scholarly journals The Impact of Acne Treatment on Skin Bacterial Microbiota: A Systematic Review

2021 ◽  
pp. 120347542110379
Author(s):  
Megan Lam ◽  
Angie Hu ◽  
Patrick Fleming ◽  
Charles W. Lynde

Background Microbial strains such as Cutibacterium acnes have been examined as contributors to the pathogenesis of acne. Given the prevalence of the disease among adolescents and adults, the overutilization of antimicrobial agents may breed resistance and alter commensal microflora. Objectives To characterize the impact of acne treatment on the diversity and relative abundance of the cutaneous microbial community, particularly of the bacterial flora Methods An electronic search was conducted of Embase, MEDLINE, and the Cochrane Central Register of Controlled Trials (CENTRAL) on June 5, 2020. Interventional and observational studies examining patients receiving acne treatment with culture-independent, community-level analysis of the cutaneous microbiome were included. Results Nine studies with 170 treated acne patients were included. Five studies reported a significant change in alpha diversity following treatment, 3 of which examining systemic antibiotics reported significant increases in diversity. Two of 3 studies examining effects of benzoyl peroxide reported a decrease in diversity. However, trends in diversity were heterogeneous among studies. Conclusions While individual variability in microbiome composition, and study-level heterogeneity in study sampling techniques may limit quantitative synthesis, our results support findings that acne treatment, including those not considered to have antimicrobial properties, alters the composition of the cutaneous microbiome. PROSPERO registration: CRD42020190629

Microbiome ◽  
2020 ◽  
Vol 8 (1) ◽  
Author(s):  
Alicia Moreno-Sabater ◽  
Gaelle Autaa ◽  
Delphine Sterlin ◽  
Amenie Jerbi ◽  
Remy Villette ◽  
...  

Abstract Background Interest for the study of gut mycobiota in relation with human health and immune homeostasis has increased in the last years. From this perspective, new tools to study the immune/fungal interface are warranted. Systemic humoral immune responses could reflect the dynamic relationships between gut mycobiota and immunity. Using a novel flow cytometry technology (Fungi-Flow) to determine immunoglobulin (Ig) responses to fungi, we studied the relationships between gut mycobiota and systemic humoral anti-commensal immunity. Results The Fungi-Flow method allows a sensitive and specific measurement of systemic IgG responses against 17 commensal and environmental fungi from the two main divisions; Ascomycota and Basidiomycota. IgG responses exhibited a high inter-individual variability. Anti-commensal IgG responses were contrasted with the relative abundance, alpha-diversity, and intra-genus richness of fungal species in gut mycobiota of twenty healthy donors. Categorization of gut mycobiota composition revealed two differentiated fungal ecosystems. Significant difference of anti-Saccharomyces systemic IgG responses were observed in healthy donors stratified according to the fungal ecosystem colonizing their gut. A positive and significant correlation was observed between the variety of IgG responses against fungal commensals and intestinal alpha-diversity. At the level of intra-genus species richness, intense IgG responses were associated with a low intra-genus richness for known pathobionts, but not commensals. Conclusions Fungi-Flow allows an easy and reliable measure of personalized humoral responses against commensal fungi. Combining sequencing technology with our novel Fungi-Flow immunological method, we propose that there are at least two defined ecosystems in the human gut mycobiome associated with systemic humoral responses. Fungi-Flow opens new opportunities to improve our knowledge about the impact of mycobiota in humoral anti-commensal immunity and homeostasis.


2021 ◽  
Vol 5 (Supplement_2) ◽  
pp. 1162-1162
Author(s):  
Abigail Johnson ◽  
Mo Houtti ◽  
Anna Saboe ◽  
Katie Koecher ◽  
Ravi Menon ◽  
...  

Abstract Objectives Health outcomes in previous fiber intervention studies have been variable, potentially due to differences in gut microbiome composition. This study aimed to determine if the effect of fiber intervention on the microbiome differs by initial microbiome or the quantity of fiber provided. Methods This study was designed as a randomized, un-blinded, cross-over trial of fiber cereal dosage. The cross-over design tested the effect of two 2-week long interventions with a High (28g) and Low (14g) level of daily supplemental fiber from whole wheat and bran cereal. Analysis was also completed on the overall study as a single arm, non-randomized, intervention of fiber cereal. The study enrolled 31 healthy adults. The microbiome was assessed at baseline and after intervention for changes in diversity, composition, and stability. Results Across all individuals, fiber intervention increased microbiome alpha-diversity (paired t-test, P = 0.047), but the microbiome was otherwise resistant to the effects of the intervention. Increasing fiber dose (High v. Low) was not associated with consistent changes in beta-diversity (linear mixed models). Approximately 20% of subjects were identified as responders based on beta diversity effect size. At baseline, responders had higher Prevotella copri and lower Bacteroides abundance than non-responders (Wilcoxon rank sum, qval < 0.05). In responders, fiber intake caused increased abundance of Bacteroides and Alistipes and reduced Prevotella (paired Wilcoxon, q < 0.2). In all subjects, fiber intervention decreased microbiome stability (paired Wilcoxon signed rank test, P = 0.006). In responders, there was a significant effect of the fiber level on stability, with higher fiber further lowering stability (linear mixed model, P = 0.05). Conclusions Our data suggest a responder/non-responder microbiome signature for this whole wheat and bran fiber cereal. We find that many effects were not additive by dosage level. Overall, microbiome diversity was increased and stability was decreased during the fiber cereal intervention and in responders this was dose dependent; the clinical implications of the impact of changes in stability remain unknown, and it is possible that the microbiome would stabilize in a longer intervention study. ClinicalTrials.gov identifier: NCT03623308. Funding Sources General Mills, Inc.


Author(s):  
Carola Venturini ◽  
Bethany Bowring ◽  
Alicia Fajardo-Lubian ◽  
Carol Devine ◽  
Jonathan Iredell

Effective antimicrobial stewardship requires a better understanding of the impact of different antibiotics on the gut microflora. Studies in humans are confounded by large inter-individual variability and difficulty in identifying control cohorts. However, controlled murine models can provide valuable information. Here, we examine the impact of a penicillin-like antibiotic (piperacillin-tazobactam, TZP) or a third-generation cephalosporin (ceftriaxone, CRO) on the murine gut microbiota by analysis of changes in fecal microbiome composition by 16S-rRNA amplicon sequencing and standard microbiology. Colonization resistance to multidrug resistant Escherichia coli ST131 and Klebsiella pneumoniae ST258 was also tested. Changes in microbiome composition and a significant (p< 0.05) decrease in diversity occurred in all treated mice, but dysbiosis was more marked and prolonged after CRO exposure with a persistent rise in Proteobacteria. Enterobacteriaceae blooms occurred in all antibiotic treated mice, but for TZP, unlike CRO, these were only significant under direct antibiotic pressure. At the height of dysbiosis after antibiotic termination, the murine gut was highly susceptible to colonization with both multidrug resistant enterobacterial pathogens. Co-habitation of treated mice with untreated individuals had a notable mitigating effect on dysbiosis of treated guts. The administration of a third-generation cephalosporin caused a more severe dysbiosis in the murine gut microflora, when compared to a penicillin/β-lactam inhibitor combination with comparable activity against medically important virulent bacteria. At the height of dysbiosis, both antibiotic treatments equally led to microbial imbalance associated with loss of resistance to gut colonization by antibiotic-resistant pathogens.


2020 ◽  
Author(s):  
Lydie Ploux ◽  
Min Jin ◽  
Sophie Hellé ◽  
Cosette Betscha ◽  
Jean-Marc Strub ◽  
...  

&lt;p&gt;L- and D-Cateslytin (CTL) are antimicrobial peptides (AMP) derived from chromogranin A, a protein of the stress response system. Their antimicrobial properties have been thoroughly characterized and already exploited in biomaterials. However, effects on biofilms of yeast and bacteria have never been specifically addressed. We have investigated the impact of both L and D configurations of CTL on the growth of biofilms formed by Candida albicans, Escherichia coli or Staphylococcus aureus microorganisms.&lt;/p&gt; &lt;p&gt;The study was conducted in different media and two strategies of treatment were tested, consisting of administrating the peptide either just at the beginning of biofilm development i.e. on just adhering pioneer microbial cells or on a biofilm already allowed to develop for 24h. We also considered whether the peptide was modified in contact with the medium or/and microbial metabolites. Planktonic and sessile populations of microbial cells were analyzed by spectrophotometry, crystal violet staining, MTT and confocal microscopy with staining by Syto9&amp;#210; and propidium iodide. Identification of the peptides and their derived fragments was investigated by HPLC and Mass-Spectroscopy.&lt;/p&gt; &lt;p&gt;In general, CTL-D exhibited higher antibiofilm performances than CTL-L. In addition, concentrations necessary to inhibit biofilm formation were found to vary from ten to eighty times the MICs determined in planktonic cultures. Nevertheless, the results also demonstrate that sessile microorganisms and biofilms are sensitive to CTL (L and D conformations) differently that planktonic populations. Significant (p-value &lt; 0.01) effects were observed on both sessile and planktonic populations and with both strategies of treatments, but they highly varied with medium, species and CTL configuration. Typically, better antibiofilm effect than common antibiotics was reached in some specific conditions, while enhancement of aggregation or biofilm formation occurred in another medium and for other doses. Nevertheless,&lt;/p&gt; &lt;p&gt;Finally, this confirms the quality of CTL peptides as new antimicrobial agents and reveals their anti-biofilm properties. This also specifies the conditions of use necessary to benefit of the highest performances.&lt;/p&gt;


Nutrients ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 881
Author(s):  
Niklas D. Aardema ◽  
Daphne M. Rodriguez ◽  
Arnaud J. Van Wettere ◽  
Abby D. Benninghoff ◽  
Korry J. Hintze

Previous work by our group using a mouse model of inflammation-associated colorectal cancer (CAC) showed that the total Western diet (TWD) promoted colon tumor development. Others have also shown that vancomycin-mediated changes to the gut microbiome increased colorectal cancer (CRC). Therefore, the objective of this study was to determine the impact of vancomycin on colon tumorigenesis in the context of a standard mouse diet or the TWD. A 2 × 2 factorial design was used, in which C57Bl/6J mice were fed either the standard AIN93G diet or TWD and with vancomycin in the drinking water or not. While both the TWD and vancomycin treatments independently increased parameters associated with gut inflammation and tumorigenesis compared to AIN93G and plain water controls, mice fed the TWD and treated with vancomycin had significantly increased tumor multiplicity and burden relative to all other treatments. Vancomycin treatment significantly decreased alpha diversity and changed the abundance of several taxa at the phylum, family, and genus levels. Conversely, basal diet had relatively minor effects on the gut microbiome composition. These results support our previous research that the TWD promotes colon tumorigenesis and suggest that vancomycin-induced changes to the gut microbiome are associated with higher tumor rates.


Gut Pathogens ◽  
2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Elizabeth A. Holzhausen ◽  
Maria Nikodemova ◽  
Courtney L. Deblois ◽  
Jodi H. Barnet ◽  
Paul E. Peppard ◽  
...  

Abstract Background New technologies like next-generation sequencing have led to a proliferation of studies investigating the role of the gut microbiome in human health, particularly population-based studies that rely upon participant self-collection of samples. However, the impact of methodological differences in sample shipping, storage, and processing are not well-characterized for these types of studies, especially when transit times may exceed 24 h. The aim of this study was to experimentally assess microbiota stability in stool samples stored at 4 °C for durations of 6, 24, 48, 72, and 96 h with no additives to better understand effects of variable shipping times in population-based studies. These data were compared to a baseline sample that was immediately stored at − 80 °C after stool production. Results Compared to the baseline sample, we found that the alpha-diversity metrics Shannon’s and Inverse Simpson’s had excellent intra-class correlations (ICC) for all storage durations. Chao1 richness had good to excellent ICC. We found that the relative abundances of bacteria in the phyla Verrucomicrobia, Actinobacteria, and Proteobacteria had excellent ICC with baseline for all storage durations, while Firmicutes and Bacteroidetes ranged from moderate to good. We interpreted the ICCs as follows: poor: ICC < 0.50, moderate: 0.50 < ICC < 0.75, good: 0.75 < ICC < 0.90, and excellent: ICC > 0.90. Using the Bray–Curtis dissimilarity index, we found that the greatest change in community composition occurred between 0 and 24 h of storage, while community composition remained relatively stable for subsequent storage durations. Samples showed strong clustering by individual, indicating that inter-individual variability was greater than the variability associated with storage time. Conclusions The results of this analysis suggest that several measures of alpha diversity, relative abundance, and overall community composition are robust to storage at 4 °C for up to 96 h. We found that the overall community richness was influenced by storage duration in addition to the relative abundances of sequences within the Firmicutes and Bacteroidetes phyla. Finally, we demonstrate that inter-individual variability in microbiota composition was greater than the variability due to changing storage durations.


2018 ◽  
Author(s):  
Simon Lax ◽  
Cesar Cardona ◽  
Dan Zhao ◽  
Valerie J. Winton ◽  
Gabriel Goodney ◽  
...  

ABSTRACTDespite considerable efforts to characterize the ecology of bacteria and fungi in the built environment (BE), the metabolic mechanisms underpinning their colonization and successional dynamics remain unclear. Here, we applied bacterial/viral particle counting, qPCR, 16S and ITS rRNA amplicon sequencing, and metabolomics to longitudinally characterize the ecological dynamics of four commonly used building materials maintained at high humidity conditions (~94% RH). We varied the natural inoculum provided to each material by placing them in different occupied spaces, and we wet the surface of half of the samples of each material to simulate a flooding event. As expected, different materials showed different bacterial and viral particle abundance, with wet materials having higher growth rates and lower alpha diversity compared to non-wetted materials. Wetting described the majority of the variance in bacterial, fungal and metabolite structure, and material type only influenced bacterial and metabolic diversity, while location of inoculation was only weakly associated with bacterial and fungal beta diversity. Metabolites indicative of microbial activity were identified, as were those that were native to the surface material. Glucose-phosphate was abundant on all materials (except mold-free gypsum) and was correlated with Enterobacteriaceae, which could indicate a potential bacterial nutrient source. A compound consistent with scopoletin, a plant metabolite with antimicrobial activity, was significantly negatively correlated with Bacillus and positively correlated with Pseudomonas and enriched in medium density fiberboard (MDF) materials. In wet samples, the alkaloids nigragillin and fumigaclavine C, both with antimicrobial properties, were significantly positively correlated with the fungal phylum Ascomycota. Nigragillin, was also negatively correlated with Bacillus and Pseudomonas abundance. Thiabendazole and azoxystrobin (anti-fungal compounds) were highly abundant on mold-resistant gypsum wallboard and likely directly influenced the decreased fungal growth observed on this material. The mold-resistant gypsum material also showed a significant increase in bacterial alpha diversity, and bacterial and viral particle abundance, as well as a decrease in metabolite diversity, likely a result of reduced fungal growth. Penicillium taxa were positively correlated with thiabendazole, which suggested the persistence of resistant strains. Also, specific to the wet samples, Bacillus abundance was positively correlated with the azoxystrobin, suggesting bi-directional competitive adaptation, and positively correlated with metabolites known to interfere with Pseudomonas biofilm formation, which could explain the anti-correlation between these taxa. As expected, high moisture conditions enabled faster growth of inoculating microorganisms, whose composition, chemistry, and competition was shaped by surface material, suggesting that both fungal and bacterial growth need to be considered when determining the impact of dampness in built environments.


Antibiotics ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 146 ◽  
Author(s):  
Matteo Cuccato ◽  
Selene Rubiola ◽  
Diana Giannuzzi ◽  
Elena Grego ◽  
Paola Pregel ◽  
...  

In poultry production, gut microbiota (GM) plays a pivotal role and influences different host functions related to the efficiency of production performances. Antimicrobial (AM) use is one of the main factors affecting GM composition and functions. Although several studies have focused their attention on the role of AMs as growth promoters in the modulation of GM in broilers, the consequences of higher AM concentrations administered during prophylactic treatments need to be better elucidated. For this purpose, 16S rRNA gene sequencing was performed to evaluate the impact of different prophylactic AM protocols on the composition and diversity of the broiler GM. Diversity analysis has shown that AM treatment significantly affects alpha diversity in ileum and beta diversity in both ileum and caecum. In ileal samples, the Enterobacteriaceae family has been shown to be particularly affected by AM treatments. AMs have been demonstrated to affect GM composition in broiler. These findings indicate that withdrawal periods were not enough for the restoral of the original GM. Further studies are needed for a better elucidation of the negative effects caused by an altered GM in broilers.


Molecules ◽  
2021 ◽  
Vol 26 (13) ◽  
pp. 4007
Author(s):  
Magdalena Ratajczak ◽  
Dorota Kaminska ◽  
Eliza Matuszewska ◽  
Elżbieta Hołderna-Kedzia ◽  
Jarosław Rogacki ◽  
...  

Bee products have been known for centuries for their versatile healing properties. In recent decades they have become the subject of documented scientific research. This review aims to present and compare the impact of bee products and their components as antimicrobial agents. Honey, propolis, royal jelly and bee venom are bee products that have antibacterial properties. Sensitivity of bacteria to these products varies considerably between products and varieties of the same product depending on their origin. According to the type of bee product, different degrees of activity were observed against Gram-positive and Gram-negative bacteria, yeasts, molds and dermatophytes, as well as biofilm-forming microorganisms. Pseudomonas aeruginosa turned out to be the most resistant to bee products. An analysis of average minimum inhibitory concentration values for bee products showed that bee venom has the strongest bacterial effectiveness, while royal jelly showed the weakest antibacterial activity. The most challenging problems associated with using bee products for medical purposes are dosage and safety. The complexity and variability in composition of these products raise the need for their standardization before safe and predictable clinical uses can be achieved.


2020 ◽  
Author(s):  
Carola Venturini ◽  
Bethany Bowring ◽  
Alicia Fajardo-Lubian ◽  
Carol Devine ◽  
Jonathan Iredell

AbstractEffective antimicrobial stewardship requires a better understanding of the impact of different antibiotics on the gut microflora. Studies in humans are confounded by large inter-individual variability and difficulty in identifying control cohorts. However, controlled murine models can provide valuable information. We examined the impact of a penicillin-like antibiotic (piperacillin/tazobactam, TZP) or a third-generation cephalosporin (ceftriaxone, CRO) on the murine gut microbiota. We analyzed gut microbiome composition by 16S-rRNA amplicon sequencing and effects on the Enterobacteriaceae by qPCR and standard microbiology. Colonization resistance to multidrug resistant Escherichia coli ST131 and Klebsiella pneumoniae ST258 was also tested. Changes in microbiome composition and a significant (p<0.001) decrease in diversity occurred in all treated mice, but were more marked and longer lasting after CRO exposure with a persistent rise in Proteobacteria levels. Increases in the Enterobacteriaceae occurred in all antibiotic treated mice, but were transient and associated with direct antibiotic pressure. Co-habitation of treated and untreated mice attenuated the detrimental effect of antibiotics on treated animals, but also caused disturbance in untreated co-habitants. At the height of dysbiosis after antibiotic termination, the murine gut was highly susceptible to colonization with both multidrug resistant pathogens. The administration of a third-generation cephalosporin caused a significantly prolonged dysbiosis in the murine gut microflora, when compared to a penicillin/β-lactam inhibitor combination with comparable activity against medically important virulent bacteria. At the height of dysbiosis, both antibiotic treatments equally led to microbial imbalance associated with loss of resistance to gut colonization by antibiotic-resistant pathogens.


Sign in / Sign up

Export Citation Format

Share Document