Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Mol Sci ; 23(12)2022 Jun 18.
Article in English | MEDLINE | ID: covidwho-2282707

ABSTRACT

The intestinal microbiota is known to influence local immune homeostasis in the gut and to shape the developing immune system towards elimination of pathogens and tolerance towards self-antigens. Even though the lung was considered sterile for a long time, recent evidence using next-generation sequencing techniques confirmed that the lower airways possess their own local microbiota. Since then, there has been growing evidence that the local respiratory and intestinal microbiota play a role in acute and chronic pediatric lung diseases. The concept of the so-called gut-lung axis describing the mutual influence of local microbiota on distal immune mechanisms was established. The mechanisms by which the intestinal microbiota modulates the systemic immune response include the production of short-chain fatty acids (SCFA) and signaling through pattern recognition receptors (PRR) and segmented filamentous bacteria. Those factors influence the secretion of pro- and anti-inflammatory cytokines by immune cells and further modulate differentiation and recruitment of T cells to the lung. This article does not only aim at reviewing recent mechanistic evidence from animal studies regarding the gut-lung axis, but also summarizes current knowledge from observational studies and human trials investigating the role of the respiratory and intestinal microbiota and their modulation by pre-, pro-, and synbiotics in pediatric lung diseases.


Subject(s)
Gastrointestinal Microbiome , Lung Diseases , Microbiota , Animals , Child , Fatty Acids, Volatile , Gastrointestinal Microbiome/physiology , Humans , Lung
2.
Biol Rev Camb Philos Soc ; 2022 Sep 15.
Article in English | MEDLINE | ID: covidwho-2237115

ABSTRACT

Circadian rhythms are present in almost every organism and regulate multiple aspects of biological and physiological processes (e.g. metabolism, immune responses, and microbial exposure). There exists a bidirectional circadian interaction between the host and its gut microbiota, and potential circadian orchestration of both host and gut microbiota in response to invading pathogens. In this review, we summarize what is known about these intestinal microbial oscillations and the relationships between host circadian clocks and various infectious agents (bacteria, fungi, parasites, and viruses), and discuss how host circadian clocks prime the immune system to fight pathogen infections as well as the direct effects of circadian clocks on viral activity (e.g. SARS-CoV-2 entry and replication). Finally, we consider strategies employed to realign normal circadian rhythmicity for host health, such as chronotherapy, dietary intervention, good sleep hygiene, and gut microbiota-targeted therapy. We propose that targeting circadian rhythmicity may provide therapeutic opportunities for the treatment of infectious diseases.

3.
Int J Mol Sci ; 23(23)2022 Nov 28.
Article in English | MEDLINE | ID: covidwho-2143228

ABSTRACT

The human coronavirus SARS-CoV-2 or COVID-19 that emerged in late 2019 causes a respiratory tract infection and has currently resulted in more than 627 million confirmed cases and over 6.58 million deaths worldwide up to October 2022. The highest death rate caused by COVID-19 is in older people, especially those with comorbidities. This evidence presents a challenge for biomedical research on aging and also identifies some key players in inflammation, including mast cells and platelets, which could represent important markers and, at the same time, unconventional therapeutic targets. Studies have shown a decrease in the diversity of gut microbiota composition in the elderly, particularly a reduced abundance of butyrate-producing species, and COVID-19 patients manifest faecal microbiome alterations, with an increase in opportunistic pathogens and a depletion of commensal beneficial microorganisms. The main purpose of this narrative review is to highlight how an altered condition of the gut microbiota, especially in the elderly, could be an important factor and have a strong impact in the lung homeostasis and COVID-19 phenomenon, jointly to the activation of mast cells and platelets, and also affect the outcomes of the pathology. Therefore, a targeted and careful control of the intestinal microbiota could represent a complementary intervention to be implemented for the management and the challenge against COVID-19.


Subject(s)
COVID-19 , Microbiota , Humans , Aged , SARS-CoV-2 , Mast Cells , Lung , Dysbiosis
4.
Microbiome ; 10(1): 162, 2022 10 03.
Article in English | MEDLINE | ID: covidwho-2053974

ABSTRACT

BACKGROUND: Mammalian intestinal microbiomes are necessary for antagonizing systemic viral infections. However, very few studies have identified whether poultry commensal bacteria play a crucial role in protecting against systemic viral infections. Nephropathogenic infectious bronchitis virus (IBV) is a pathogenic coronavirus that causes high morbidity and multiorgan infection tropism in chickens. RESULTS: In this study, we used broad-spectrum oral antibiotics (ABX) to treat specific pathogen free (SPF) chickens to deplete the microbiota before infection with nephropathogenic IBV to analyze the impact of microbiota on IBV infections in vivo. Depletion of the SPF chicken microbiota increases pathogenicity and viral burden following IBV infection. The gnotobiotic chicken infection model further demonstrated that intestinal microbes are resistant to nephropathogenic IBV infection. In addition, ABX-treated chickens showed a severe reduction in macrophage activation, impaired type I IFN production, and IFN-stimulated gene expression in peripheral blood mononuclear cells and the spleen. Lactobacillus isolated from SPF chickens could restore microbiota-depleted chicken macrophage activation and the IFNAR-dependent type I IFN response to limit IBV infection. Furthermore, exopolysaccharide metabolites of Lactobacillus spp. could induce IFN-ß. CONCLUSIONS: This study revealed the resistance mechanism of SPF chicken intestinal microbiota to nephropathogenic IBV infection, providing new ideas for preventing and controlling nephropathogenic IBV. Video abstract.


Subject(s)
Gastrointestinal Microbiome , Infectious bronchitis virus , Poultry Diseases , Animals , Anti-Bacterial Agents , Chickens , Infectious bronchitis virus/genetics , Leukocytes, Mononuclear , Mammals
5.
Cells ; 11(16)2022 08 18.
Article in English | MEDLINE | ID: covidwho-1997525

ABSTRACT

Clinical and experimental data indicate that severe acute respiratory syndrome coronavirus (SARS-CoV)-2 infection is associated with significant changes in the composition and function of intestinal microbiota. However, the relevance of these effects for SARS-CoV-2 pathophysiology is unknown. In this study, we analyzed the impact of microbiota depletion after antibiotic treatment on the clinical and immunological responses of K18-hACE2 mice to SARS-CoV-2 infection. Mice were treated with a combination of antibiotics (kanamycin, gentamicin, metronidazole, vancomycin, and colistin, Abx) for 3 days, and 24 h later, they were infected with SARS-CoV-2 B lineage. Here, we show that more than 80% of mice succumbed to infection by day 11 post-infection. Treatment with Abx had no impact on mortality. However, Abx-treated mice presented better clinical symptoms, with similar weight loss between infected-treated and non-treated groups. We observed no differences in lung and colon histopathological scores or lung, colon, heart, brain and kidney viral load between groups on day 5 of infection. Despite some minor differences in the expression of antiviral and inflammatory markers in the lungs and colon, no robust change was observed in Abx-treated mice. Together, these findings indicate that microbiota depletion has no impact on SARS-CoV-2 infection in mice.


Subject(s)
COVID-19 Drug Treatment , Microbiota , Angiotensin-Converting Enzyme 2 , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Melphalan , Mice , Mice, Transgenic , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2 , gamma-Globulins
6.
Int J Environ Res Public Health ; 19(16)2022 08 17.
Article in English | MEDLINE | ID: covidwho-1987814

ABSTRACT

The intestinal microbiota plays an important role in the immune response against viral infections, modulating both innate and adaptive immune responses. The cytokine storm is associated with COVID-19 severity, and the patient's immune status is influenced by the intestinal microbiota in a gut-lung bidirectional interaction. In this study, we evaluate the intestinal microbiota of Brazilian patients in different post-COVID-19 periods, and correlate this with clinical data and the antibiotic therapy used during the acute phase. DNA extracted from stool samples was sequenced and total anti-SARS-CoV-2 antibodies and C-reactive protein were quantified. Compared with controls, there were significant differences in the microbiota diversity in post-COVID-19 patients, suggesting an intestinal dysbiosis even several months after acute disease resolution. Additionally, we detected some genera possibly associated with the post-COVID-19 dysbiosis, including Desulfovibrio, Haemophillus, Dialister, and Prevotella, in addition to decreased beneficial microbes, associated with antibiotic-induced dysbiosis, such as Bifidobacterium and Akkermansia. Therefore, our hypothesis is that dysbiosis and the indiscriminate use of antibiotics during the pandemic may be associated with post-COVID-19 clinical manifestations. In our study, 39% (n = 58) of patients reported symptoms, including fatigue, dyspnea, myalgia, alopecia, anxiety, memory loss, and depression. These data suggest that microbiota modulation may represent a target for recovery from acute COVID-19 and a therapeutic approach for post-COVID-19 sequelae.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Acute Disease , Dysbiosis/microbiology , Humans , Pandemics
7.
Profilakticheskaya Meditsina ; 25(7):106-115, 2022.
Article in Russian | Academic Search Complete | ID: covidwho-1955160

ABSTRACT

The literature review summarizes the main aspects of the impact of SARS-CoV-2 on the gastrointestinal tract, which must be taken into account during a difficult epidemiological situation, and shows the main clinical and functional changes that affect the condition of patients and the activities of the healthcare system. The clinical manifestations caused by the action of SARS-CoV-2 on the organs of the gastrointestinal tract are described in detail. Clinical and laboratory methods established the fact of the development of a multi-inflammatory syndrome, which determines the systemic profile of the lesion and determines the nature and severity of the course of the disease, its complications and outcome. The histological picture of the lesion of the digestive system by the virus is described. The necessity of further studies of the mechanisms of interaction of the virus with the cells of the intestine and the immune system in order to develop methods for early diagnosis, effective treatment and prevention of complications of a new coronavirus infection is substantiated. The E-library, MedLine, National Center for Biotechnology Information databases were used to search for literature sources. (English) [ FROM AUTHOR] В обзоре литературы обобщены основные аспекты влияния SARS-CoV-2 на желудочно-кишечный тракт и связанные с этим особенности организации медицинской помощи, которые необходимо учитывать во время сложной эпидемиологической ситуации. Приведены основные клинико-функциональные изменения в состоянии пациентов. Клинико-лабораторными методами установлен факт развития мультивоспалительного синдрома, обусловливающего системный профиль поражения, определяющего характер и тяжесть течения болезни, ее осложнения и исход. Описана гистологическая картина поражения вирусом органов пищеварения. Обоснована необходимость дальнейших исследований механизмов взаимодействия вируса с клетками кишечника и иммунной системы для разработки методов ранней диагностики, эффективного лечения и профилактики осложнений новой коронавирусной инфекции. Для поиска источников литературы использованы базы данных e-Library, MedLine, National Center for Biotechnology Information. (Russian) [ FROM AUTHOR] Copyright of Profilakticheskaya Meditsina is the property of Media Sphere Publishing House and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full . (Copyright applies to all s.)

8.
Nutrients ; 14(11)2022 May 28.
Article in English | MEDLINE | ID: covidwho-1869723

ABSTRACT

Berry consumption has beneficial effects on blood pressure. Intestinal microbiota transform berry phytochemicals into more bioactive forms. Thus, we performed a systematic review of randomized clinical trials to determine whether berry polyphenols in foods, extracts or supplements have effects on both the profile of gut microbiota and systolic and diastolic blood pressure in humans. PubMed, Cochrane Library, Scopus, and CAB Abstracts (EBSCOhost) were searched for randomized clinical trials in humans published from 1 January 2011 to 29 October 2021. Search results were imported into Covidence for screening and data extraction by two blinded reviewers, who also performed bias assessment independently. The literature search identified 216 publications; after duplicates were removed, 168 publications were screened with 12 full-text publications assessed for eligibility. Ultimately three randomized clinical trials in humans met the eligibility criteria. One randomized clinical trial showed a low risk of bias while the other two randomized clinical trials included low, high or unclear risk of bias. Together the randomized clinical trials showed that berry consumption (Aronia berry, strawberries, raspberries, cloudberries and bilberries) for 8-12 weeks had no significant effect on both blood pressure and the gut microbiota. More randomized clinical trials are needed to determine the effects of berry consumption on the profile of gut microbiota and blood pressure in humans.


Subject(s)
Gastrointestinal Microbiome , Polyphenols , Blood Pressure , Fruit , Humans , Polyphenols/pharmacology , Randomized Controlled Trials as Topic
9.
Front Cell Infect Microbiol ; 12: 848650, 2022.
Article in English | MEDLINE | ID: covidwho-1834359

ABSTRACT

Clinical and experimental studies indicate that the bacterial and fungal gut microbiota modulates immune responses in distant organs including the lungs. Immune dysregulation is associated with severe SARS-CoV-2 infection, and several groups have observed gut bacterial dysbiosis in SARS-CoV-2 infected patients, while the fungal gut microbiota remains poorly defined in these patients. We analyzed the fungal gut microbiome from rectal swabs taken prior to anti-infective treatment in 30 SARS-CoV-2 positive (21 non-severe COVID-19 and 9 developing severe/critical COVID-19 patients) and 23 SARS-CoV-2 negative patients by ITS2-sequencing. Pronounced but distinct interconnected fungal communities distinguished SARS-CoV-2 positive and negative patients. Fungal gut microbiota in severe/critical COVID-19 illness was characterized by a reduced diversity, richness and evenness and by an increase of the relative abundance of the Ascomycota phylum compared with non-severe COVID-19 illness. A dominance of a single fungal species with a relative abundance of >75% was a frequent feature in severe/critical COVID-19. The dominating fungal species were highly variable between patients even within the groups. Several fungal taxa were depleted in patients with severe/critical COVID-19.The distinct compositional changes of the fungal gut microbiome in SARS-CoV-2 infection, especially in severe COVID-19 illness, illuminate the necessity of a broader approach to investigate whether the differences in the fungal gut microbiome are consequences of SARS-CoV-2 infection or a predisposing factor for critical illness.


Subject(s)
Ascomycota , COVID-19 , Gastrointestinal Microbiome , Mycobiome , Bacteria , Dysbiosis , Humans , SARS-CoV-2
10.
Vopr Pitan ; 91(1): 86-97, 2022.
Article in Russian | MEDLINE | ID: covidwho-1744381

ABSTRACT

The problem of increasing the population antiviral immunity is of particular importance during the third year of the SARS-CoV-2 pandemic. Concomitant intestinal dysbiosis is known to play an significant role in immune cell dysfunction. Therefore, it is very important to take measures to maintain the gut microbiota using the most affordable nutritional remedies, which include fermented milk and probiotic products designed for mass population consumption and capable of enhancing their immune defence when added to the daily diet. The aim of the study was to analyze scientific evidence highlighting the role of intestinal microbiota in maintaining the macro-organism immunological balance, and to evaluate modern fermented milk and probiotic products in terms of their effect on normalising the gut microbiota and their importance in the prevention and treatment of SARS-CoV-2. Material and methods. The presented scientific and analytical review analyzed the data of electronic resources of the Global Health platform, scientific libraries eLIBRARY.RU, Cochrane Library and CyberLeninka, the search system Google Academy¼, specialized sites for scientific publications ScienceDirect and Elsevier, bibliographic databases of articles on medical sciences MEDLINE, CDC infection diseases, Embase and PubMed- NCBI. The structural-logical, analytical and axiomatic methods were used. Results. It has been shown that normal intestinal microbiota takes part in maintaining metabolism in the digestive tract, increases the body's immune reactivity and regulates the functioning of all organs and systems. The severity of dysbiotic disorders can determine susceptibility to SARS-CoV-2, the severity of this infection course, as well as the level of post-infection and post-vaccination anti-COVID-19 immunity. The high prevalence of gut dysbacteriosis indicates the need to strengthen measures of correcting dysbiotic disorders, including the inclusion of fermented and probiotic products in the daily population diet. Conclusion. Fermented milk and probiotic products, as sources of easily digestible macronutrients, essential micronutrients, biologically active substances and beneficial live microorganisms, should be included in the daily diet during the SARS-CoV-2 pandemic to increase the adaptive capacity and immunity of the population.


Subject(s)
COVID-19 , Diet , Gastrointestinal Microbiome , Milk , Probiotics , Animals , COVID-19/immunology , COVID-19/prevention & control , Fermentation , Gastrointestinal Microbiome/immunology , Humans , Milk/microbiology , Pandemics , Probiotics/administration & dosage , SARS-CoV-2
11.
Viruses ; 14(3)2022 02 25.
Article in English | MEDLINE | ID: covidwho-1737037

ABSTRACT

Coronavirus disease 2019, or COVID-19, is a major challenge facing scientists worldwide. Alongside the lungs, the system of organs comprising the GI tract is commonly targeted by COVID-19. The dysbiotic modulations in the intestine influence the disease severity, potentially due to the ability of the intestinal microbiota to modulate T lymphocyte functions, i.e., to suppress or activate T cell subpopulations. The interplay between the lungs and intestinal microbiota is named the gut-lung axis. One of the most usual comorbidities in COVID-19 patients is type 2 diabetes, which induces changes in intestinal microbiota, resulting in a pro-inflammatory immune response, and consequently, a more severe course of COVID-19. However, changes in the microbiota in this comorbid pathology remain unclear. Metformin is used as a medication to treat type 2 diabetes. The use of the type 2 diabetes drug metformin is a promising treatment for this comorbidity because, in addition to its hypoglycemic action, it can increase amount of intestinal bacteria that induce regulatory T cell response. This dual activity of metformin can reduce lung damage and improve the course of the COVID-19 disease.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Dysbiosis , Humans , Immunity
12.
Archives of Neuroscience ; 8(4):3, 2021.
Article in English | Web of Science | ID: covidwho-1689858
13.
Clin Microbiol Infect ; 28(4): 502-512, 2022 Apr.
Article in English | MEDLINE | ID: covidwho-1605010

ABSTRACT

BACKGROUND: Vulnerable patients with intestinal colonization of multidrug-resistant organisms (MDROs) are recognized to be at increased risk of invasive MDRO-driven infection. Intestinal microbiota transplantation (IMT, also called faecal microbiota transplant) is the transfer of healthy screened donor stool to an affected recipient, and recent interest has focused on its impact on the reduction of invasive MDRO infection. OBJECTIVES: To describe how to establish a clinical IMT pathway for patients at risk of MDRO invasive infection, with special considerations for optimizing administration and assessment of endpoints. SOURCES: Expert guidelines and peer-reviewed clinical studies are encompassed and discussed. CONTENT: IMT is offered to patients with MDROs detected on rectal or stool screening and either at risk of MDRO invasive infection due to altered immune status or those with recurrent MDRO-mediated invasive disease and considered at risk of further disease. Donor screening should include pathogens with theoretical or demonstrated risk of transmission (including MDROs themselves and SARS-CoV-2) and take into consideration the relative immunosuppressed state of potential recipients. Delivery of IMT is timed for when the patient is free from active infection, but no additional antibiotics are indicated. If administered when future immunosuppression is to take place, IMT is aligned at least 2 weeks beforehand to ensure sufficient time for engraftment. Patients are followed up in terms of adverse effects from IMT and clinicians are advised to discuss with the IMT multidisciplinary team on choice of antibiotics if needed to take into consideration the impact upon the intestinal microbiome. Prevention of invasive disease is the primary measure of success, rather than using intestinal decolonization as a binary outcome. Repeat IMT is considered case by case. IMPLICATIONS: Future research areas should include randomized studies that consider clinical outcomes and cost-effectiveness, and better understanding of mechanisms to identify markers of treatment success and functional microbiome components that could be used therapeutically.


Subject(s)
Drug Resistance, Multiple, Bacterial , Fecal Microbiota Transplantation , COVID-19 , Gastrointestinal Microbiome , Humans , SARS-CoV-2
14.
Front Pharmacol ; 12: 758159, 2021.
Article in English | MEDLINE | ID: covidwho-1581234

ABSTRACT

Background: The world has been unprecedentedly hit by a global pandemic which broke the record of deadly pandemics that faced humanity ever since its existence. Even kids are well-versed in the terminologies and basics of the SARS-CoV-2 virus and COVID-19 now. The vaccination program has been successfully launched in various countries, given that the huge global population of concern is still far behind to be vaccinated. Furthermore, the scarcity of any potential drug against the COVID-19-causing virus forces scientists and clinicians to search for alternative and complementary medicines on a war-footing basis. Aims and Objectives: The present review aims to cover and analyze the etiology and epidemiology of COVID-19, the role of intestinal microbiota and pro-inflammatory markers, and most importantly, the natural products to combat this deadly SARS-CoV-2 virus. Methods: A primary literature search was conducted through PubMed and Google Scholar using relevant keywords. Natural products were searched from January 2020 to November 2020. No timeline limit has been imposed on the search for the biological sources of those phytochemicals. Interactive mapping has been done to analyze the multi-modal and multi-target sources. Results and Discussion: The intestinal microbiota and the pro-inflammatory markers that can serve the prognosis, diagnosis, and treatment of COVID-19 were discussed. The literature search resulted in yielding 70 phytochemicals and ten polyherbal formulations which were scientifically analyzed against the SARS-CoV-2 virus and its targets and found significant. Retrospective analyses led to provide information about 165 biological sources that can also be screened if not done earlier. Conclusion: The interactive analysis mapping of biological sources with phytochemicals and targets as well as that of phytochemical class with phytochemicals and COVID-19 targets yielded insights into the multitarget and multimodal evidence-based complementary medicines.

15.
Front Cell Infect Microbiol ; 11: 747816, 2021.
Article in English | MEDLINE | ID: covidwho-1556003

ABSTRACT

The gut microbiota contributes to maintaining human health and regulating immune responses. Severe COVID-19 illness is associated with a dysregulated pro-inflammatory immune response. The effect of SARS-CoV-2 on altering the gut microbiome and the relevance of the gut microbiome on COVID-19 severity needs to be clarified. In this prospective study, we analyzed the gut microbiome of 212 patients of a tertiary care hospital (117 patients infected with SARS-CoV-2 and 95 SARS-CoV-2 negative patients) using 16S rRNA gene sequencing of the V3-V4 region. Inflammatory markers and immune cells were quantified from blood. The gut microbiome in SARS-CoV-2 infected patients was characterized by a lower bacterial richness and distinct differences in the gut microbiome composition, including an enrichment of the phyla Proteobacteria and Bacteroidetes and a decrease of Actinobacteria compared to SARS-CoV-2 negative patients. The relative abundance of several genera including Bifidobacterium, Streptococcus and Collinsella was lower in SARS-CoV-2 positive patients while the abundance of Bacteroides and Enterobacteriaceae was increased. Higher pro-inflammatory blood markers and a lower CD8+ T cell number characterized patients with severe COVID-19 illness. The gut microbiome of patients with severe/critical COVID-19 exhibited a lower abundance of butyrate-producing genera Faecalibacterium and Roseburia and a reduction in the connectivity of a distinct network of anti-inflammatory genera that was observed in patients with mild COVID-19 illness and in SARS-CoV-2 negative patients. Dysbiosis of the gut microbiome associated with a pro-inflammatory signature may contribute to the hyperinflammatory immune response characterizing severe COVID-19 illness.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Anti-Inflammatory Agents , Humans , Prospective Studies , RNA, Ribosomal, 16S/genetics , SARS-CoV-2
17.
MMW Fortschr Med ; 163(Suppl 5): 17-20, 2021 09.
Article in German | MEDLINE | ID: covidwho-1353739

ABSTRACT

BACKGROUND: There is a bidirectional interaction between the intestines and lungs, the so-called lung-intestinal axis. METHOD: The review article reports on studies that deal with a possible influence of the intestinal microbiota on the immune response to a SARS-CoV-2 infection. RESULTS AND CONCLUSIONS: Studies have shown that COVID-19 is accompanied by dysbiosis that persists even after successful virus conversion (negative PCR). One study found that the severity of COVID-19 is associated with the intestinal microbiota. A dysbiosis could thus favor the so-called cytokine storm. There is indication that pre- and probiotics could boost the immune response in both the guts and lungs.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Dysbiosis , Humans , Intestines , SARS-CoV-2
18.
Front Immunol ; 12: 660179, 2021.
Article in English | MEDLINE | ID: covidwho-1264332

ABSTRACT

The complex interplay between the gut microbiota, the intestinal barrier, the immune system and the liver is strongly influenced by environmental and genetic factors that can disrupt the homeostasis leading to disease. Among the modulable factors, diet has been identified as a key regulator of microbiota composition in patients with metabolic syndrome and related diseases, including the metabolic dysfunction-associated fatty liver disease (MAFLD). The altered microbiota disrupts the intestinal barrier at different levels inducing functional and structural changes at the mucus lining, the intercellular junctions on the epithelial layer, or at the recently characterized vascular barrier. Barrier disruption leads to an increased gut permeability to bacteria and derived products which challenge the immune system and promote inflammation. All these alterations contribute to the pathogenesis of MAFLD, and thus, therapeutic approaches targeting the gut-liver-axis are increasingly being explored. In addition, the specific changes induced in the intestinal flora may allow to characterize distinctive microbial signatures for non-invasive diagnosis, severity stratification and disease monitoring.


Subject(s)
Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Liver/immunology , Metabolic Syndrome/immunology , Non-alcoholic Fatty Liver Disease/immunology , Animals , CCR5 Receptor Antagonists/therapeutic use , Dysbiosis/immunology , Dysbiosis/microbiology , Humans , Imidazoles/therapeutic use , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Liver/metabolism , Liver/pathology , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Sulfoxides/therapeutic use
19.
Front Immunol ; 12: 676232, 2021.
Article in English | MEDLINE | ID: covidwho-1247868

ABSTRACT

The intestinal microbiota is thought to be an important biological barrier against enteric pathogens. Its depletion, however, also has curative effects against some viral infections, suggesting that different components of the intestinal microbiota can play both promoting and inhibitory roles depending on the type of viral infection. The two primary mechanisms by which the microbiota facilitates or inhibits viral invasion involve participation in the innate and adaptive immune responses and direct or indirect interaction with the virus, during which the abundance and composition of the intestinal microbiota might be changed by the virus. Oral administration of probiotics, faecal microbiota transplantation (FMT), and antibiotics are major therapeutic strategies for regulating intestinal microbiota balance. However, these three methods have shown limited curative effects in clinical trials. Therefore, the intestinal microbiota might represent a new and promising supplementary antiviral therapeutic target, and more efficient and safer methods for regulating the microbiota require deeper investigation. This review summarizes the latest research on the relationship among the intestinal microbiota, anti-viral immunity and viruses and the most commonly used methods for regulating the intestinal microbiota with the goal of providing new insight into the antiviral effects of the gut microbiota.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/immunology , Probiotics/therapeutic use , SARS-CoV-2/physiology , Virus Diseases/therapy , Animals , Host-Pathogen Interactions , Humans
20.
Clin Infect Dis ; 71(10): 2669-2678, 2020 12 17.
Article in English | MEDLINE | ID: covidwho-1059703

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) is an emerging serious global health problem. Gastrointestinal symptoms are common in COVID-19 patients, and severe acute respiratory syndrome coronavirus 2 RNA has been detected in stool specimens. However, the relationship between the gut microbiome and disease remains to be established. METHODS: We conducted a cross-sectional study of 30 patients with COVID-19, 24 patients with influenza A(H1N1), and 30 matched healthy controls (HCs) to identify differences in the gut microbiota by 16S ribosomal RNA gene V3-V4 region sequencing. RESULTS: Compared with HCs, COVID-19 patients had significantly reduced bacterial diversity; a significantly higher relative abundance of opportunistic pathogens, such as Streptococcus, Rothia, Veillonella, and Actinomyces; and a lower relative abundance of beneficial symbionts. Five biomarkers showed high accuracy for distinguishing COVID-19 patients from HCs with an area under the curve (AUC) up to 0.89. Patients with H1N1 displayed lower diversity and different overall microbial composition compared with COVID-19 patients. Seven biomarkers were selected to distinguish the 2 cohorts (AUC = 0.94). CONCLUSIONS: The gut microbial signature of patients with COVID-19 was different from that of H1N1 patients and HCs. Our study suggests the potential value of the gut microbiota as a diagnostic biomarker and therapeutic target for COVID-19, but further validation is needed.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Influenza A Virus, H1N1 Subtype , Influenza, Human , Cross-Sectional Studies , Dysbiosis , Feces , Humans , Influenza A Virus, H1N1 Subtype/genetics , RNA, Ribosomal, 16S/genetics , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL