Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Acta Biomater ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39368722

ABSTRACT

Oral biotherapeutics hold significant promise, but their lack of controllability and targeting poses a major challenge, particularly for intestinal bacterial biotherapeutics. In response, we have developed a nanoencapsulation approach that responds to the release of enzyme activity in the organism and activates the enzyme in situ, allowing for controlled colonization of microbes in the gut. The nano-coating comprises a two-layer structure: an inner layer of polydopamine with photothermal and adhesive properties, and an outer layer of gelatin-sodium carboxymethylcellulose, which is hydrolyzed by cellulases in the gut following photothermal interaction with dopamine. We have successfully achieved controlled colonization of a wide range of microorganisms. Furthermore, in a diabetes model, this approach has had a profound impact on regulating glucagon-like peptide-1 (GLP-1) production, ß-cell physiology, and promoting insulin secretion. This nanocoating is achieved by in situ activation of cellulase without the need for genetic or targeted molecular modification, representing a new paradigm and alternative strategy for microbial therapy. It not only enables precise and controlled colonization of probiotics but also demonstrates great potential for broader application in the field of oral biotherapy. STATEMENT OF SIGNIFICANCE: We have developed a nano-encapsulation method that triggers enzyme activity in response to enzymatic activity, resulting in the controlled release and adhesion of a wide range of microorganisms in the gut. The nano coating comprises two layers: an inner layer of polydopamine with photothermal and adhesion properties, and an outer layer of a gelatin-sodium carboxymethylcellulose polymer, which can be hydrolyzed by cellulases in the intestine. Additionally, this method allows for the preparation of various microbial coatings. This approach holds significant promise for regulating GLP-1 production, the physiological function of pancreatic ß-cells, and promoting insulin secretion in diabetes models.

2.
J Dent Res ; : 220345241272011, 2024 Oct 12.
Article in English | MEDLINE | ID: mdl-39394772

ABSTRACT

Childhood is considered crucial in the establishment of future oral microbiota. However, the precise period of oral microbiota development remains unclear. This study aimed to identify the progression of oral microbiota formation in children. We longitudinally investigated the salivary microbiota of 54 children across 13 time points from 1 wk to 60 mo (5 y) old and their parents at 2 time points as a representative sample of the adult microbiota. Using next-generation sequencing, we obtained 10,000 gene sequences of the 16s rRNA V1-V2 region for each sample. The detection rate in children of 110 operational taxonomic units commonly detected in more than 85% of mothers and fathers, defined as the main constituent bacteria, was 25% at 1 wk old, increased to 80% between 6 and 18 mo old, and reached approximately 90% by 36 mo old. Early main constituent bacteria detected at 1 wk old were limited to Streptococcus, Rothia, and Gemella. At 6 to 18 mo old, the detection rates of various main constituent bacteria, including Neisseria, Haemophilus, and Fusobacterium, increased. UniFrac distance analysis showed that the oral microbiota of children approached that of adults at 6 to 18 mo old. In the weighted UniFrac distance index, unlike the unweighted index, there were no significant changes in children between 36 and 60 mo old from adults, and microbiota formation at 60 mo old was sufficiently advanced to be included within the range of adult individual differences. Our findings suggest that the initial 36 mo, particularly the period from 6 to 18 mo old, consists of a time window for oral microbiota maturation. In addition, the development of microbiota during this period may be critical for future oral disease prevention.

3.
Article in English | MEDLINE | ID: mdl-39347930

ABSTRACT

BACKGROUND: The gut microbiota, comprising billions of microorganisms, plays a pivotal role in health and disease. This study aims to investigate the effect of sepsis on gut microbiome of neonates admitted to the Neonatal Intensive Care Unit. METHODS: A prospective cohort study was carried out in the NICU of tertiary care hospital in Karnataka, India, from January 2021 to September 2023. Preterm neonates with birth weight < 1500 g and gestational age < 37 weeks were recruited, excluding those with congenital gastrointestinal anomalies, necrotizing enterocolitis, or blood culture-negative infections. The study population was divided into three groups: healthy neonates (Group A), neonates with drug-sensitive GNB sepsis (Group B), and neonates with pan drug-resistant GNB sepsis (Group C). Stool samples were collected aseptically, snapped in liquid nitrogen, and stored at -80°C for extraction of DNA and microbiome analysis. RESULTS: The gut microbiota of healthy neonates (Group A) was dominated by Proteobacteria (24.04%), Actinobacteria (27.13%), Firmicutes (12.74%), and Bacteroidetes (3%). Predominant genera included Bifidobacterium (55.17%), Enterobacter (12.55%), Enterococcus (50.69%), Streptococcus (7.92%), and Bacteroides (3.58%).Groups B and C, the microbiota exhibited higher Proteobacteria abundance (57.16% and 66.58%, respectively) and reduced diversity of beneficial bacteria. Notably, the presence of sepsis was associated with an increase in pathogenic bacteria and a decrease in beneficial commensal bacteria. CONCLUSION: Neonates with sepsis exhibited significant gut microbiome dysbiosis, characterized by increased Proteobacteria and reduced beneficial bacteria diversity. These findings highlight the potential of microbiome profiling as a diagnostic tool and underscore the importance of gut microbiota modulation in managing neonatal sepsis.

4.
Cureus ; 16(7): e65220, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39184586

ABSTRACT

Introduction The oral cavity hosts diverse microorganisms affected by factors like pH, temperature, and oxygen levels, influencing disease potential. Dentists manage oral diseases and improve aesthetics using durable restorations. Understanding periodontal response to crowns and fixed partial dentures (FPDs) is essential for effective treatment. This study aims to assess the impact of FPDs on periodontal health by comparing microbial colonies in individuals with and without FPDs. The hypothesis is that there will be no difference in microbial flora and gingival health between the two groups. Materials and methods This in vitro study utilized 40 salivary samples divided into two groups: 20 from patients with FPDs/crowns (Group 1) and 20 from patients without (Group 2). Unstimulated saliva was collected, diluted, and cultured on nutrient agar and Sabouraud Dextrose Agar to quantify anaerobic bacteria and Candida colonies. Colony counts were scored from 0 to IV based on colony-forming unit (CFU), and microscopic examination identified the types of microbes present. Data were analyzed using an unpaired t-test with IBM SPSS Statistics for Windows, Version 26 (Released 2019; IBM Corp., Armonk, New York, United States), with significance set at p < 0.05. Results The independent t-test analysis showed significantly higher mean CFUs of anaerobic microbes in Group 1 (experimental) compared to Group 2 (control) (p = 0.000). However, mean CFUs of Candida did not significantly differ between groups (p = 0.194). Microscopic examination identified Enterococcus faecalis, Pseudomonas aeruginosa, Candida albicans, Staphylococcus aureus, and Streptococcus mutans in the experimental group, whereas the control group contained only Staphylococcus aureus and Streptococcus mutans. Conclusion Individuals with FPDs exhibit higher concentrations of anaerobic microbes and specific bacteria, suggesting an increased risk of gingival inflammation and emphasizing the importance of maintaining good oral hygiene.

5.
J Control Release ; 375: 20-46, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39214316

ABSTRACT

The microbiota at different sites in the body is closely related to disease. The intake of probiotics is an effective strategy to alleviate diseases and be adjuvant in their treatment. However, probiotics may suffer from harsh environments and colonization resistance, making it difficult to maintain a sufficient number of live probiotics to reach the target sites and exert their original probiotic effects. Encapsulation of probiotics is an effective strategy. Therefore, probiotic delivery systems, as effective methods, have been continuously developed and innovated to ensure that probiotics are effectively delivered to the targeted site. In this review, initially, the design of probiotic delivery systems is reviewed from four aspects: probiotic characteristics, processing technologies, cell-derived wall materials, and interactions between wall materials. Subsequently, the review focuses on the effects of probiotic delivery systems that target four main microbial colonization sites: the oral cavity, skin, intestine, and vagina, as well as disease sites such as tumors. Finally, this review also discusses the safety concerns of probiotic delivery systems in the treatment of disease and the challenges and limitations of implementing this method in clinical studies. It is necessary to conduct more clinical studies to evaluate the effectiveness of different probiotic delivery systems in the treatment of diseases.

6.
J Clin Med ; 13(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999378

ABSTRACT

Introduction: Patients with chronic pancreatitis (CP) as well as with pancreatic head carcinoma (CA) undergo the surgical intervention named "pylorus-preserving pancreatoduodenectomy according to Traverso-Longmire (PPPD)", which allowed a comparative analysis of the postoperative courses. The hypothesis was that patients with CA would have worse general as well as immune status than patients with CP due to the severity of the tumor disease and that this would be reflected in the more disadvantageous early postoperative outcome after PPPD. Methods: With the aim of eliciting the influence of the different diagnoses, the surgical outcome of all consecutive patients who underwent surgery at the Dept. of General, Abdominal, Vascular and Transplant Surgery at the University Hospital at Magdeburg between 2002 and 2015 (inclusion criterion) was recorded and comparatively evaluated. Early postoperative outcome was characterized by general and specific complication rate indicating morbidity, mortality, and microbial colonization rate, in particular surgical site infection (SSI, according to CDC criteria). In addition, microbiological findings of swabs and cultures from all compartments as well as preoperative and perioperative parameters from patient records were retrospectively documented and used for statistical comparison in this systematic retrospective unicenter observational study (design). Results: In total, 192 cases with CA (68.1%) and 90 cases with CP (31.9%) met the inclusion criteria of this study. Surprisingly, there were similar specific complication rates of 45.3% (CA) vs. 45.6% (CP; p = 0.97) and in-hospital mortality, which differed only slightly at 3.65% (CA) vs. 3.3% (CP; p = 0.591); the overall complication rate tended to be higher for CA at 23.4% vs. 14.4% (CP; p = 0.082). Overall, potentially pathogenic germs were detected in 28.9% of all patients in CP compared to 32.8% in CA (p = 0.509), and the rate of SSI was 29.7% (CA) and 24.4% (CP; p = 0.361). In multivariate analysis, CA was found to be a significant risk factor for the development of SSI (OR: 2.025; p = 0.048); the underlying disease had otherwise no significant effect on early postoperative outcome. Significant risk factors in the multivariate analysis were also male sex for SSI and microbial colonization, and intraoperatively transfused red cell packs for mortality, general and specific complications, and surgical revisions. Conclusions: Based on these results, a partly significant, partly trending negative influence of the underlying disease CA, compared to CP, on the early postoperative outcome was found, especially with regard to SSI after PPPD. This influence is corroborated by the international literature.

7.
Indian J Otolaryngol Head Neck Surg ; 76(3): 2411-2416, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38883486

ABSTRACT

Tracheostomy is a surgical procedure in an emergency setting to relieve the upper airway obstruction by creating an opening in the anterior part of trachea. It can also be done electively to wean off from a ventilator, during an elective surgery and clearance of pulmonary secretions. This study was a retrospective analysis of microbiological profile, antibiotic sensitivity & resistance pattern in patients with a tracheostomized wound. A retrospective review of the microbiological profiles of all adult patients who underwent a tracheostomy was conducted between May 2022 and May 2023 at our hospital. Based on the tracheostomy indications, patients were allocated under obstructed and non-obstructed group. Any patient with at least one positive sample was followed up quarterly for a year. The first culture result obtained was recorded at least one month following the last antibiotic dose in each quarter. Out of the 65 tracheal aspirate results obtained from 58 patients (mean age, 57.5 ± 16.48 years), the most common procedure and indications were surgical tracheostomy (72.4%) and non-obstructed causes (74.1%), respectively. Moreover, 47.7% of the culture results indicated Pseudomonas aeruginosa, which showed significantly different proportions across the quarters (p = 0.006). Among obstructed patients, P. aeruginosa was the most common (35%), followed by methicillin-resistant Staphylococcus aureus (MRSA; 23.5%). The colonization was predominantly by gram negative bacteria Acinetobacter species, P. aeruginosa & Klebsiella pneumoniae and fungal species like Candida albicans followed by Aspergillus niger and non-Albicans candida.

8.
Cell Rep ; 43(4): 114087, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38583152

ABSTRACT

Microbial invasions underlie host-microbe interactions resulting in pathogenesis and probiotic colonization. In this study, we explore the effects of the microbiome on microbial invasion in Drosophila melanogaster. We demonstrate that gut microbes Lactiplantibacillus plantarum and Acetobacter tropicalis improve survival and lead to a reduction in microbial burden during infection. Using a microbial interaction assay, we report that L. plantarum inhibits the growth of invasive bacteria, while A. tropicalis reduces this inhibition. We further show that inhibition by L. plantarum is linked to its ability to acidify its environment via lactic acid production by lactate dehydrogenase, while A. tropicalis diminishes the inhibition by quenching acids. We propose that acid from the microbiome is a gatekeeper to microbial invasions, as only microbes capable of tolerating acidic environments can colonize the host. The methods and findings described herein will add to the growing breadth of tools to study microbe-microbe interactions in broad contexts.


Subject(s)
Drosophila melanogaster , Animals , Drosophila melanogaster/microbiology , Microbiota , Acetobacter/metabolism , Gastrointestinal Microbiome/drug effects , Lactobacillus plantarum/metabolism , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Lactic Acid/pharmacology
9.
Microorganisms ; 12(3)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38543622

ABSTRACT

This comprehensive review delves into the forefront of research on the human oral microbiome, exploring recent advancements that span microbial colonization, state-of-the-art detection methodologies, and the complex interplay involved in disease progression. Through an exhaustive analysis of the contemporary literature, we illuminate the dynamic orchestration of microbial communities within the oral cavity, underscoring their pivotal role in health and disease. Cutting-edge detection techniques, including metagenomics and high-throughput sequencing, are discussed regarding their transformative impact on understanding the intricacies of oral microbial ecosystems. As we stand on the cusp of a new decade, this review anticipates a paradigm shift in the field, emphasizing the potential for rapid identification and targeted management of detrimental oral microorganisms. Insights gained from this exploration not only contribute to our fundamental understanding of the oral microbiome but also hold promise for the development of innovative therapeutic strategies to maintain oral health. This article aims to serve as a valuable resource for researchers, clinicians, and public health professionals engaged in unraveling the mysteries of the microbial symphony within the human oral cavity.

10.
FEMS Microbiol Lett ; 3712024 Jan 09.
Article in English | MEDLINE | ID: mdl-38553956

ABSTRACT

Habitat type is a strong determinant of microbial composition. Habitat interfaces, such as the boundary between aquatic and terrestrial systems, present unique combinations of abiotic factors for microorganisms to contend with. Aside from the spillover of certain harmful microorganisms from agricultural soils into water (e.g. fecal coliform bacteria), we know little about the extent of soil-water habitat switching across microbial taxa. In this study, we developed a proof-of-concept system to facilitate the capture of putatively generalist microorganisms that can colonize and persist in both soil and river water. We aimed to examine the phylogenetic breadth of putative habitat switchers and how this varies across different source environments. Microbial composition was primarily driven by recipient environment type, with the strongest phylogenetic signal seen at the order level for river water colonizers. We also identified more microorganisms colonizing river water when soil was collected from a habitat interface (i.e. soil at the side of an intermittently flooded river, compared to soil collected further from water sources), suggesting that environmental interfaces could be important reservoirs of microbial habitat generalists. Continued development of experimental systems that actively capture microorganisms that thrive in divergent habitats could serve as a powerful tool for identifying and assessing the ecological distribution of microbial generalists.


Subject(s)
Ecosystem , Fresh Water , Phylogeny , Fresh Water/microbiology , Soil , Water
11.
Proc Biol Sci ; 291(2016): 20232036, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38320611

ABSTRACT

Early life microbial colonizers shape and support the immature vertebrate immune system. Microbial colonization relies on the vertical route via parental provisioning and the horizontal route via environmental contribution. Vertical transmission is mostly a maternal trait making it hard to determine the source of microbial colonization in order to gain insight into the establishment of the microbial community during crucial development stages. The evolution of unique male pregnancy in pipefishes and seahorses enables the disentanglement of both horizontal and vertical transmission, but also facilitates the differentiation of maternal versus paternal provisioning ranging from egg development, to male pregnancy and early juvenile development. Using 16S rRNA amplicon sequencing and source-tracker analyses, we revealed how the distinct origins of transmission (maternal, paternal and horizontal) shaped the juvenile internal and external microbiome establishment in the broad-nosed pipefish Syngnathus typhle. Our data suggest that transovarial maternal microbial contribution influences the establishment of the juvenile gut microbiome whereas paternal provisioning mainly shapes the juvenile external microbiome. The identification of juvenile key microbes reveals crucial temporal shifts in microbial development and enhances our understanding of microbial transmission routes, colonization dynamics and their impact on lifestyle evolution.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Smegmamorpha , Animals , Male , RNA, Ribosomal, 16S/genetics , Immune System , Smegmamorpha/genetics
12.
Cureus ; 16(1): e52972, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38406113

ABSTRACT

This comprehensive review explores the intricate landscape of the neonatal skin microbiome, shedding light on its dynamic composition, developmental nuances, and influential factors. The neonatal period represents a critical window during which microbial colonization significantly impacts local skin health and the foundational development of the immune system. Factors such as mode of delivery and gestational age underscore the vulnerability of neonates to disruptions in microbial establishment. Key findings emphasize the broader systemic implications of the neonatal skin microbiome, extending beyond immediate health outcomes to influence susceptibility to infections, allergies, and immune-related disorders. This review advocates for a paradigm shift in neonatal care, proposing strategies to preserve and promote a healthy skin microbiome for long-term health benefits. The implications of this research extend to public health, where interventions targeting the neonatal skin microbiome could potentially mitigate diseases originating in early life. As we navigate the intersection of research and practical applications, bridging the gap between knowledge and implementation becomes imperative for translating these findings into evidence-based practices and improving neonatal well-being on a broader scale.

13.
Environ Pollut ; 342: 123136, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38092341

ABSTRACT

Municipal sewer systems have received increasing attention due to the magnitude of the microplastic stock and its potential ecological impacts. However, as a critical aspect of the adverse impacts, little is known about the plastisphere that forms in these engineered environments. Using high-throughput absolute quantification sequencing, we conducted a systemic study combining field survey and laboratory batch test to explain the general plastisphere pattern and the role of environmental and polymeric factors in driving plastisphere succession and assembly there. We demonstrated the capacity of microplastics to support high levels of microbial colonization, increasing by 8.7-56.0 and 1.26-5.62 times at field and laboratory scales, respectively, despite the less diverse communities hosted in the resulting plastisphere. Sediment communities exhibited higher diversity but greater loss of specific operational taxonomic units in their plastisphere than in the wastewater. The former plastisphere had primarily an enhanced methanogenesis-oriented metabolic network linked to hydrolysis fermentation, hydrogen-producing acetogenesis, and denitrification, while the latter had a pronounced niche partitioning and competitive interaction network. Exogenous substrate flux and composition were key in stimulating plastisphere community growth and succession. Furthermore, the high nitrogen baseline facilitated alternative niche formation for plastisphere nitrifiers and denitrifiers, and the plastisphere pathogens associated with denitrification and plastic biodegradation functions increased significantly. The aerobic state also promoted a 1.71 times higher colonizer load and a denser interaction pattern than the anaerobic state. Selective filtering by polymers was evident: polyethylene supported higher plastisphere diversity than polypropylene. This study provides new insights into the mechanisms driving colonizer loads and the adaptive succession and assembly of the plastisphere in such a typically hydrodynamic and highly contaminated environment. The results help to fill the knowledge gap in understanding the potential role of microplastics in shaping the microecology of sewers and increasing health risks and substrate loss during sewer transfer.


Subject(s)
Microplastics , Plastics , Biodegradation, Environmental , Fermentation , High-Throughput Nucleotide Sequencing
14.
Wien Med Wochenschr ; 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37861874

ABSTRACT

Diaper dermatitis (DD) is an umbrella term with different clinical presentations, pathophysiology, treatments, and outcomes. The major subtypes include irritant contact dermatitis and candida-associated DD. In case of atypical presentation or unresponsiveness to treatment, other differential diagnoses should be considered. Although DD can occur at any age, it peaks in newborns and infants < 2 years of age. We will focus on this age group in our narrative review.

15.
Front Microbiol ; 14: 1129250, 2023.
Article in English | MEDLINE | ID: mdl-37795296

ABSTRACT

Neonatal calves have a limited capacity to initiate immune responses due to a relatively immature adaptive immune system, which renders them susceptible to many on-farm diseases. At birth, the mucosal surfaces of the intestine are rapidly colonized by microbes in a process that promotes mucosal immunity and primes the development of the adaptive immune system. In a companion study, our group demonstrated that supplementation of a live yeast probiotic, Saccharomyces cerevisiae boulardii (SCB) CNCM I-1079, to calves from birth to 1 week of age stimulates secretory IgA (sIgA) production in the intestine. The objective of the study was to evaluate how SCB supplementation impacts the intestinal microbiota of one-week-old male calves, and how changes in the bacterial community in the intestine relate to the increase in secretory IgA. A total of 20 calves were randomly allocated to one of two treatments at birth: Control (CON, n = 10) fed at 5 g/d of carrier with no live yeast; and SCB (n = 10) fed at 5 g of live SCB per day (10 × 109 CFU/d). Our study revealed that supplementing calves with SCB from birth to 1 week of age had its most marked effects in the ileum, increasing species richness and phylogenetic diversity in addition to expediting the transition to a more interconnected bacterial community. Furthermore, LEfSe analysis revealed that there were several differentially abundant taxa between treatments and that SCB increased the relative abundance the family Eubacteriaceae, Corynebacteriaceae, Eggerthellaceae, Bacillaceae, and Ruminococcaceae. Furthermore, network analysis suggests that SCB promoted a more stable bacterial community and appears to reduce colonization with Shigella. Lastly, we observed that the probiotic-driven increase in microbial diversity was highly correlated with the enhanced secretory IgA capacity of the ileum, suggesting that the calf's gut mucosal immune system relies on the development of a stable and highly diverse microbial community to provide the necessary cues to train and promote its proper function. In summary, this data shows that supplementation of SCB promoted establishment of a diverse and interconnected microbiota, prevented colonization of Escherichia Shigella and indicates a possible role in stimulating humoral mucosal immunity.

16.
Animals (Basel) ; 13(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37684982

ABSTRACT

The consumption of maternal feces (coprophagy) is commonly observed in healthy foals and is a proposed contributor to microbial colonization of the foal's gastrointestinal tract (GIT). This study investigated the role of coprophagy in the establishment of fibrolytic bacteria in the foal GIT. Nine thoroughbred mares were dosed with chromic oxide, an indigestible marker, as a method to detect the occurrence of coprophagy by their foals. Foal fecal samples were collected from 12 h to 21 d after birth to measure chromic oxide and neutral detergent fiber (NDF) and to enumerate cellulolytic bacteria using culture-based techniques. Milk yield was estimated at 7 and 14 d postpartum. Coprophagy was detected as early as 3 d after birth and detected in all foals by 7 d of age. There were strong relationships between coprophagy and cellulolytic bacteria and NDF in foal feces at 7 d of age (r = 0.9703 and r = 0.7878, respectively; p < 0.05). Fecal NDF and chromic oxide concentrations were negatively related to milk yield (r = -0.8144 and r = -0.6966, respectively; p < 0.05), suggesting milk availability affected the incidence of coprophagy. Based on the relationships identified, maternal feces are an important source of fiber and live microbes for the foal, contributing to the development of the microbial community.

17.
Sci Total Environ ; 897: 165318, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37422225

ABSTRACT

The development of diagnostic methods to accurately assess the effects of treatments on lithobiont colonization remains a challenge for the conservation of Cultural Heritage monuments. In this study, we tested the efficacy of biocide-based treatments on microbial colonization of a dolostone quarry, in the short and long-term, using a dual analytical strategy. We applied a metabarcoding approach to characterize fungal and bacterial communities over time, integrated with microscopy techniques to analyze the interactions of microorganisms with the substrate and evaluate the effectiveness. These communities were dominated by the bacterial phyla Actinobacteriota, Proteobacteria and Cyanobacteria, and the fungal order Verrucariales, which include taxa previously reported as biodeteriogenic agents and observed here associated with biodeterioration processes. Following the treatments, changes over time in the abundance profiles depend on taxa. While Cyanobacteriales, Cytophagales and Verrucariales decreased in abundance, other groups, such as Solirubrobacteriales, Thermomicrobiales and Pleosporales increased. These patterns could be related not only to the specific effects of the biocide on the different taxa, but also to different recolonization abilities of those organisms. The different susceptibility to treatments could be associated with the inherent cellular properties of different taxa, but differences in biocide penetration to endolithic microhabitats could be involved. Our results demonstrate the importance of both removing epilithic colonization and applying biocides to act against endolithic forms. Recolonization processes could also explain some of the taxon-dependent responses, especially in the long-term. Taxa showing resistance, and those benefiting from nutrient accumulation in the form of cellular debris following treatments, may have an advantage in colonizing treated areas, pointing to the need for long-term monitoring of a wide range of taxa. This study highlights the potential utility of combining metabarcoding and microscopy to analyze the effects of treatments and design appropriate strategies to combat biodeterioration and establish preventive conservation protocols.


Subject(s)
Ascomycota , Cyanobacteria , Disinfectants , Disinfectants/pharmacology , Microscopy
18.
Animals (Basel) ; 13(14)2023 Jul 09.
Article in English | MEDLINE | ID: mdl-37508029

ABSTRACT

Feces enable frequent samplings for the same animal, which is valuable in studies investigating the development of the gut microbiome in piglets. Creep feed should prepare the piglet's gut for the postweaning period and shape the microbiome accordingly. Little is known about the variation that is caused by differences in fecal color and consistency and different sample types (feces versus swab samples). Therefore, this study evaluated the age-related alterations in the microbiome composition (16S rRNA gene) in feces of suckling and newly weaned piglets in the context of nutrition and fecal consistency, color and sample type from day 2 to 34 of life. Feces from 40 healthy piglets (2 each from 20 litters) were collected on days 2, 6, 13, 20, 27, 30 and 34. Weaning occurred on day 28. Half of the litters only drank sow milk during the suckling phase, whereas the other half had access to creep feed from day 10. Creep feeding during the suckling phase influenced the age-related total bacterial and archaeal abundances but had less of an influence on the relative bacterial composition. Results further showed different taxonomic compositions in feces of different consistency, color and sample type, emphasizing the need to consider these characteristics in comprehensive microbiome studies.

19.
Mol Med ; 29(1): 90, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37407941

ABSTRACT

Abnormal microbial colonization in the gut at an early stage of life affects growth, development, and health, resulting in short- and long-term adverse effects. Microbial colonization patterns of preterm infants differ from those of full-term infants in that preterm babies and their mothers have more complicated prenatal and postnatal medical conditions. Maternal complications, antibiotic exposure, delivery mode, feeding type, and the use of probiotics may significantly shape the gut microbiota of preterm infants at an early stage of life; however, these influences subside with age. Although some factors and processes are difficult to intervene in or avoid, understanding the potential factors and determinants will help in developing timely strategies for a healthy gut microbiota in preterm infants. This review discusses potential determinants of gut microbial colonization in preterm infants and their underlying mechanisms.


Subject(s)
Gastrointestinal Microbiome , Infant, Premature , Infant , Pregnancy , Female , Infant, Newborn , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
20.
Nutrients ; 15(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37375672

ABSTRACT

BACKGROUND: An inadequate perinatal nutritional environment can alter the maturation of the intestinal barrier and promote long-term pathologies such as metabolic syndrome or chronic intestinal diseases. The intestinal microbiota seems to play a determining role in the development of the intestinal barrier. In the present study, we investigated the impact of consuming an early postnatal prebiotic fiber (PF) on growth, intestinal morphology and the microbiota at weaning in postnatal-growth-restricted mice (PNGR). METHODS: Large litters (15 pups/mother) were generated from FVB/NRj mice to induce PNGR at postnatal day 4 (PN4) and compared to control litters (CTRL, 8 pups/mother). PF (a resistant dextrin) or water was orally administered once daily to the pups from PN8 to PN20 (3.5 g/kg/day). Intestinal morphology was evaluated at weaning (PN21) using the ileum and colon. Microbial colonization and short-chain fatty acid (SCFA) production were investigated using fecal and cecal contents. RESULTS: At weaning, the PNGR mice showed decreased body weight and ileal crypt depth compared to the CTRL. The PNGR microbiota was associated with decreased proportions of the Lachnospiraceae and Oscillospiraceae families and the presence of the Akkermansia family and Enterococcus genus compared to the CTRL pups. The propionate concentrations were also increased with PNGR. While PF supplementation did not impact intestinal morphology in the PNGR pups, the proportions of the Bacteroides and Parabacteroides genera were enriched, but the proportion of the Proteobacteria phylum was reduced. In the CTRL pups, the Akkermansia genus (Verrucomicrobiota phylum) was present in the PF-supplemented CTRL pups compared to the water-supplemented ones. CONCLUSIONS: PNGR alters intestinal crypt maturation in the ileum at weaning and gut microbiota colonization. Our data support the notion that PF supplementation might improve gut microbiota establishment during the early postnatal period.


Subject(s)
Dietary Supplements , Prebiotics , Female , Pregnancy , Animals , Mice , Intestines , Lactation , Mice, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL