chaetomium globosum Combined with Liquid Chromatography Tandem Mass Spectrometry Chemical Profiling" Biomolecules 13, no. 12: 1683. https://doi.org/10.3390/biom13121683

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

Article Metrics

Back to TopTop