Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
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
2.
Chin Med J (Engl) ; 135(16): 1927-1939, 2022 Aug 20.
Article in English | MEDLINE | ID: covidwho-2119069

ABSTRACT

ABSTRACT: Fecal microbiota transplantation (FMT) has been used as a core therapy for treating dysbiosis-related diseases by remodeling gut microbiota. The methodology and technology for improving FMT are stepping forward, mainly including washed microbiota transplantation (WMT), colonic transendoscopic enteral tubing (TET) for microbiota delivery, and purified Firmicutes spores from fecal matter. To improve the understanding of the clinical applications of FMT, we performed a systematic literature review on FMT published from 2011 to 2021. Here, we provided an overview of the reported clinical benefits of FMT, the methodology of processing FMT, the strategy of using FMT, and the regulations on FMT from a global perspective. A total of 782 studies were included for the final analysis. The present review profiled the effectiveness from all clinical FMT uses in 85 specific diseases as eight categories, including infections, gut diseases, microbiota-gut-liver axis, microbiota-gut-brain axis, metabolic diseases, oncology, hematological diseases, and other diseases. Although many further controlled trials will be needed, the dramatic increasing reports have shown the promising future of FMT for dysbiosis-related diseases in the gut or beyond the gut.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Fecal Microbiota Transplantation/methods , Dysbiosis/therapy , Feces
3.
BMJ Open ; 12(6): e056594, 2022 06 27.
Article in English | MEDLINE | ID: covidwho-1978555

ABSTRACT

INTRODUCTION: Several studies have demonstrated dysbiosis in irritable bowel syndrome (IBS). Therefore, faecal microbiota transplantation, whose effect and safety have been proven in Clostridioides difficile infections, may hold promise in other conditions, including IBS. Our study will examine the effectiveness of stool transfer with artificially increased microbial diversity in IBS treatment. METHODS AND ANALYSIS: A three-group, double-blind,randomised, cross-over, placebo-controlled study of two pairs of gut microbiota transfer will be conducted in 99 patients with diarrhoeal or mixed type of IBS. Patients aged 18-65 will be randomised into three equally sized groups: group A will first receive two enemas of study microbiota mixture (deep-frozen stored stool microbiota mixed from eight healthy donors); after 8 weeks, they will receive two enemas with placebo (autoclaved microbiota mixture), whereas group B will first receive placebo, then microbiota mixture. Finally, group C will receive placebos only. The IBS Severity Symptom Score (IBS-SSS) questionnaires will be collected at baseline and then at weeks 3, 5, 8, 11, 13, 32. Faecal bacteriome will be profiled before and regularly after interventions using 16S rDNA next-generation sequencing. Food records, dietary questionnaires, anthropometry, bioimpedance, biochemistry and haematology workup will be obtained at study visits during the follow-up period. The primary outcome is the change in the IBS-SSS between the baseline and 4 weeks after the intervention for each patient compared with placebo. Secondary outcomes are IBS-SSS at 2 weeks after the intervention and 32 weeks compared with placebo and changes in the number of loose stools, Bristol stool scale, abdominal pain and bloating, anthropometric parameters, psychological evaluation and the gut microbiome composition. ETHICS AND DISSEMINATION: The study was approved by the Ethics Committee of Thomayer University Hospital, Czechia (G-18-26); study results will be published in peer-reviewed journals and presented at international conferences and patient group meetings. TRIAL REGISTRATION NUMBER: NCT04899869.


Subject(s)
Irritable Bowel Syndrome , Microbiota , Cross-Over Studies , Diarrhea/therapy , Dysbiosis/therapy , Fecal Microbiota Transplantation/methods , Humans , Irritable Bowel Syndrome/therapy , Randomized Controlled Trials as Topic
4.
Folia Microbiol (Praha) ; 67(6): 811-824, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-1943173

ABSTRACT

The unique functionality of Akkermansia muciniphila in gut microbiota indicates it to be an indispensable microbe for human welfare. The importance of A. muciniphila lies in its potential to convert mucin into beneficial by-products, regulate intestinal homeostasis and maintain gut barrier integrity. It is also known to competitively inhibit other mucin-degrading bacteria and improve metabolic functions and immunity responses in the host. It finds a pivotal perspective in various diseases and their treatment. It has future as a promising probiotic, disease biomarker and therapeutic agent for chronic diseases. Disease-associated dysbiosis of A. muciniphila in the gut microbiome makes it a potential candidate as a biomarker for some diseases and can provide future theranostics by suggesting ways of diagnosis for the patients and best treatment method based on the screening results. Manipulation of A. muciniphila in gut microbiome may help in developing a novel personalized therapeutic action and can be a suitable next generation medicine. However, the actual pathway governing A. muciniphila interaction with hosts remains to be investigated. Also, due to the limited availability of products containing A. muciniphila, it is not exploited to its full potential. The present review aims at highlighting the potential of A. muciniphila in mucin degradation, contribution towards the gut health and host immunity and management of metabolic diseases such as obesity and type 2 diabetes, and respiratory diseases such as cystic fibrosis and COVID-19.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Humans , Dysbiosis/therapy , Verrucomicrobia/metabolism , Mucins/metabolism , Mucus
5.
J Clin Gastroenterol ; 56(4): 285-298, 2022 04 01.
Article in English | MEDLINE | ID: covidwho-1865009

ABSTRACT

BACKGROUND: Gastrointestinal symptoms are common in Coronavirus Disease 2019 (COVID-19), related to infection of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) of intestinal cells through the angiotensin converting enzyme 2 (ACE2) receptor in the brush border. Also, patients are treated with multiple antibiotics. Therefore, an increase in gut dysbiosis and in the prevalence of Clostridium difficile infection (CDI) is expected in patients with COVID-19. METHODS: A PubMed search was conducted using the terms "gut microbiota," "gut mycobiota," "dysbiosis" AND "COVID-19"; "Clostridium difficile," "Clostridioides difficile" AND "COVID-19"; "probiotics," "bacteriotherapy AND COVID-19." Only case series, observational and experimental studies were included. RESULTS: A total of 384 papers were retrieved and 21 fulfilled selection criteria. Later, a new paper was identified, thus 22 papers were reviewed. Main findings: (1) gut bacterial dysbiosis has been found in fecal samples of COVID-19 patients, with enrichment of opportunistic organisms and decrease of beneficial commensals such as Faecalibacterium prausnitizii. Dysbiosis is related to inflammatory markers and illness severity. (2) There is evidence for abnormal gut barrier and bacterial translocation with a negative impact in the lungs. (3) Fungal dysbiosis correlating with pulmonary mycobiota, has also been found. (4) There is controversy in the CDI rates among COVID-19 patients versus controls and pandemic versus prepandemic era. (5) There is no available evidence yet to support bacteriotherapy in COVID-19. (6) Fecal microbiota transplantation (FMT) has been proposed for COVID-19, although there is no evidence to support it. Also, FMT can be safely used during the pandemic for CDI if strict screening protocols for donors and fecal product are implemented. CONCLUSIONS: In COVID-19 there is bacterial and fungal dysbiosis that correlates with systemic and pulmonary inflammation, and illness severity. Further investigations are warranted to determine the efficacy of bacteriotherapy and FMT for modulating gut dysbiosis in COVID-19.


Subject(s)
COVID-19 , Clostridioides difficile , Clostridium Infections , COVID-19/therapy , Clostridium Infections/therapy , Dysbiosis/microbiology , Dysbiosis/therapy , Fecal Microbiota Transplantation/methods , Humans , SARS-CoV-2
6.
Front Cell Infect Microbiol ; 12: 804644, 2022.
Article in English | MEDLINE | ID: covidwho-1753360

ABSTRACT

Introduction: The Coronavirus Disease 2019 (COVID-19) pandemic caused by Severe Acute Respiratory Coronavirus 2 (SARS-CoV-2) emerged in late December 2019. Considering the important role of gut microbiota in maturation, regulation, and induction of the immune system and subsequent inflammatory processes, it seems that evaluating the composition of gut microbiota in COVID-19 patients compared with healthy individuals may have potential value as a diagnostic and/or prognostic biomarker for the disease. Also, therapeutic interventions affecting gut microbial flora may open new horizons in the treatment of COVID-19 patients and accelerating their recovery. Methods: A systematic search was conducted for relevant studies published from December 2019 to December 2021 using Pubmed/Medline, Embase, and Scopus. Articles containing the following keywords in titles or abstracts were selected: "SARS-CoV-2" or "COVID-19" or "Coronavirus Disease 19" and "gastrointestinal microbes" or "dysbiosis" or "gut microbiota" or "gut bacteria" or "gut microbes" or "gastrointestinal microbiota". Results: Out of 1,668 studies, 22 articles fulfilled the inclusion criteria and a total of 1,255 confirmed COVID-19 patients were examined. All included studies showed a significant association between COVID-19 and gut microbiota dysbiosis. The most alteration in bacterial composition of COVID-19 patients was depletion in genera Ruminococcus, Alistipes, Eubacterium, Bifidobacterium, Faecalibacterium, Roseburia, Fusicathenibacter, and Blautia and enrichment of Eggerthella, Bacteroides, Actinomyces, Clostridium, Streptococcus, Rothia, and Collinsella. Also, some gut microbiome alterations were associated with COVID-19 severity and poor prognosis including the increment of Bacteroides, Parabacteroides, Clostridium, Bifidobacterium, Ruminococcus, Campylobacter, Rothia, Corynebacterium, Megasphaera, Enterococcus, and Aspergillus spp. and the decrement of Roseburia, Eubacterium, Lachnospira, Faecalibacterium, and the Firmicutes/Bacteroidetes ratio. Conclusion: Our study showed a significant change of gut microbiome composition in COVID-19 patients compared with healthy individuals. This great extent of impact has proposed the gut microbiota as a potential diagnostic, prognostic, and therapeutic strategy for COVID-19. There is much evidence about this issue, and it is expected to be increased in near future.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , COVID-19/diagnosis , COVID-19/therapy , Dysbiosis/diagnosis , Dysbiosis/therapy , Gastrointestinal Microbiome/physiology , Humans , Prognosis , SARS-CoV-2
7.
Curr Med Sci ; 41(6): 1087-1095, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1540258

ABSTRACT

OBJECTIVE: Coronavirus disease 2019 (COVID-19) is often accompanied by gastrointestinal symptoms, which are related to gut microbiota dysbiosis (GMD). Whether washed microbiota transplantation (WMT) is an effective treatment for COVID-19 patients suspected of having GMD by restoring the gut microbiota is unknown. This study is designed to explore the efficacy and safety of WMT in COVID-19 patients suspected of having GMD. METHODS: This is a randomized, multicenter, single-blind prospective study. COVID-19 patients suspected of having GMD will be randomly divided to receive routine treatment only or to receive routine treatment and WMT. The frequency of WMT will be once a day for three consecutive days. Laboratory and imaging examinations will be performed at admission, 1 and 2 weeks after treatment, and on the day of discharge. Then a telephone follow-up will be conducted at 1st week, 2nd week, and 6th month after discharge. The clinical efficacy and safety of WMT in COVD-19 patients suspected of having GMD and the effects of WMT on the organ function, homeostasis, inflammatory response, intestinal mucosal barrier function, and immunity of the patients will be evaluated. RESULTS: By following the proposed protocol, WMT is expected to be efficacious and safe for the treatment of COVID-19 patients suspected of having GMD, and the therapeutic effect is expected to be associated with improvement of the intestinal mucosal barrier function, inflammatory response, and immunity. CONCLUSION: The findings from this study may offer a new approach for the prevention and treatment of COVID-19 patients suspected of having GMD.


Subject(s)
COVID-19/microbiology , COVID-19/therapy , Dysbiosis/microbiology , Dysbiosis/therapy , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome , SARS-CoV-2 , Adult , Aged , COVID-19/complications , China , Clinical Protocols , Dysbiosis/etiology , Fecal Microbiota Transplantation/adverse effects , Female , Humans , Living Donors , Male , Middle Aged , Prospective Studies , Safety , Single-Blind Method , Treatment Outcome , Young Adult
8.
Nutr Health ; 28(2): 163-175, 2022 Jun.
Article in English | MEDLINE | ID: covidwho-1501915

ABSTRACT

Background: Despite strategies based on social distancing, the coronavirus disease 2019 (COVID-19) expands globally, and so far, many attempts have been made to achieve effective treatment for patients with COVID-19. This disease infects the lower respiratory tract and may lead to severe acute respiratory syndrome coronavirus (SARS-CoV). COVID-19 also can cause gastrointestinal infections. Therefore, COVID-19 patients with gastrointestinal symptoms are more likely to be complicated by SARS-CoV. In this disease, acquired immune responses are impaired, and uncontrolled inflammatory responses result in cytokine storms, leading to acute lung injury and thrombus formation. Probiotics are living microorganisms that contribute to the health of the host if administered in appropriate doses. Aim: This study aimed to provide evidence to show the importance of gut dysbiosis in viral disease, especially COVID-19. Therefore, we have focused on the impact of probiotics consumption on preventing severe symptoms of the disease. Methods: We have entirely searched SCOPUS, PubMed, and Google Scholar databases to collect evidence regarding the relationship between probiotics and viral infections to expand this relationship to the COVID-19. Results: It has been shown that probiotics directly counteract SARS-CoV in the gastrointestinal and respiratory tracts. Moreover, probiotics suppress severe immune responses and prevent cytokine storms to inhibit pathologic inflammatory conditions in the body via modulation of immune responses. Conclusion: According to available evidence based on their antiviral and respiratory activities, using probiotics might be an adjuvant therapy to reduce the burden and severity of this disease.


Subject(s)
COVID-19 , Probiotics , Cytokine Release Syndrome , Dysbiosis/therapy , Humans , Probiotics/therapeutic use , SARS-CoV-2
9.
Ann Rheum Dis ; 80(9): 1158-1167, 2021 09.
Article in English | MEDLINE | ID: covidwho-1356912

ABSTRACT

OBJECTIVES: Although causality remains to be established, targeting dysbiosis of the intestinal microbiota by faecal microbiota transplantation (FMT) has been proposed as a novel treatment for inflammatory diseases. In this exploratory, proof-of-concept study, we evaluated the safety and efficacy of FMT in psoriatic arthritis (PsA). METHODS: In this double-blind, parallel-group, placebo-controlled, superiority trial, we randomly allocated (1:1) adults with active peripheral PsA (≥3 swollen joints) despite ongoing treatment with methotrexate to one gastroscopic-guided FMT or sham transplantation into the duodenum. Safety was monitored throughout the trial. The primary efficacy endpoint was the proportion of participants experiencing treatment failure (ie, needing treatment intensification) through 26 weeks. Key secondary endpoints were change in Health Assessment Questionnaire Disability Index (HAQ-DI) and American College of Rheumatology (ACR20) response at week 26. RESULTS: Of 97 screened, 31 (32%) underwent randomisation (15 allocated to FMT) and 30 (97%) completed the 26-week clinical evaluation. No serious adverse events were observed. Treatment failure occurred more frequently in the FMT group than in the sham group (9 (60%) vs 3 (19%); risk ratio, 3.20; 95% CI 1.06 to 9.62; p=0.018). Improvement in HAQ-DI differed between groups (0.07 vs 0.30) by 0.23 points (95% CI 0.02 to 0.44; p=0.031) in favour of sham. There was no difference in the proportion of ACR20 responders between groups (7 of 15 (47%) vs 8 of 16 (50%)). CONCLUSIONS: In this first preliminary, interventional randomised controlled trial of FMT in immune-mediated arthritis, we did not observe any serious adverse events. Overall, FMT appeared to be inferior to sham in treating active peripheral PsA. TRIAL REGISTRATION NUMBER: NCT03058900.


Subject(s)
Arthritis, Psoriatic/therapy , Dysbiosis/therapy , Fecal Microbiota Transplantation/methods , Adult , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/microbiology , Female , Humans , Male , Methotrexate/therapeutic use , Middle Aged , Proof of Concept Study , Treatment Outcome
10.
Int J Food Sci Nutr ; 72(3): 293-299, 2021 May.
Article in English | MEDLINE | ID: covidwho-1169469

ABSTRACT

Coronavirus disease 2019 (COVID-19) has become pandemic very rapidly at the beginning of 2020. In the rush to possible therapeutic options, probiotics administration has been proposed mainly based on indirect observation. Some evidence of COVID-19 effects on intestinal microbiota dysbiosis has been shown and probiotics have been considered for their efficacy in the management of respiratory tract viral infections. These observations could be reinforced by the more and more evident existence of a lung-gut axis, suggesting the modulation of gut microbiota among the approaches to the COVID-19 prevention and treatment. As different possible roles of probiotics in this extremely severe illness have been contemplated, the aim of this work is to collect all the currently available information related to this topic, providing a starting point for future studies focussing on it.


Subject(s)
COVID-19/therapy , Dysbiosis/therapy , Gastrointestinal Microbiome , Probiotics/therapeutic use , COVID-19/complications , Dysbiosis/virology , Humans
11.
Curr Opin Allergy Clin Immunol ; 21(3): 245-251, 2021 06 01.
Article in English | MEDLINE | ID: covidwho-1153254

ABSTRACT

PURPOSE OF REVIEW: Data regarding the effects of coronavirus disease 2019 (COVID-19) on host-microbiome alteration and subsequent effects on susceptibility and clinical course of COVID-19, especially in atopic patients, are currently limited. Here, we review the studies regarding the microbiome of atopic patients with other respiratory infections and discuss the potential role of probiotics as therapeutic targets for COVID-19 to decrease its susceptibility and severity of COVID-19. RECENT FINDINGS: Respiratory tract virus infection affects the gut and airway microbiome structures and host's immune function. Diverse factors in atopic diseases affect the airway and gut microbiome structures, which are expected to negatively influence host health. However, response to respiratory virus infection in atopic hosts depends on the preexisting microbiome and immune responses. This may explain the inconclusiveness of the effects of COVID-19 on the susceptibility, morbidity, and mortality of patients with atopic diseases. Beneficial probiotics may be a therapeutic adjuvant in COVID-19 infection as the beneficial microbiome can decrease the viral load in the early phase of respiratory virus infection and improve the morbidity and mortality. SUMMARY: Application of probiotics can be a potential adjuvant treatment in respiratory virus infection to improve host immune responses and disturbed microbiome structures in atopic patients. Further related studies involving COVID-19 are warranted in near future.


Subject(s)
COVID-19 , Dysbiosis , Gastrointestinal Microbiome/immunology , Hypersensitivity , Pandemics , Probiotics/therapeutic use , SARS-CoV-2/immunology , COVID-19/epidemiology , COVID-19/immunology , COVID-19/microbiology , COVID-19/therapy , Dysbiosis/epidemiology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/therapy , Humans , Hypersensitivity/epidemiology , Hypersensitivity/immunology , Hypersensitivity/microbiology , Hypersensitivity/therapy
12.
Chemotherapy ; 66(1-2): 24-32, 2021.
Article in English | MEDLINE | ID: covidwho-1147336

ABSTRACT

Respiratory and gastrointestinal symptoms are the predominant clinical manifestations of the coronavirus disease 2019 (COVID-19). Infecting intestinal epithelial cells, the severe acute respiratory syndrome coronavirus-2 may impact on host's microbiota and gut inflammation. It is well established that an imbalanced intestinal microbiome can affect pulmonary function, modulating the host immune response ("gut-lung axis"). While effective vaccines and targeted drugs are being tested, alternative pathophysiology-based options to prevent and treat COVID-19 infection must be considered on top of the limited evidence-based therapy currently available. Addressing intestinal dysbiosis with a probiotic supplement may, therefore, be a sensible option to be evaluated, in addition to current best available medical treatments. Herein, we summed up pathophysiologic assumptions and current evidence regarding bacteriotherapy administration in preventing and treating COVID-19 pneumonia.


Subject(s)
COVID-19 , Dysbiosis , Gastrointestinal Microbiome/immunology , Probiotics/pharmacology , COVID-19/immunology , COVID-19/physiopathology , COVID-19/prevention & control , Dietary Supplements , Dysbiosis/therapy , Dysbiosis/virology , Humans , SARS-CoV-2
13.
J Med Case Rep ; 15(1): 60, 2021 Feb 08.
Article in English | MEDLINE | ID: covidwho-1069584

ABSTRACT

BACKGROUND: To investigate the potential beneficial effect of fecal microbiota transplantation (FMT) on gastrointestinal symptoms, gut dysbiosis and immune status in discharged COVID-19 patients. CASE PRESENTATION: A total of 11 COVID-19 patients were recruited in April, 2020, about one month on average after they were discharged from the hospital. All subjects received FMT for 4 consecutive days by oral capsule administrations with 10 capsules for each day. In total, 5 out of 11 patients reported to be suffered from gastrointestinal symptoms, which were improved after FMT. After FMT, alterations of B cells were observed, which was characterized as decreased naive B cell (P = 0.012) and increased memory B cells (P = 0.001) and non-switched B cells (P = 0.012).The microbial community richness indicated by operational taxonomic units number, observed species and Chao1 estimator was marginally increased after FMT. Gut microbiome composition of discharged COVID-19 patients differed from that of the general population at both phylum and genera level, which was characterized with a lower proportion of Firmicutes (41.0%) and Actinobacteria (4.0%), higher proportion of Bacteroidetes (42.9%) and Proteobacteria (9.2%). FMT can partially restore the gut dysbiosis by increasing the relative abundance of Actinobacteria (15.0%) and reducing Proteobacteria (2.8%) at the phylum level. At the genera level, Bifidobacterium and Faecalibacterium had significantly increased after FMT. CONCLUSIONS: After FMT, altered peripheral lymphocyte subset, restored gut microbiota and alleviated gastrointestinal disorders were observe, suggesting that FMT may serve as a potential therapeutic and rehabilitative intervention for the COVID-19.


Subject(s)
B-Lymphocyte Subsets , COVID-19/complications , Dysbiosis/therapy , Fecal Microbiota Transplantation , Gastrointestinal Diseases/therapy , Gastrointestinal Microbiome , Aged , Bacteroidetes , Bifidobacterium , COVID-19/immunology , Dysbiosis/microbiology , Faecalibacterium , Female , Gastrointestinal Diseases/microbiology , Humans , Male , Middle Aged , Patient Discharge , Proteobacteria , SARS-CoV-2 , Young Adult
14.
Virus Res ; 291: 198203, 2021 01 02.
Article in English | MEDLINE | ID: covidwho-885454

ABSTRACT

Infectious diseases related to viruses, as well as bacterial pathogens, abound in all parts of the world, burdening health and economy. Thus, there is a dire need to find new prevention and treatment strategies to improve clinical practices related to viral infections. Human gut contains trillions of bacteria which have regulatory roles in immune development, homeostasis, and body metabolism. Today, it is difficult to find any prominent viral infection that hasn't had any link with the human gut microbiota. In this opinion-based review article, I argued the significance of manipulating human gut microbiota as novel therapeutics through probiotics or FMT in alleviating complexities related to viral infections, and pinpointed bottlenecks involved in this research.


Subject(s)
Microbiota/genetics , Probiotics/therapeutic use , Virus Diseases/therapy , Animals , Dysbiosis/etiology , Dysbiosis/therapy , Dysbiosis/virology , Gastrointestinal Microbiome , Humans , Mice , Microbiota/immunology , Virus Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL