Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
N Engl J Med ; 386(3): 220-229, 2022 01 20.
Article in English | MEDLINE | ID: covidwho-1632249

ABSTRACT

BACKGROUND: Current therapies for recurrent Clostridioides difficile infection do not address the disrupted microbiome, which supports C. difficile spore germination into toxin-producing bacteria. SER-109 is an investigational microbiome therapeutic composed of purified Firmicutes spores for the treatment of recurrent C. difficile infection. METHODS: We conducted a phase 3, double-blind, randomized, placebo-controlled trial in which patients who had had three or more episodes of C. difficile infection (inclusive of the qualifying acute episode) received SER-109 or placebo (four capsules daily for 3 days) after standard-of-care antibiotic treatment. The primary efficacy objective was to show superiority of SER-109 as compared with placebo in reducing the risk of C. difficile infection recurrence up to 8 weeks after treatment. Diagnosis by toxin testing was performed at trial entry, and randomization was stratified according to age and antibiotic agent received. Analyses of safety, microbiome engraftment, and metabolites were also performed. RESULTS: Among the 281 patients screened, 182 were enrolled. The percentage of patients with recurrence of C. difficile infection was 12% in the SER-109 group and 40% in the placebo group (relative risk, 0.32; 95% confidence interval [CI], 0.18 to 0.58; P<0.001 for a relative risk of <1.0; P<0.001 for a relative risk of <0.833). SER-109 led to less frequent recurrence than placebo in analyses stratified according to age stratum (relative risk, 0.24 [95% CI, 0.07 to 0.78] for patients <65 years of age and 0.36 [95% CI, 0.18 to 0.72] for those ≥65 years) and antibiotic received (relative risk, 0.41 [95% CI, 0.22 to 0.79] with vancomycin and 0.09 [95% CI, 0.01 to 0.63] with fidaxomicin). Most adverse events were mild to moderate and were gastrointestinal in nature, with similar numbers in the two groups. SER-109 dose species were detected as early as week 1 and were associated with bile-acid profiles that are known to inhibit C. difficile spore germination. CONCLUSIONS: In patients with symptom resolution of C. difficile infection after treatment with standard-of-care antibiotics, oral administration of SER-109 was superior to placebo in reducing the risk of recurrent infection. The observed safety profile of SER-109 was similar to that of placebo. (Funded by Seres Therapeutics; ECOSPOR III ClinicalTrials.gov number, NCT03183128.).


Subject(s)
Clostridioides difficile , Clostridium Infections/therapy , Firmicutes , Aged , Anti-Bacterial Agents/adverse effects , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Humans , Intention to Treat Analysis , Male , Microbiota/drug effects , Middle Aged , Recurrence , Secondary Prevention , Spores, Bacterial
3.
PLoS One ; 17(1): e0262057, 2022.
Article in English | MEDLINE | ID: covidwho-1622351

ABSTRACT

Respiratory tract infections (RTIs) are extremely common and can cause gastrointestinal tract symptoms and changes to the gut microbiota, yet these effects are poorly understood. We conducted a systematic review to evaluate the reported evidence of gut microbiome alterations in patients with a RTI compared to healthy controls (PROSPERO: CRD42019138853). We systematically searched Medline, Embase, Web of Science, Cochrane and the Clinical Trial Database for studies published between January 2015 and June 2021. Studies were eligible for inclusion if they were human cohorts describing the gut microbiome in patients with an RTI compared to healthy controls and the infection was caused by a viral or bacterial pathogen. Dual data screening and extraction with narrative synthesis was performed. We identified 1,593 articles and assessed 11 full texts for inclusion. Included studies (some nested) reported gut microbiome changes in the context of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) (n = 5), influenza (H1N1 and H7N9) (n = 2), Tuberculosis (TB) (n = 4), Community-Acquired Pneumonia CAP (n = 2) and recurrent RTIs (rRTI) (n = 1) infections. We found studies of patients with an RTI compared to controls reported a decrease in gut microbiome diversity (Shannon) of 1.45 units (95% CI, 0.15-2.50 [p, <0.0001]) and a lower abundance of taxa (p, 0.0086). Meta-analysis of the Shannon value showed considerable heterogeneity between studies (I2, 94.42). Unbiased analysis displayed as a funnel plot revealed a depletion of Lachnospiraceae, Ruminococcaceae and Ruminococcus and enrichment of Enterococcus. There was an important absence in the lack of cohort studies reporting gut microbiome changes and high heterogeneity between studies may be explained by variations in microbiome methods and confounder effects. Further human cohort studies are needed to understand RTI-induced gut microbiome changes to better understand interplay between microbes and respiratory health.


Subject(s)
Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Respiratory Tract Infections/microbiology , Animals , Bacteria/growth & development , Humans
4.
Acta Biochim Pol ; 68(3): 393-398, 2021 Aug 25.
Article in English | MEDLINE | ID: covidwho-1547880

ABSTRACT

The time of COVID-19 pandemic focused the attention of scientist to recognise the complex medical symptoms of the disease, modes of infection and possible therapies. The organisms' response towards SARS-CoV-2 infection depends on many individual factors and the course of disease is described as unprecedented and complex. Numerous symptoms from the respiratory system, abnormalities in the gastrointestinal tract, stroke, liver damage and coagulopathy, among others, are accompanied by negative side effects of the pandemic lifestyle, including immunity depletion, overall fitness impairment, skin condition worsening, psychological and psychiatric consequences. There is an urgent need to seek all possible routes for assuring favouring conditions to build and support the organisms' microbiological barriers and enhance immunity, which will also help during the ongoing vaccination action. Probiotic Lactic Acid Bacteria (LAB) and environmental Bacillus species are microorganisms typically found in food products or dietary supplements, but also applied on body surfaces or technological surfaces at home and in the industry. Since the contemporary definition of probiotics points to positive health effects, it is of highest importance to follow strict regulations and standards of product manufacturing, especially in the times of biohazard risks and rising public distrust of therapies. There is an urgent need to seek all possible routes for assuring the favouring conditions to build and support the organisms' microbiological barriers and enhance the immunity, that will serve also during the ongoing vaccination action. Probiotic LAB and environmental Bacillus species are microorganisms typically found in food products or dietary supplements, but also applied on body surface or technological surfaces in household and industry. Since the contemporary definition of probiotics points out the positive health effects, it is of highest importance to follow strict regulations and standards of product manufacturing, especially in the times of biohazard and rising public distrust of therapies.


Subject(s)
COVID-19/immunology , Probiotics/therapeutic use , COVID-19/microbiology , COVID-19/virology , Dietary Supplements , Gastrointestinal Tract/microbiology , Humans , SARS-CoV-2/isolation & purification
6.
PLoS One ; 16(6): e0251727, 2021.
Article in English | MEDLINE | ID: covidwho-1282294

ABSTRACT

BACKGROUND: The emergence of vancomycin resistant Enterococci (VRE) has alarmed the global community due to its tendency for colonization of the gastrointestinal tract. Human Immunodeficiency Virus (HIV) patients are colonized by vancomycin resistant Enterococci than other groups. The aim of this study was to determine the incidence of vancomycin resistant Enterococci and its associated factors among HIV infected patients on Anti-Retroviral Therapy (ART). METHODS: Institution based cross sectional study was conducted among HIV infected patients on ART at from June 1 to August 30, 2020. Socio-demographic and clinical data were collected by pre-tested structured questionnaire. Stool sample was collected and processed by standard microbiological techniques. Kirby Bauer Disc diffusion method was used to perform antimicrobial susceptibility testing. Data were entered by Epi data version 4.6.0.2 and analyzed by SPSS version 25. Bivariable and multivariable logistic regression model was used to analyze the association between dependent and independent variables. P-values in the multivariable analysis, adjusted odds ratio (AOR) and 95% confidence interval (CI) were used to determine the strength of association. P-value ≤0.05 was considered as significant. RESULTS: Enterococci spp was isolated on 123/200 (61.50%) patients. Among these isolates, the incidence of vancomycin resistant Enterococci was 11.4% [95% CI: (6.0-17.0)]. Antimicrobial susceptibility patterns against Enterococci showed highest rate of resistance to ampicillin (69.9%). Multidrug resistances were observed in 49.59% of Enterococci isolates. Study participants who had prior antibioticexposurer more than two weeks [AOR = 7.35; 95% CI: (1.2144.64)] and hospitalization for the last six months [AOR = 5.68; 95% CI: (1.09 29.74)] were significantly associated with vancomycin resistant Enterococci. CONCLUSIONS: In our study high incidence of vancomycin resistant Enterococci was found. Previous exposure to antibiotics for more than two weeks and hospitalization for more than six months were significantly associated with vancomycin resistant Enterococci. The isolated Enterococci had variable degrees of resistance to commonly prescribed antibiotics. Therefore, periodic surveillance on antimicrobial resistance pattern, adhering to rational use of antibiotics and implementing infection prevention protocols may reduce colonization by VRE.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Gastrointestinal Tract/microbiology , Gram-Positive Bacterial Infections/epidemiology , HIV Infections/complications , HIV/isolation & purification , Vancomycin-Resistant Enterococci/isolation & purification , Vancomycin/pharmacology , Adolescent , Adult , Aged , Anti-Bacterial Agents/pharmacology , Cross-Sectional Studies , Ethiopia/epidemiology , Female , Gram-Positive Bacterial Infections/microbiology , HIV Infections/drug therapy , HIV Infections/microbiology , HIV Infections/virology , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Young Adult
7.
Front Cell Infect Microbiol ; 11: 590874, 2021.
Article in English | MEDLINE | ID: covidwho-1158345

ABSTRACT

Gut microbiome alterations may play a paramount role in determining the clinical outcome of clinical COVID-19 with underlying comorbid conditions like T2D, cardiovascular disorders, obesity, etc. Research is warranted to manipulate the profile of gut microbiota in COVID-19 by employing combinatorial approaches such as the use of prebiotics, probiotics and symbiotics. Prediction of gut microbiome alterations in SARS-CoV-2 infection may likely permit the development of effective therapeutic strategies. Novel and targeted interventions by manipulating gut microbiota indeed represent a promising therapeutic approach against COVID-19 immunopathogenesis and associated co-morbidities. The impact of SARS-CoV-2 on host innate immune responses associated with gut microbiome profiling is likely to contribute to the development of key strategies for application and has seldom been attempted, especially in the context of symptomatic as well as asymptomatic COVID-19 disease.


Subject(s)
COVID-19/pathology , Dysbiosis/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Immunity, Innate/immunology , Angiotensin-Converting Enzyme 2/biosynthesis , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Bacteria/metabolism , COVID-19/therapy , Cardiovascular Diseases/pathology , Diabetes Mellitus, Type 2/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gene Expression/genetics , Humans , Leukocyte L1 Antigen Complex/biosynthesis , Obesity/pathology , Probiotics/pharmacology , SARS-CoV-2/immunology , Severity of Illness Index
8.
Front Cell Infect Microbiol ; 10: 619075, 2020.
Article in English | MEDLINE | ID: covidwho-1084083

ABSTRACT

Albeit the lungs were thought to be sterile, recent scientific data reported a microbial microbiota in the lungs of healthy individuals. Apparently, new developments in technological approachesincluding genome sequencing methodologies contributed in the identification of the microbiota and shed light on the role of the gut and lung microbiomes in the development of respiratory diseases. Moreover, knowledge of the human microbiome in health may act as a tool for evaluating characteristic shifts in the case of disease. This review paper discusses the development of respiratory disease linked to the intestinal dysbiosis which influences the lung immunity and microbiome. The gastrointestinal-lung dialogue provides interesting aspects in the pathogenesis of the respiratory diseases. Lastly, we were further interested on the role of this interconnection in the progression and physiopathology of newly emergedCOVID-19.


Subject(s)
Bacteria/isolation & purification , Lung/immunology , Lung/microbiology , Microbiota/physiology , Bacteria/classification , COVID-19/pathology , Gastrointestinal Tract/microbiology , Humans , SARS-CoV-2/growth & development
9.
Gut ; 70(4): 698-706, 2021 04.
Article in English | MEDLINE | ID: covidwho-1024254

ABSTRACT

OBJECTIVE: Although COVID-19 is primarily a respiratory illness, there is mounting evidence suggesting that the GI tract is involved in this disease. We investigated whether the gut microbiome is linked to disease severity in patients with COVID-19, and whether perturbations in microbiome composition, if any, resolve with clearance of the SARS-CoV-2 virus. METHODS: In this two-hospital cohort study, we obtained blood, stool and patient records from 100 patients with laboratory-confirmed SARS-CoV-2 infection. Serial stool samples were collected from 27 of the 100 patients up to 30 days after clearance of SARS-CoV-2. Gut microbiome compositions were characterised by shotgun sequencing total DNA extracted from stools. Concentrations of inflammatory cytokines and blood markers were measured from plasma. RESULTS: Gut microbiome composition was significantly altered in patients with COVID-19 compared with non-COVID-19 individuals irrespective of whether patients had received medication (p<0.01). Several gut commensals with known immunomodulatory potential such as Faecalibacterium prausnitzii, Eubacterium rectale and bifidobacteria were underrepresented in patients and remained low in samples collected up to 30 days after disease resolution. Moreover, this perturbed composition exhibited stratification with disease severity concordant with elevated concentrations of inflammatory cytokines and blood markers such as C reactive protein, lactate dehydrogenase, aspartate aminotransferase and gamma-glutamyl transferase. CONCLUSION: Associations between gut microbiota composition, levels of cytokines and inflammatory markers in patients with COVID-19 suggest that the gut microbiome is involved in the magnitude of COVID-19 severity possibly via modulating host immune responses. Furthermore, the gut microbiota dysbiosis after disease resolution could contribute to persistent symptoms, highlighting a need to understand how gut microorganisms are involved in inflammation and COVID-19.


Subject(s)
Bacteria , COVID-19 , Dysbiosis , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract , Immunity , SARS-CoV-2 , Adult , Bacteria/genetics , Bacteria/immunology , Bacteria/isolation & purification , C-Reactive Protein/analysis , COVID-19/blood , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/immunology , Cytokines/analysis , DNA, Bacterial/isolation & purification , Dysbiosis/epidemiology , Dysbiosis/etiology , Dysbiosis/immunology , Dysbiosis/virology , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/virology , Hong Kong , Humans , Male , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Severity of Illness Index , Transferases/analysis
10.
Med Hypotheses ; 147: 110476, 2021 Feb.
Article in English | MEDLINE | ID: covidwho-1002911

ABSTRACT

At the end of 2019, an emerging outbreak caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that first reported from Wuhan, China. The first manifestations of patients infected with SARS-CoV-2 was flu-like symptoms, while other type of manifestations, especially gastrointestinal manifestations were discovered recently. As of June 2020, there is no specific drug or treatment strategy for COVID-19, a disease caused by SARS-CoV-2, so different combination of antiviral drugs is currently being used. Gut microbiota mostly consists of four phyla, including Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. The interaction between gut microbiota and immune system through releasing some cytokines such as IL-1ß, IL-2, IL-10, TNF-α, and IFN-γ that play roles in the severity of COVID-19. In this article, a new potential treatment for COVID-19 by fecal microbiota transplantation (FMT) is described. FMT revealed promising results in different diseases, especially recurrent clostridium difficile infection, and it might reduce length of hospital admission and severity of the disease by modification of gut microbiota composition.


Subject(s)
COVID-19/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/virology , Bacteria/classification , COVID-19/virology , China , Cost-Benefit Analysis , Feces/virology , Humans , Immune System , Lung/microbiology , Models, Theoretical
11.
Molecules ; 25(21)2020 Oct 22.
Article in English | MEDLINE | ID: covidwho-983191

ABSTRACT

Inflammation is a biological response to the activation of the immune system by various infectious or non-infectious agents, which may lead to tissue damage and various diseases. Gut commensal bacteria maintain a symbiotic relationship with the host and display a critical function in the homeostasis of the host immune system. Disturbance to the gut microbiota leads to immune dysfunction both locally and at distant sites, which causes inflammatory conditions not only in the intestine but also in the other organs such as lungs and brain, and may induce a disease state. Probiotics are well known to reinforce immunity and counteract inflammation by restoring symbiosis within the gut microbiota. As a result, probiotics protect against various diseases, including respiratory infections and neuroinflammatory disorders. A growing body of research supports the beneficial role of probiotics in lung and mental health through modulating the gut-lung and gut-brain axes. In the current paper, we discuss the potential role of probiotics in the treatment of viral respiratory infections, including the COVID-19 disease, as major public health crisis in 2020, and influenza virus infection, as well as treatment of neurological disorders like multiple sclerosis and other mental illnesses.


Subject(s)
Coronavirus Infections/therapy , Influenza, Human/therapy , Mental Disorders/therapy , Multiple Sclerosis/therapy , Pneumonia, Viral/therapy , Probiotics/therapeutic use , Respiratory Tract Infections/therapy , Betacoronavirus/drug effects , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , Brain/immunology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/microbiology , Coronavirus Infections/virology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Humans , Immunomodulation , Influenza, Human/immunology , Influenza, Human/microbiology , Influenza, Human/virology , Lung/immunology , Mental Disorders/immunology , Mental Disorders/microbiology , Microbial Consortia/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology , Orthomyxoviridae/drug effects , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/microbiology , Pneumonia, Viral/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , SARS-CoV-2 , Symbiosis/immunology
12.
Front Cell Infect Microbiol ; 10: 576551, 2020.
Article in English | MEDLINE | ID: covidwho-979016

ABSTRACT

Infection with the SARS-CoV-2 virus causes cardiopulmonary and vascular complications, ranging in severity. Understanding the pathogenic mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for its prevention and/or treatment. Virus microbiota reciprocal interactions have been studied in a variety of viral infections. For example, the integrity of Coronavirus particles can be disrupted by surfactin, a bacterial surface molecule that targets other viruses, including that of influenza A. In this light, intestinal microbiota likely influences COVID-19 virulence, while from its side SARS-CoV-2 may affect the intestinal microbiome promoting dysbiosis and other deleterious consequences. Hence, the microbiota pre-existing health status and its alterations in the course of SARS-CoV-2 infection, are likely to play an important, still underscored role in determining individual susceptibility and resilience to COVID-19. Indeed, the vast majority of COVID-19 worst clinical conditions and fatalities develop in subjects with specific risk factors such as aging and the presence of one or more comorbidities, which are intriguingly characterized also by unhealthy microbiome status. Moreover, these comorbidities require complex pharmacological regimens known as "polypharmacy" that may further affect microbiota integrity and worsen the resilience to viral infections. This complex situation may represent a further and underestimated risk with regard to COVID-19 clinical burden for the elderly and comorbid people. Here, we discuss the possible biological, physiopathological, and clinical implications of gut microbiota in COVID-19 and the strategies to improve/maintain its healthy status as a simple and adjunctive strategy to reduce COVID-19 virulence and socio-sanitary burden.


Subject(s)
COVID-19/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , SARS-CoV-2/physiology , Age Factors , COVID-19/physiopathology , COVID-19/virology , Dysbiosis/microbiology , Dysbiosis/virology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/virology , Humans , Microbial Interactions , Risk Factors , Virulence , COVID-19 Drug Treatment
14.
Eur Rev Med Pharmacol Sci ; 24(20): 10853-10859, 2020 10.
Article in English | MEDLINE | ID: covidwho-914960

ABSTRACT

OBJECTIVE: The aim of this review paper was to discuss the gut microbiota-related aspects of COVID-19 patients. We presented the faecal-oral transmission of SARS-CoV-2, gut microbiota imbalance, and fecal microbiota transplantation as a hidden source of this virus. MATERIALS AND METHODS: We analyzed the available literature (PubMed, Embase, Google Scholar databases) regarding COVID-19 and gut microbiota related aspects. RESULTS: The gastrointestinal symptoms, such as nausea, vomiting, diarrhea, abdominal discomfort/pain, may occur in these patients. Notably, these symptoms may contribute to the severity of COVID-19. Recent several studies have revealed a new SARS-CoV-2 transmission possibility, opening a fresh view on COVID-19. It is observed the possibility of SARS-CoV-2 transmission via faecal-oral route. Fecal microbiota transplantation may be a hidden source of SARS-CoV-2. Additionally, the pharmacological treatment of COVID-19 and other factors may significantly alter the composition of gut microbiota. Among others, loss of bacterial diversity, the decrease of commensal microbes as well as the increase of opportunistic pathogens are observed. CONCLUSIONS: The alterations of gut microbiota in COVID-19 patients consequently may lead to the development of gut dysbiosis-related diseases even after recovery from COVID-19. Therefore, it is recommended to screen stool samples taken from recovered patients at least 35 days after clearance of virus from respiratory tract. Before 35 days period, SARS-CoV-2 may still be detected in feces. It is also recommended to screen the composition as well as the activity of gut microbiota to assess its balance. In the case of gut dysbiosis, there should be introduced an appropriate method of its modulation. Additionally, all the fecal samples which are prepared for fecal microbiota transplantation should be tested for SARS-CoV-2 to provide protection for its recipients.


Subject(s)
Coronavirus Infections/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Tract/microbiology , Pneumonia, Viral/microbiology , COVID-19 , Diarrhea/virology , Feces/virology , Gastrointestinal Diseases/virology , Gastrointestinal Microbiome , Gastrointestinal Tract/virology , Humans , Pandemics , Severity of Illness Index , Vomiting/virology
15.
Indian J Med Microbiol ; 38(3 & 4): 261-264, 2020.
Article in English | MEDLINE | ID: covidwho-914619

ABSTRACT

Although children with novel coronavirus infection (COVID-19) typically present with fever and respiratory symptoms, some children have reported gastrointestinal (GI) symptoms including vomiting and diarrhoea during the course of the disease. The continuous positive detection of the viral RNA from faeces in children even after nasopharyngeal swabs turned negative suggests that the GI tract may shed virus and a tentative faecal-oral transmission. The presence of angiotensin-converting enzyme 2 receptor and transmembrane serine protease 2, which are the key proteins of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cell entry process, in the GI tract can explain the digestive symptoms in COVID-19. COVID-19 has implications for the management of children with chronic luminal diseases. There is increasing concern regarding the risk that children with inflammatory bowel disease being infected with SARS-CoV-2.


Subject(s)
Coronavirus Infections/pathology , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Pneumonia, Viral/pathology , Betacoronavirus , COVID-19 , Child , Coronavirus Infections/diagnosis , Feces/virology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Humans , Inflammatory Bowel Diseases/pathology , Pandemics , Pneumonia, Viral/diagnosis , RNA, Viral/isolation & purification , SARS-CoV-2
16.
Benef Microbes ; 11(5): 477-488, 2020 Sep 01.
Article in English | MEDLINE | ID: covidwho-740509

ABSTRACT

Neonatal calf diarrhoea is one of the challenges faced by intensive farming, and probiotics are considered a promising approach to improve calves' health. The objective of this study was to evaluate the effect of potential probiotic lactobacilli on new-born dairy calves' growth, diarrhoea incidence, faecal score, cytokine expression in blood cells, immunoglobulin A (IgA) levels in plasma and faeces, and pathogen abundance in faeces. Two in vivo assays were conducted at the same farm in two annual calving seasons. Treated calves received one daily dose of the selected lactobacilli (Lactobacillus reuteri TP1.3B or Lactobacillus johnsonii TP1.6) for 10 consecutive days. A faecal score was recorded daily, average daily gain (ADG) was calculated, and blood and faeces samples were collected. Pathogen abundance was analysed by absolute qPCR in faeces using primers directed at Salmonella enterica, rotavirus, coronavirus, Cryptosporidium parvum and three Escherichia coli virulence genes (eae, clpG and Stx1). The faecal score was positively affected by the administration of both lactobacilli strains, and diarrhoea incidence was significantly lower in treated calves. No differences were found regarding ADG, cytokine expression, IgA levels and pathogen abundance. Our findings showed that oral administration of these strains could improve gastrointestinal health, but results could vary depending on the calving season, which may be related to pathogen seasonality and other environmental effects.


Subject(s)
Cattle Diseases/therapy , Diarrhea , Lactobacillus johnsonii/metabolism , Limosilactobacillus reuteri/metabolism , Probiotics/therapeutic use , Animals , Animals, Newborn , Cattle , Cattle Diseases/microbiology , Cattle Diseases/prevention & control , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Cryptosporidiosis/prevention & control , Cytokines/blood , Dairying , Diarrhea/prevention & control , Diarrhea/therapy , Diarrhea/veterinary , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Feces/virology , Gastrointestinal Tract/microbiology , Immunoglobulin A/blood , Rotavirus Infections/prevention & control , Rotavirus Infections/veterinary , Salmonella Infections, Animal/prevention & control
17.
Med Hypotheses ; 144: 110206, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-726782

ABSTRACT

COVID-19 is associated with acute and lethal pneumonia, causing the severe acute respiratory syndrome (SARS), which is not confined to the respiratory tract, as demonstrated by clinical evidence of the involvement of multiple organs, including the central nervous system (CNS). In this context, we hypothesized that both oligosymptomatic and symptomatic patients present an imbalance in the microbiota-gut (immune system) and nervous system axis, worsening the clinical picture. The brain constantly receives a direct and indirect influence from the intestine, more specifically from the immune system and intestinal microbiota. The presence of SARS-CoV-2 in the intestine and CNS, can contribute to both neurological disorders and gut immune system imbalance, events potentialized by an intestinal microbiota dysbiosis, aggravating the patient's condition and causing more prolonged harmful effects.


Subject(s)
COVID-19/immunology , COVID-19/microbiology , Gastrointestinal Microbiome , Brain/physiopathology , COVID-19/physiopathology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/physiopathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Homeostasis , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Humans , Models, Biological , Neuroimmunomodulation/physiology , Pandemics , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
18.
Appl Microbiol Biotechnol ; 104(19): 8089-8104, 2020 Oct.
Article in English | MEDLINE | ID: covidwho-723331

ABSTRACT

Interspecies transmissions of viruses between animals and humans may result in unpredictable pathogenic potential and new transmissible diseases. This mechanism has recently been exemplified by the discovery of new pathogenic viruses, such as the novel severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) pandemic, Middle-East respiratory syndrome-coronavirus epidemic in Saudi Arabia, and the deadly outbreak of Ebola in West Africa. The. SARS-CoV-2 causes coronavirus disease-19 (COVID-19), which is having a massive global impact in terms of economic disruption, and, above all, human health. The disease is characterized by dry cough, fever, fatigue, myalgia, and dyspnea. Other symptoms include headache, sore throat, rhinorrhea, and gastrointestinal disorders. Pneumonia appears to be the most common and severe manifestation of the infection. Currently, there is no vaccine or specific drug for COVID-19. Further, the development of new antiviral requires a considerable length of time and effort for drug design and validation. Therefore, repurposing the use of natural compounds can provide alternatives and can support therapy against COVID-19. In this review, we comprehensively discuss the prophylactic and supportive therapeutic role of probiotics for the management of COVID-19. In addition, the unique role of probiotics to modulate the gut microbe and assert gut homeostasis and production of interferon as an antiviral mechanism is described. Further, the regulatory role of probiotics on gut-lung axis and mucosal immune system for the potential antiviral mechanisms is reviewed and discussed.Key points• Gut microbiota role in antiviral diseases• Factors influencing the antiviral mechanism• Probiotics and Covid-19.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/therapy , Pneumonia, Viral/therapy , Probiotics/therapeutic use , Animals , COVID-19 , Coronavirus Infections/prevention & control , Coronavirus Infections/transmission , Gastrointestinal Tract/microbiology , Humans , Immunity, Mucosal , Lung/immunology , Lung/microbiology , Lung/virology , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Pneumonia, Viral/transmission , Probiotics/metabolism , Respiratory Tract Infections/microbiology , SARS-CoV-2 , Virus Diseases/prevention & control , Virus Diseases/therapy , Virus Diseases/transmission , Vitamin D/physiology , Zinc/metabolism
19.
Virus Res ; 286: 198103, 2020 09.
Article in English | MEDLINE | ID: covidwho-669613

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has evolved into a major pandemic called coronavirus disease 2019 (COVID-19) that has created unprecedented global health emergencies, and emerged as a serious threat due to its strong ability for human-to-human transmission. The reports indicate the ability of SARS-CoV-2 to affect almost any organ due to the presence of a receptor known as angiotensin converting enzyme 2 (ACE2) across the body. ACE2 receptor is majorly expressed in the brush border of gut enterocytes along with the ciliated cells and alveolar epithelial type II cells in the lungs. The amino acid transport function of ACE2 has been linked to gut microbial ecology in gastrointestinal (GI) tract, thereby suggesting that COVID-19 may, to some level, be linked to the enteric microbiota. The significant number of COVID-19 patients shows extra-pulmonary symptoms in the GI tract. Many subsequent studies revealed viral RNA of SARS-CoV-2 in fecal samples of COVID-19 patients. This presents a new challenge in the diagnosis and control of COVID-19 infection with a caution for proper sanitation and hygiene. Here, we aim to discuss the immunological co-ordination between gut and lungs that facilitates SARS-CoV-2 to infect and multiply in the inflammatory bowel disease (IBD) and non-IBD patients.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/immunology , Cytokine Release Syndrome/immunology , Dysbiosis/immunology , Gastrointestinal Tract/immunology , Inflammatory Bowel Diseases/immunology , Lung/immunology , Pneumonia, Viral/immunology , Angiotensin-Converting Enzyme 2 , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/microbiology , Coronavirus Infections/virology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/microbiology , Cytokine Release Syndrome/virology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Dysbiosis/drug therapy , Dysbiosis/microbiology , Dysbiosis/virology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/virology , Gene Expression , Host-Pathogen Interactions/immunology , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/virology , Lung/drug effects , Lung/microbiology , Lung/virology , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/microbiology , Pneumonia, Viral/virology , Receptors, Virus/genetics , Receptors, Virus/immunology , SARS-CoV-2
20.
Food Res Int ; 136: 109577, 2020 Oct.
Article in English | MEDLINE | ID: covidwho-662726

ABSTRACT

The year 2020 will be remembered by a never before seen, at least by our generation, global pandemic of COVID-19. While a desperate search for effective vaccines or drug therapies is on the run, nutritional strategies to promote immunity against SARS-CoV-2, are being discussed. Certain fermented foods and probiotics may deliver viable microbes with the potential to promote gut immunity. Prebiotics, on their side, may enhance gut immunity by selectively stimulating certain resident microbes in the gut. Different levels of evidence support the use of fermented foods, probiotics and prebiotics to promote gut and lungs immunity. Without being a promise of efficacy against COVID-19, incorporating them into the diet may help to low down gut inflammation and to enhance mucosal immunity, to possibly better face the infection by contributing to diminishing the severity or the duration of infection episodes.


Subject(s)
Coronavirus Infections/therapy , Fermented Foods , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammation , Pneumonia, Viral/therapy , Prebiotics , Probiotics , Betacoronavirus , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/microbiology , Coronavirus Infections/virology , Diet , Gastrointestinal Tract/immunology , Humans , Inflammation/etiology , Inflammation/microbiology , Inflammation/prevention & control , Inflammation/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/microbiology , Pneumonia, Viral/virology , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL