Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(15)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39125628

RESUMEN

Staphylococcus aureus, particularly drug-resistant strains, poses significant challenges in healthcare due to its ability to form biofilms, which confer increased resistance to antibiotics and immune responses. Building on previous knowledge that several flavonoids exhibit antibiofilm activity, this study sought to identify a novel flavonoid capable of effectively inhibiting biofilm formation and virulence factor production in S. aureus strains including MRSA. Among the 19 flavonoid-like compounds tested, 3,2'-dihydroxyflavone (3,2'-DHF) was identified for the first time as inhibiting biofilm formation and virulence factors in S. aureus with an MIC 75 µg/mL. The antibiofilm activity was further confirmed by microscopic methods. Notably, 3,2'-DHF at 5 µg/mL was effective in inhibiting both mono- and polymicrobial biofilms involving S. aureus and Candida albicans, a common co-pathogen. 3,2'-DHF reduces hemolytic activity, slime production, and the expression of key virulence factors such as hemolysin gene hla and nuclease gene nuc1 in S. aureus. These findings highlight the potential of 3,2'-DHF as a novel antibiofilm and antivirulence agent against both bacterial and fungal biofilms, offering a promising alternative to traditional antibiotics in the treatment of biofilm-associated infections.


Asunto(s)
Antibacterianos , Biopelículas , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus , Factores de Virulencia , Biopelículas/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Factores de Virulencia/genética , Antibacterianos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/patogenicidad , Flavonas/farmacología , Flavonoides/farmacología , Virulencia/efectos de los fármacos , Humanos
2.
Biofilm ; 8: 100218, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39175909

RESUMEN

Effective management of microbial biofilms holds significance within food and medical environments. Candida albicans, an opportunistic fungus, forms mucosal biofilms closely linked to candidiasis and drug-resistant infections due to their drug tolerance. Morphologic change from yeast to filamentous cells is a key virulence factor and a prerequisite for biofilm development. This study investigated the anti-fungal and antibiofilm activities of 20 flavonoids against C. albicans. With their known antioxidant capabilities, flavonoids hold promise in combating infections associated with biofilms. Among them, flavone and its derivatives exhibited moderate antifungal activity, 3,2'-dihydroxyflavone (3,2'-DHF) at 1 µg/mL exhibited strong antibiofilm activity (MIC 50 µg/mL). In addition, 3,2'-DHF dramatically inhibited cell aggregation and germ tube/hyphae formation. Transcriptomic analyses revealed that flavone and 3,2'-DHF behaved differently, as 3,2'-DHF downregulated the expressions of germ tube/hyphae-forming and biofilm-related genes (ECE1, HWP1, TEC1, and UME6) but upregulated the biofilm/hyphal regulators (CHK1, IFD6, UCF1, and YWP1). Tests evaluating toxicity with plant and nematode models revealed that flavone and 3,2'-DHF exhibited mild toxicity. Current results indicate that hydroxylated flavone derivatives can enhance anti-fungal and antibiofilm activities and provide a source of potential anti-fungal agents against drug-resistant C. albicans.

3.
Front Cell Infect Microbiol ; 14: 1414618, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903941

RESUMEN

Candida species comprise a ubiquitous pathogenic fungal genus responsible for causing candidiasis. They are one of the primary causatives of several mucosal and systemic infections in humans and can survive in various environments. In this study, we investigated the antifungal, anti-biofilm, and anti-hyphal effects of six N-substituted phthalimides against three Candida species. Of the derivatives, N-butylphthalimide (NBP) was the most potent, with a minimum inhibitory concentration (MIC) of 100 µg/ml and which dose-dependently inhibited biofilm at sub-inhibitory concentrations (10-50 µg/ml) in both the fluconazole-resistant and fluconazole-sensitive Candida albicans and Candida parapsilosis. NBP also effectively inhibited biofilm formation in other pathogens including uropathogenic Escherichia coli, Staphylococcus epidermidis, Staphylococcus aureus, and Vibrio parahaemolyticus, along with the polymicrobial biofilms of S. epidermidis and C. albicans. NBP markedly inhibited the hyphal formation and cell aggregation of C. albicans and altered its colony morphology in a dose-dependent manner. Gene expression analysis showed that NBP significantly downregulated the expression of important hyphal- and biofilm-associated genes, i.e., ECE1, HWP1, and UME6, upon treatment. NBP also exhibited mild toxicity at concentrations ranging from 2 to 20 µg/ml in a nematode model. Therefore, this study suggests that NBP has anti-biofilm and antifungal potential against various Candida strains.


Asunto(s)
Antifúngicos , Biopelículas , Candida albicans , Hifa , Pruebas de Sensibilidad Microbiana , Ftalimidas , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Antifúngicos/farmacología , Ftalimidas/farmacología , Candida albicans/efectos de los fármacos , Hifa/efectos de los fármacos , Hifa/crecimiento & desarrollo , Candida/efectos de los fármacos , Candidiasis/microbiología , Candidiasis/tratamiento farmacológico , Animales , Humanos , Candida parapsilosis/efectos de los fármacos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Fluconazol/farmacología
4.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397101

RESUMEN

Skin microbiota, such as acne-related Cutibacterium acnes, Staphylococcus aureus, and fungal Candida albicans, can form polymicrobial biofilms with greater antimicrobial tolerance to traditional antimicrobial agents and host immune systems. In this study, the phytopigment shikonin was investigated against single-species and multispecies biofilms under aerobic and anaerobic conditions. Minimum inhibitory concentrations of shikonin were 10 µg/mL against C. acnes, S. aureus, and C. albicans, and at 1-5 µg/mL, shikonin efficiently inhibited single biofilm formation and multispecies biofilm development by these three microbes. Shikonin increased porphyrin production in C. acnes, inhibited cell aggregation and hyphal formation by C. albicans, decreased lipase production, and increased hydrophilicity in S. aureus. In addition, shikonin at 5 or 10 µg/mL repressed the transcription of various biofilm-related genes and virulence-related genes in C. acnes and downregulated the gene expression levels of the quorum-sensing agrA and RNAIII, α-hemolysin hla, and nuclease nuc1 in S. aureus, supporting biofilm inhibition. In addition, shikonin prevented multispecies biofilm development on porcine skin, and the antimicrobial efficacy of shikonin was recapitulated in a mouse infection model, in which it promoted skin regeneration. The study shows that shikonin inhibits multispecies biofilm development by acne-related skin microbes and might be useful for controlling bacterial infections.


Asunto(s)
Acné Vulgar , Antiinfecciosos , Naftoquinonas , Infecciones Estafilocócicas , Animales , Ratones , Candida albicans/genética , Staphylococcus aureus , Biopelículas , Antiinfecciosos/farmacología
5.
Phytomedicine ; 124: 155306, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176270

RESUMEN

BACKGROUND: Most bacteria and fungi form biofilms that attach to living or abiotic surfaces. These biofilms diminish the efficacy of antimicrobial agents and contribute to chronic infections. Furthermore, multispecies biofilms composed of bacteria and fungi are often found at chronic infection sites. PURPOSE: In this study, lawsone (2­hydroxy-1,4-naphthoquinone) and its parent 1,4-naphthoquinone were studied for antimicrobial and antibiofilm activities against single-species and multispecies biofilms of enterohemorrhagic Escherichia coli O157:H7 (EHEC) and Candida albicans. METHODS: Biofilm formation assays, biofilm eradication assays, antimicrobial assays, live cell imaging microscopy, confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM), extracellular polymeric substances and indole production, cell surface hydrophilicity assay, cell motility, cell aggregation, hyphal growth, dual species biofilm formation, quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), and toxicity assays on plant seed germination and nematode model were utilized to investigate how lawsone affect biofilm development. RESULTS: Sub-inhibitory concentrations of lawsone (35 µg/ml) significantly inhibited single-and multispecies biofilm development. Lawsone reduced the production of curli and indole, and the swarming motility of EHEC, efficiently inhibited C. albicans cell aggregation and hyphal formation, and increased the cell surface hydrophilicity of C. albicans. Transcriptomic analysis showed that lawsone suppressed the expression of the curli-related genes csgA and csgB in EHEC, and the expression of several hypha- and biofilm-related genes (ALS3, ECE1, HWP1, and UME6) in C. albicans. In addition, lawsone up to 100 µg/ml was nontoxic to the nematode Caenorhabditis elegans and to the seed growth of Brassica rapa and Triticum aestivum. CONCLUSION: These results show that lawsone inhibits dual biofilm development and suggest that it might be useful for controlling bacterial or fungal infections and multispecies biofilms.


Asunto(s)
Antiinfecciosos , Escherichia coli O157 , Naftoquinonas , Candida albicans , Biopelículas , Indoles/farmacología
6.
Microbiol Spectr ; 11(6): e0173723, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37874140

RESUMEN

IMPORTANCE: The persistence of Candida infections is due to its ability to form biofilms that enable it to resist antifungals and host immune systems. Hence, inhibitions of the biofilm formation and virulence characteristics of Candida sp. provide potential means of addressing these infections. Among various chromone derivatives tested, four chromone-3-carbonitriles showed antifungal, antibiofilm, and antivirulence activities against several Candida species. Their mode of action has been partially revealed, and their toxicity is reported here using nematode and plant models.


Asunto(s)
Antifúngicos , Candidiasis , Antifúngicos/farmacología , Candida , Candida albicans , Candidiasis/tratamiento farmacológico , Biopelículas , Pruebas de Sensibilidad Microbiana
7.
Microbiol Spectr ; 10(5): e0253622, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36190417

RESUMEN

Candida albicans is an opportunistic pathogenic fungus responsible for candidiasis. The pathogen readily forms antifungal agent-resistant biofilms on implanted medical devices or human tissue. Morphologic transition from yeast to filamentous cells and subsequent biofilm formation is a key virulence factor and a prerequisite for biofilm development by C. albicans. We investigated the antibiofilm and antifungal activities of 18 hydroquinones against fluconazole-resistant C. albicans. Tetrachlorohydroquinone (TCHQ) at subinhibitory concentrations (2 to 10 µg/mL) significantly inhibited C. albicans biofilm formation with an MIC of 50 µg/mL, whereas the backbone hydroquinone did not (MIC > 400 µg/mL), and it markedly inhibited cell aggregation and hyphal formation. Transcriptomic analyses showed that TCHQ downregulated the expressions of several hyphae-forming and biofilm-related genes (ALS3, ECE1, HWP1, RBT5, and UME6) but upregulated hyphae- and biofilm-inhibitory genes (IFD6 and YWP1). Furthermore, it prevented C. albicans biofilm development on porcine skin and at concentrations of 20 to 50 µg/mL was nontoxic to the nematode Caenorhabditis elegans and did not adversely affect Brassica rapa seed germination and growth. This study indicates that hydroquinones, particularly TCHQ, diminish the virulence, biofilm formation, and animal tissue adhesion of C. albicans, which suggests hydroquinones should be considered potential candidate antifungal agents against drug-resistant C. albicans strains. IMPORTANCE Persistence in chronic infections by Candida albicans is due to its ability of biofilm formation that endures conventional antifungals and host immune systems. Hence, the inhibition of biofilm formation and virulence characteristics is another mean of addressing infections. This study is a distinctive one since 18 hydroquinone analogues were screened and TCHQ efficiently inhibited the biofilm formation by C. albicans with significantly changed expressional profile of hyphae-forming and biofilm-related genes. The antibiofilm efficacy was confirmed using a porcine skin model and chemical toxicity was investigated using plant seed germination and nematode models. Our findings reveal that TCHQ can efficiently control the C. albicans biofilms and virulence characteristics.


Asunto(s)
Candida albicans , Hifa , Animales , Humanos , Candida albicans/genética , Hifa/genética , Antifúngicos/farmacología , Hidroquinonas/farmacología , Fluconazol/farmacología , Biopelículas , Factores de Virulencia/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología
8.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36142597

RESUMEN

Staphylococcus aureus is one of the major pathogens responsible for antimicrobial resistance-associated death. S. aureus can secrete various exotoxins, and staphylococcal biofilms play critical roles in antibiotic tolerance and the persistence of chronic infections. Here, we investigated the inhibitory effects of 18 hydroquinones on biofilm formation and virulence factor production by S. aureus. It was found that 2,5-bis(1,1,3,3-tetramethylbutyl) hydroquinone (TBHQ) at 1 µg/mL efficiently inhibits biofilm formation by two methicillin-sensitive and two methicillin-resistant S. aureus strains with MICs of 5 µg/mL, whereas the backbone compound hydroquinone did not (MIC > 400 µg/mL). In addition, 2,3-dimethylhydroquinone and tert-butylhydroquinone at 50 µg/mL also exhibited antibiofilm activity. TBHQ at 1 µg/mL significantly decreased the hemolytic effect and lipase production by S. aureus, and at 5−50 µg/mL was non-toxic to the nematode Caenorhabditis elegans and did not adversely affect Brassica rapa seed germination or growth. Transcriptional analyses showed that TBHQ suppressed the expression of RNAIII (effector of quorum sensing). These results suggest that hydroquinones, particularly TBHQ, are potentially useful for inhibiting S. aureus biofilm formation and virulence.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Antibacterianos/farmacología , Biopelículas , Exotoxinas/farmacología , Humanos , Hidroquinonas/farmacología , Lipasa , Meticilina/farmacología , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus , Factores de Virulencia/farmacología
9.
Microbiol Spectr ; 10(3): e0133022, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35647620

RESUMEN

Staphylococcus aureus is a major human pathogen that secretes several toxins associated with the pathogenesis of sepsis and pneumonia. Its antibiotic resistance is notorious, and its biofilms play a critical role in antibiotic tolerance. We hypothesized fatty acids might inhibit S. aureus biofilm formation and the expressions of its virulence factors. Initially, the antibiofilm activities of 27 fatty acids against a methicillin-sensitive S. aureus strain were investigated. Of the fatty acids tested, three C18 unsaturated fatty acids, that is, petroselinic, vaccenic, and oleic acids at 100 µg/mL, inhibited S. aureus biofilm formation by more than 65% without affecting its planktonic cell growth (MICs were all > 400 µg/mL). Notably, petroselinic acid significantly inhibited biofilm formation of two methicillin-resistant S. aureus strains and two methicillin-sensitive S. aureus strains. In addition, petroselinic acid significantly suppressed the production of three virulence factors, namely, staphyloxanthin, lipase, and α-hemolysin. Transcriptional analysis showed that petroselinic acid repressed the gene expressions of quorum sensing regulator agrA, effector of quorum sensing RNAIII, α-hemolysin hla, nucleases nuc1 and nuc2, and the virulence regulator saeR. Furthermore, petroselinic acid dose-dependently inhibited S. aureus biofilm formation on abiotic surfaces and porcine skin. These findings suggest that fatty acids, particularly petroselinic acid, are potentially useful for controlling biofilm formation by S. aureus. IMPORTANCE Fatty acids with a long carbon chain have recently attracted attention because of their antibiofilm activities against microbes. Here, we report the antibiofilm activities of 27 fatty acids against S. aureus. Of the fatty acids tested, three C18 unsaturated fatty acids (petroselinic, vaccenic, and oleic acids) significantly inhibited biofilm formation by S. aureus. Furthermore, petroselinic acid inhibited the production of several virulence factors in S. aureus. The study also reveals that the action mechanism of petroselinic acid involves repression of quorum-sensing-related and virulence regulator genes. These findings show that natural and nontoxic petroselinic acid has potential use as a treatment for S. aureus infections, including infections by methicillin-resistant S. aureus strains, and in food processing facilities.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Antibacterianos/farmacología , Biopelículas , Ácidos Grasos Insaturados , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Humanos , Meticilina/metabolismo , Ácidos Oléicos/metabolismo , Staphylococcus aureus/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
Pharmaceutics ; 14(5)2022 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-35631633

RESUMEN

Acne vulgaris is a common chronic inflammatory skin disease involving Cutibacterium acnes with other skin commensals such as Staphylococcus aureus and Candida albicans in the anaerobic and lipid-rich conditions of pilosebaceous units. These microbes readily form multispecies biofilms that are tolerant of traditional antibiotics as well as host immune systems. The phytopigment alizarin was previously found to prevent biofilm formation by S. aureus and C. albicans strains under aerobic conditions. Hence, we hypothesized that alizarin might control C. acnes and multispecies biofilm development. We found that under anaerobic conditions, alizarin efficiently inhibited single biofilm formation and multispecies biofilm development by C. acnes, S. aureus, and C. albicans without inhibiting planktonic cell growth. Alizarin increased the hydrophilicities of S. aureus and C. albicans cells, decreased lipase production by S. aureus, diminished agglutination by C. acnes, and inhibited the aggregation of C. albicans cells. Furthermore, the co-administration of alizarin and antibiotics enhanced the antibiofilm efficacies of alizarin against C. acnes. A transcriptomic study showed that alizarin repressed the transcriptions of various biofilm-related genes such as lipase, hyaluronate lyase, adhesin/invasion-related, and virulence-related genes of C. acnes. Furthermore, alizarin at 100 µg/mL prevented C. acnes biofilm development on porcine skin. Our results show that alizarin inhibits multispecies biofilm development by acne-causing microbes and suggest it might be a useful agent for treating or preventing C. acnes-causing skin diseases.

11.
Front Microbiol ; 13: 876932, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35633672

RESUMEN

Fatty acids have diverse functions in the vast majority of cells. At high doses, they act as antimicrobials while, at low doses, they exhibit antibiofilm and antivirulence activities. In this study, the synergistic antibacterial and antibiofilm activities of 30 fatty acids and 11 antibiotics were investigated against methicillin-sensitive and methicillin-resistant Staphylococcus aureus strains. Of the 15 saturated and 15 unsaturated fatty acids examined, 16 enhanced the antibacterial activity of tobramycin. Combinatorial treatment with myristoleic acid (the most active) at 10 µg/ml and tobramycin at 10 µg/ml decreased cell survival by >4 log as compared with tobramycin treatment alone. Notably, aminoglycoside antibiotics, such as tobramycin, kanamycin, gentamicin, and streptomycin exhibited antimicrobial synergy with myristoleic acid. Co-treatment with myristoleic acid and antibiotics markedly decreased biofilm formation. Interestingly, co-treatment with tobramycin and myristoleic acid induced a reduction in S. aureus cell size. These results suggest that fatty acids, particularly myristoleic acid, can be used as aminoglycoside antibiotic adjuvants against recalcitrant S. aureus infections.

12.
Front Microbiol ; 13: 818165, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35369516

RESUMEN

Cinnamaldehyde has a broad range of biological activities, which include antibiofilm and anthelmintic activities. The ever-growing problem of drug resistance and limited treatment options have created an urgent demand for natural molecules with antibiofilm and anthelmintic properties. Hence, we hypothesized that molecules with a scaffold structurally similar to that of cinnamaldehyde might act as dual inhibitors against fungal biofilms and helminths. In this regard, eleven cinnamaldehyde analogs were tested to determine their effects on fungal Candida albicans biofilm and nematode Caenorhabditis elegans. α-Methyl and trans-4-methyl cinnamaldehydes efficiently inhibited C. albicans biofilm formation (>90% inhibition at 50 µg/mL) with minimum inhibitory concentrations (MICs) of ≥ 200 µg/mL and 4-bromo and 4-chloro cinnamaldehydes exhibited anthelmintic property at 20 µg/mL against C. elegans. α-Methyl and trans-4-methyl cinnamaldehydes inhibited hyphal growth and cell aggregation. Scanning electron microscopy was employed to determine the surface architecture of C. albicans biofilm and cuticle of C. elegans, and confocal laser scanning microscopy was used to determine biofilm characteristics. The perturbation in gene expression of C. albicans was investigated using qRT-PCR analysis and α-methyl and trans-4-methyl cinnamaldehydes exhibited down-regulation of ECE1, IFD6, RBT5, UCF1, and UME6 and up-regulation of CHT4 and YWP1. Additionally, molecular interaction of these two molecules with UCF1 and YWP1 were revealed by molecular docking simulation. Our observations collectively suggest α-methyl and trans-4-methyl cinnamaldehydes are potent biofilm inhibitors and that 4-bromo and 4-chloro cinnamaldehydes are anthelmintic agents. Efforts are required to determine the range of potential therapeutic applications of cinnamaldehyde analogs.

13.
Carbohydr Polym ; 284: 118959, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35287925

RESUMEN

Biofilm formation by microorganisms is a serious clinical problem that leads to drug failure. Nanocarriers (NCs) have shown good potential for controlling drug-resistant biofilms, although the effective penetration and retention of NCs in biofilms is still a big task. The issue was overcome by selecting alizarin as a natural antibiofilm agent, but its low water solubility restricts its further use. Thus, in present study, chitosan-gum arabic-coated liposomes-alizarin nanocarriers (CGL-Alz NCs) were synthesized using an ionotropic gelation method to improve drug release and penetration of alizarin inside biofilm cells. CGL-Alz NCs acted against biofilms caused by Candida albicans or Staphylococcus aureus and improved penetration of alizarin inside biofilms exerting long-term antibiofilm effects caused by sustained release of alizarin from NCs. Furthermore, significant biofilm and hyphae reduction was observed at a 5 µg/mL concentration of NCs. This research work opens a new avenue of an innovative strategy to treat biofilm-associated multispecies infections.


Asunto(s)
Quitosano , Antraquinonas , Biopelículas , Candida albicans , Quitosano/farmacología , Goma Arábiga/farmacología
14.
Microbiol Spectr ; 10(1): e0205621, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107361

RESUMEN

The Gram-positive anaerobic bacterium Cutibacterium acnes is a major inhabitant of human skin and has been implicated in acne vulgaris formation and in the formation of multispecies biofilms with other skin-inhabiting organisms like Staphylococcus aureus and Candida albicans. Indoles are widespread in nature (even in human skin) and function as important signaling molecules in diverse prokaryotes and eukaryotes. In the present study, we investigated the antibacterial and antibiofilm activities of 20 indoles against C. acnes. Of the indoles tested, indole-3-carbinol at 0.1 mM significantly inhibited biofilm formation by C. acnes without affecting planktonic cell growth, and the anticancer drug 3,3'-diindolylmethane (DIM) at 0.1 mM (32 µg/mL) also significantly inhibited planktonic cell growth and biofilm formation by C. acnes, whereas the other indoles and indole itself were less effective. Also, DIM at 0.1 mM successfully inhibited multispecies biofilm formation by C. acnes, S. aureus, and C. albicans. Transcriptional analyses showed that DIM inhibited the expressions of several biofilm-related genes in C. acnes, and at 0.05 mM, DIM inhibited hyphal formation and cell aggregation by C. albicans. These results suggest that DIM and other indoles inhibit biofilm formation by C. acnes and have potential use for treating C. acnes associated diseases. IMPORTANCE Since indoles are widespread in nature (even in human skin), we hypothesized that indole and its derivatives might control biofilm formation of acne-causing bacteria (Cutibacterium acnes and Staphylococcus aureus) and fungal Candida albicans. The present study reports for the first time the antibiofilm and antimicrobial activities of several indoles on C. acnes. Of the indoles tested, two anticancer agents, indole-3-carbinol and 3,3'-diindolylmethane found in cruciferous vegetables, significantly inhibited biofilm formation by C. acnes. Furthermore, the most active 3,3'-diindolylmethane successfully inhibited multispecies biofilm formation by C. acnes, S. aureus, and C. albicans. Transcriptional analyses showed that 3,3'-diindolylmethane inhibited the expressions of several biofilm-related genes including lipase, hyaluronate lyase, and virulence-related genes in C. acnes, and 3,3'-diindolylmethane inhibited hyphal formation and cell aggregation by C. albicans. Our findings show that 3,3'-diindolylmethane offers a potential means of controlling acne vulgaris and multispecies biofilm-associated infections due to its antibiofilm and antibiotic properties.


Asunto(s)
Acné Vulgar/microbiología , Antineoplásicos/farmacología , Bacterias/efectos de los fármacos , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Candida albicans/efectos de los fármacos , Indoles/farmacología , Antibacterianos/farmacología , Bacterias/crecimiento & desarrollo , Candida albicans/crecimiento & desarrollo , Humanos , Pruebas de Sensibilidad Microbiana , Filogenia , Propionibacteriaceae/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Virulencia
15.
Microb Biotechnol ; 15(2): 590-602, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34156757

RESUMEN

Biofilms are communities of bacteria, fungi or yeasts that form on diverse biotic or abiotic surfaces, and play important roles in pathogenesis and drug resistance. A generic saw palmetto oil inhibited biofilm formation by Staphylococcus aureus, Escherichia coli O157:H7 and fungal Candida albicans without affecting their planktonic cell growth. Two main components of the oil, lauric acid and myristic acid, are responsible for this antibiofilm activity. Their antibiofilm activities were observed in dual-species biofilms as well as three-species biofilms of S. aureus, E. coli O157:H7 and C. albicans. Transcriptomic analysis showed that lauric acid and myristic acid repressed the expressions of haemolysin genes (hla and hld) in S. aureus, several biofilm-related genes (csgAB, fimH and flhD) in E. coli and hypha cell wall gene HWP1 in C. albicans, which supported biofilm inhibition. Also, saw palmetto oil, lauric acid and myristic acid reduced virulence of three microbes in a nematode infection model and exhibited minimal cytotoxicity. Furthermore, combinatorial treatment of fatty acids and antibiotics showed synergistic antibacterial efficacy against S. aureus and E. coli O157:H7. These results demonstrate that saw palmetto oil and its main fatty acids might be useful for controlling bacterial infections as well as multispecies biofilms.


Asunto(s)
Escherichia coli O157 , Staphylococcus aureus , Antibacterianos/farmacología , Biopelículas , Candida albicans , Ácidos Láuricos/farmacología , Ácido Mirístico/farmacología , Extractos Vegetales , Serenoa
16.
Biomedicines ; 9(9)2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34572317

RESUMEN

Acinetobacter baumannii is a nosocomial pathogen, and its biofilms are tolerant to desiccation, nutrient starvation, and antimicrobial treatment on biotic and abiotic surfaces, tissues, and medical devices. Biofilm formation by A. baumannii is triggered by a quorum sensing cascade, and we hypothesized that fatty acids might inhibit its biofilm formation by interfering with quorum sensing. Initially, we investigated the antibiofilm activities of 24 fatty acids against A. baumannii ATCC 17978 and two clinical isolates. Among these fatty acids, two unsaturated fatty acids, nervonic and oleic acid, at 20 µg/mL significantly inhibited A. baumannii biofilm formation without affecting its planktonic cell growth (MICs were >500 µg/mL) and markedly decreased the motility of A. baumannii but had no toxic effect on the nematode Caenorhabditis elegans. Interestingly, molecular dynamic simulations showed that both fatty acids bind to the quorum sensing acyl homoserine lactone synthase (AbaI), and decent conformational stabilities of interactions between the fatty acids and AbaI were exhibited. Our results demonstrate that nervonic and oleic acid inhibit biofilm formation by A. baumannii strains and may be used as lead molecules for the control of persistent A. baumannii infections.

17.
Phytomedicine ; 91: 153710, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34461422

RESUMEN

BACKGROUND: Cutibacterium acnes is a major colonizer and inhabitant of human skin and contributes to the pathogenesis of acne vulgaris. C. acnes either alone or with Staphylococcus aureus, which also inhabits skin, readily forms biofilms that are often tolerant of conventional antibiotics and the host immune system. It was hypothesized that the amphiphilic nature of some fatty acids (FAs) inhibit C. acnes or mixed biofilm formation. PURPOSE: The antibacterial and antibiofilm activities of 24 saturated and unsaturated FAs were investigated against C. acnes as well as a mixture of the bacteria C. acnes and S. aureus. METHODS: Anti-biofilm assays, antimicrobial assays, confocal laser scanning microscopy, scanning electron microscopy, extracellular polymeric substance production, and microbial adherence to hydrocarbon assay were utilized to elucidate how active FAs influence biofilm development. RESULTS: Seventeen FAs at 20 µg/ml inhibited C. acnes biofilm formation by 60-99%. The minimum inhibitory concentrations (MICs) of 20 FAs were ≥ 500 µg/ml but 4 medium-chain FAs had MICs in a range 15 to 200 µg/ml. Interestingly, myristoleic acid inhibited biofilm formation at 1 µg/ml. Myristoleic acid also inhibited the formation of S. aureus and mixed C. acnes/S. aureus biofilms. FAs reduced C. acnes hydrophobicity and we found this was generally correlated with their antibiofilm forming efficacies. Transcriptional analyses showed that myristoleic acid modulates the expression of several biofilm-related genes such as lipase, hyaluronate lyase, and virulence-related genes. CONCLUSION: This study shows myristoleic acid and other FAs inhibit biofilm formation by C. acnes and mixed biofilm formation by C. acnes and S. aureus. Hence, myristoleic acid might be useful for treating or preventing acne and C. acnes associated diseases.


Asunto(s)
Antibacterianos , Biopelículas/efectos de los fármacos , Ácidos Grasos Monoinsaturados/farmacología , Propionibacterium acnes/efectos de los fármacos , Staphylococcus aureus , Antibacterianos/farmacología , Matriz Extracelular de Sustancias Poliméricas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus/efectos de los fármacos
18.
Mar Drugs ; 19(6)2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34073814

RESUMEN

Biofilm formation by Staphylococcus aureus plays a critical role in the persistence of chronic infections due to its tolerance against antimicrobial agents. Here, we investigated the antibiofilm efficacy of six phorbaketals: phorbaketal A (1), phorbaketal A acetate (2), phorbaketal B (3), phorbaketal B acetate (4), phorbaketal C (5), and phorbaketal C acetate (6), isolated from the Korean marine sponge Phorbas sp. Of these six compounds, 3 and 5 were found to be effective inhibitors of biofilm formation by two S. aureus strains, which included a methicillin-resistant S. aureus. In addition, 3 also inhibited the production of staphyloxanthin, which protects microbes from reactive oxygen species generated by neutrophils and macrophages. Transcriptional analyses showed that 3 and 5 inhibited the expression of the biofilm-related hemolysin gene hla and the nuclease gene nuc1.


Asunto(s)
Antibacterianos/farmacología , Poríferos/química , Sesterterpenos/farmacología , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/aislamiento & purificación , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genes Bacterianos , Sesterterpenos/aislamiento & purificación , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiología , Xantófilas/metabolismo
19.
Microb Biotechnol ; 14(4): 1353-1366, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33252828

RESUMEN

Candida biofilms are tolerant to conventional antifungal therapeutics and the host immune system. The transition of yeast cells to hyphae is considered a key step in C. albicans biofilm development, and this transition is inhibited by the quorum-sensing molecule farnesol. We hypothesized that fatty acids mimicking farnesol might influence hyphal and biofilm formation by C. albicans. Among 31 saturated and unsaturated fatty acids, six medium-chain saturated fatty acids, that is, heptanoic acid, octanoic acid, nonanoic acid, decanoic acid, undecanoic acid and lauric acid, effectively inhibited C. albicans biofilm formation by more than 75% at 2 µg ml-1 with MICs in the range 100-200 µg ml-1 . These six fatty acids at 2 µg ml-1 and farnesol at 100 µg ml-1 inhibited hyphal growth and cell aggregation. The addition of fatty acids to C. albicans cultures decreased the productions of farnesol and sterols. Furthermore, down-regulation of several hyphal and biofilm-related genes caused by heptanoic or nonanoic acid closely resembled the changes caused by farnesol. In addition, nonanoic acid, the most effective compound diminished C. albicans virulence in a Caenorhabditis elegans model. Our results suggest that medium-chain fatty acids inhibit more effectively hyphal growth and biofilm formation than farnesol.


Asunto(s)
Candida albicans , Farnesol , Antifúngicos/farmacología , Biopelículas , Farnesol/farmacología , Ácidos Grasos , Hifa
20.
Front Microbiol ; 11: 584812, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193228

RESUMEN

Indole and its derivatives have been shown to interfere with the quorum sensing (QS) systems of a wide range of bacterial pathogens. While indole has been previously shown to inhibit QS in Serratia marcescens, the effects of various indole derivatives on QS, biofilm formation, and virulence of S. marcescens remain unexplored. Hence, in the present study, we investigated the effects of 51 indole derivatives on S. marcescens biofilm formation, QS, and virulence factor production. The results obtained revealed that several indole derivatives (3-indoleacetonitrile, 5-fluoroindole, 6-fluoroindole, 7-fluoroindole, 7-methylindole, 7-nitroindole, 5-iodoindole, 5-fluoro-2-methylindole, 2-methylindole-3-carboxaldehyde, and 5-methylindole) dose-dependently interfered with quorum sensing (QS) and suppressed prodigiosin production, biofilm formation, swimming motility, and swarming motility. Further assays showed 6-fluoroindole and 7-methylindole suppressed fimbria-mediated yeast agglutination, extracellular polymeric substance production, and secretions of virulence factors (e.g., proteases and lipases). QS assays on Chromobacterium violaceum CV026 confirmed that indole derivatives interfered with QS. The current results demonstrate the antibiofilm and antivirulence properties of indole derivatives and their potentials in applications targeting S. marcescens virulence.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...