Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
Gut Microbes ; 14(1): 2018899, 2022.
Article in English | MEDLINE | ID: covidwho-2323446

ABSTRACT

Intestinal bacteria may influence lung homeostasis via the gut-lung axis. We conducted a single-center, quadruple-blinded, randomized trial in adult symptomatic Coronavirus Disease 2019 (Covid19) outpatients. Subjects were allocated 1:1 to probiotic formula (strains Lactiplantibacillus plantarum KABP022, KABP023, and KAPB033, plus strain Pediococcus acidilactici KABP021, totaling 2 × 109 colony-forming units (CFU)) or placebo, for 30 days. Co-primary endpoints included: i) proportion of patients in complete symptomatic and viral remission; ii) proportion progressing to moderate or severe disease with hospitalization, or death; and iii) days on Intensive Care Unit (ICU). Three hundred subjects were randomized (median age 37.0 years [range 18 to 60], 161 [53.7%] women, 126 [42.0%] having known metabolic risk factors), and 293 completed the study (97.7%). Complete remission was achieved by 78 of 147 (53.1%) in probiotic group compared to 41 of 146 (28.1%) in placebo (RR: 1.89 [95 CI 1.40-2.55]; P < .001), significant after multiplicity correction. No hospitalizations or deaths occurred during the study, precluding the assessment of remaining co-primary outcomes. Probiotic supplementation was well-tolerated and reduced nasopharyngeal viral load, lung infiltrates and duration of both digestive and non-digestive symptoms, compared to placebo. No significant compositional changes were detected in fecal microbiota between probiotic and placebo, but probiotic supplementation significantly increased specific IgM and IgG against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) compared to placebo. It is thus hypothesized this probiotic primarily acts by interacting with the host's immune system rather than changing colonic microbiota composition. Future studies should replicate these findings and elucidate its mechanism of action (Registration: NCT04517422).Abbreviations: AE: Adverse Event; BMI: Body Mass Index; CONSORT: CONsolidated Standards of Reporting Trials; CFU: Colony-Forming Units; eDRF: Electronic Daily Report Form; GLA: Gut-Lung Axis; GSRS: Gastrointestinal Symptoms Rating Scale; hsCRP: High-sensitivity C-Reactive Protein; HR: Hazard Ratio; ICU: Intensive Care Unit; OR: Odds Ratio; PCoA: Principal Coordinate Analysis; RR: Relative Risk; RT-qPCR: Real-Time Quantitative Polymerase Chain Reaction; SARS-CoV2: Severe acute respiratory syndrome coronavirus 2; SpO2: Peripheral Oxygen Saturation; WHO: World Health Organization.


Subject(s)
COVID-19/therapy , Probiotics/pharmacology , SARS-CoV-2 , Adult , COVID-19/immunology , COVID-19/virology , Female , Gastrointestinal Microbiome , Humans , Male , Middle Aged , Placebos
2.
Nutrients ; 15(2)2023 Jan 05.
Article in English | MEDLINE | ID: covidwho-2318718

ABSTRACT

INTRODUCTION: Prebiotics, probiotics and synbiotics are known to have major beneficial effects on human health due to their ability to modify the composition and the function of the gut mucosa, the gut microbiota and the immune system. These components largely function in a healthy population throughout different periods of life to confer homeostasis. Indeed, they can modulate the composition of the gut microbiota by increasing bacteria strands that are beneficial for health, such as Firmicute and Bifidobacteria, and decreasing harmful bacteria, such as Enteroccocus. Their immunomodulation properties have been extensively studied in different innate cells (dendritic cells, macrophages, monocytes) and adaptive cells (Th, Treg, B cells). They can confer a protolerogenic environment but also modulate pro-inflammatory responses. Due to all these beneficial effects, these compounds have been investigated to prevent or to treat different diseases, such as cancer, diabetes, allergies, autoimmune diseases, etc. Regarding the literature, the effects of these components on dendritic cells, monocytes and T cells have been studied and presented in a number of reviews, but their impact on B-cell response has been less widely discussed. CONCLUSIONS: For the first time, we propose here a review of the literature on the immunomodulation of B-lymphocytes response by prebiotics, probiotics and synbiotics, both in healthy conditions and in pathologies. DISCUSSION: Promising studies have been performed in animal models, highlighting the potential of prebiotics, probiotics and synbiotics intake to treat or to prevent diseases associated with B-cell immunomodulation, but this needs to be validated in humans with a full characterization of B-cell subsets and not only the humoral response.


Subject(s)
Probiotics , Synbiotics , Animals , Humans , Prebiotics , Probiotics/pharmacology , Immunomodulation , B-Lymphocytes , Macrophages
3.
Trials ; 23(1): 603, 2022 Jul 27.
Article in English | MEDLINE | ID: covidwho-2317113

ABSTRACT

BACKGROUND: Some probiotics appear to improve athletic performance, endurance, and recovery after intense exercise. Other formulations may provide performance-related benefits via immune and gastrointestinal functions in athletic individuals. However, few formulations have been studied for both types of effects among non-elite athletes. The primary objective of this study is to assess the ergogenic effects of a probiotic on high-intensity endurance running performance in non-elite runners. Secondary objectives include assessment of perceived exertion, blood chemistry, immune and stress biomarkers, cold and flu symptoms, and gastrointestinal health after the probiotic intervention. METHODS: This 9-week randomized, placebo-controlled, double-blind, parallel trial will assess the ergogenic effects of a probiotic (5 billion colony-forming units/day, for 6 weeks) in healthy, non-elite runners (N=32; 18-45 years). Participants will be monitored via daily and weekly questionnaires during the 2-week pre-baseline, 6-week intervention, and 1-week washout. Questionnaires will inquire about activity, muscle soreness, gastrointestinal symptoms, cold and flu symptoms, stool form and frequency, and adverse events. During the pre-baseline visit, maximal oxygen uptake (V̇O2 max) is assessed to set appropriate individualized workload settings for the treadmill time-to-exhaustion endurance tests. These time-to-exhaustion endurance running tests will be completed at an intensity of 85% VO2max at baseline and final visits. During these tests, self-perceived exercise effort will be rated via the Borg Rating of Perceived Exertion scale and finger sticks assessing capillary blood glucose and lactate concentrations will be collected every 3 min. Additional questionnaires will assess diet and motivation to exercise. Body composition will be assessed using air displacement plethysmography at the baseline and final visits. Hypotheses will be tested using two-sided tests, and a linear model and with a type I error rate of α=0.05. Primary and secondary outcomes will be tested by comparing results between the intervention groups, adjusting for baseline values. DISCUSSION: These results will build evidence documenting the role of probiotics on running endurance performance and physiological responses to exercise in non-elite athletes. Understanding the potential mechanisms of probiotic effects and how they mitigate the intestinal or immune discomforts caused by running could provide additional strategy means to help runners improve their performance. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov NCT04588142 . Posted on October 19, 2020. PROTOCOL VERSION: July 2, 2021, version 1.2.


Subject(s)
Performance-Enhancing Substances , Probiotics , Athletes , Double-Blind Method , Exercise/physiology , Humans , Performance-Enhancing Substances/pharmacology , Physical Endurance/physiology , Probiotics/adverse effects , Randomized Controlled Trials as Topic
4.
Molecules ; 28(7)2023 Apr 04.
Article in English | MEDLINE | ID: covidwho-2300303

ABSTRACT

Depression is a common and complex mental and emotional disorder that causes disability, morbidity, and quite often mortality around the world. Depression is closely related to several physical and metabolic conditions causing metabolic depression. Studies have indicated that there is a relationship between the intestinal microbiota and the brain, known as the gut-brain axis. While this microbiota-gut-brain connection is disturbed, dysfunctions of the brain, immune system, endocrine system, and gastrointestinal tract occur. Numerous studies show that intestinal dysbiosis characterized by abnormal microbiota and dysfunction of the microbiota-gut-brain axis could be a direct cause of mental and emotional disorders. Traditional treatment of depression includes psychotherapy and pharmacotherapy, and it mainly targets the brain. However, restoration of the intestinal microbiota and functions of the gut-brain axis via using probiotics, their metabolites, prebiotics, and healthy diet may alleviate depressive symptoms. Administration of probiotics labeled as psychobiotics and their metabolites as metabiotics, especially as an adjuvant to antidepressants, improves mental disorders. It is a new approach to the prevention, management, and treatment of mental and emotional illnesses, particularly major depressive disorder and metabolic depression. For the effectiveness of antidepressant therapy, psychobiotics should be administered at a dose higher than 1 billion CFU/day for at least 8 weeks.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Probiotics , Humans , Depression/drug therapy , Probiotics/therapeutic use , Prebiotics , Brain
5.
Int J Mol Sci ; 24(7)2023 Apr 01.
Article in English | MEDLINE | ID: covidwho-2298486

ABSTRACT

Gut microbiota (GM) modulation can be investigated as possible solution to enhance recovery after COVID-19. An open-label, single-center, single-arm, pilot, interventional study was performed by enrolling twenty patients recently recovered from COVID-19 to investigate the role of a mixed probiotic, containing Lactobacilli, Bifidobacteria and Streptococcus thermophilus, on gastrointestinal symptoms, local and systemic inflammation, intestinal barrier integrity and GM profile. Gastrointestinal Symptom Rating Scale, cytokines, inflammatory, gut permeability, and integrity markers were evaluated before (T0) and after 8 weeks (T1) of probiotic supplementation. GM profiling was based on 16S-rRNA targeted-metagenomics and QIIME 2.0, LEfSe and PICRUSt computational algorithms. Multiple machine learning (ML) models were trained to classify GM at T0 and T1. A statistically significant reduction of IL-6 (p < 0.001), TNF-α (p < 0.001) and IL-12RA (p < 0.02), citrulline (p value < 0.001) was reported at T1. GM global distribution and microbial biomarkers strictly reflected probiotic composition, with a general increase in Bifidobacteria at T1. Twelve unique KEGG orthologs were associated only to T0, including tetracycline resistance cassettes. ML classified the GM at T1 with 100% score at phylum level. Bifidobacteriaceae and Bifidobacterium spp. inversely correlated to reduction of citrulline and inflammatory cytokines. Probiotic supplementation during post-COVID-19 may trigger anti-inflammatory effects though Bifidobacteria and related-metabolism enhancement.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Probiotics , Humans , Gastrointestinal Microbiome/genetics , Citrulline , Probiotics/therapeutic use , Probiotics/pharmacology , Cytokines , Bifidobacterium , Machine Learning
6.
Arch Microbiol ; 205(5): 182, 2023 Apr 09.
Article in English | MEDLINE | ID: covidwho-2302375

ABSTRACT

COVID-19, an acute respiratory viral infection conveyed by pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected millions of individuals globally, and is a public health emergency of international concern. Till now, there are no highly effective therapies for this infection without vaccination. As they can evolve quickly and cross the strain level easily, these viruses are causing epidemics or pandemics that are allied with more severe clinical diseases. A new approach is needed to improve immunity to confirm the protection against emerging viral infections. Probiotics can modify gut microbial dysbiosis, improve the host immune system, and stimulate immune signaling, increasing systemic immunity. Several probiotic bacterial therapies have been proven to decrease the period of bacterial or viral infections. Superinduction of inflammation, termed cytokine storm, has been directly linked with pneumonia and severe complications of viral respiratory infections. In this case, probiotics as potential immunomodulatory agents can be an appropriate candidate to improve the host's response to respiratory viral infections. During this COVID-19 pandemic, any approach that can induce mucosal and systemic immunity could be helpful. Here, we summarize contexts regarding the effectiveness of various probiotics for preventing virus-induced respiratory infectious diseases, especially those that could be employed for COVID-19 patients. In addition, the effects of probiotics, their mechanisms on different aspects of immune responses against respiratory viral infection, and their antiviral properties in clinical findings have been described in detail.


Subject(s)
COVID-19 , Probiotics , Respiratory Tract Infections , Virus Diseases , Humans , COVID-19/therapy , SARS-CoV-2 , Pandemics/prevention & control , Probiotics/therapeutic use , Respiratory Tract Infections/microbiology
7.
World J Gastroenterol ; 29(11): 1708-1720, 2023 Mar 21.
Article in English | MEDLINE | ID: covidwho-2290749

ABSTRACT

Coronavirus disease 2019 (COVID-19) infection caused by the severe acute respiratory syndrome coronavirus 2 virus, its symptoms, treatment, and post-COVID-19 effects have been a major focus of research since 2020. In addition to respiratory symptoms, different clinical variants of the virus have been associated with dynamic symptoms and multiorgan diseases, including liver abnormalities. The release of cytokines by the activation of innate immune cells during viral infection and the high doses of drugs used for COVID-19 treatment are considered major drivers of liver injury in COVID-19 patients. The degree of hepatic inflammation in patients suffering from chronic liver disease and having COVID-19 could be severe and can be estimated through different liver chemistry abnormality markers. Gut microbiota influences liver chemistry through its metabolites. Gut dysbiosis during COVID-19 treatment can promote liver inflammation. Here, we highlighted the bidirectional association of liver physiology and gut microbiota (gut-liver axis) and its potential to manipulate drug-induced chemical abnormalities in the livers of COVID-19 patients.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Liver Diseases , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , COVID-19 Drug Treatment , Liver Diseases/metabolism , Inflammation , Dysbiosis/therapy
8.
Int Arch Allergy Immunol ; 184(3): 291-301, 2023.
Article in English | MEDLINE | ID: covidwho-2290461

ABSTRACT

INTRODUCTION: The favorable effects of probiotics have been demonstrated in allergic disorders. However, the underlying immunological mechanisms are poorly understood. In the present study, we investigated the improvement of clinical symptoms and immunological balance after receiving probiotics in patients with asthma. METHODS: The present study was a randomized, double-blind, placebo-controlled trial in which 40 patients with asthma were enrolled. They were treated with probiotics or placebo: 1 capsule/day for 8 weeks. Pulmonary function test, percentage of CD4+ CD25+ FoxP3+ Tregs, and gene expression of T-bet, GATA-3, RORγt, and Foxp3 in PBMCs were assessed at baseline and after treatment. RESULTS: Our results showed a significant increase in the expression of FoxP3 and CD4+ CD25+ FoxP3+ Tregs population, while RORγt and GATA3 expression were reduced. In addition, pulmonary function tests showed a significant improvement in forced expiratory volume and forced vital capacity after receiving probiotics. DISCUSSION/CONCLUSION: Our findings demonstrate that 8-week treatment with probiotic supplementation can control T-helper 2-predominant and Th17 pro-inflammatory responses and improve forced vital and forced expiratory volume in asthmatic patients. It seems probiotics can be used besides common treatments for patients with asthma.


Subject(s)
Asthma , Probiotics , Humans , T-Lymphocytes, Regulatory , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Dietary Supplements , Probiotics/therapeutic use , Forkhead Transcription Factors/genetics
9.
Int J Mol Sci ; 24(7)2023 Mar 31.
Article in English | MEDLINE | ID: covidwho-2293786

ABSTRACT

Microbial contamination in the hospital environment is a major concern for public health, since it significantly contributes to the onset of healthcare-associated infections (HAIs), which are further complicated by the alarming level of antimicrobial resistance (AMR) of HAI-associated pathogens. Chemical disinfection to control bioburden has a temporary effect and can favor the selection of resistant pathogens, as observed during the COVID-19 pandemic. Instead, probiotic-based sanitation (probiotic cleaning hygiene system, PCHS) was reported to stably abate pathogens, AMR, and HAIs. PCHS action is not rapid nor specific, being based on competitive exclusion, but the addition of lytic bacteriophages that quickly and specifically kill selected bacteria was shown to improve PCHS effectiveness. This study aimed to investigate the effect of such combined probiotic-phage sanitation (PCHSφ) in two Italian hospitals, targeting staphylococcal contamination. The results showed that PCHSφ could provide a significantly higher removal of staphylococci, including resistant strains, compared with disinfectants (-76%, p < 0.05) and PCHS alone (-50%, p < 0.05). Extraordinary sporadic chlorine disinfection appeared compatible with PCHSφ, while frequent routine chlorine usage inactivated the probiotic/phage components, preventing PCHSφ action. The collected data highlight the potential of a biological sanitation for better control of the infectious risk in healthcare facilities, without worsening pollution and AMR concerns.


Subject(s)
Bacteriophages , COVID-19 , Cross Infection , Probiotics , Humans , Sanitation/methods , Chlorine , Pandemics , Cross Infection/prevention & control , Cross Infection/microbiology , Staphylococcus , Delivery of Health Care , Probiotics/therapeutic use
10.
Gut Microbes ; 15(1): 2201157, 2023.
Article in English | MEDLINE | ID: covidwho-2306573

ABSTRACT

The epidemic of coronavirus disease-19 (COVID-19) has grown to be a global health threat. Gastrointestinal symptoms are thought to be common clinical manifestations apart from a series of originally found respiratory symptoms. The human gut harbors trillions of microorganisms that are indispensable for complex physiological processes and homeostasis. Growing evidence demonstrate that gut microbiota alteration is associated with COVID-19 progress and severity, and post-COVID-19 syndrome, characterized by decrease of anti-inflammatory bacteria like Bifidobacterium and Faecalibacterium and enrichment of inflammation-associated microbiota including Streptococcus and Actinomyces. Therapeutic strategies such as diet, probiotics/prebiotics, herb, and fecal microbiota transplantation have shown positive effects on relieving clinical symptoms. In this article, we provide and summarize the recent evidence about the gut microbiota and their metabolites alterations during and after COVID-19 infection and focus on potential therapeutic strategies targeting gut microbiota. Understanding the connections between intestinal microbiota and COVID-19 would provide new insights into COVID-19 management in the future.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Probiotics , Humans , Post-Acute COVID-19 Syndrome , Prebiotics , Probiotics/therapeutic use
11.
Lett Appl Microbiol ; 71(3): 229-234, 2020 Sep.
Article in English | MEDLINE | ID: covidwho-2248530

ABSTRACT

Understanding mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for prevention and/or treatment. This could be achieved via the inhibition of viral entry and/or replication, or by suppression of the immunologic response that is provoked by the infection (known as the cytokine storm). Probiotics are defined as 'live microorganisms that, when administered in adequate amounts, confer a health benefit on the host'. There is scarcity of evidence about the relationship between COVID-19 and gut microbiota. So, whether or not these supplements can prevent or ameliorate COVID-19-associated symptoms is not fully understood. The aim of this study is to provide an indirect evidence about the utility of probiotics in combating COVID-19 or its associated symptoms, through the review of its antiviral and anti-inflammatory properties in vitro, animal models and human trials. SIGNIFICANCE AND IMPACT OF THE STUDY: The role of probiotics in alleviation of the novel COVID-19 has not been established. This review provides an insight about the anti-inflammatory, antiviral effects of probiotics in vitro, animal models and human. The latter can provide an indirect evidence and/or hypothesis-driven approach to investigate the use of probiotics as adjunctive therapy in the prophylaxis and/or alleviation of COVID-19 symptoms.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19/diet therapy , Gastrointestinal Microbiome/drug effects , Probiotics/therapeutic use , SARS-CoV-2/drug effects , Animals , Cytokines/blood , Dietary Supplements , Humans , Rats
12.
PLoS One ; 18(3): e0278356, 2023.
Article in English | MEDLINE | ID: covidwho-2289020

ABSTRACT

BACKGROUND AND AIMS: Oral probiotic supplementation may be a beneficial adjunctive therapy for patients with symptomatic COVID-19. However, its safety and efficacy are unclear. We aimed to investigate how probiotic supplementation impacts COVID-19 symptom trajectory and patient outcomes by conducting a systematic review and meta-analysis of randomized controlled trials (RCTs). METHODS: RCTs randomizing patients with COVID-19 to probiotics were searched in PubMed Central, Embase, CINAHL, and Cochrane Library from inception to July 31, 2022. We performed a random-effects pairwise meta-analysis for all outcomes using the restricted maximum likelihood (REML) estimator. We used the GRADE approach to assess the certainty of the evidence. RESULTS: A total of 1027 participants from eight RCT studies were included in the meta-analysis. Probiotic supplements probably reduce the incidence of diarrhea (RR 0.61 [0.43 to 0.87]; moderate certainty) and probably reduce cough or dyspnea compared to placebo/standard care (RR 0.37 [0.19 to 0.73]; moderate certainty). Probiotic supplements may improve composite endpoint measured by clinical escalation or mortality compared to placebo (RR 0.41 [0.18 to 0.93]; low certainty evidence); however, they may not significantly reduce the need for clinical escalation (RR 0.57 [0.31 to 1.07]; low certainty evidence) or mortality (RR 0.50 [0.20 to 1.29]; low certainty evidence). In addition, the probiotic supplement is associated with reduced adverse events (RR 0.62 [0.46 to 0.83]; moderate certainty). CONCLUSION: Early probiotic supplement is a safe and effective adjunctive therapy that reduces the risk of symptoms and health care burden related to COVID-19 across all severity types.


Subject(s)
COVID-19 , Probiotics , Humans , COVID-19/complications , Probiotics/adverse effects , Diarrhea/etiology , Dental Care
13.
Expert Rev Anti Infect Ther ; 21(6): 667-674, 2023 06.
Article in English | MEDLINE | ID: covidwho-2287033

ABSTRACT

OBJECTIVES: This study was conducted to assess the clinical efficacy of probiotics in the treatment of patients with COVID19. METHODS: PubMed, Embase, Cochrane Library, and ClinicalTrials.gov were searched for studies from their inception to 8 February 2022. Randomized controlled trials (RCTs) that compared the clinical efficacy of probiotics with usual care or standard care for patients with COVID19 were included. The primary outcome was all-cause mortality. Random-effects model using MantelHaenszel and inverse variance methods were performed to analyze the data. RESULTS: Eight RCTs with 900 patients were included. The study group receiving probiotics had a non-significantly lower rate of mortality than the control group had, but this difference was not significant (risk ratio [RR], 0.51; 95% CI, 0.22 to 1.16). However, the study group had significantly lower rates of dyspnea (RR, 0.11; 95% CI, 0.02 to 0.60), fever (RR, 0.37; 95% CI, 0.16 to 0.85) and headache (RR, 0.19; 95% CI, 0.05 to 0.65). Higher complete remission of COVID-19-associated symptoms was observed in the study group than the control group (RR, 1.89; 95% CI, 1.40-2.55). CONCLUSIONS: Although probiotics use did not improve clinical outcomes or reduce inflammatory markers, it may relieve COVID-19-associated symptoms.


Subject(s)
COVID-19 , Probiotics , Humans , Randomized Controlled Trials as Topic , Treatment Outcome , Probiotics/adverse effects
14.
Nutrients ; 15(5)2023 Feb 22.
Article in English | MEDLINE | ID: covidwho-2268342

ABSTRACT

I read with interest the paper by Sung et al. entitled "Body Fat Reduction Effect of Bifidobacterium breve B-3: A Randomized, Double-Blind, Placebo Comparative Clinical Trial" where a reduction in body fat mass after Bifidobacterium breve B-3 (BB-3) ingestion for 12 weeks was reported [...].


Subject(s)
Bifidobacterium breve , Probiotics , Double-Blind Method , Adipose Tissue
15.
Microbiome ; 11(1): 64, 2023 03 30.
Article in English | MEDLINE | ID: covidwho-2255969

ABSTRACT

BACKGROUND: The COVID-19 pandemic has highlighted the extent to which the public transportation environment, such as in subways, may be important for the transmission of potential pathogenic microbes among humans, with the possibility of rapidly impacting large numbers of people. For these reasons, sanitation procedures, including massive use of chemical disinfection, were mandatorily introduced during the emergency and remain in place. However, most chemical disinfectants have temporary action and a high environmental impact, potentially enhancing antimicrobial resistance (AMR) of the treated microbes. By contrast, a biological and eco-sustainable probiotic-based sanitation (PBS) procedure was recently shown to stably shape the microbiome of treated environments, providing effective and long-term control of pathogens and AMR spread in addition to activity against SARS-CoV-2, the causative agent of COVID-19. Our study aims to assess the applicability and impact of PBS compared with chemical disinfectants based on their effects on the surface microbiome of a subway environment. RESULTS: The train microbiome was characterized by both culture-based and culture-independent molecular methods, including 16S rRNA NGS and real-time qPCR microarray, for profiling the train bacteriome and its resistome and to identify and quantify specific human pathogens. SARS-CoV-2 presence was also assessed in parallel using digital droplet PCR. The results showed a clear and significant decrease in bacterial and fungal pathogens (p < 0.001) as well as of SARS-CoV-2 presence (p < 0.01), in the PBS-treated train compared with the chemically disinfected control train. In addition, NGS profiling evidenced diverse clusters in the population of air vs. surface while demonstrating the specific action of PBS against pathogens rather than the entire train bacteriome. CONCLUSIONS: The data presented here provide the first direct assessment of the impact of different sanitation procedures on the subway microbiome, allowing a better understanding of its composition and dynamics and showing that a biological sanitation approach may be highly effective in counteracting pathogens and AMR spread in our increasingly urbanized and interconnected environment. Video Abstract.


Subject(s)
COVID-19 , Disinfectants , Microbiota , Probiotics , Railroads , Humans , SARS-CoV-2/genetics , Sanitation/methods , RNA, Ribosomal, 16S/genetics , Pandemics/prevention & control , Case-Control Studies , Disinfectants/pharmacology
16.
PLoS One ; 18(3): e0279118, 2023.
Article in English | MEDLINE | ID: covidwho-2269932

ABSTRACT

The Covid-19 associated mucormycosis (CAM) is an emerging disease affecting immunocompromised patients. Prevention of such infections using probiotics and their metabolites persist as effective therapeutic agents. Therefore, the present study emphasizes on assessment of their efficacy and safety. Samples from different sources like human milk, honey bee intestine, toddy, and dairy milk were collected, screened and characterized for potential probiotic lactic acid bacteria (LAB) and their metabolites to be used as effective antimicrobial agents to curtail CAM. Three isolates were selected based on probiotic properties and characterized as Lactobacillus pentosus BMOBR013, Lactobacillus pentosus BMOBR061 and Pediococcus acidilactici BMOBR041 by 16S rRNA sequencing and MALDI TOF-MS. The antimicrobial activity against standard bacterial pathogens showed ˃9 mm zone of inhibition. Furthermore, the antifungal activity of three isolates was tested against Aspergillus flavus MTCC 2788, Fusarium oxysporum, Candida albicans and Candida tropicalis where the results showed significant inhibition of each fungal strain. Further studies were carried out on lethal fungal pathogens like Rhizopus sp. and two Mucor sp. which are associated with post Covid-19 infection in immunosuppressed diabetic patients. Our studies on CAM inhibitory effect of LAB revealed the efficient inhibition against Rhizopus sp. and two Mucor sp. The cell free supernatants of three LAB showed varied inhibitory activity against these fungi. Following the antimicrobial activity, the antagonistic metabolite 3-Phenyllactic acid (PLA) in culture supernatant was quantified and characterized by HPLC and LC-MS using standard PLA (Sigma Aldrich). The isolate L. pentosus BMOBR013 produced highest PLA (0.441 g/L), followed by P. acidilactici BMOBR041 (0.294 g/L) and L. pentosus BMOBR061 (0.165 g/L). The minimum inhibitory concentration of HPLC eluted PLA on the Rhizopus sp. and two Mucor sp. was found to be 180 mg/ml which was further confirmed by inhibition of total mycelia under live cell imaging microscope.


Subject(s)
Anti-Infective Agents , COVID-19 , Lactobacillales , Mucormycosis , Probiotics , Humans , Animals , Bees/genetics , Mucormycosis/drug therapy , RNA, Ribosomal, 16S/genetics , Lactobacillales/genetics , Fungi/genetics , Probiotics/pharmacology , Polyesters
17.
Curr Nutr Rep ; 12(2): 263-269, 2023 06.
Article in English | MEDLINE | ID: covidwho-2251720

ABSTRACT

PURPOSE OF REVIEW: Our goal is to provide the most recent and accurate scientific evidence available regarding COVID-19's interaction with the human gut and the role of nutrition/nutritional supplementation in the prevention and treatment of the disease. RECENT FINDINGS: Gastrointestinal symptoms of COVID-19 are common and often persist even after classically defined illness resolution. Nutritional status and content have been shown to impact infection risk and severity. Well-balanced diets are associated with decreased infection risk/severity, and early nutrition is associated with better outcomes in the critically ill. No specific vitamin supplementation regimen has shown consistent benefit for infection treatment or prevention. The impact of COVID-19 extends far past the pulmonary system, and its impact on the gut should not be ignored. For those interested in adopting lifestyle modifications to prevent severe COVID-19 infection/side effects, consideration should be made for adoption of a well-balanced diet (e.g., Mediterranean style), utilization of probiotics, and addressing nutritional/vitamin deficiencies. Future, high-quality research is needed in this arena.


Subject(s)
COVID-19 , Malnutrition , Probiotics , Humans , SARS-CoV-2 , Dietary Supplements , Nutritional Status , Probiotics/therapeutic use
18.
Ter Arkh ; 94(10): 1163-1170, 2022 Nov 22.
Article in Russian | MEDLINE | ID: covidwho-2270273

ABSTRACT

AIM: To evaluate the efficacy of Saccharomyces boulardii (S. boulardii) CNCM I-745 probiotic drug in preventing and treating diarrhea in hospitalized patients with COVID-19. MATERIALS AND METHODS: A prospective comparative study was conducted in two parallel groups. The study included males and females aged 18 to 60 with the following diagnosis confirmed by polymerase chain reaction: U07.2 Coronavirus infection COVID-19, caused by SARS-CoV-2 virus (grade 1-3 pneumonia according to CT scan). All patients received antibiotic therapy. The patients were subdivided into two equal groups (n=60) depending on the administration of S. boulardii CNCM I-745 probiotic drug in addition to standard treatment. The probiotic was prescribed by the attending physician; the dose was 2 capsules per day (500 mg/day) 30 min before the meal for 10 days. All patients were monitored for main clinical, laboratory, and instrumental parameters during the study. In addition, the symptom of diarrhea (stool with a frequency of more than 3 times a day of type 6 and 7 according to the Bristol stool scale), including its frequency, duration, and the number of bowel movements of loose stool per day were precisely evaluated in both groups. RESULTS: In the overall patient pool, diarrhea was reported in 21.7% of in-patients during the observation period (95% confidence interval [CI] 14.2-29.1) with a mean duration of 4.6154 days (95% CI 3.7910-5.4398). The incidence of diarrhea in group 1 was 13.3% (95% CI 4.5-22.2), and in group 2, it was 30.0% (95% CI 18.1-41.9). Relative risk showed that the use of the S. boulardii CNCM I-745 probiotic drug leads to a significant reduction in the risk of diarrhea in hospitalized patients with COVID-19 infection receiving antibiotic therapy (odds ratio [OR] 0.3590, 95% CI 0.1421-0.9069; p=0.0303). In group 1, the duration of diarrhea was 3.1250 days (95% CI 2.5892-3.6608) versus 5.2778 days (95% CI 4.2290-6.3265) in group 2, p=0.0112. The mean daily frequency of loose stools in patients with diarrhea in group 1 was 3.2500 (95% CI 2.6588-3.8412) versus 4.3889 (95% CI 3.7252-5.0525) in group 2, p=0.0272. The secondary endpoint, duration of hospital stay, was also significantly shorter in group 1 patients - 11.6833 days (95% CI 11.2042-12.1625) versus 12.7333 days (95% CI 12.1357-13.3309) in group 2, p=0.0120. CONCLUSION: The present prospective comparative study demonstrated that adding S. boulardii CNCM I-745 probiotic drug into the standard treatment regimen of patients with new coronavirus infection COVID-19 receiving antibiotic therapy helps reduce the incidence of diarrhea and its severity during hospitalization, as well as the duration of hospital stay.


Subject(s)
COVID-19 , Probiotics , Saccharomyces boulardii , Male , Female , Humans , Pharmaceutical Preparations , Prospective Studies , SARS-CoV-2 , Diarrhea/epidemiology , Diarrhea/etiology , Diarrhea/prevention & control , Probiotics/therapeutic use , Anti-Bacterial Agents/therapeutic use
20.
Clin Nutr ; 42(2): 216-226, 2023 02.
Article in English | MEDLINE | ID: covidwho-2245406

ABSTRACT

Vaccination is the most effective way to confer potent and long-term protection from infectious diseases. However, poorer responses to immunization are common in young adults with sub-optimal immune health and the elderly because of immunosenescence and increased comorbidities. Recent mechanistic studies have highlighted that the microbiota and its compounds modulate many molecular pathways that can influence the host immune system. Consequently, altering the microbiota composition or activity with immunonutrition, specifically with biotic interventions (probiotics, prebiotics, synbiotics, or postbiotics), may enhance the immune response and vaccine efficacy. This review aims to examine the available data for these biotic strategies to provide clinicians, researchers, and vaccine developers with a mechanistically driven synthesis of how biotic interventions could modulate the immune responses to vaccination. The article describes some postulated mechanistic pathways involved in immunological responses to vaccines and immunomodulation with biotic interventions. Randomized clinical trials were also reviewed to evaluate the impact of specific biotic interventions on vaccination outcomes in different age groups. Few strains and formulations significantly increased antigen-specific antibody titers in individual of all ages. However, studies have also pointed to a substantial heterogeneity that can be attributed to the difference in biotic intervention, strain, dose, viability, type of vaccine antigen, study location, as well as duration, and timing of administration. Future investigations should focus on establishing optimal strains, doses, and timing of administration with respect to vaccination, especially in the elderly and children, where vaccine effectiveness and duration of immunization matter.


Subject(s)
Probiotics , Synbiotics , Vaccines , Child , Young Adult , Humans , Aged , Prebiotics , Immunity
SELECTION OF CITATIONS
SEARCH DETAIL