Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.374
Filter
1.
Int J Food Microbiol ; 422: 110809, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38955023

ABSTRACT

Sterigmatocystin (STC) is an emerging mycotoxin that poses a significant threat to the food security of cereal crops. To mitigate STC contamination in maize, this study employed selected lactic acid bacteria as biocontrol agents against Aspergillus versicolor, evaluating their biocontrol potential and analyzing the underlying mechanisms. Lactiplantibacillus plantarum HJ10, isolated from pickle, exhibited substantial in vitro antifungal activity and passed safety assessments, including antibiotic resistance and hemolysis tests. In vivo experiments demonstrated that L. plantarum HJ10 significantly reduced the contents of A. versicolor and STC in maize (both >84 %). The impact of heat, enzymes, alkali, and other treatments on the antifungal activity of cell-free supernatant (CFS) was investigated. Integrated ultra-high-performance liquid chromatography (UPLC) and gas chromatography-mass spectrometry (GC-MS) analysis revealed that lactic acid, acetic acid, and formic acid are the key substances responsible for the in vitro antifungal activity of L. plantarum HJ10. These metabolites induced mold apoptosis by disrupting cell wall structure, increasing cell membrane fluidity, reducing enzyme activities, and disrupting energy metabolism. However, in vivo antagonism by L. plantarum HJ10 primarily occurs through organic acid production and competition for growth space and nutrients. This study highlights the potential of L. plantarum HJ10 in reducing A. versicolor and STC contamination in maize.


Subject(s)
Aspergillus , Lactobacillales , Sterigmatocystin , Zea mays , Zea mays/microbiology , Aspergillus/metabolism , Aspergillus/growth & development , Lactobacillales/metabolism , Antifungal Agents/pharmacology , Food Contamination/prevention & control , Antibiosis
2.
J Diabetes Complications ; 38(8): 108780, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38968867

ABSTRACT

BACKGROUND-AIM: Non-alcoholic fatty liver disease (NAFLD1) is the most frequent chronic liver disorder worldwide. Currently, no pharmacological treatment has been approved for NAFLD. Probiotics have been suggested as a potential therapy for NAFLD. The aim of this systematic review and meta-analysis was to assess the impact of probiotic intake on liver tests, lipids, glycemic parameters and inflammatory markers in NAFLD patients. METHODS: We searched electronic databases using related terms. Meta-analysis was performed using random-effects models. Clinical outcomes were presented as standard mean difference (SMD2) with a 95 % confidence interval (CI3). Publication bias and heterogeneity were evaluated in eligible studies. RESULTS: Fifteen randomized clinical trials comprising 899 participants were included in our meta-analysis. Probiotic supplementation improved alanine transaminase [SMD -0.796; 95 % CI (-1.419, -0.172); p = 0.012], Homeostatic Model Assessment for Insulin Resistance (HOMA-IR4) [SMD -0.596; 95 % CI (-1.071, -0.121); p = 0.01] and insulin levels [SMD -1.10; 95 % CI (-2.121, -0.087); p = 0.03]. No significant effects were observed on fasting glucose, hemoglobin A1c, aspartate transaminase, lipid profile, interleukin-6 and tumor necrosis factor-α. CONCLUSIONS: Probiotic intake may improve insulin sensitivity and alanine transaminase in NAFLD patients.


Subject(s)
Lipids , Non-alcoholic Fatty Liver Disease , Probiotics , Randomized Controlled Trials as Topic , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/complications , Probiotics/therapeutic use , Probiotics/administration & dosage , Humans , Lipids/blood , Glycemic Index , Liver/metabolism , Biomarkers/blood , Insulin Resistance , Liver Function Tests , Blood Glucose/metabolism , Blood Glucose/analysis , Inflammation Mediators/blood
3.
Mol Med ; 30(1): 105, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030525

ABSTRACT

Radiotherapy is a widely used cancer treatment that utilizes powerful radiation to destroy cancer cells and shrink tumors. While radiation can be beneficial, it can also harm the healthy tissues surrounding the tumor. Recent research indicates that the microbiota, the collection of microorganisms in our body, may play a role in influencing the effectiveness and side effects of radiation therapy. Studies have shown that specific species of bacteria living in the stomach can influence the immune system's response to radiation, potentially increasing the effectiveness of treatment. Additionally, the microbiota may contribute to adverse effects like radiation-induced diarrhea. A potential strategy to enhance radiotherapy outcomes and capitalize on the microbiome involves using probiotics. Probiotics are living microorganisms that offer health benefits when consumed in sufficient quantities. Several studies have indicated that probiotics have the potential to alter the composition of the gut microbiota, resulting in an enhanced immune response to radiation therapy and consequently improving the efficacy of the treatment. It is important to note that radiation can disrupt the natural balance of gut bacteria, resulting in increased intestinal permeability and inflammatory conditions. These disruptions can lead to adverse effects such as diarrhea and damage to the intestinal lining. The emerging field of radiotherapy microbiome research offers a promising avenue for optimizing cancer treatment outcomes. This paper aims to provide an overview of the human microbiome and its role in augmenting radiation effectiveness while minimizing damage.


Subject(s)
Gastrointestinal Microbiome , Neoplasms , Probiotics , Radiotherapy , Humans , Gastrointestinal Microbiome/radiation effects , Neoplasms/radiotherapy , Neoplasms/microbiology , Neoplasms/immunology , Neoplasms/therapy , Probiotics/therapeutic use , Radiotherapy/adverse effects , Radiotherapy/methods , Animals , Microbiota/radiation effects , Radiation Injuries/microbiology , Radiation Injuries/therapy , Radiation Injuries/etiology , Treatment Outcome
5.
Article in English | MEDLINE | ID: mdl-39046670

ABSTRACT

The mammalian gut microbiota plays a crucial role in promoting host health, and lactic acid bacteria (LAB) are commonly employed as probiotics for their beneficial effects. The Hangul deer (Cervus hanglu hanglu), a critically endangered red deer subspecies found in the Indian subcontinent, requires meticulous health management for its conservation. This pioneering study aimed to isolate, identify, and evaluate the in-vitro probiotic functional properties of LAB strains from the faeces of Hangul deer. A total of 27 LAB strains were isolated and identified using 16S rDNA gene sequencing, followed by comprehensive probiotic characterization and safety assessment. Remarkably, four species exhibited robust resistance and survivability against varying pH levels and bile salts, along with high aggregation and co-aggregation capacities. Notably, Lactobacillus acidophilus and Enterococcus mundtii strains displayed antibacterial activities. Safety assessment revealed the absence of hemolytic activity and virulence genes in all four strains. Antibiotic susceptibility testing showed that Lactobacillus acidophilus and Enterococcus casseliflavus were susceptible to all tested antibiotics, while Enterococcus mundtii exhibited resistance to clindamycin, and Enterococcus gallinarum exhibited resistance to erythromycin. These findings suggest that the isolated LAB strains possess advantageous probiotic characteristics and hold potential as dietary supplements for promoting the health and disease management of Hangul deer.

6.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-39038938

ABSTRACT

With the increasing prevalence of age-related chronic diseases burdening healthcare systems, there is a pressing need for innovative management strategies. Our study focuses on the gut microbiota, essential for metabolic, nutritional, and immune functions, which undergoes significant changes with aging. These changes can impair intestinal function, leading to altered microbial diversity and composition that potentially influence health outcomes and disease progression. Using advanced metagenomic sequencing, we explore the potential of personalized probiotic supplements in 297 older adults by analyzing their gut microbiota. We identified distinctive Lactobacillus and Bifidobacterium signatures in the gut microbiota of older adults, revealing probiotic patterns associated with various population characteristics, microbial compositions, cognitive functions, and neuroimaging results. These insights suggest that tailored probiotic supplements, designed to match individual probiotic profile, could offer an innovative method for addressing age-related diseases and functional declines. Our findings enhance the existing evidence base for probiotic use among older adults, highlighting the opportunity to create more targeted and effective probiotic strategies. However, additional research is required to validate our results and further assess the impact of precision probiotics on aging populations. Future studies should employ longitudinal designs and larger cohorts to conclusively demonstrate the benefits of tailored probiotic treatments.


Subject(s)
Aging , Dietary Supplements , Gastrointestinal Microbiome , Probiotics , Probiotics/therapeutic use , Probiotics/administration & dosage , Humans , Aged , Female , Male , Aged, 80 and over , Middle Aged , Lactobacillus/genetics , Metagenomics/methods , Bifidobacterium
7.
Article in English | MEDLINE | ID: mdl-39058454

ABSTRACT

BACKGROUND AND OBJECTIVES: Newborns, particularly premature ones, are vulnerable to bacterial infections and often receive antibiotics, disrupting their normal intestinal microbiota. Probiotics have multiple health benefits and are recommended for restoring balance, but caution is needed due to potential consequences, such as promoting antibiotic resistance. Therefore, this study aimed to conduct a systematic review to assess current knowledge regarding the impact of probiotics on antibiotic resistance in newborns. METHODS: This systematic review is aligned with the PRISMA 2020 statement. The search involved three electronic databases (PubMed, Scopus, and Web of Science) and the keywords used were "newborn", "probiotic", and "antibiotic resistance". Titles/abstracts were independently analyzed, followed by full-text assessments. Data extraction and synthesis were conducted, assessing outcomes like antibiotic resistance gene and mobile gene elements quantity and quality, vancomycin resistant enterococci colonization, antibiotic-resistant bacteria colonization, and microbiota changes. It was only included randomized controlled trials and cohort studies focusing on human newborns. RESULTS: In the study selection process, 1,970 articles were identified and six were included in this review. The results varied, with two studies suggesting a positive impact of probiotics on antibiotic resistance, one a negative impact, while three others found no significant impact. Microbiota changes were also addressed across four studies, where the findings included promotion of beneficial bacteria and reduction of potential pathogens. Bias risk was evaluated using Joanna Briggs Institute tools. CONCLUSION: Our findings did not allow for clear conclusions to be drawn, due to the observed heterogeneity between studies, highlighting the need for further studies.

8.
Fish Shellfish Immunol ; 152: 109788, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39053586

ABSTRACT

In the process of screening for probiotic strains, there are no clearly established bacterial phenotypic markers which could be used for the prediction of their in vivo mechanism of action. In this work, we demonstrate for the first time that Machine Learning (ML) methods can be used for accurately predicting the in vivo immunomodulatory activity of probiotic strains based on their cell surface phenotypic features using a snail host-microbe interaction model. A broad range of snail gut presumptive probiotics, including 240 new lactic acid bacterial strains (Lactobacillus, Leuconostoc, Lactococcus, and Enterococcus), were isolated and characterized based on their capacity to withstand snails' gastrointestinal defense barriers, such as the pedal mucus, gastric mucus, gastric juices, and acidic pH, in association with their cell surface hydrophobicity, autoaggregation, and biofilm formation ability. The implemented ML pipeline predicted with high accuracy (88 %) strains with a strong capacity to enhance chemotaxis and phagocytic activity of snails' hemolymph cells, while also revealed bacterial autoaggregation and cell surface hydrophobicity as the most important parameters that significantly affect host immune responses. The results show that ML approaches may be useful to derive a predictive understanding of host-probiotic interactions, while also highlighted the use of snails as an efficient animal model for screening presumptive probiotic strains in the light of their interaction with cellular innate immune responses.

9.
Dent J (Basel) ; 12(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39057009

ABSTRACT

S. salivarius M18 administration has been proven to provide positive effects on periodontal health; however, there is still no consensus on the optimum duration of probiotic administration. This study aimed to evaluate the effect of three months of probiotic supplementation on bleeding on probing, signs of gingival inflammation, and dental biofilm. Sixty-two eligible individuals with gingivitis were enrolled in this placebo-controlled, double-blind trial and randomly allocated to the M18 or control groups. Primary outcomes were changes in gingival condition (gingival index, GI; gingival bleeding index, GBI) after 1, 2, and 3 months of lozenges administration and after a one-month washout. Secondary outcomes included changes in the Quigley-Hein plaque index (modified by Turesky et al.) after 1, 2, and 3 months of lozenges administration and after a washout. In total, 60 individuals completed the study (31 and 29 in the M18 group and the control group, respectively). No severe adverse events were reported. Probiotic supplementation resulted in a significant decrease in gingival bleeding at 1 month (effect size 1.09 [CI95%: 0.55-1.63]), 2 months (effect size 0.78 [CI95%: 0.26-1.30]), and 3 months (effect size 0.67 [CI95%: 0.15-1.18]) and a significant reduction in dental plaque accumulation at 2 months (effect size 0.63 [CI95%: 0.12-1.14]) and 3 months (effect size 0.55 [CI95%: 0.03-1.05]). A three-month supplementation with the probiotic resulted in a significant reduction in gingival bleeding and biofilm accumulation; however, a long-lasting effect is not expected, indicating the need for probiotic intake on a long-term basis.

10.
Curr Issues Mol Biol ; 46(7): 7447-7472, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39057083

ABSTRACT

Even with the intensive efforts by public health programs to control and prevent it, non-typhoidal Salmonella (NTS) infection remains an important public health challenge. It is responsible for approximately 150 million illnesses and 60,000 deaths worldwide annually. NTS infection poses significant risks with high rates of morbidity and mortality, leading to potential short- and long-term complications. There is growing concern among health authorities about the increasing incidence of antimicrobial resistance, with multidrug resistance totaling 22.6% in Europe, highlighting an urgent need for new therapeutic approaches. Our review aims to provide a comprehensive overview of NTS infection. We outline the molecular mechanisms involved in the pathogenesis of NTS infection, as well as the events leading to invasive NTS infection and the subsequent complications associated with it. Given the widespread implications of antimicrobial resistance, our review also presents the global landscape of resistance, including multidrug resistance, and delve into the underlying mechanisms driving this resistance. The rising rates of antibiotic resistance frequently lead to treatment failures, emphasizing the importance of investigating alternative therapeutic options. Therefore, in this review we also explore potential alternative therapies that could offer promising approaches to treating NTS infections.

11.
J Fungi (Basel) ; 10(7)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39057374

ABSTRACT

Yeasts are gaining increasing attention for their potential health benefits as probiotics in recent years. Researchers are actively searching for new yeast strains with probiotic properties (i.e, Debaryomyces hansenii; Kluyveromyces marxianus; Yarrowia lipolytica; Pichia hudriavzevii; and Torulaspora delbrueckii) from various sources, including traditional fermented foods, the human gut, and the environment. This exploration is expanding the pool of potential probiotic yeasts beyond the well-studied Saccharomyces boulardii. Research suggests that specific yeast strains possess properties that could be beneficial for managing conditions like inflammatory bowel disease, irritable bowel syndrome, skin disorders, and allergies. Additionally, probiotic yeasts may compete with pathogenic bacteria for adhesion sites and nutrients, thereby inhibiting their growth and colonization. They might also produce antimicrobial compounds that directly eliminate harmful bacteria. To achieve these goals, the approach that uses probiotics for human health is changing. Next-generation yeast probiotics are emerging as a powerful new approach in the field of live biotherapeutics. By using genetic engineering, scientists are able to equip these tools with specialized capabilities. However, most research on these probiotic yeasts is still in its early stages, and more clinical trials are needed to confirm their efficacy and safety for various health conditions. This review could provide a brief overview of the situation in this field.

12.
Metabolites ; 14(7)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39057689

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) poses an emerging threat topublic health. Nonalcoholic steatohepatitis (NASH) is reported to be the most rapidly rising cause of hepatocellular carcinoma in the western world. Recently, a new term has been proposed: metabolic dysfunction-associated steatotic liver disease (MASLD). The introduction of this new terminology has sparked a debate about the interchangeability of these terms. The pathogenesis of NAFLD/MASLD is thought to be multifactorial, involving both genetic and environmental factors. Among these factors, alterations in gut microbiota and gut dysbiosis have recently garnered significant attention. In this context, this review will further discuss the gut-liver axis, which refers to the bidirectional interaction between the human gut microbiota and the liver. Additionally, the therapeutic potential of probiotics, particularly next-generation probiotics and genetically engineered bacteria, will be explored. Moreover, the role of prebiotics, synbiotics, postbiotics, and phages as well as fecal microbiota transplantation will be analyzed. Particularly for lean patients with NAFLD/MASLD, who have limited treatment options, approaches that modify the diversity and composition of the gut microbiota may hold promise. However, due to ongoing safety concerns with approaches that modulate gut microbiota, further large-scale studies are necessary to better assess their efficacy and safety in treating NAFLD/MASLD.

13.
Toxics ; 12(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39058179

ABSTRACT

Microcystin-LR (MCLR) poses a significant threat to aquatic ecosystems and public health. This study investigated the protective effects of the probiotic Lactobacillus rhamnosus against MCLR-induced developmental toxicity in zebrafish larvae. Zebrafish larvae were exposed to various concentrations of MCLR (0, 0.9, 1.8, and 3.6 mg/L) with or without L. rhamnosus from 72 to 168 h post-fertilization (hpf). Probiotic supplementation significantly improved survival, hatching, and growth rates and reduced malformation rates in MCLR-exposed larvae. L. rhamnosus alleviated MCLR-induced oxidative stress by reducing reactive oxygen species (ROS) levels and enhancing glutathione (GSH) content and catalase (CAT) activity. Probiotics also mitigated MCLR-induced lipid metabolism disorders by regulating key metabolites (triglycerides, cholesterol, bile acids, and free fatty acids) and gene expression (ppara, pparb, srebp1, and nr1h4). Moreover, 16S rRNA sequencing revealed that L. rhamnosus modulated the gut microbiome structure and diversity in MCLR-exposed larvae, promoting beneficial genera like Shewanella and Enterobacter and inhibiting potential pathogens like Vibrio. Significant correlations were found between gut microbiota composition and host antioxidant and lipid metabolism parameters. These findings suggest that L. rhamnosus exerts protective effects against MCLR toxicity in zebrafish larvae by alleviating oxidative stress, regulating lipid metabolism, and modulating the gut microbiome, providing insights into probiotic-based strategies for mitigating MCLR toxicity in aquatic organisms.

14.
Adv Exp Med Biol ; 1449: 59-78, 2024.
Article in English | MEDLINE | ID: mdl-39060731

ABSTRACT

Infantile colic (IC) is c is a self-limiting functional gastrointestinal disorder (FGID) with a favorable natural history. Worldwide, IC has a significant impact on many newborns and their families. Although not an indication of an illness, its symptoms are wide and generic and may indicate a potentially serious underlying issue in a tiny percentage of newborns who may require a medical evaluation. The pathogenesis appears to be multifactorial implying a complex relationship between the infant and the environment. One of the most studied theories attributes a key role to the gut microbiota in the pathogenesis of IC. A variety of approaches have been suggested for the clinical management of IC, and several randomized controlled trials have been reported in the literature. Probiotics can change the host's microbiota and positively impact health. They may be able to restore balance and create a better intestinal microbiota landscape since there is mounting evidence that the gut microbial environment of colicky newborns differs from that of healthy infants. In this review, we revise the most commonly studied probiotics and mixtures to treat and prevent IC and the most recent recommendations.


Subject(s)
Colic , Gastrointestinal Microbiome , Probiotics , Humans , Colic/therapy , Colic/prevention & control , Colic/microbiology , Probiotics/therapeutic use , Infant, Newborn , Infant
15.
Adv Exp Med Biol ; 1449: 1-28, 2024.
Article in English | MEDLINE | ID: mdl-39060728

ABSTRACT

Given that the host-microbe interaction is shaped by the immune system response, it is important to understand the key immune system-microbiota relationship during the period from conception to the first years of life. The present work summarizes the available evidence concerning human reproductive microbiota, and also, the microbial colonization during early life, focusing on the potential impact on infant development and health outcomes. Furthermore, we conclude that some dietary strategies including specific probiotics and other-biotics could become potentially valuable tools to modulate the maternal-neonatal microbiota during this early critical window of opportunity for targeted health outcomes throughout the entire lifespan.


Subject(s)
Microbiota , Probiotics , Humans , Infant , Infant, Newborn , Female , Microbiota/physiology , Microbiota/immunology , Pregnancy , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Child Development/physiology , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology
16.
Adv Exp Med Biol ; 1449: 29-41, 2024.
Article in English | MEDLINE | ID: mdl-39060729

ABSTRACT

Preterm infants differ significantly from their term infant counterparts regarding bacterial colonization patterns related to maternal microbiota diversity, mode of delivery, feeding type, antibiotic exposure, and the environmental influences related to prolonged hospitalization in the neonatal intensive care unit (NICU). Necrotizing enterocolitis (NEC), a multifactorial intestinal disorder characterized by ischemic bowel disease, disproportionately impacts preterm infants and has a high disease burden. Recent studies in the basic, translational, and clinical scientific literature have advanced knowledge into this complex disease process. Despite the explosion of research into NEC, however, there is a still a great deal unknown about this devastating illness. Additionally, the disease morbidity and mortality for NEC remain high despite advances in therapy options. This chapter reviews the current literature into the preterm infant microbiome, pathogenesis of NEC, potential targets for altering preterm microbiome, influence of microbiome on other organ systems, long-term implications of microbiome dysbiosis, and future directions of study.


Subject(s)
Dysbiosis , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Premature , Humans , Enterocolitis, Necrotizing/microbiology , Infant, Newborn , Dysbiosis/microbiology
17.
Adv Exp Med Biol ; 1449: 187-193, 2024.
Article in English | MEDLINE | ID: mdl-39060739

ABSTRACT

The chapter provides an overview of the current regulatory challenges surrounding the use of probiotics. It explores the global regulatory landscape, noting the need for uniform regulations across various regions. It emphasizes that these inconsistencies pose challenges for consumers, healthcare professionals, and industry stakeholders. Furthermore, the chapter highlights the ongoing efforts at the Codex Alimentarius to establish harmonized probiotic guidelines. The chapter also discusses labeling regulations, stressing the need for more accurate and comprehensive information on probiotic products to aid in evidence-based decision-making. Finally, it addresses safety concerns, particularly for vulnerable populations like children, and calls for a multi-layered approach to safety assessments. The chapter concludes by advocating harmonizing regulations and guidelines to facilitate probiotics' safer and more effective use.


Subject(s)
Probiotics , Probiotics/therapeutic use , Probiotics/adverse effects , Humans , Food Labeling/legislation & jurisprudence , Food Labeling/standards
18.
Adv Exp Med Biol ; 1449: 113-133, 2024.
Article in English | MEDLINE | ID: mdl-39060734

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major health problem worldwide, and the strongest determinant of liver disease in children. The possible influence of high-fat/low-fiber dietary patterns with microbiota (e.g., increased Firmicutes/Bacteroidetes ratio), and ultimately with MASLD occurrence and progression has been elucidated by several association studies. The possible mechanisms through which microbes exert their detrimental effects on MASLD include gut vascular barrier damage, a shift towards non-tolerogenic immunologic environment, and the detrimental metabolic changes, including a relative reduction of propionate and butyrate in favor of acetate, endogenous ethanol production, and impairment of the unconjugated bile acid-driven FXR-mediated gut-liver axis. The impact of nutritional and probiotic interventions in children with MASLD is described.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Probiotics , Synbiotics , Humans , Probiotics/therapeutic use , Probiotics/administration & dosage , Synbiotics/administration & dosage , Child , Fecal Microbiota Transplantation/methods , Fatty Liver/therapy , Fatty Liver/microbiology , Fatty Liver/pathology , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/metabolism
19.
Adv Exp Med Biol ; 1449: 143-156, 2024.
Article in English | MEDLINE | ID: mdl-39060736

ABSTRACT

Acute infectious diarrhea (AID) is one of the most common diseases in pediatric age with relevant burden both in high and in low-income countries. Thanks to their direct action on enterocyte functions and indirect actions on the mucosal and systemic immune system and on intestinal microbiome, probiotics are an ideal intervention to treat AID in childhood. However, their efficacy is strictly related to strains and indications, and practitioners should take this information into account in clinical practice. This chapter summarizes the main mechanisms of action of probiotics in AID, with a focus on proof of efficacy supporting their use in prevention and treatment of childhood AID. The use of selected strains in appropriate doses is strongly recommended by guidelines of AID, based on compelling proofs of efficacy and safety. At present, therapy with probiotics of AID is probably the strongest indication for probiotic use in medicine. Their role in prevention of AID is however questionable in healthy population, whereas it should be considered in at-risk population. Evidence for prevention of diarrhea in day-care centers and communities is lacking, but consistent evidence supports efficacy in prevention of hospital acquired diarrhea. Finally, this chapter presents novelties on this topic, in particular the role of rotavirus immunization on probiotics effectiveness and the effect of probiotics and postbiotics on Covid-associated diarrhea.Overall: AID is the most convincing area for probiotic use in children with gastrointestinal disorders, and effective strains should be used early on after onset of symptoms.


Subject(s)
COVID-19 , Diarrhea , Probiotics , Humans , Probiotics/therapeutic use , Diarrhea/microbiology , Diarrhea/therapy , Diarrhea/prevention & control , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19/therapy , Acute Disease , Gastrointestinal Microbiome , SARS-CoV-2/immunology , Child
20.
Adv Exp Med Biol ; 1449: 175-186, 2024.
Article in English | MEDLINE | ID: mdl-39060738

ABSTRACT

Clostridioides difficile is the most common causative agent of antibiotic-associated diarrhea. This spore forming, obligate anaerobic, gram-positive bacillus is becoming responsible for an increasing number of infections worldwide, both in community and in hospital settings, whose severity can vary widely from an asymptomatic infection to a lethal disease. While discontinuation of antimicrobial agents and antibiotic treatment of the infection remain the cornerstone of therapy, more recent fecal microbiota transplantation has also been valid as a therapy. The use of probiotics, especially Saccharomyces boulardii CNCM I-745 have become valid forms of prevention therapy. Although there are studies in adults with microbiota-targeted new generation therapies and Clostridium difficile vaccines, there are no data in the paediatric age group yet.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Fecal Microbiota Transplantation , Probiotics , Humans , Clostridium Infections/prevention & control , Clostridium Infections/microbiology , Clostridium Infections/therapy , Clostridioides difficile/pathogenicity , Clostridioides difficile/physiology , Probiotics/therapeutic use , Anti-Bacterial Agents/therapeutic use , Gastrointestinal Microbiome , Diarrhea/prevention & control , Diarrhea/microbiology , Diarrhea/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...