[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Antimicrobial and Anti-Infective Activity of Natural Products, 2nd Edition

A special issue of Antibiotics (ISSN 2079-6382). This special issue belongs to the section "Plant-Derived Antibiotics".

Deadline for manuscript submissions: 30 April 2025 | Viewed by 11430

Special Issue Editors


E-Mail Website
Guest Editor
Centro de Ciências Biológicas e da Saúde, Universidade Federal do Maranhão, São Luís 65080-805, MA, Brazil
Interests: antimicrobial activity; bioactive compounds; mechanisms of action; natural products
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
Interests: antimicrobial compounds; models of infection
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The first volume of the Special Issue “Antimicrobial and Anti-infective Activity of Natural Products” was published in 2022. It was a successful issue with 20 published papers and has encouraged us to open a second volume with the same topic.

As a continuation of the Special Issue published in 2022, this second volume will also search for new antimicrobial compounds from natural products, mechanisms of action of pure compounds, in silico evidence of antimicrobial activity, synergistic associations with antibiotics, antimicrobial effects of probiotics and other microorganisms (and their products), and compounds with antivirulence activity or capable of neutralizing microbial resistance. We also welcome the submission of research papers that study alternative methods of demonstrating the in vivo efficacy of compounds or natural extracts.

Manuscripts reporting on the screening of crude plant extracts will not normally be considered for publication in this Section. Manuscripts dealing with zone-of-inhibition assays will not normally be considered for publication; biological activities should be further quantified via MIC, IC50, or other determinations.

Dr. Valério Monteiro-Neto
Dr. Elizabeth S. Fernandes
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Antibiotics is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • antimicrobial activity
  • antivirulence activity
  • natural compounds and extracts
  • probiotics
  • Streptomyces spp. and other antibiotic-producing microorganisms
  • antimicrobial resistance inhibition
  • betalactamase inhibition
  • efflux pump inhibition
  • synergistic associations
  • models of infection
  • clinical assays

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Related Special Issue

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

18 pages, 3963 KiB  
Article
Vismia guianensis Improves Survival of Tenebrio molitor and Mice During Lethal Infection with Candida albicans
by Arthur André Castro Costa, Elizangela Pestana Motta, Aluísio Silva Oliveira, Pamela Gomes Santos, Josivan Regis Farias, Danielle Cristine Gomes Franco, Mayara Cristina Pinto Silva, Nicolle Teixeira Barbosa, Simone Batista Muniz, Luís Douglas Miranda. Silva, Lucilene Amorim Silva, Claudia Quintino Rocha, Flavia Raquel Fernandes Nascimento and Rosane Nassar Meireles Guerra
Antibiotics 2025, 14(1), 72; https://doi.org/10.3390/antibiotics14010072 (registering DOI) - 11 Jan 2025
Viewed by 249
Abstract
Background/Objectives: Vismia guianensis is a vegetal species popularly used to treat fungal infections. This study evaluated the anti-Candida effect of V. guianensis extract after C. albicans lethal infection in Tenebrio molitor larvae and mice. Methods and Results: The chemical profile [...] Read more.
Background/Objectives: Vismia guianensis is a vegetal species popularly used to treat fungal infections. This study evaluated the anti-Candida effect of V. guianensis extract after C. albicans lethal infection in Tenebrio molitor larvae and mice. Methods and Results: The chemical profile analysis of a hydroethanolic extract of the leaves of V. guianensis (EHVG) identified 14 compounds. Two sets of experiments used T. molitor larvae. To evaluate toxicity, the uninfected larvae were treated with EHVG or anthraquinone. We considered the following groups: the controls received PBS; ANFO B received amphotericin B (600 mg/mL); EHVG received the extract; and ANTQ received anthraquinone. The extract and anthraquinone resulted in low-level toxicity in the T. molitor larvae. Another set of experiments evaluated the EHVG effect during lethal infection with Candida albicans. The T. molitor larvae were treated intracelomically (ic/10 μL). Treatment with EHVG efficiently improved the survival of the larvae after lethal infection (60%), probably due to the reduction in CFUs. In the mice, the antifungal effect of EHVG was determined in three groups of immunosuppressed Swiss mice (cyclophosphamide, 50 mg/kg/ip) infected with C. albicans (1 × 107 CFU/ip). The control animals were infected and untreated; the ANFO B animals were infected and treated with amphotericin B (600 µg/kg/ip); and the EHVG animals were infected and treated with the extract (5 mg/kg/orally). A SHAM group (uninfected and untreated) was also included. Survival was assessed for 5 days. The extract increased the mice’s survival (60%) and life expectancy, reducing the CFU counts in the peritoneum and blood. EHVG also increased the number of blood neutrophils and peritoneal macrophages. These systemic activities are likely associated with the presence of flavonoids in the extract. Conclusions: The beneficial effects of EHVG in lethal sepsis are related to an antifungal effect, with the number of CFUs decreasing in the larvae and the mice. In addition, EHVG showed immunological activity in the mice, considering immune cell distribution and cytokine production. Full article
Show Figures

Figure 1

Figure 1
<p>The first-order spectrum of direct flow injection analysis (FIA-ESI-IT-MS) was obtained in the negative mode for EHVG. (<span class="html-fig-inline" id="antibiotics-14-00072-i001"><img alt="Antibiotics 14 00072 i001" src="/antibiotics/antibiotics-14-00072/article_deploy/html/images/antibiotics-14-00072-i001.png"/></span>) Fragments of chemical compounds identified.</p>
Full article ">Figure 2
<p>A visual representation of the toxicity of the hydroethanolic extract of <span class="html-italic">Vismia guianensis</span> (EHVG) in the <span class="html-italic">Tenebrio molitor</span> larvae. The green arrows indicate a chrysalid or chrysalid formation, and the red arrows indicate the dead larvae (n = 15/group).</p>
Full article ">Figure 3
<p>The treatment with <span class="html-italic">V. guianensis</span> hydroethanolic extract (EHVG) and anthraquinone increased the survival rate of the <span class="html-italic">T. molitor</span> larvae lethally infected with <span class="html-italic">C. albicans</span>. The animals were infected with 1 × 10<sup>7</sup> CFU/mL (10 μL) via an intracelomic route (ic) and treated using the same route with 10 μL of EHVG (5 mg/kg, 10 μL) or anthraquinone (5 mg/kg). The EHVG-treated groups were compared to those larvae infected and treated with amphotericin B (ANFO B; 0.6 mg/kg, 10 μL) and with a control group that received PBS (10 μL). The data expressed as percentages were obtained from the Log-Rank (Mantel–Cox) test and a Kaplan–Meier curve considering 15 animals/group. (*) <span class="html-italic">p</span> &lt; 0.05 in comparison to the control.</p>
Full article ">Figure 4
<p>The hydroethanolic extract of <span class="html-italic">V. guianensis</span> leaves (EHVG) reduced the number of <span class="html-italic">C. albicans</span> colonies in <span class="html-italic">T. molitor</span>. The animals were infected with sub-lethal concentrations of <span class="html-italic">C. albicans</span> (5 × 10<sup>4</sup> CFU/mL, ic, 10 μL), and the CFUs were determined 3 days later. The larvae were treated concomitantly (via ic route, 10 μL) with EHVG (5 mg/kg) or amphotericin B (0.6 mg/kg, 10 μL), and then compared with the control group (receiving PBS, 10 μL). The mean ± standard deviation data correspond to 15 animals/group. (*) <span class="html-italic">p</span> &lt; 0.05 in comparison to the control group.</p>
Full article ">Figure 5
<p>The treatment with EHVG improved the survival rate of the mice infected with <span class="html-italic">C. albicans</span>. The animals were immunosuppressed with cyclophosphamide (50 mg/kg, ip), infected 48 h later with <span class="html-italic">C. albicans</span> (1 × 10<sup>7</sup> CFU, ip, 200 μL), and distributed into groups according to treatment (200 μL): EHVG, oral treatment with EHVG (5 mg/kg, 200 μL); ANFO B, treatment with amphotericin B (600 μg/kg/100 μL); and control, receiving PBS. A SHAM group without infection and treatment was also included. The data are expressed as mean ± standard deviation considering 5 animals/group. (*) <span class="html-italic">p</span> &lt; 0.05 compared to PBS.</p>
Full article ">Figure 6
<p>EHVG efficiently reduced the number of CFUs in the blood (<b>A</b>) and the peritoneum (<b>B</b>). The animals were immunosuppressed with cyclophosphamide (50 mg/kg, ip, 200 μL) 48 h before <span class="html-italic">C. albicans</span> infection (1 × 10<sup>7</sup> CFU, ip, 200 μL) and divided into groups according to the treatments. EHVG, the animals were treated orally (immediately after infection) with hydroalcoholic extract of <span class="html-italic">V. guianensis</span> (EHGV; 5 mg/kg, 200 μL); ANFO B, the animals were treated subcutaneously with amphotericin B (600 μg/kg, 100 μL); control, the animals received PBS; SHAM, there was no infection and no treatment. The data are expressed as mean ± standard deviation (n = 5 animals/group). (*) <span class="html-italic">p</span> &lt; 0.05; (**) <span class="html-italic">p</span> &lt; 0.01; and (***) <span class="html-italic">p</span> &lt; 0.001 when compared to the control group.</p>
Full article ">Figure 7
<p>Different effects of EHVG on blood (<b>A</b>), splenic (<b>B</b>), and peritoneal cells (<b>C</b>) from infected mice. Animals were immunosuppressed with cyclophosphamide (50 mg/kg, ip, 200 μL) 48 h before <span class="html-italic">C. albicans</span> infection (1 × 10<sup>7</sup> CFU, ip, 200 μL). Animals were distributed into following groups according to treatment that occurred immediately after infection: EHVG, received extract by oral route (5 mg/kg, 200 μL); ANFO B: received amphotericin B (600 μg/kg, 100 μL) subcutaneously; control group, received sterile PBS; and SHAM, no infection and no treatment. Cells were counted under ordinary light optical microscope (X40). Data represent mean ± standard deviation (n = 5 animals/group). (*) <span class="html-italic">p</span> &lt; 0.05; (**) <span class="html-italic">p</span> &lt; 0.01; and (***) <span class="html-italic">p</span> &lt; 0.001 when comparing EHVG with other groups. (**) <span class="html-italic">p</span> &lt; 0.01 comparing Anfo B and control group.</p>
Full article ">Figure 8
<p>Cell populations in blood (<b>A</b>) and peritoneal fluid (<b>B</b>) from animals immunosuppressed with cyclophosphamide (50 mg/kg, ip, 200 μL) 48 h before <span class="html-italic">C. albicans</span> infection (1 × 10<sup>7</sup>, ip, 200 μL). EHVG, animals infected and treated (immediately after infection) orally with hydroalcoholic extract of <span class="html-italic">V. guianensis</span> (5 mg/kg, 200 μL); ANFO B, animals infected and treated subcutaneously with amphotericin B (600 μg/kg, 100 μL); PBS, infected animals that received PBS (control). Data are expressed as mean ± standard deviation (n = 5 animals/group). (*) <span class="html-italic">p</span> &lt; 0.05 when compared to control, and (#) <span class="html-italic">p</span> &lt; 0.05 when compared to the ANFO B group.</p>
Full article ">Figure 9
<p>Treatment with EHVG modulates cytokine levels: IFN-γ (<b>A</b>), IL-10 (<b>B</b>), and TNF-α (<b>C</b>). Animals were immunosuppressed with cyclophosphamide (50 mg/kg, ip, 200 μL) and lethally infected 48 h later with <span class="html-italic">C. albicans</span> (1 × 10<sup>7</sup> CFU, ip, 200 μL). Animals were divided and treated immediately after infection according to the following groups: EHVG, treated orally with hydroalcoholic extract of <span class="html-italic">Vismia guianensis</span> (5 mg/kg, 200 μL); ANFOB, treated subcutaneously with amphotericin B (600 μg/kg, 100 μL); PBS, infected animals that received sterile PBS (control). Data are expressed as mean ± standard deviation. (**) <span class="html-italic">p</span> &lt; 0.01, and (***) <span class="html-italic">p</span> &lt; 0.001 compared to control, and (#) <span class="html-italic">p</span> &lt; 0.05 compared to ANFOB group.</p>
Full article ">Figure 10
<p>Schematic representation of <span class="html-italic">Tenebrio molitor larvae</span> showing site for infection and treatment between 4th and 5th metamers (<sup>®</sup>Biorender). To evaluate <span class="html-italic">T. molitor</span> lethality, the following aspects were considered: Activity—movement without stimuli (0) or the absence of movement (2). Melanization—those without melanization (0), or black larvae (2). Survival—whether they were alive (0) or dead (2).</p>
Full article ">
19 pages, 4130 KiB  
Article
Identification and Bioactivity Analysis of a Novel Bacillus Species, B. maqinnsis sp. nov. Bos-x6-28, Isolated from Feces of the Yak (Bos grunniens)
by Qiang Ma, Xin Xiang, Yan Ma, Guangzhi Li, Xingyu Liu, Boai Jia, Wenlin Yang, Hengxia Yin and Benyin Zhang
Antibiotics 2024, 13(12), 1238; https://doi.org/10.3390/antibiotics13121238 - 23 Dec 2024
Viewed by 495
Abstract
Background: The identification of novel bacterial species from the intestines of yaks residing on the Qinghai–Tibet Plateau is pivotal in advancing our understanding of host–microbiome interactions and represents a promising avenue for microbial drug discovery. Methods: In this study, we conducted a polyphasic [...] Read more.
Background: The identification of novel bacterial species from the intestines of yaks residing on the Qinghai–Tibet Plateau is pivotal in advancing our understanding of host–microbiome interactions and represents a promising avenue for microbial drug discovery. Methods: In this study, we conducted a polyphasic taxonomic analysis and bioactive assays on a Bacillus strain, designated Bos-x6-28, isolated from yak feces. Results: The findings revealed that strain Bos-x6-28 shares a high 16S rRNA gene sequence similarity (98.91%) with B. xiamenensis HYC-10T and B. zhangzhouensis DW5-4T, suggesting close phylogenetic affinity. Physiological and biochemical characterizations demonstrated that Bos-x6-28 could utilize nine carbon sources, including D-galactose, inositol, and fructose, alongside nine nitrogen sources, such as threonine, alanine, and proline. Analysis of biochemical markers indicated that Bos-x6-28’s cell wall hydrolysates contained mannose, glucose, and meso-2,6-diaminopimelic acid, while menaquinone-7 (MK-7), phosphatidylethanolamine (PE), phosphatidylcholine (PC), and phosphatidylglycerol (DPG) were found in the cell membrane. The primary cellular fatty acids included C16:0 (28.00%), cyclo-C17:0 (19.97%), C14:0 (8.75%), cyclo-C19:0 (8.52%), iso-C15:0 (5.49%), anteiso-C15:0 (4.61%), and C12:0 (3.15%). Whole-genome sequencing identified a genome size of 3.33 Mbp with 3353 coding genes. Digital DNA–DNA hybridization (dDDH) and average nucleotide identity (ANI) analyses confirmed Bos-x6-28 as a novel species, hereby named B. maqinnsis Bos-x6-28 (MCCC 1K09379). Further genomic analysis unveiled biosynthetic gene clusters encoding bioactive natural compounds, including β-lactones, sactipeptides, fengycin, and lichenysin analogs. Additionally, in vitro assays demonstrated that this strain exhibits antibacterial and cytotoxic activities. Conclusions: These findings collectively indicate the novel Bacillus species B. maqinnsis Bos-x6-28 as a promising source for novel antibiotic and antitumor agents. Full article
Show Figures

Figure 1

Figure 1
<p>Phylogenetic analysis of strain Bos-x6-28 based on 16S rRNA gene sequence using the neighbor-joining (NJ) method. The sequence numbers in parentheses represent the GenBank accession numbers of the corresponding strains. The scale bar indicates a 0.02 nucleotide divergence per site.</p>
Full article ">Figure 2
<p>Microscopic structure of strain Bos-x6-28. (<b>A</b>) Scanning electron microscopy (SEM) at 20,000× magnification, showing the detailed ultrastructure of Bos-x6-28. (<b>B</b>) Light microscopy at 1000× magnification after Gram staining, illustrating cellular morphology and Gram characteristics.</p>
Full article ">Figure 3
<p>Two-dimensional TLC analysis of phospholipid components in strain Bos-x6-28. (<b>A</b>) Ninhydrin staining; (<b>B</b>) Anisaldehyde staining; (<b>C</b>) Phosphomolybdic acid staining.</p>
Full article ">Figure 4
<p>Circular genome map of strain Bos-x6-28. The outermost ring denotes the genomic size, with each tick mark representing 5 kb. The second and third rings display genes on the positive and negative strands, respectively, with different colors indicating various COG functional classifications. The fourth ring represents repeat sequences. The fifth ring shows tRNA (blue) and rRNA (purple) genes. The sixth ring illustrates GC content, where light yellow areas indicate regions with GC content higher than the genome’s average, with peak heights corresponding to the extent of deviation from the mean; blue areas denote regions with GC content below the genomic average. The innermost ring represents the GC-skew, with dark gray indicating regions where G content exceeds C content, and red indicating regions where C content exceeds G content.</p>
Full article ">Figure 5
<p>Comparative genomic analysis of strain Bos-x6-28 and its closely related species. The outermost ring represents the strain origin and shared core gene clusters, followed by the geometric homogeneity index and functional homogeneity index. Subsequent rings display data for <span class="html-italic">B. safensis</span> subsp. <span class="html-italic">safensis</span> FO-36b, <span class="html-italic">B. pumilus</span> NCTC10337, <span class="html-italic">B. australimaris</span> NH7I-1, <span class="html-italic">B. altitudinis</span> 41KF2b, <span class="html-italic">B. zhangzhouensis</span> DW5-4, <span class="html-italic">B. xiamenensis</span> HYC-10, <span class="html-italic">B. safensis</span> subsp. <span class="html-italic">osmophilus</span> BC09, and Bos-x6-28. The heat map shows the ANI values among these strains, with all ANI values below 95% (indicated in pink), highlighting the genomic divergence among the strains.</p>
Full article ">Figure 6
<p>Bioactivity of secondary metabolites from strain Bos-x6-28. (<b>A</b>) Inhibitory activity against <span class="html-italic">B. subtilis</span>, with ampicillin as a positive control and methanol as a negative control. (<b>B</b>) Inhibitory activity against human liver cancer cells (HepG2), using DMSO as the blank control. Data are presented as mean ± SD, derived from three independent experiments conducted in triplicate. Statistical significance is indicated as *** <span class="html-italic">p</span> &lt; 0.001 compared to control cells, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to control cells (Wilcoxon <span class="html-italic">t</span>-test).</p>
Full article ">
20 pages, 3131 KiB  
Article
Ellagic Acid Potentiates the Inhibitory Effects of Fluconazole Against Candida albicans
by Amanda Graziela Gonçalves Mendes, Carmem Duarte Lima Campos, José Lima Pereira-Filho, Aleania Polassa Almeida Pereira, Gabriel Silva Abrantes Reis, Árlon Wendel de Marinho Silva Araújo, Pablo de Matos Monteiro, Flávia Castello Branco Vidal, Silvio Gomes Monteiro, Isabella Fernandes da Silva Figueiredo, Elizabeth Soares Fernandes, Cristina de Andrade Monteiro and Valério Monteiro-Neto
Antibiotics 2024, 13(12), 1174; https://doi.org/10.3390/antibiotics13121174 - 4 Dec 2024
Viewed by 613
Abstract
Background/Objectives: Antifungal resistance to azoles, coupled with the increasing prevalence of Candida albicans infections, represents a significant public health challenge and has driven the search for new natural compounds that can act as alternatives or adjuvants to the current antifungals. Ellagic acid (EA) [...] Read more.
Background/Objectives: Antifungal resistance to azoles, coupled with the increasing prevalence of Candida albicans infections, represents a significant public health challenge and has driven the search for new natural compounds that can act as alternatives or adjuvants to the current antifungals. Ellagic acid (EA) has demonstrated antifungal activity; however, its effects are not fully understood. In this study, we investigated the in vitro anti-Candida activity of EA and its ability to potentiate the effects of fluconazole (FLZ) on C. albicans. Methods: The Minimum Inhibitory Concentration (MIC) of EA was determined by broth microdilution and its interaction with FLZ was assessed using a checkerboard assay. Additionally, we examined the effects of EA on yeast-to-hypha transition, inhibition of biofilm formation, time–kill kinetics, hemolytic activity, and cytotoxicity in HeLa ATCC® CCL-2™ cells. Results: EA exhibited MIC values ranging from 250 to 2000 µg/mL and showed synergistic and additive interactions with FLZ, resulting in a marked reduction in the MIC values of FLZ (up to 32-fold) and EA (up to 16-fold). In the time–kill assay, the most effective combinations were 4× EA MIC, 2× EA MIC, and FIC EA + FLZ, which showed fungicidal activity. Furthermore, EA did not show hemolytic activity and demonstrated low and dose-dependent cytotoxicity in HeLa cells, with no cytotoxic effects observed in combination with FLZ. EA and the synergistic combination of EA and FLZ interfered with both the yeast-to-hypha transition process in C. albicans cells and biofilm formation. In addition to its antifungal efficacy, EA demonstrated a favorable safety profile at the concentrations used. Conclusions: This study presents promising results regarding the potential use of EA in combination with FLZ for the treatment of C. albicans infections. Full article
Show Figures

Figure 1

Figure 1
<p>Time-kill curve of <span class="html-italic">C. albicans</span> ATCC 90028 under the action of EA (<b>A</b>) and FLZ (<b>B</b>) and their combination (FIC EA + FLZ) (<b>C</b>) showing the medians. The Shapiro–Wilk test was used to assess the normality of the sample. Significant differences between groups were determined by Kruskal–Wallis analysis followed by Dunn’s test with <span class="html-italic">p</span> &gt; 0.05 considered significant. Legend: FIC = fractional inhibitory combination, MIC = minimal inhibitory concentration, EA = ellagic acid, FLZ = fluconazole.</p>
Full article ">Figure 2
<p>Time-kill curve of <span class="html-italic">C. albicans</span> CA 08 under the action of EA (<b>A</b>), FLZ (<b>B</b>) and in combination (FIC EA + FLZ) (<b>C</b>), at different concentrations and control. The Shapiro–Wilk test was applied to analyze normality. Significant differences between groups were determined by Kruskal-Walli’s analysis followed by Dunn’s test, presenting the medians test with (<span class="html-italic">p</span> &gt; 0.05) considered significant. Legend: FIC = fractional inhibitory combinations, MIC = minimal inhibitory concentration, EA = ellagic acid, FLZ = fluconazole.</p>
Full article ">Figure 3
<p><span class="html-italic">C. albicans</span> hyphae formation. Clinical isolates: (<b>A</b>) CA 90028; (<b>B</b>) CA 013; (<b>C</b>) CA 010; (<b>D</b>) CA08. The Shapiro–Wilk normality test was applied. The results are presented as mean and standard deviation; significant differences between groups were determined by one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison where the value of <span class="html-italic">p</span> &lt; 0.05 (*) was considered significant.</p>
Full article ">Figure 4
<p>Cell viability of young <span class="html-italic">C. albicans</span> biofilms. Clinical isolates: (<b>A</b>) CA 90028; (<b>B</b>) CA 013; (<b>C</b>) CA 010; (<b>D</b>) CA08. The Shapiro–Wilk normality test was applied. Results are presented as mean and standard deviation; significant differences between groups were determined by one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison where the value of <span class="html-italic">p</span> &lt; 0.05 (*) was significant.</p>
Full article ">Figure 5
<p>Cell viability of mature <span class="html-italic">C. albicans</span> biofilms. Clinical isolates: (<b>A</b>) CA 90028; (<b>B</b>) CA 013; (<b>C</b>) CA 010; (<b>D</b>) CA08. The Shapiro–Wilk normality test was applied. Significant differences between groups were determined by Kruskal–Wallis analysis of variance followed by Dunn’s test, where the value of <span class="html-italic">p</span> &lt; 0.05 (*) was significant.</p>
Full article ">Figure 6
<p>Hemolytic activity. The Shapiro–Wilk normality test was applied. The test results are presented with mean and standard deviation; significant differences between specific groups were determined by one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison, where the value of (* <span class="html-italic">p</span> &lt; 0.05) was significant.</p>
Full article ">Figure 7
<p>Cytotoxicity assay in HeLa. Cell viability was evaluated after (<b>A</b>) 24 h, (<b>B</b>) 48 h, and (<b>C</b>) 72 h. The Shapiro–Wilk normality test was applied. Significant differences between groups were determined by Kruskal–Wallis analysis followed by Dunn’s multiple comparisons test, where the value of (* <span class="html-italic">p</span> &lt; 0.05) was significant. EA = ellagic acid; FLZ = fluconazole.</p>
Full article ">
23 pages, 6586 KiB  
Article
Studies Regarding Antimicrobial Properties of Some Microbial Polyketides Derived from Monascus Strains
by Daniela Albisoru, Nicoleta Radu, Lucia Camelia Pirvu, Amalia Stefaniu, Narcisa Băbeanu, Rusandica Stoica and Dragos Paul Mihai
Antibiotics 2024, 13(11), 1092; https://doi.org/10.3390/antibiotics13111092 - 16 Nov 2024
Viewed by 1028
Abstract
Finding new molecules to prevent the growth of antimicrobial resistance is a hot topic for scientists worldwide. It has been reported that some raw bioproducts containing Monascus polyketides have antimicrobial activities, but extensive studies on this effect have not been conducted. In this [...] Read more.
Finding new molecules to prevent the growth of antimicrobial resistance is a hot topic for scientists worldwide. It has been reported that some raw bioproducts containing Monascus polyketides have antimicrobial activities, but extensive studies on this effect have not been conducted. In this context, our studies aimed to evaluate the antimicrobial properties of six raw bioproducts containing three classes of microbial polyketides biosynthesized by three Monascus strains through solid-state biosynthesis. As a methodology, we performed in silico predictions using programs such as PyMOL v3.0.4 and employed ESI-MS techniques to provide evidence of the presence of the six studied compounds in our bioproducts. The results obtained in silico were validated through in vitro studies using the Kirby-Bauer diffusion method on bacteria and fungi. The test performed in silico showed that Monascorubramine has the highest affinity for both Gram-positive and Gram-negative bacteria, followed by yellow polyketides such as Ankaflavin and Monascin. The estimated pharmacokinetic parameters indicated high gastrointestinal absorption and the potential to cross the blood-brain barrier for all studied compounds. However, the compounds also inhibit most enzymes involved in drug metabolism, presenting some level of toxicity. The best in vitro results were obtained for S. aureus, with an extract containing yellow Monascus polyketides. Predictions made for E. coli were validated in vitro for P. aeruginosa, S. enterica, and S. marcescens, as well as for fungi. Significant antibacterial properties were observed during this study for C. albicans, S. aureus, and fungal dermatophytes for crude bioproducts containing Monascus polyketides. In conclusion, the antimicrobial properties of Monascus polyketides were validated both in silico and in vitro. However, due to their potential toxicity, these bioproducts would be safer to use as topical formulations. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental study design.</p>
Full article ">Figure 2
<p>Molecular docking validation—superposition of predicted poses (pink) of co-crystallized inhibitors on initial conformations (green): (<b>a</b>) trimethoprim in saDHFR binding site (PDB ID: 2w9s, RMSD 0.6535 Å); (<b>b</b>) trimethoprim in ecDHFR binding site (PDB ID: 7mym, RMSD 0.3521 Å); (<b>c</b>) UCP11E in caDHFR binding site (PDB ID: 4hoe, RMSD 0.4389 Å); (<b>d</b>) trimethoprim in hDHFR binding site (PDB ID: 2w3a, RMSD 0.9559 Å).</p>
Full article ">Figure 3
<p>Predicted binding poses of Monascorubramine in DHFR active sites. (<b>a</b>) saDHFR; (<b>b</b>) ecDHFR; (<b>c</b>) caDHFR; (<b>d</b>) hDHFR.</p>
Full article ">Figure 4
<p>2D diagrams of predicted molecular interactions between Monascorubramine and active sites of DHFR homologues. (<b>a</b>) saDHFR; (<b>b</b>) ecDHFR; (<b>c</b>) caDHFR; (<b>d</b>) hDHFR.</p>
Full article ">Figure 5
<p>“Boiled egg” diagram illustrating the distribution of the investigated compounds in the chemical space of molecules that are absorbed in the gastrointestinal (GI) tract or passively permeate the blood–brain barrier (BBB) based on calculated WlogP (octanol/water partition coefficient) and TPSA (topological polar surface area) values. Molecules located in the “egg yolk” are predicted to passively permeate through the BBB. Molecules located in the white area are predicted to be passively absorbed in the GI tract.</p>
Full article ">Figure 6
<p>ESI-MS analysis of a total alcoholic extract of the following: (<b>a</b>) <span class="html-italic">Monascus purpureus</span>; (<b>b</b>) <span class="html-italic">Monascus ruber</span>; (<b>c</b>) <span class="html-italic">Monascus</span> sp. 3 <span class="html-italic">(Monascus ruber</span>; highly productive).</p>
Full article ">Figure 7
<p>Antibacterial properties of polyketides obtained from Monascus-derived bioproducts: (<b>a</b>) antibacterial properties for <span class="html-italic">S. aureus</span> (yellow polyketides exhibit the best activities); (<b>b</b>) antibacterial properties for <span class="html-italic">S. aureus</span> MRSA (yellow polyketides exhibit moderate activities); (<b>c</b>) antibacterial properties for <span class="html-italic">S. marcescens</span> (red polyketides exhibit the best activities); (<b>d</b>) antibacterial properties for <span class="html-italic">P. aeruginosa</span> (red polyketides exhibit moderate antimicrobial activities); (<b>e</b>) antibacterial properties for <span class="html-italic">S. enterica</span> (red polyketides exhibit local-moderate antimicrobial activities).</p>
Full article ">Figure 7 Cont.
<p>Antibacterial properties of polyketides obtained from Monascus-derived bioproducts: (<b>a</b>) antibacterial properties for <span class="html-italic">S. aureus</span> (yellow polyketides exhibit the best activities); (<b>b</b>) antibacterial properties for <span class="html-italic">S. aureus</span> MRSA (yellow polyketides exhibit moderate activities); (<b>c</b>) antibacterial properties for <span class="html-italic">S. marcescens</span> (red polyketides exhibit the best activities); (<b>d</b>) antibacterial properties for <span class="html-italic">P. aeruginosa</span> (red polyketides exhibit moderate antimicrobial activities); (<b>e</b>) antibacterial properties for <span class="html-italic">S. enterica</span> (red polyketides exhibit local-moderate antimicrobial activities).</p>
Full article ">Figure 8
<p>Antifungal properties of polyketides obtained from Monascus-derived bioproducts for the following: (<b>a</b>) <span class="html-italic">Candida albicans</span>; (<b>b</b>) <span class="html-italic">S. brevicaulis</span>, (<b>c</b>) <span class="html-italic">M. gypseum</span>; (<b>d</b>) <span class="html-italic">T. mentagrophytes</span>.</p>
Full article ">Figure 9
<p>Flow diagram used to obtain enhanced extracts of yellow, orange, and red polyketides: (<b>a</b>) Solid-state biosynthesis of <span class="html-italic">Monascus</span> bioproducts (RYR); (<b>b</b>) Sample preparation of <span class="html-italic">Monascus</span> bioproducts for analysis; (<b>c</b>) Obtaining <span class="html-italic">Monascus</span> extract with yellow polyketides; (<b>d</b>) Obtaining <span class="html-italic">Monascus</span> extract with orange polyketides; (<b>e</b>) Obtaining <span class="html-italic">Monascus</span> extract with red polyketides.</p>
Full article ">Figure 9 Cont.
<p>Flow diagram used to obtain enhanced extracts of yellow, orange, and red polyketides: (<b>a</b>) Solid-state biosynthesis of <span class="html-italic">Monascus</span> bioproducts (RYR); (<b>b</b>) Sample preparation of <span class="html-italic">Monascus</span> bioproducts for analysis; (<b>c</b>) Obtaining <span class="html-italic">Monascus</span> extract with yellow polyketides; (<b>d</b>) Obtaining <span class="html-italic">Monascus</span> extract with orange polyketides; (<b>e</b>) Obtaining <span class="html-italic">Monascus</span> extract with red polyketides.</p>
Full article ">
15 pages, 3630 KiB  
Article
Anti-Biofilm Action of Cineole and Hypericum perforatum to Combat Pneumonia-Causing Drug-Resistant P. aeruginosa
by Sourav Chakraborty, Piyush Baindara, Pralay Sharma, Austin Jose T, Kumaravel V, Raja Manoharan and Santi M. Mandal
Antibiotics 2024, 13(8), 689; https://doi.org/10.3390/antibiotics13080689 - 24 Jul 2024
Viewed by 1636
Abstract
Hospital-acquired antibiotic-resistant pneumonia is one of the major causes of mortality around the world that pose a catastrophic threat. Pseudomonas aeruginosa is one of the most significant opportunistic pathogens responsible for hospital-acquired pneumonia and gained resistance to the majority of conventional antibiotics. There [...] Read more.
Hospital-acquired antibiotic-resistant pneumonia is one of the major causes of mortality around the world that pose a catastrophic threat. Pseudomonas aeruginosa is one of the most significant opportunistic pathogens responsible for hospital-acquired pneumonia and gained resistance to the majority of conventional antibiotics. There is an urgent need for antibiotic alternatives to control drug-resistant pneumonia and other related respiratory infections. In the present study, we explored the antibacterial potential of cineole in combination with homeopathic medicines against biofilm-forming drug-resistant P. aeruginosa. Out of 26 selected and screened homeopathic medicines, Hypericum Perforatum (HyPer) was found to eradicate biofilm-forming drug-resistant P. aeruginosa most effectively when used in combination with cineole. Interestingly, the synergistic action of HyPer and cineole was also found to be similarly effective against planktonic cells of P. aeruginosa. Further, the potential synergistic killing mechanisms of cineole and HyPer were determined by analyzing zeta membrane potential, outer membrane permeability, and DNA release from P. aeruginosa cells upon treatment with cineole and HyPer. Additionally, molecular docking analysis revealed strong binding affinities of hypericin (an active ingredient of HyPer) with the PqsA (a quorum sensing protein) of P. aeruginosa. Overall, our findings revealed the potential synergistic action of cineole and HyPer against biofilm-forming drug-resistant P. aeruginosa. Cineole and HyPer could be used in combination with other bronchodilators as inhalers to control the biofilm-forming drug-resistant P. aeruginosa. Full article
Show Figures

Figure 1

Figure 1
<p>Primary screening of 26 homeopathic medicines against drug-resistant <span class="html-italic">P. aeruginosa</span>. <span class="html-italic">Hypericum perforatum</span> displayed the highest anti-biofilm activity, represented in a maroon color bar. Error bars show a standard deviation (SD) while statistical significance is considered at the level of <span class="html-italic">p</span> &lt; 0.05 (indicated as red stars above bar). The experiment was performed three times independently in triplicate.</p>
Full article ">Figure 2
<p>Synergistic antibacterial activity of HyPer and cineole. (<b>A</b>) Well diffusion assay displaying the synergistic action of HyPer and cineole. Well numbers 1, 2, 3, 4, and 5 indicate 5 μL, 10 μL, 20 μL, 40 μL, and 60 μL of HyPer (30 CH), respectively, in combination with 125 μg/mL of Cn. Cn alone was used as a control. (<b>B</b>) MIC determination of HyPer in combination with 125 μg/mL of cineole. T1, T2, T3, T4, and T5 indicate 5 μL, 10 μL, 20 μL, 40 μL, and 60 μL of HyPer (30 CH), respectively, in combination with 125 μg/mL of Cn. <span class="html-italic">P. aeruginosa</span> treated with Cn alone and 30% ethanol in PBS (UT) were used as controls. Error bars show a standard deviation (SD) while statistical significance is considered at the level of <span class="html-italic">p</span> &lt; 0.05 (indicated as red stars above bars). All experiments were performed three times independently in triplicate.</p>
Full article ">Figure 3
<p>Membrane zeta potential of untreated and treated <span class="html-italic">P. aeruginosa</span> cells with cineole, HyPer, and a combination of both. <span class="html-italic">P. aeruginosa</span> treated with 30% ethanol in PBS was used as a control. Error bars show a standard deviation (SD) while statistical significance is considered at the level of <span class="html-italic">p</span> &lt; 0.05 (indicated as red stars above bars). The experiment was performed three times independently in triplicate.</p>
Full article ">Figure 4
<p>Outer membrane permeability of untreated and treated <span class="html-italic">P. aeruginosa</span> cells with cineole, HyPer, and a combination of both for different time points of treatment: (<b>A</b>) 1 h, (<b>B</b>) 2 h, and (<b>C</b>) 3 h. <span class="html-italic">P. aeruginosa</span> cells treated with 30% ethanol in PBS were used as a control. Error bars show a standard deviation (SD) while statistical significance is considered at the level of <span class="html-italic">p</span> &lt; 0.05 (indicated as red stars). All experiments were performed three times independently in triplicate.</p>
Full article ">Figure 5
<p>Intracellular leakage of untreated and treated <span class="html-italic">P. aeruginosa</span> cells with cineole, HyPer, and a combination of both for the time points of 0, 5, 10, 20, and 30 min. <span class="html-italic">P. aeruginosa</span> cells treated with 30% ethanol in PBS were used as a control. Error bars show a standard deviation (SD) while statistical significance is considered at the level of <span class="html-italic">p</span> &lt; 0.05 (indicated as red stars). The experiment was performed three times independently in triplicate.</p>
Full article ">Figure 6
<p>Molecular docking and interaction of cineole and hypericin with PqsA, a biofilm-forming protein of <span class="html-italic">P. aeruginosa</span>. (<b>A</b>) Docked complex of cineole and PqsA. The dotted red line circle highlights the position of cineole in the complex. (<b>B</b>) Three-dimensional representation of interacting amino acid residues of PqsA with cineole upon docking. (<b>C</b>) Docked complex of hypericin and PqsA. The dotted red line circle highlights the position of hypericin in the complex. (<b>D</b>) Three-dimensional representation of interacting amino acid residues of PqsA with hypercin upon docking. PqsA is shown as a yellow ribbon while cineole, hypercin, and interactive amino acid residues are shown as sticks. Interacting bonds are represented as dotted green, purple, and violet lines.</p>
Full article ">
17 pages, 3725 KiB  
Article
Artemisinin May Disrupt Hyphae Formation by Suppressing Biofilm-Related Genes of Candida albicans: In Vitro and In Silico Approaches
by Esra Sumlu, Merve Aydin, Emine Nedime Korucu, Saliha Alyar and Ahmed Moustapha Nsangou
Antibiotics 2024, 13(4), 310; https://doi.org/10.3390/antibiotics13040310 - 28 Mar 2024
Cited by 1 | Viewed by 2403
Abstract
This study aimed to assess the antifungal and antibiofilm efficacy of artemisinin against Candida (C.) species, analyze its impact on gene expression levels within C. albicans biofilms, and investigate the molecular interactions through molecular docking. The antifungal efficacy of artemisinin on a variety [...] Read more.
This study aimed to assess the antifungal and antibiofilm efficacy of artemisinin against Candida (C.) species, analyze its impact on gene expression levels within C. albicans biofilms, and investigate the molecular interactions through molecular docking. The antifungal efficacy of artemisinin on a variety of Candida species, including fluconazole-resistant and -susceptible species, was evaluated by the microdilution method. The effect of artemisinin on C. albicans biofilm formation was investigated by MTT and FESEM. The mRNA expression of the genes related to biofilm was analyzed by qRT-PCR. In addition, molecular docking analysis was used to understand the interaction between artemisinin and C. albicans at the molecular level with RAS1-cAMP-EFG1 and EFG1-regulated genes. Artemisinin showed higher sensitivity against non-albicans Candida strains. Furthermore, artemisinin was strongly inhibitory against C. albicans biofilms at 640 µg/mL. Artemisinin downregulated adhesion-related genes ALS3, HWP1, and ECE1, hyphal development genes UME6 and HGC1, and hyphal CAMP-dependent protein kinase regulators CYR1, RAS1, and EFG1. Furthermore, molecular docking analysis revealed that artemisinin and EFG1 had the highest affinity, followed by UME6. FESEM analysis showed that the fluconazole- and artemisinin-treated groups exhibited a reduced hyphal network, unusual surface bulges, and the formation of pores on the cell surfaces. Our study suggests that artemisinin may have antifungal potential and showed a remarkable antibiofilm activity by significantly suppressing adhesion and hyphal development through interaction with key proteins involved in biofilm formation, such as EFG1. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of artemisinin on the expression of biofilm-related genes in strain (<b>a</b>) 7A, (<b>b</b>) 66A, and (<b>c</b>) <span class="html-italic">C. albicans</span> ATCC 10231. 18S ribosomal RNA was used for normalization of gene expression levels. Values are expressed as mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05, significantly different from the control; # <span class="html-italic">p</span> &lt; 0.05, significantly different from the fluconazole.</p>
Full article ">Figure 2
<p>Molecular association of artemisinin with EFG1, UME6, HGC1, RAS1, CYR1, HWP1, ECE1, and ALS3 in <span class="html-italic">C. albicans</span> ATCC 10231. Each graph shows (<b>A</b>) molecular binding mode (dashed thick orange line, hydrogen bonds; unbound amino acids, steric interactions); (<b>B</b>) 3D interaction docked pose view (green area: active site); (<b>C</b>) two-dimensional representation of hydrogen bonds made by artemisinin and related protein.</p>
Full article ">Figure 2 Cont.
<p>Molecular association of artemisinin with EFG1, UME6, HGC1, RAS1, CYR1, HWP1, ECE1, and ALS3 in <span class="html-italic">C. albicans</span> ATCC 10231. Each graph shows (<b>A</b>) molecular binding mode (dashed thick orange line, hydrogen bonds; unbound amino acids, steric interactions); (<b>B</b>) 3D interaction docked pose view (green area: active site); (<b>C</b>) two-dimensional representation of hydrogen bonds made by artemisinin and related protein.</p>
Full article ">Figure 3
<p>(<b>A</b>) Molecular binding mode of EFG1 and FLC (dashed thick orange line, hydrogen bonds; unbound amino acids, steric interactions); (<b>B</b>) the 3D interaction docked pose view (green area: active site); (<b>C</b>) two-dimensional representation of hydrogen bonds made by FLC and EFG1.</p>
Full article ">Figure 4
<p>Root mean square deviation (RMSD) values of genes–artemisinin.</p>
Full article ">Figure 5
<p>FESEM images of <span class="html-italic">C. albicans</span> ATCC 10231 biofilms after control and 24 h exposure to artemisinin and fluconazole. (<b>A</b>–<b>C</b>)<b>.</b> Untreated biofilm culture. (<b>D</b>–<b>F</b>)<b>.</b> Biofilm culture treated with 640 μg/mL artemisinin. (<b>G</b>–<b>I</b>)<b>.</b> Biofilm culture treated with 4 μg/mL fluconazole. Magnification scales of 500×, 2000×, and 50,000× were used for imaging. Arrows indicate unusual surfaces and pore formation. The frame shows the selected region.</p>
Full article ">
20 pages, 9595 KiB  
Article
Antimicrobial Activity of Bacillus amyloliquefaciens BS4 against Gram-Negative Pathogenic Bacteria
by Ana Paula Palacios-Rodriguez, Abraham Espinoza-Culupú, Yerson Durán and Tito Sánchez-Rojas
Antibiotics 2024, 13(4), 304; https://doi.org/10.3390/antibiotics13040304 - 28 Mar 2024
Cited by 1 | Viewed by 2668
Abstract
Worldwide, bacterial resistance is one of the most severe public health problems. Currently, the failure of antibiotics to counteract superbugs highlights the need to search for new molecules with antimicrobial potential to combat them. The objective of this research was to evaluate the [...] Read more.
Worldwide, bacterial resistance is one of the most severe public health problems. Currently, the failure of antibiotics to counteract superbugs highlights the need to search for new molecules with antimicrobial potential to combat them. The objective of this research was to evaluate the antimicrobial activity of Bacillus amyloliquefaciens BS4 against Gram-negative bacteria. Thirty yeasts and thirty-two Bacillus isolates were tested following the agar well-diffusion method. Four Bacillus sp. strains (BS3, BS4, BS17, and BS21) showed antagonistic activity against E. coli ATCC 25922 using bacterial culture (BC) and the cell-free supernatant (CFS), where the BS4 strain stood out, showing inhibitory values of 20.50 ± 0.70 mm and 19.67 ± 0.58 mm for BC and CFS, respectively. The Bacillus sp. BS4 strain can produce antioxidant, non-hemolytic, and antimicrobial metabolites that exhibit activity against several microorganisms such as Salmonella enterica, Klebsiella pneumoniae, Shigella flexneri, Enterobacter aerogenes, Proteus vulgaris, Yersinia enterocolitica, Serratia marcescens, Aeromonas sp., Pseudomonas aeruginosa, Candida albicans, and Candida tropicalis. According to the characterization of the supernatant, the metabolites could be proteinaceous. The production of these metabolites is influenced by carbon and nitrogen sources. The most suitable medium to produce antimicrobial metabolites was TSB broth. The one-factor-at-a-time method was used to standardize parameters such as pH, agitation, temperature, carbon source, nitrogen source, and salts, resulting in the best conditions of pH 7, 150 rpm, 28 °C, starch (2.5 g/L), tryptone (20 g/L), and magnesium sulfate (0.2 g/L), respectively. Moreover, the co-culture was an excellent strategy to improve antimicrobial activity, achieving maximum antimicrobial activity with an inhibition zone of 21.85 ± 1.03 mm. These findings position the Bacillus amyloliquefaciens BS4 strain as a promising candidate for producing bioactive molecules with potential applications in human health. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Phenotypic characterization of the <span class="html-italic">Bacillus</span> sp. BS4 strain: (<b>a</b>) strain growth on TSA agar at 24 h reveals irregular size and shape, with a raised, butyrous appearance and mucoid consistency, observed using a stereomicroscope with 10× magnification; and (<b>b</b>) Gram stain (100× magnification).</p>
Full article ">Figure 2
<p>Effect of agitation, pH, and temperature on antimicrobial metabolite production of the BS4 strain against <span class="html-italic">E. coli</span> ATCC 25922.</p>
Full article ">Figure 3
<p>Effects of various carbon sources on antimicrobial activity of the <span class="html-italic">Bacillus</span> sp. BS4 strain. (<b>a</b>) Bar graph represents the effect of carbon sources and (<b>b</b>) Inhibition zones of CFS. Distinct letters represent statistically significant differences between treatments based on the LSD test (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Effects of various nitrogen sources on antimicrobial activity of the <span class="html-italic">Bacillus</span> sp. BS4 strain. (<b>a</b>) Bar graph represents the effect of nitrogen sources and (<b>b</b>) Inhibition zones of CFS Distinct letters represent statistically significant differences between treatments based on the LSD test (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Effects of various mineral salts on antimicrobial activity of the <span class="html-italic">Bacillus</span> sp. BS4 strain. (<b>a</b>) Bar graph represents the effect of mineral salts sources and (<b>b</b>) Inhibition zones of CFS. Distinct letters represent statistically significant differences between treatments based on the LSD test (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Comparison of the growth kinetics of the BS4 strain in TSB (<span class="html-fig-inline" id="antibiotics-13-00304-i001"><img alt="Antibiotics 13 00304 i001" src="/antibiotics/antibiotics-13-00304/article_deploy/html/images/antibiotics-13-00304-i001.png"/></span>) and MOD broth (<span class="html-fig-inline" id="antibiotics-13-00304-i002"><img alt="Antibiotics 13 00304 i002" src="/antibiotics/antibiotics-13-00304/article_deploy/html/images/antibiotics-13-00304-i002.png"/></span>). The error bars in the figure indicate the standard deviations from three replicates.</p>
Full article ">Figure 7
<p>Comparisons of the antimicrobial activity of the BS4 strain in TSB broth and MOD broth. Distinct letters represent statistically significant differences between treatments based on the LSD test where ns, not significant; a, <span class="html-italic">p</span> &lt; 0.05; b, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Evaluation of antioxidant (<b>a</b>) and anti-hemolytic (<b>b</b>) activity of the <span class="html-italic">Bacillus</span> sp. BS4 strain. FC, entire fraction of cell-free supernatant; F3K, 3 kDa fraction; MC, culture medium; T, Trolox; ABTS, solution of ABTS<sup>+</sup> (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid).</p>
Full article ">Figure 9
<p>MIC of different Gram-negative bacteria in the presence of different concentrations of CFS. These data represent the mean (±SEM) of three independent experiments (bs, bronchial secretion; ci, clinical isolation).</p>
Full article ">Figure 10
<p>Phylogenetic relationship of strains BS3 and BS4 (Δ) with the type strains of the <span class="html-italic">Bacillus</span> genus. Sequences were aligned using MUSCLE (MEGA software version 11), and phylogenetic inferences were obtained using neighbor joining with 1000 bootstrap replicates in MEGA 11. <span class="html-italic">Brevibacillus laterosporus</span> DSM25T (AB112720) and <span class="html-italic">Alicyclobacillus acidocaldarius</span> (AB089859) were used as outgroups. Scale bar = 1% nucleotide sequence divergence.</p>
Full article ">

Review

Jump to: Research

21 pages, 1602 KiB  
Review
A Comprehensive Review on the Antibacterial, Antifungal, Antiviral, and Antiparasitic Potential of Silybin
by José Lima Pereira-Filho, Amanda Graziela Gonçalves Mendes, Carmem Duarte Lima Campos, Israel Viegas Moreira, Cinara Regina Aragão Vieira Monteiro, Suzany Hellen da Silva Soczek, Elizabeth Soares Fernandes, Rafael Cardoso Carvalho and Valério Monteiro-Neto
Antibiotics 2024, 13(11), 1091; https://doi.org/10.3390/antibiotics13111091 - 15 Nov 2024
Viewed by 1383
Abstract
Silybin, a flavonolignan extracted from the seeds of the plant species Silybum marianum (L.) Gaertn., has a variety of pharmacological activities, including antimicrobial activity against several microorganisms of clinical interest. This review analyzes the existing studies on silybin’s antimicrobial activity and possible mechanisms [...] Read more.
Silybin, a flavonolignan extracted from the seeds of the plant species Silybum marianum (L.) Gaertn., has a variety of pharmacological activities, including antimicrobial activity against several microorganisms of clinical interest. This review analyzes the existing studies on silybin’s antimicrobial activity and possible mechanisms of action. Silybin has been shown to inhibit the growth of Gram-positive and Gram-negative bacteria, as well as some fungi, viruses, and protozoa. In general, possible mechanisms of antimicrobial action include the inhibition of efflux pumps, prevention of biofilm formation, reduction of the expression of virulence factors, induction of apoptosis-like effects, and plasma membrane damage, as well as the inhibition of nucleic acid and protein synthesis. Silybin has been shown to have synergistic effects when combined with conventional antibiotics against both drug-sensitive and drug-resistant microorganisms. However, the low bioavailability observed for this flavonolignan has been a challenge to its clinical use. In this context, nanotechnology has been used to increase silybin’s bioavailability while enhancing its antimicrobial activity. Furthermore, certain structural modifications have been able to enhance its antimicrobial activity in comparison to that of the natural molecule. Overall, this review provides insights into the scientific understanding of the mechanism of action of silybin and its desired properties for the effective treatment of infections. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of silybin A and silybin B. Flavonolignans contain a flavonoid moiety linked to a lignan and phenylpropanoid moiety.</p>
Full article ">Figure 2
<p>Antibacterial mechanism of silybin. Silybin exerts its antibacterial action through different mechanisms, including (<b>a</b>) inhibition of biofilm formation and biofilm formation, disrupting adherent bacterial communities; (<b>b</b>) inhibition of the expression of efflux pump genes, such as NorA, ABC and AcrABZ-TolC pumps, increasing the intracellular concentration of silybin; (<b>c</b>) inhibition of quorum sensing, limiting bacterial communication; (<b>d</b>) reduction of virulence factors, such as adhesins that are essential for the cell adhesion process; (<b>e</b>) DNA fragmentation, resulting in damage to the genetic material; (<b>f</b>) inhibition of RNA and protein synthesis; and (<b>g</b>) apoptosis-like death, promoting DNA fragmentation and cell death.</p>
Full article ">Figure 3
<p>Antifungal mechanism of silybin. Silybin exerts its antifungal action through different mechanisms, including (<b>a</b>) inhibition of biofilm formation, disrupting adherent bacterial communities; (<b>b</b>) damage to the plasma membrane, causing cell rupture; (<b>c</b>) mitochondrial alterations, generating an increase in reactive oxygen species (ROS), intensifying oxidative stress; (<b>d</b>) DNA fragmentation, resulting in damage to the genetic material; and (<b>e</b>) induction of apoptosis, promoting cell disintegration and death.</p>
Full article ">
Back to TopTop