Review Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 7, 2022; 28(25): 2802-2822
Published online Jul 7, 2022. doi: 10.3748/wjg.v28.i25.2802
Altered gut microbiota patterns in COVID-19: Markers for inflammation and disease severity
Chiranjib Chakraborty, School of Life Science and Biotechnology, Adamas University, Kolkata 700126, India
Ashish Ranjan Sharma, Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University, Chuncheon-si 24252, South Korea
Manojit Bhattacharya, Department of Zoology, Fakir Mohan University, Balasore 756020, India
Kuldeep Dhama, Division of Pathology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Bareilly 243122, Uttar Pradesh, India
Sang-Soo Lee, Institute for Skeletal Aging & Orthopedic Surgery, Hallym University, Chuncheon-si 24252, South Korea
ORCID number: Chiranjib Chakraborty (0000-0002-3958-239X); Ashish Ranjan Sharma (0000-0003-3973-6755); Manojit Bhattacharya (0000-0001-9669-1835); Kuldeep Dhama (0000-0001-7469-4752.); Sang-Soo Lee (0000-0001-5074-7581).
Author contributions: Chakraborty C, Sharma AR, and Bhattacharya M contributed equally, Chakraborty C designed the research study and wrote the main manuscript draft; Sharma AR reviewed and edited the manuscript; Bhattacharya M developed the figures and tables; Dhama KD performed the English editing and validation; Lee SS did the funding acquisition; all authors have read and approved the final manuscript.
Supported by the Hallym University Research Fund and the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1C1C1008694 & NRF-2020R1I1A3074575).
Conflict-of-interest statement: There are no conflicts of interest to report.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Sang-Soo Lee, MD, PhD, Full Professor, Institute for Skeletal Aging & Orthopedic Surgery, Hallym University, Chuncheon-si 24252, South Korea. 123sslee@gmail.com
Received: January 12, 2022
Peer-review started: January 12, 2022
First decision: March 10, 2022
Revised: March 19, 2022
Accepted: May 13, 2022
Article in press: May 13, 2022
Published online: July 7, 2022

Abstract

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to a severe respiratory illness and alters the gut microbiota, which dynamically interacts with the human immune system. Microbiota alterations include decreased levels of beneficial bacteria and augmentation of opportunistic pathogens. Here, we describe critical factors affecting the microbiota in coronavirus disease 2019 (COVID-19) patients. These include, such as gut microbiota imbalance and gastrointestinal symptoms, the pattern of altered gut microbiota composition in COVID-19 patients, and crosstalk between the microbiome and the gut-lung axis/gut-brain-lung axis. Moreover, we have illustrated the hypoxia state in COVID-19 associated gut microbiota alteration. The role of ACE2 in the digestive system, and control of its expression using the gut microbiota is discussed, highlighting the interactions between the lungs, the gut, and the brain during COVID-19 infection. Similarly, we address the gut microbiota in elderly or co-morbid patients as well as gut microbiota dysbiosis of in severe COVID-19. Several clinical trials to understand the role of probiotics in COVID-19 patients are listed in this review. Augmented inflammation is one of the major driving forces for COVID-19 symptoms and gut microbiome disruption and is associated with disease severity. However, understanding the role of the gut microbiota in immune modulation during SARS-CoV-2 infection may help improve therapeutic strategies for COVID-19 treatment.

Key Words: COVID-19, Inflammation, Gut microbiota, Therapeutic

Core Tip: The gut microbiota of coronavirus disease 2019 (COVID-19) patients is altered compared to that of healthy individuals, with a reduction in the count of beneficial bacteria and an increase in the count of opportunistic fungi. In this review, we elucidate the components governing immune modulation. Additionally, we explore the effect of changes in the microbial ecosystem in COVID-19 patients, with an aim to help develop precise therapeutics and expand our knowledge regarding the pattern of changes in the gut microbiota of COVID-19 patients.



INTRODUCTION

The coronavirus disease 2019 (COVID-19) pandemic has stimulated research on several medical conditions and on individual patient variations during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection to unfold underlying disease mechanisms. Scientists have determined the inflammatory response and cellular injury mediated by acute SARS-CoV-2 infection. Moreover, several studies have revealed the involvement of the gastrointestinal (GI) tract and its associated gut microbiome during COVID-19, motivating research in this field. Increasing evidence has surfaced confirming the association of the GI tract and COVID-19, including[1,2] a severe state of gut dysbiosis in COVID-19 patients[3,4]. Similarly, GI symptoms such as vomiting, abdominal pain, and diarrhea have been noted in many COVID-19 patients[5-7]. Moreover, high expression of ACE2 receptor was reported in epithelial cells of the GI tract[8]. SARS-CoV-2 RNA has been identified in rectal and anal swabs, as well as stool specimens[7,9,10]. Finally, liver damage, loss of appetite, and irritable inflammatory diseases have been reported as post-COVID-19 illnesses[11]. These all data strongly indicate a correlation between the GI including the gut microbiome, and COVID-19.

The gut microbiota plays an important role in controlling gut health and acts as a health modulator (Figure 1)[12] aidings in different metabolic activities and extensively impacting health and disease[13,14]. Ongoing research aims to better understand the gut microbiota and provide insights into the mechanistic conditions required to implement normal health functions. The gut microbiota controls specific functions in the host, such as drug and xenobiotic metabolism and nutrient metabolism[15]. Simultaneously, it helps maintain the structural integrity of the gut mucosal barrier, protects against pathogens, and regulates immunomodulation, as well as health and disease conditions[16,17]. Several other studies suggest a possible link between COVID-19 and gut microbiota composition[18,19]. Additionally, an association has been shown between altered gut microbial composition and increased risk factors in COVID-19 patients (Figure 1)[20,21].

Figure 1
Figure 1 The schematic diagram shows normal healthy gut and the incidence in gut microbiota and gut virome in coronavirus disease 2019 patients. COVID-19: Coronavirus disease 2019.

Inflammation is a major risk factor in COVID-19 patients[22-24]. During uncontrolled inflammation, abnormal levels of cytokines such as interleukin-1 beta (IL-1β), IL-6, IL-8, IL-10, IL-12, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ) are found in the patients[23,25-27]. Certain abnormal levels of cytokines are substantial related to the severity of COVID-19 and are probably responsible for the “cytokine storm” syndrome manifested during the disease[28-30]. Research has correlated the inflammation during COVID-19 with GI and hepatic manifestations of the disease[31].

Interactions between the gut microbiota and the lungs, known as the gut-lung axis, have sparked interest for gastroenterology studies focusing on COVID-19 as these interactions affect disease severity. Changes in the gut microbiome certainly affect homeostasis and may lead to increased infections[32,33]. Similarly, in addition to the gut, COVID-19 can also have a detrimental effect on the central nervous system (CNS) and the blood-brain barrier (BBB) and disrupt the gut-brain-lung axis. Studies have explored therapeutic options (nicotinic cholinergic agonists and vagus nerve stimulation) to minimize the damage caused to this axis[34].

Research is necessary to understand how the gut microbiome is altered during COVID-19 infection and the factors that influence the microbiome during mild to moderate and severe disease. Studies have been conducted to understand the GI symptoms during COVID-19 and to detect viral shedding using the fecal matter of SARS-CoV-2 patients. The gut microbiota of COVID-19 patients has been mapped to obtain evidence regarding inflammation, disease severity, and therapeutic development.

Using these studies, we explore the following critical factors: (1) The gut microbiota imbalance and GI symptoms in COVID-19 patients; (2) fecal viral shedding in COVID-19 patients and restoration of the gut microbiota; (3) the pattern of altered gut microbiota composition in COVID-19 patients; (4) alterations in gut biosynthesis during COVID-19 infection; (5) the role of ACE2 in the digestive system and the gut microbiome; (6) crosstalk between the microbiome and the gut-lung axis during COVID-19 infection; (7) crosstalk between the microbiome and the gut-brain-lung axis during COVID-19 infection; and (8) hypoxia during COVID-19 associated with altered gut microbiota. We also discuss how immune responses and inflammation due to COVID-19 drive the changes in the microbial ecosystem and summarize therapeutic options currently in development.

GI SYMPTOMS IN COVID-19 PATIENTS

Along with respiratory symptoms and fever, GI symptoms have also been observed in COVID-19 patients (Table 1). A study by Redd et al[35] reported abdominal pain (14.5%), nausea (26.4%), diarrhea (33.7%), and vomiting (15.4%) in patients from the United States. Three hundred and eighteen hospitalized COVID-19 patients were evaluated to understand their symptoms. In another study with 204 COVID-19 patients, 50.5% (103 patients) exhibited GI symptoms. Among these 103 patients, 78.6% showed a lack of appetite, 34% had diarrhea, 3.9% vomited, and 1.9% complained of abdominal pain. The authors correlated patients describing GI symptoms with other measurements such as prothrombin time, monocyte count, and liver enzyme levels. Patients with GI symptoms had elevated mean liver enzyme levels, extended prothrombin times, and lower monocyte counts[36]. In a much larger cohort study involving 1099 COVID-19 patients from 552 different hospitals spread to over 30 provinces, only 3.8% of patients experienced diarrhea. The authors concluded that fever and cough are common symptoms, unlike diarrhea, among the COVID-19 patient population[37].

Table 1 Different gastrointestinal symptoms in coronavirus disease 2019 patients.
SI. No.
Total number of human subjects involved in study
Demographics of the study populations
Vomiting
Diarrhea
Nausea
Remarks/study summary
Ref.           
1191Adults (46-67 years) hospitalised, Chinese peoples, 91 patients having comorbidity7 (3.7%)9 (4.7%)7 (3.7%)Identification of several risk factors and a detailed clinical course of illness for mortality of COVID-19 patients[121]
2171Minor aged (1 d-15 years, hospitalised, Chinese children, no such comorbidity11 (6.4%)15 (8.8%)NAReport of a spectrum of illness from children infected with SARS-CoV-2 virus[122]
31099Median age group (35-58 years), hospitalised, Chinese patients without any comorbidity55 (5.0%)42 (3.8%)55 (5.0%)Identification and definition of clinical characteristics and disease severity of hospitalized COVID-19 patients[37]
4140Adults (25-87 year), hospitalised Chinese patients with high comorbidity7 (5.0%)18 (12.9%)24 (17.3%)Report on hospitalized patients having COVID-19 with abnormal clinical manifestations (fever, fatigue, gastrointestinal symptoms, allergy)[123]
573Adults hospitalised Chinese patients, comorbidity reportedNA26 (35.6%)NAClinical significance of SARS-CoV-2 by examining viral RNA in feces of COVID-19 patients during hospitalizations[124]
652Adults (mean age 59.7 year), critically ill ICU- admitted Chinese patients, comorbidity reported2 (3.8%)NANARetrospective, single-centered, observational study on critically ill, ICU-admitted adult COVID-19 patients[125]
7138Adult (median age 56 years), hospitalised Chinese patients with comorbidities5 (3.6%)14 (10.1%)14 (10.1%)Clinical characteristics of COVID-19 patients in hospitalized conditions[126]
841Middle age group (41-58 years) hospitalised Chinese patients with comorbiditiesNA1 (2.6%)NAEpidemiological, laboratory, clinical, and radiological features and treatment with clinical outcomes of hospitalized COVID-19 patients[46]
962Studied patients (median age 41 years) were hospitalised, Chinese ethnicity and comorbidity reportedNA3 (4.8%)NAMost common symptoms at onset of illness with clinical data in confirmed COVID-19 patients[127]
10137Studied patients ( mean age 57-55) ware Chinese and hospitalised, comorbidity was also notedNA11 (8%)NAInvestigation of epidemiological history, clinical characteristics, treatment, and prognosis of COVID-19 patients[128]
1181Chinese patients (mean age was 49.5 years), hospitalised with high comorbidities4 (4.9%)3 (3.7%)NAReport of confirmed COVID-19 patients with chest computer tomography imaging anomalies[129]
1299Hospitalised, Chinese patients (average age of the patients was 55.5 years), comorbidity was reported1 (1%)2 (2.0%)1 (1%)Inclusive exploration of epidemiology and clinical features of COVID-19 patients[130]

These findings suggest that the virus might be present for a period in the GI tract, which may cause a GI infection (Figure 2). Importantly, fecal viral shedding was noted after clearing SARS-CoV-2 from the respiratory tract, suggesting that the virus can persist for a long time in the GI tract, especially in patients who manifest GI symptoms. During COVID-19 infection, gut microbiota composition is altered, possibly explaining the GI imbalance and manifestations of the different GI symptoms such as abdominal pain, nausea, vomiting, and diarrhea, as described above. This change in the gut microbiota includes reduced levels of commensals microbes and is observed in patient samples even after 30 d of disease remission[38-40]. Additional studies addressed the imbalance of the gut microbiota and its association with different GI-related aspects of COVID-19[41]. The gut microbiota population in COVID-19 patients with low to moderate GI symptoms should also be analyzed. Evaluating these diverse patient populations will enable a thorough description of this phenomenon.

Figure 2
Figure 2 The schematic diagram illustrates the severe acute respiratory syndrome coronavirus 2 entry in the body, causes of gut microbiota imbalance which assists in manifesting the gastrointestinal symptoms in coronavirus disease 2019 patients. GI: Gastrointestinal; SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2.
FECAL VIRAL SHEDDING IN COVID-19 PATIENTS

Table 2 lists various cohort studies reporting fecal viral shedding by COVID-19 patients and detecting SARS-CoV-2 RNA in the fecal matter[42,43]. SARS-CoV-2 RNA-positive fecal matter was detected in 66.67% of COVID-19 patients (42 patients) in China[43]. Researchers attempted to evaluate the viral shedding period in stool samples, and noted viral shedding in asymptomatic patients. For example, SARS-CoV-2 RNA was detected from a stool sample of an asymptomatic child 17 d after viral exposure[9].

Table 2 Fecal viral shedding in coronavirus disease 2019 patients.
SI. No.
Total number of human subjects in study
Demographics of the study populations
Gastrointestinal symptoms
Confirmed cases of fecal shedding
Remarks/study summary
Ref.           
1205Patients (mean age of 44 years) were hospitalised, Chinese without any comorbiditiesNo symptoms44Evidence-based study for gastrointestinal infection of SARS-CoV-2 virus and its possible fecal-oral transmission route in humans[131]
273Different age group (10 mo to 78 years old), hospitalised Chinese patients without report any comorbiditiesGastrointestinal bleeding, diarrhea39Description of epidemiological and clinical characteristics of COVID-19 patients[124]
310Chinese patients have aged 19-40 years, hospitalised and no such comorbidity was reportedHemoptysis, diarrhea, cough8Report of median aged COVID-19 confirmed patients in ICU[127]
414Patients (18-87 years) were hospitalized, Chinese individuals without any comorbiditiesNo symptoms5Retrospective analysis of laboratory-confirmed COVID-19 cases in hospitalized conditions[132]
566Chinese patients (median age of 44 years) were hospitalised, comorbidity was not reportedNo symptoms11Viral RNA detection was performed from throat swabs, stool, urine, and serum samples in different clinical conditions in COVID-19 patients[133]
618Adults patients (median age, 47 years) from Singapore were hospitalised and comorbidities was notedNo symptoms4COVID-19 patient case series using clinical, laboratory, and radiological data[134]
774Studied paients belonged from China and were hospitalised with comorbiditiesNo symptoms41Analysis of respiratory and fecal samples to determine clinical symptoms and medical treatments of COVID-19 patients[135]
89Adults Chinese patients were hospitalised without any comorbiditiesDiarrhea and urinary irritation2Detection of SARS-CoV-2 RNA in urine and blood samples, and anal, oropharyngeal swabs of confirmed COVID-19 patients[136]

Certain studies have reported that virus separation from stool samples is difficult. For example, Wölfel et al[44] detected viral RNA in stool samples but attempts to isolate the virus were unsuccessful, most likely due to the mild nature of the infection. A viral load below 106 copies per milliliter often hampers viral isolation[36]. The viral load also varies widely from one sample to another, including stool, serum, and respiratory samples[44-46]. However, understanding the correlation between the altered gut microbiota and the viral load in patient samples is essential for advancing therapeutic strategies centered around restoring the microbiota.

Additionally, efforts should focus on determining the possible correlation between fecal viral shedding and altered gut microbiota composition at different stages the infection, i.e., mild to moderate or severe COVID-19.

ALTERED GUT MICROBIOTA COMPOSITION IN COVID-19 PATIENTS

SARS-CoV-2 infections have led to changes in the ecology of the gut microbiota in patients (compared to that seen in controls). These changes are influenced by the immune responses elicited during COVID-19 (Table 3). Different studies have revealed the growth of unusual microorganisms and depletion of common gut microbes (bacterial, viral, and fungal populations) in COVID-19 patients (Figure 3).

Figure 3
Figure 3 The diagram illustrates increased or decreased gut microbiota in coronavirus disease 2019 patients, including bacterial, viral, and fungal populations. COVID-19: Coronavirus disease 2019.
Table 3 Analysis of gut microbiota in coronavirus disease 2019 patients in different cohorts.
SI. No.
Cohort composition
No of Patients
Demographics of the study populations
Country
Significant gut microbiota found
Study conclusion
Reference
1A pilot study with 15 healthy individuals (controls) and 15 patients with COVID-19 15Study performed with hospitalised patients (median age 55 years), Chinese ethnicity and comorbidities were reportedHong KongAbundance of Clostridium hathewayi, Clostridium ramosum, Coprobacillus, which are correlated with COVID-19 severityChange in the fecal microbiome of COVID-19 patients during hospitalization, compared to healthy individuals (controls)[48]
2The two-hospitals cohort, serial stool samples collected from 27 COVID-19 patients among 10027Adults hospitalised Chinese patients, comorbidities were notedHong KongFaecalibacterium prausnitzii, Eubacteriumrectale and bifidobacteriaGut microbiome involved in COVID-19 severity[38]
3United States cohort (majority African American)50Studied patients (mean age 62.3 years) were hospitalised with comorbidities, American ethnicityUnited StatesSome of the significant genera (Corynebacterium Peptoniphilus, Campylobacter, etc.) No significant associations found between the composition microbiome and disease severity from COVID-19 patient gut microbiota[50]
4The study used 53 COVID-19 patients and 76 healthy individuals. 81 fecal samples collected during hospitalization53Adults Chinase hospitalised patients, no such comorbidities were notedChinaElevated gut microbes such as Rothia mucilaginosa, Granulicatella spp, etc.COVID-19 infection linked with change of the microbiome in COVID-19 patients[137]
515 patients Cohort15Study performed adults hospitalised patients with comorbidities, Chinese ethnicityHong KongElevated bacterial species Collinsella tanakaei, Collinsella aerofaciens, Morganella morganii, Streptococcus infantisThe study found fecal viral (SARS-CoV-2) activity[54]
6Two-hospital cohort with a total of 100 patients. Stool samples collected from 27 patients27Hospitalised adults patients were from China, comorbidities were notedHong KongSeveral gut microbiota such as Faecalibacterium prausnitzii, Eubacterium rectale, and bifidobacteriaGut microbiota associated disease severity and inflammation in COVID-19 patients [38]
798 COVID-19 patients (3 asymptomatic, 34 moderate, 53 mild, 3 critical, 5 severe), serial fecal samples collected from 37 COVID-19 patients37Adults (mean age 37 years) patients, hospitalised condition from Chinese ethnicity, comorbidities were reportedHong KongA total of 10 virus species in fecal matter (9 DNA virus species and 1 RNA virus, pepper chlorotic spot virus)Analysis of gut virome (RNA and DNA virome) in COVID-19 patients[47]
8Study of fecal samples from 30 COVID-19 patients30Patients (mean age 46 years) were hospitalised from Chinese groups, comorbidities were notedHong KongIncreased proportions of fungal pathogens (Candida albicans, Candida auris, Aspergillus flavus, Aspergillus niger) in fecal samplesAnalysis of fecal fungal microbiome of COVID-19 patients[48]

To understand the severity of disease in COVID-19 patients, the gut microbiota composition of 100 COVID-19 patients was analyzed in two hospital cohorts. Stool samples were collected from 27 of the 100 patients. The gut microbiome compositions were characterized using total DNA extracted from stool samples. The authors demonstrated that the number of gut commensals and Bifidobacteria was low and correlated with several factors of disease severity, such as high concentrations of inflammatory cytokines and C-reactive protein (CRP). These data suggests that the composition of the microbiota is associated with disease severity[38]. Another study carried out RNA and DNA profiling by sequencing of the virome using fecal matter from COVID-19 patients. The fecal matter of 98 COVID-19 patients was analyzed to understand COVID-19 severity and its association with the gut virome. The study showed that COVID-19 severity is inversely correlated with gut viruses, and older patients are more prone to severe COVID-19 outcomes[47]. Alterations in fungal microbiomes during COVID-19 have also been investigated. Analysis of the fecal mycobiome using the deep shotgun method showed heterogeneous microbial profiles, with enrichment of fungal genera such as Aspergillus and Candida. Two species of Aspergillus (Aspergillus flavus and Aspergillus niger) were identified in fecal samples after clearance of SARS-CoV-2 from nasopharyngeal samples[48]. Additionally, there is evidence of abundant symbionts among COVID-19 patients including Clostridium ramosum, Coprobacillus, and Clostridium hathewayi, which directly correlated with disease severity. Conversely, Faecalibacterium prausnitzii, which was also abundant among the patients, was inversely correlated with disease severity[49].

Similarly, in a study by Yeoh et al[38], stool samples from 27 patients were correlated with blood markers and inflammatory cytokines. The study concluded that the scale of COVID-19 severity might be associated with the gut microbiome and linked it to COVID-19 inflammation[46]. In another study containing a greater number of African Americans, enriched genera (Campylobacter, Corynebacterium, and Peptoniphilus) were mapped in the COVID-19 patient population, the gut microbial composition was markedly different between positive and negative samples. However, the study did not identify any considerable association between COVID-19 severity and microbiome composition[50].

Certain studies even noted a reduction in fiber-utilizing bacteria such as Prevotella, Bacteroides plebius, and Faecalibacterium prausnitzii (F. prausnitzii), and a low Firmicute/Bacteroidetes ratio[51]. Poor outcomes were noted in special populations, such as hypertensive, diabetic, and elderly patients[52,53]. Research is still underway to ascertain the different types of gut microbial populations (pro-inflammatory, opportunistic, beneficial, or anti-inflammatory) present depending on COVID-19 severity (Figure 4).

Figure 4
Figure 4 The diagram illustrates different types of mapped gut microbiota in coronavirus disease 2019 patients. Pro-inflammatory microbiota, opportunistic microbiota, the microbiome in severe coronavirus disease 2019 (COVID-19) patients, and the microbiome in low to moderate COVID-19 patients, anti-inflammatory microbiota, and beneficial microbiota. COVID-19: Coronavirus disease 2019.

These studies help us understand how gut microbiota composition affects patients with moderate to severe COVID-19 and how gut microbiota diversity might alter immunity in COVID-19 patients.

ALTERATIONS IN THE BIOSYNTHESIS OF BIOLOGICAL COMPOUNDS IN THE GUT DURING COVID-19 INFECTION

Other than compositional changes in gut microbiota, functional changes during SARS-CoV-2 infection were observed in some patients. The gut microbiota aids in different biosynthetic pathways, such as amino acid biosynthesis, carbohydrate metabolism, nucleotide de novo biosynthesis, and glycolysis. This might be due to the abundance of bacterial components such as Collinsella tanakaei, Streptococcus infantis, Morganella morganii, and Collinsella aerofaciens, etc. Apart from these microbes, many short-chain fatty acid (SCFA) synthesis bacteria, such as Lachnospiraceae bacteria, Bacteroides stercoris, Alistipes onderdonkii, and Parabacteroides merdae were present in COVID-19 samples with mild symptoms and in non-COVID-19 samples[54]. In a study using non-human primate models, β diversity analysis and 16S rRNA gene profiling were carried out to understand the gut microbiota composition during SARS-CoV-2 infection. The study revealed substantial changes in the gut microbiota composition and metabolism and a reduction in the concentration of SCFAs as well as a difference in the concentrations of bile acids. The study also found alterations in tryptophan metabolites during SARS-CoV-2 infection in the animal models[55].

Shotgun metagenomic sequencing using fecal samples has also been performed to profile the gut microbiome in SARS-CoV-2 infected patients. Researchers observed prolonged impairment of L-isoleucine biosynthesis and SCFAs due to alterations in the gut microbiome of patients with COVID-19[56].

ROLE OF ACE2 IN THE DIGESTIVE SYSTEM AND THE GUT MICROBIOME

The ACE2 (angiotensin-converting enzyme 2) receptor acts as a binding site by which SARS-CoV-2 enters host cells[57,58]. A higher expression of ACE2 in the cell favors SARS-CoV-2 infection. Despite this, ACE2 deficiency can play a vital role in SARS-CoV-2 infection[59]. Increased ACE2 expression is found in the epithelial cells of the respiratory tract (nasal mucosa, nasopharynx, and lungs), in different parts of the intestine, and in different types of epithelial cells, including nasal, corneal, and intestinal epithelial cells in humans[60]. In addition, this protein is expressed in different parts of the digestive system, such as the small intestine, stomach, colon, and liver[61]. However, ACE2 expression is controlled by distinct microbial communities in several body tissues. Mouse model studies suggest an association between certain microbial communities and overexpression of ACE2. This overexpression may prevent detrimental changes in hypoxia-induced gut pathophysiology and pulmonary pathophysiology[62]. ACE2 expression is controlled in the GI and respiratory tract[63]. Additionally, it can also be controlled by some bacterial species from important phyla. Downregulation of ACE2 expression was associated with the Bacteroidetes phylum. Among all species of this phylum, Bacteroides dorei has been shown to inhibit ACE2 expression in the colon, whereas the Firmicutes phylum plays a variable role in its modulation[20,49,64]. These findings are supported by other studies describing the modulation of ACE2 expression in the gut by the microbiota[65,66].

GUT-LUNG AXIS CROSSTALK DURING COVID-19 INFECTION

Several reports indicate that manipulation of the gut microbiota may be used to treat pulmonary diseases[67]. Therefore, the gut-lung axis crosstalk can help to elucidate these respiratory and digestive system interactions (Figure 5). Dysbiosis occurs when there are detrimental changes in the microbial composition of the gut or respiratory tract. It often leads to altered immune responses and the development of diseases, such as COVID-19. Nonetheless, of gut dysbiosis can be manipulated for treatment purposes[32,67-69]. Studies suggest that SARS-CoV-2 from the lungs travels to the gut via the lymphatic system leading to disrupted gut permeability[70,71]. Furthermore, the extent of dysbiosis is associated with COVID-19 severity[4,72]. Therefore, understanding the crosstalk between the microbiome and the gut-lung axis during COVID-19 infection may provide therapeutic approaches.

Figure 5
Figure 5 The diagram points out the normal gut and its microbial association. The figure also illustrates the crosstalk between the microbiome and gut-lung axis. SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2.
GUT-BRAIN-LUNG AXIS CROSSTALK DURING COVID-19 INFECTION

Like the gut-lung axis, crosstalk between the microbiome and the gut-brain axis has been recognized and remains the topic[73-75]. Several studies have illustrated the role of the microbiome-gut-brain axis in different neurological disorders[76,77].

The interaction between the brain and the gut (also called the gut-brain axis) is bidirectional, with several pathways involved, including bacterial metabolites, neuroanatomical communications, neurotransmitters, and hormones[78]. The vagus nerve is primarily involved in such communication, and these molecules (neurotransmitters/hormones) are produced in the GI tract. During communication between neurotransmitters and hormones, they might interact with the receptors on the vagus nerve, relaying information to the brain[78-81]. Many hormones can cross the BBB and affect the CNS directly. Additionally, neuroendocrine pathways which operate via the hypothalamic-pituitary-adrenal (HPA) axis associated with stress also affect the BBB. The stress-HPA axis is associated with the release of glucocorticoids such as cortisol from the adrenal cortex. Cortisol, is associated with augmented intestinal permeability and GI motility, affecting the gut microbiota[78,82-84]. The stress-HPA axis may also lead to inflammation and bacteria-derived impaired metabolite production, especially SCFAs[78,84]. Therefore, a thorough understanding of the gut-brain axis can help the development of therapeutic approaches via modulation of the gut microbial composition.

The gut microbiota might play a distinct role in controlling the host immune system, and research is underway to uncover more in this field[85,86]. The involvement of the lungs (gut-brain-lung axis) occurs when inflammation and neurodegeneration in the brain stem due to COVID-19 prevent cranial nerve signaling, disrupting anti-inflammatory pathways and normal respiratory and GI functions. Recently, the lungs have been associated in the crosstalk among the microbiota-gut-brain axis components, and this axis was also noted during COVID-19 (Figure 6)[34,78]. Moreover, in COVID-19 patients, alterations in the gut microbiota have been shown to reduce live microbes (Bifidobacterium and Lactobacillus) during intestinal microbial dysbiosis[87].

Figure 6
Figure 6 The diagram describes the normal gut and its microbial association. The figure also illustrates the crosstalk between the microbiome and gut-brain-lung axis. BDNF: Brain-derived neurotrophic factor; HPA: Hypothalamic-pituitary-adrenal; SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2.

The microbial translocation to the gut and its subsequent damage may play a vital role in inferior clinical outcomes for the disease. The gut-brain-lung axis during COVID-19 infection can also offer clues indicate viable directions for therapeutic development[34].

HYPOXIA IN COVID-19 AND GUT MICROBIOTA

Abnormal cytokine release (cytokine storms) and inflammatory responses may be associated with hypoxia during severe COVID-19. Viral replication in the lungs leads to a cytokine storm, destroying normal lung function and causing hypoxemia, i.e., low oxygen levels in the blood. Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that regulates cellular functions such as cell proliferation and angiogenesis. In hypoxic conditions, HIF-1α binds to the hypoxemic response element and induces the production of cytokines such as IL-6 and TNF-α, leading to hypoxia[88]. There are other collective causes of hypoxia, including pulmonary infiltration and thrombosis. The COVID-19 virus induces pneumonia that causes atelectasis (collapsing of air sacs), leading to low oxygen levels in the body[89]. Additionally, COVID-19 leads to mitochondrial damage, production of reactive oxygen species, and subsequently HIF-1α, further promoting viral infections and inflammation[90].

As part of its normal metabolic functions, the gut microbiota produces neurotropic metabolites, neurotransmitters, peptides, and SCFA, whose levels are disrupted due to COVID-19. SCFA such as butyrate confer neuroprotection. Modulation of gut microbes (responsible for such metabolite production) by SARS-CoV-2 alters hypoxia-sensing, negatively impacting the CNS[91]. Therefore, an association between gut microbiota and hypoxia in COVID-19 patients can be speculated, and may be linked to the CNS (Figure 7).

Figure 7
Figure 7 The figure illustrates an association between gut microbiota and hypoxia in coronavirus disease 2019 patients, and it is connected with central nervous system. SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2.
ALTERATION OF GUT MICROBIOTA IN COVID-19: EVIDENCE FOR INFLAMMATION OR DIEASE, SEVERITY?

Under normal conditions, colonization of the normal microbiota in the gut causes resistance to pathogen[92,93]. Much of the normal gut microbiota belongs to Clostridia., which produces butyric acid. This SCFA is produced during dietary fiber fermentation along with acetic acid and propionic acid, which play a critical role in gut health (Figure 8A)[94,95]. Butyric acid helps in maintain the integrity of the gut barrier by providing a vital energy resource for colonocytes. This SCFA also hinders histone deacetylase activity and inhibits the activation of the nuclear factor (NF)-κB signaling pathway activation. This phenomenon may activate the G protein-coupled receptor pair (GPR41 /GPR43). These events help exert an anti-inflammatory response in normal gut health and stimulate regulatory T cells (Treg cells)[96-100]. Treg cells play a central role in suppressing inflammatory responses[97,101]. However, in COVID-19 patients, typical microbiota dysbiosis causes an imbalance in all these events.

Figure 8
Figure 8 The figure illustrates normal gut microbiota and immunological consequences, and coronavirus disease 2019 related altered gut microbiota associated inflammation. A: Normal gut microbiota and immunological consequences for healthy gut; B: Coronavirus disease 2019 (COVID-19) related altered gut microbiota associated inflammation. The inflammatory condition in COVID-19 patients causes the abnormal release of different cytokines, such as interleukin-1 beta (IL-1β), IL-6, IL-8, IL-10, IL-12, granulocyte-macrophage colony-stimulating factor, tumor necrosis factor-alpha, and interferon-gamma. PSA: Polysaccharide A; SCFA: Short-chain fatty acid; IL-1β: Interleukin-1 beta; IL-6: Interleukin-6; GMCSF: Granulocyte-macrophage colony-stimulating factor; TNF-α: Tumor necrosis factor-alpha; IFN-γ: Interferon-gamma.

There is a distinct connection between dysbiosis of the gut microbiota and hyper-inflammatory responses, especially cytokine release, in some COVID-19 patients[102] (Figure 8). Researchers noted that gut microbiota composition is related to the COVID-19 severity of and observed an association between altered cytokine levels and gut microbiota composition[38]. Cytokines/inflammatory factors, such as IL-1β, IL-6, and TNF-α, are usually associated with inflammation during disease[103]. In the case of severe COVID-19, the levels of certain cytokines, such as IL-6, IL-10, TNF-α, and IFN-are raised abnormally, and in some cases, cytokine storms are observed (Figure 8B)[23]. In pilot study, the quality of gut microbial composition was associated with the severity COVID-19 in 15 patients at the time of hospitalization in Hong Kong. The study showed an abundance of microbes such as Clostridium hathewayi, Clostridium ramosum, and Coprobacillus in COVID-19 patients. Moreover, an anti-inflammatory bacteria, Faecalibacterium prausnitzii, was be inversely correlated with disease severity[49].

Nonetheless, more detailed studies are needed to understand the impaired gut health during COVID-19, especially in extreme forms of the disease. Another study confirmed microbiota dysbiosis in COVID-19 patients. This study found differential bacterial populations with a decrease in F. prausnitzii and Clostridium spp and an association of IL-21 in mild to severe COVID-19 patients[51].

A gut microbiota richness analysis in COVID-19 patients was conducted over through a six-month evaluation using 16S rDNA sequencing. This study showed that, patients with decreased post-convalescence richness in bacterial microbiota had high disease severity with increased CRP. Additionally, the authors observed increased incidence of intensive care unit admissions with worse pulmonary functions in these patients[104]. The study suggested an association between the hyper-inflammatory response in COVID-19 and gut dysbiosis. However, a greater number of studies testing patients well after recovery are required to fully illustrate gut dysbiosis, associated factors, and the hyper-inflammatory response during COVID-19.

GUT MICROBIOTA IN ELDERLY OR CO-MORBID COVID-19 PATIENTS

Researchers have attempted to understand the role of the gut microbiota in elderly or co-morbid COVID-19 patients. A recent study evaluated the association of the gut microbiota and its modulation in COVID-19 patients. In this study, the cohort comprised approximately 200 severe COVID-19 patients hospitalized with pneumonia. Researchers considered elderly patients (age 62 years to 64 years) and their comorbidity. Patients in this study received two types of treatments: one group was treated with only the best available therapy (BAT), and the other group was treated with oral bacteriotherapy and BAT. Researchers found a decline in mortality and decreased progress in severe disease. Finally, researchers concluded that oral bacteriotherapy might be helpful in the management of hospitalized COVID-19 patients[105]. Similarly, Rao et al[106] noted that people with the comorbidities are more prone to COVID-19-related complications. In this case, immune system deregulation and deaths were also noted. However, researchers used-glucan to enhance the immune system in COVID-19 patients. This glucan was used to augment the activity of macrophages, natural killer cells, and IL-8, implicating that it might enhance the defense mechanisms to combat the virus[106].

Recently, Liu et al[1] evaluated the role of the gut microbiota composition and its association with the post-acute COVID-19 syndrome (PACS). In this study, researchers considered the comorbidities and dietary patterns during patient selection compare gut microbiota compositions. However, no considerable differences were observed in age, comorbidities, gender, antibiotics, or antiviral drug use between patients with PACS or without PACS[1].

Therefore, in cases of elderly or co-morbid COVID-19 patients, the gut microbiota might play an important role in immune system deregulation, although further studies are required to validate the findings.

GUT MICROBIOTA BASED ON ANTIBIOTIC USAGE IN COVID-19 PATIENTS

In COVID-19 patients, the use of antibiotics is relatively common. The frequently used antibiotics in COVID-19 patients are Azithromycin, Amoxicillin Clavulanate, Cephalosporin, Tetracycline[49,107], etc. The composition of the gut microbiota is hampered in COVID-19 patients due to the usage of antibiotics, occasionally causing antibiotic-associated diarrhea (AAD)[108]. Antibiotics usages in COVID-19 patients were increased the number of opportunistic pathogens compared with that detected in an untreated control group. Zuo et al[49] reported that the gut of COVID-19 patients, using antibiotics contains opportunistic bacterial pathogens such as Bacteroides nordii, Actinomyces viscosus, and Clostridium hathewayi. Additional studies also reported this phenomenon[22,109]. An increase of opportunistic bacterial pathogens causes dysbiosis of the gut. Rafiqul Islam et al[110] also noted that the abundance of opportunistic pathogens in COVID-19 patients in Bangladesh could cause dysbiosis, with 46 genera of opportunistic bacteria being identified patient GI samples. However, a study demonstrated that particular strains of probiotics may be useful for AAD[111]. Scientists have shown that the administration of oral probiotics can recover gut health and have antiviral effects[112,53]. For probiotic strain identification, Mak et al[113] highlight the need for effective research to easily recognize the probiotic strains of therapeutic use. In this case, the probiotics should be specific for COVID-19, and help reduce the susceptibility to COVID-19 preventing severe COVID-19 disease.

GUT MICROBIOTA DYSBIOSIS DURING COVID-19 AND USE OF PROBIOTICS

Scientists identified an association between the gut microbiota dysbiosis and the severity of COVID-19. Magalhães et al[52] noted that gut microbiota dysbiosis causes poor outcomes in elderly COVID-19 patients with hypertension and diabetes. Additionally, co-morbid elderly COVID-19 patients were prone to increased inflammatory situations due to the dysbiosis. The elevated amount of bacterial products in the gut might translocate into the blood due to the increased permeability across the intestinal epithelium. Bacterial toxin products, such as lipopolysaccharides (LPS), may accumulate in blood, aggravating TLR4 and subsequent downstream signaling. This could contribute to the “cytokine storm”, and result in complications in elderly COVID-19 patients[54]. Researchers also found a different route of activation of toll-like receptor (TLR4/TLR5) in COVID-19 patients[114-116]. Hung et al[53] also reported that gut microbiota dysbiosis increases COVID-19 severity in the elderly. However, the use of probiotics is a novel way to reduce COVID-19 severity in elderly populations.

THERAPEUTIC IMPLICATIONS AND CLINICAL TRIALS TO UNDERSTAND THE ROLE OF THE GUT MICROBIOTA DURING COVID-19

A careful analysis of the microbiome-gut-lung axis during COVID-19 infection can direct research towards therapeutic options for restoring gut health. As an altered gut microbiota is strongly associated with COVID-19 and its severity, supplementation of bacterial metabolites or commensals and prebiotics to enrich the microbial ecosystem is a path toward effective therapeutic options.

However, very few studies have explored this. A randomized clinical trial with 300 registered participants assessed the effectiveness of combination therapy using Lactobacillus plantarum (L. plantarum) CECT 7484, L. plantarum CECT 30292, Pediococcus acidilactici (P. acidilactici) CECT 7483, and L. plantarum CECT 7485, in adult COVID-19 patients (ClinicalTrials.gov; Clinical trial no. NCT04517422). Nonetheless, a deficiency of well-established data calls for more studies of this nature[41]. An open-label, randomized clinical trial with 350 participants conducted by Kaleido Biosciences sought to determine the effectiveness of a novel glycan molecule (KB109) in patients with mild to moderate COVID-19 (ClinicalTrials.gov; Clinical trial no. NCT04414124)[117]. The synthetic glycan molecule reduced the number of acute care visits by COVID-19 patients. Additionally, disease resolution in patients with comorbidities was improved, compared to that in patients relying solely on supportive self-care.

A similar study attempted to evaluate the glycan molecule’s effectiveness (KB109) associated with gut microbiota function in COVID-19 patients. The same organization conducted the clinical study, an open-label, randomized clinical trial in 49 participants in the United States (ClinicalTrials.gov; Clinical trial no. NCT04486482)[118]. There were no conclusive results; however, more studies are likely to be conducted in this sense. A complete list of the clinical trials initiated to understand the role of the gut microbiota in COVID-19 and its therapeutic implications are shown in Table 4.

Table 4 List of clinical trials initiated to understand the role of gut microbiota in coronavirus disease 2019 and its therapeutic implications.
SI. No.
Objective of clinical trials
Clinical trials No.
Description of clinical trials
Remarks
1Evaluate the combination of probiotics (P. acidilactici and L. plantarum) to reduce the viral load of moderate or severe COVID-19 patientsNCT04517422It was a randomized controlled trial, 300 participants, treatment by dietary supplement (probiotics)Observational study of adult and older adult, trial completed
2To explore the natural history of mild-to-moderate COVID-19 illness and safety of a novel glycan (KB109) and self-supportive careNCT04414124It was a randomized, prospective, open-label, parallel-group controlled clinical study of 350 participantsObservational study of adults (both male and female), trial completed
3Investigate the physiologic effects of the novel glycan (KB109) on patients with COVID-19 illness on gut microbiota structure and function in the outpatientNCT04486482It was a randomized, open-label clinical study of 49 participantsObservational study of adults patients with mild-to-moderate COVID-19 infections, trial completed
4Evaluate the clinical contribution of the gut microbiota and its diversity on the COVID-19 disease severity and the viral loadNCT05107245It was case-control, diagnostic study of 143 participantsObservational study on the diagnostic evaluation of the human intestinal microbiota, trial completed
5Studied the effects of Lactobacillus coryniformis K8 intake on the prevalence and severity of COVID-19 in health professionalNCT04366180A randomized, interventional study of 314 participantsInvestigation of probiotic effects to healthcare personnel exposed to COVID-19 infection
6Investigate to exploring the role of nutritional support by probiotics to COVID-19 outpatients (adult)NCT04907877Randomized, evidence based study of 300 participantsUsed of probiotics as dietary supplement that enhance specific immune response of patients having COVID-19 respiratory infection
7Use of dietary supplement (Omni-Biotic® 10 AAD) can decrease the intestinal inflammation and improves dysbiosis for COVID-19 patientsNCT04420676It was a randomized Interventional study of 30 participantsThis study performed as double blind, placebo-controlled study
8Evaluate the probiotics efficacy to decrease the COVID-19 infection symptoms and duration of COVID-19 positive patientsNCT04621071The double-blind, randomized, controlled trial of 17 participantsThis study performed to explored the effects of dietary supplement: Probiotics (2 strains 10 × 109 UFC), trial completed
9Impact analysis of probiotic strain L. reuteri DSM 17938 for specific Abs response against SARS-CoV-2 infectionNCT04734886It was control, randomized trial of 161 participantsTo assess the upon and after COVID-19 infection in healthy adults, trial completed
10To evaluate the primary efficacy of live microbials (probiotics) for boosting up the immunity of SARS-CoV-2 infected persons (unvaccinated)NCT04847349It was double-blind, randomized, controlled trial of 54 participantsEfficacy analysis of dietary supplement (combination of live microbials) as anti COVID-19 infection, trial completed
11Evaluate the follow -up of Symprove (probiotic) to COVID-19 positive patientsNCT04877704The randomized clinical trial was performed with 60 patientsObservational study to supervision of hospitalized COVID-19 patients
12Study was performed to evaluate the possible effect of a probiotic mixtures in the improvement of COVID-19 infection symptomsNCT04390477It was randomized case control, clinical trial of 41 participantsObservational study of dietary supplement: Probiotic to COVID-19 patients
13The probiotic ( Omni-Biotic Pro Vi 5) use for investigate the side effect of post-COVID syndromeNCT04813718It was a randomized trial of 20 participantsIt was a therapeutic target study of probiotic for treatment of acute COVID-19 and prevention of post COVID infections
14To evaluate the effect of a probiotic strain on the occurrence and severity of COVID-19 in hospitalised elderly populationNCT04756466Randomized control trial of 201 participantsIt was observational study, probiotic sued for improve the immune response of elderly patients
15This study assesses the beneficial effects of the nutritional supplementation (ABBC1) to individuals taken the COVID-10 vaccineNCT04798677It was a double-blinded, placebo-controlled, randomized clinical study of 90 participantsUsed as knowing the microbiome modulating properties, observational study
16To investigate the consequence of Ligilactobacillus salivarius MP101 to hospitalised elderly individualsNCT04922918Non-randomised study of 25 participantsObservational study of aged patients having highly affected by COVID-19
17Study was performed to explored the effect of the probiotic Lactobacillus rhamnosus GGNCT04399252It was a randomized double-blind, placebo-controlled trail of 182 participantsObservational study of individuals microbiome of household contacts exposed to COVID-19
18Treatment approaches by probiotics to human gut microbiome and growing the anti-inflammatory response for COVID-19 infected patientsNCT04854941It was a randomized controlled open-label study of 200 participantsThe optimizing treatment approaches based observational study, trial completed
19To evaluate the capability of the novel nutritional supplement (probiotics and other vitamins) to COVID-19 infected and hospitalised patientsNCT04666116Randomized, single blind clinical trial of 96 participantsUsed of dietary supplementation with probiotics aims to reduce the viral load
20Using of probiotics for COVID 19 transmission reduction to health care professionalsNCT04462627It was a non-randomized trial of 500 participantsAnalysis and reduction of COVID-19 viral load to health care professionals

As the pandemic persists, it is critical to assess the effect of next-generation probiotics, prebiotics, synbiotics, and increased fiber intake on changes in gut microbiota composition in patients with mild to moderate and severe COVID-19.

FUTURE PERSPECTIVE

In several cases, complex pathophysiological and immunological responses are reported in the host due to SARS-CoV-2 infection. However, very little is known regarding the changes in gut virome in the COVID-19 patients, and this should be explored in future studies should explore it further. Moreover, the possible role of the gut microbiota in COVID-19 should be explored in future research. Likewise, population-based cohorts should be generated to illustrate the function of the altered gut microbiota during COVID-19 in different populations. This will enable the design of diagnostics and therapeutics for COVID-19 in different population types. Simultaneously, population-specific changes need to be described as this can help resolve severe conditions in COVID-19 patients. In the future, researchers should attempt to understand population-specific gut microbiota alteration during COVID-19 to design therapeutic interventions as required. Moreover, research could focus on the population specific changes in the immune response generated against the two altered gut microbiota during COVID-19.

CONCLUSION

Presently, abundant research has described the marked changes in the gut microbiomes of COVID-19 patients. Therefore, an apparent association exsists between the overall health of the gut microbiome and the progression of COVID-19[119]. Furthermore, the altered gut microbiota has been shown to persist in patients even after several days of recovery from COVID-19.

However, poor outcome were observed in elderly or co-morbid patients[97,120]. Recently, several studies discussed the factors associated with the modified gut microbiota in COVID-19 patients manifesting GI symptoms. According to some reports, increased inflammation may lead to a leaky gut, which enables the translocation of bacterial metabolites and toxins into the systemic circulation[97,120]. This might cause further complications to the severe COVID-19 patients.

In this review, we have illustrated various GI aspects of COVID-19 patients including the gut microbiota imbalance and GI symptoms, the patterns of altered gut microbiota composition, the crosstalk between the microbiome and the gut-lung axis, the crosstalk between the microbiome and the gut-brain-lung axis, as well as hypoxia associated with altered gut microbiota. We also highlighted the association between the gut microbiota and elderly or co-morbid COVID-19 patients, as well as that of gut microbiota dysbiosis and COVID-19 severity. Additionally, we explored the correlation between, probiotics usage and the gut microbiota based on antibiotic usage in COVID-19 patients. Therefore, our review will provide a distinct outline for researchers working in the field. Also, it will provide valuable insight into the role of gut microbiomes in COVID-19 patients.

Currently, therapeutics are in development to combat COVID-19. In addition to antiviral therapeutics, probiotics might be effective for improving gut health through the gut-lung axis. Recently, several clinical trials have been initiated to understand the role of probiotics in COVID-19 patients. The ongoing clinical trials will elucidate the role of probiotic therapeutics or for COVID-19 patients, and offer new alternatives in COVID-19 treatment.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Biotechnology and applied microbiology

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B, B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Gupta S, United States; Kamran M, Pakistan; Li S, China; Tan JK, Malaysia; Yu L, Singapore S-Editor: Chen YL L-Editor: A P-Editor: Wu RR

References
1.  Liu Q, Mak JWY, Su Q, Yeoh YK, Lui GC, Ng SSS, Zhang F, Li AYL, Lu W, Hui DS, Chan PK, Chan FKL, Ng SC. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut. 2022;71:544-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 223]  [Article Influence: 111.5]  [Reference Citation Analysis (0)]
2.  Troisi J, Venutolo G, Pujolassos Tanyà M, Delli Carri M, Landolfi A, Fasano A. COVID-19 and the gastrointestinal tract: Source of infection or merely a target of the inflammatory process following SARS-CoV-2 infection? World J Gastroenterol. 2021;27:1406-1418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 29]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
3.  Al Kassaa I, El Omari S, Abbas N, Papon N, Drider D, Kassem II, Osman M. High association of COVID-19 severity with poor gut health score in Lebanese patients. PLoS One. 2021;16:e0258913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
4.  Donati Zeppa S, Agostini D, Piccoli G, Stocchi V, Sestili P. Gut Microbiota Status in COVID-19: An Unrecognized Player? Front Cell Infect Microbiol. 2020;10:576551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
5.  Gu J, Han B, Wang J. COVID-19: Gastrointestinal Manifestations and Potential Fecal-Oral Transmission. Gastroenterology. 2020;158:1518-1519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 954]  [Cited by in F6Publishing: 907]  [Article Influence: 226.8]  [Reference Citation Analysis (1)]
6.  Ramachandran P, Onukogu I, Ghanta S, Gajendran M, Perisetti A, Goyal H, Aggarwal A. Gastrointestinal Symptoms and Outcomes in Hospitalized Coronavirus Disease 2019 Patients. Dig Dis. 2020;38:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
7.  Han C, Duan C, Zhang S, Spiegel B, Shi H, Wang W, Zhang L, Lin R, Liu J, Ding Z, Hou X. Digestive Symptoms in COVID-19 Patients With Mild Disease Severity: Clinical Presentation, Stool Viral RNA Testing, and Outcomes. Am J Gastroenterol. 2020;115:916-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 374]  [Cited by in F6Publishing: 360]  [Article Influence: 90.0]  [Reference Citation Analysis (0)]
8.  Ng SC, Tilg H. COVID-19 and the gastrointestinal tract: more than meets the eye. Gut. 2020;69:973-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 139]  [Article Influence: 34.8]  [Reference Citation Analysis (0)]
9.  Tang A, Tong ZD, Wang HL, Dai YX, Li KF, Liu JN, Wu WJ, Yuan C, Yu ML, Li P, Yan JB. Detection of Novel Coronavirus by RT-PCR in Stool Specimen from Asymptomatic Child, China. Emerg Infect Dis. 2020;26:1337-1339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 349]  [Article Influence: 87.3]  [Reference Citation Analysis (0)]
10.  Wong SH, Lui RN, Sung JJ. Covid-19 and the digestive system. J Gastroenterol Hepatol. 2020;35:744-748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 423]  [Article Influence: 105.8]  [Reference Citation Analysis (0)]
11.  Mehandru S, Merad M. Pathological sequelae of long-haul COVID. Nat Immunol. 2022;23:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 300]  [Article Influence: 150.0]  [Reference Citation Analysis (0)]
12.  Holmes E, Li JV, Marchesi JR, Nicholson JK. Gut microbiota composition and activity in relation to host metabolic phenotype and disease risk. Cell Metab. 2012;16:559-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 335]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
13.  Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336:1262-1267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2838]  [Cited by in F6Publishing: 2900]  [Article Influence: 241.7]  [Reference Citation Analysis (0)]
14.  Joyce SA, Gahan CG. The gut microbiota and the metabolic health of the host. Curr Opin Gastroenterol. 2014;30:120-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
15.  Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21:8787-8803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 1421]  [Cited by in F6Publishing: 1392]  [Article Influence: 154.7]  [Reference Citation Analysis (43)]
16.  Corfield AP. The Interaction of the Gut Microbiota with the Mucus Barrier in Health and Disease in Human. Microorganisms. 2018;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 79]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
17.  Tang WH, Kitai T, Hazen SL. Gut Microbiota in Cardiovascular Health and Disease. Circ Res. 2017;120:1183-1196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 757]  [Cited by in F6Publishing: 880]  [Article Influence: 125.7]  [Reference Citation Analysis (0)]
18.  Dhar D, Mohanty A. Gut microbiota and Covid-19-possible link and implications. Virus Res. 2020;285:198018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 356]  [Article Influence: 89.0]  [Reference Citation Analysis (0)]
19.  Zuo T, Wu X, Wen W, Lan P. Gut Microbiome Alterations in COVID-19. Genomics Proteomics Bioinformatics. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 51]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
20.  Chen J, Hall S, Vitetta L. Altered gut microbial metabolites could mediate the effects of risk factors in Covid-19. Rev Med Virol. 2021;31:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
21.  Yamamoto S, Saito M, Tamura A, Prawisuda D, Mizutani T, Yotsuyanagi H. The human microbiome and COVID-19: A systematic review. PLoS One. 2021;16:e0253293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 65]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
22.  Chiappetta S, Sharma AM, Bottino V, Stier C. COVID-19 and the role of chronic inflammation in patients with obesity. Int J Obes (Lond). 2020;44:1790-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 136]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
23.  Del Valle DM, Kim-Schulze S, Huang HH, Beckmann ND, Nirenberg S, Wang B, Lavin Y, Swartz TH, Madduri D, Stock A, Marron TU, Xie H, Patel M, Tuballes K, Van Oekelen O, Rahman A, Kovatch P, Aberg JA, Schadt E, Jagannath S, Mazumdar M, Charney AW, Firpo-Betancourt A, Mendu DR, Jhang J, Reich D, Sigel K, Cordon-Cardo C, Feldmann M, Parekh S, Merad M, Gnjatic S. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med. 2020;26:1636-1643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1725]  [Cited by in F6Publishing: 1521]  [Article Influence: 380.3]  [Reference Citation Analysis (0)]
24.  Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9:7204-7218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2209]  [Cited by in F6Publishing: 2057]  [Article Influence: 342.8]  [Reference Citation Analysis (0)]
25.  Tang Y, Liu J, Zhang D, Xu Z, Ji J, Wen C. Cytokine Storm in COVID-19: The Current Evidence and Treatment Strategies. Front Immunol. 2020;11:1708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 647]  [Article Influence: 161.8]  [Reference Citation Analysis (0)]
26.  Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, Ruiz C, Melguizo-Rodríguez L. SARS-CoV-2 infection: The role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 2020;54:62-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 552]  [Cited by in F6Publishing: 694]  [Article Influence: 173.5]  [Reference Citation Analysis (0)]
27.  García LF. Immune Response, Inflammation, and the Clinical Spectrum of COVID-19. Front Immunol. 2020;11:1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 450]  [Cited by in F6Publishing: 430]  [Article Influence: 107.5]  [Reference Citation Analysis (0)]
28.  Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ; HLH Across Speciality Collaboration, UK. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033-1034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6366]  [Cited by in F6Publishing: 6332]  [Article Influence: 1583.0]  [Reference Citation Analysis (0)]
29.  Sinha P, Matthay MA, Calfee CS. Is a "Cytokine Storm" Relevant to COVID-19? JAMA Intern Med. 2020;180:1152-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 478]  [Article Influence: 119.5]  [Reference Citation Analysis (0)]
30.  Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 Cytokine Storm; What We Know So Far. Front Immunol. 2020;11:1446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1044]  [Cited by in F6Publishing: 963]  [Article Influence: 240.8]  [Reference Citation Analysis (0)]
31.  Perisetti A, Gajendran M, Mann R, Elhanafi S, Goyal H. COVID-19 extrapulmonary illness -special gastrointestinal and hepatic considerations. Dis Mon. 2020;66:101064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
32.  de Oliveira GLV, Oliveira CNS, Pinzan CF, de Salis LVV, Cardoso CRB. Microbiota Modulation of the Gut-Lung Axis in COVID-19. Front Immunol. 2021;12:635471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 112]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
33.  Zhou D, Wang Q, Liu H. Coronavirus disease 2019 and the gut-lung axis. Int J Infect Dis. 2021;113:300-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
34.  Johnson SD, Olwenyi OA, Bhyravbhatla N, Thurman M, Pandey K, Klug EA, Johnston M, Dyavar SR, Acharya A, Podany AT, Fletcher CV, Mohan M, Singh K, Byrareddy SN. Therapeutic implications of SARS-CoV-2 dysregulation of the gut-brain-lung axis. World J Gastroenterol. 2021;27:4763-4783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
35.  Redd WD, Zhou JC, Hathorn KE, McCarty TR, Bazarbashi AN, Thompson CC, Shen L, Chan WW. Prevalence and Characteristics of Gastrointestinal Symptoms in Patients With Severe Acute Respiratory Syndrome Coronavirus 2 Infection in the United States: A Multicenter Cohort Study. Gastroenterology. 2020;159:765-767.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 268]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
36.  Pan L, Mu M, Yang P, Sun Y, Wang R, Yan J, Li P, Hu B, Wang J, Hu C, Jin Y, Niu X, Ping R, Du Y, Li T, Xu G, Hu Q, Tu L. Clinical Characteristics of COVID-19 Patients With Digestive Symptoms in Hubei, China: A Descriptive, Cross-Sectional, Multicenter Study. Am J Gastroenterol. 2020;115:766-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1160]  [Cited by in F6Publishing: 1130]  [Article Influence: 282.5]  [Reference Citation Analysis (0)]
37.  Guan W-j, Ni Z-y, Hu Y, Liang W-h, Ou C-q, He J-x, Liu L, Shan H, Lei C-l, Hui DS. Clinical characteristics of coronavirus disease 2019 in China. New Engl J of Med. 2020;382 (18):1708-1720.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Yeoh YK, Zuo T, Lui GC, Zhang F, Liu Q, Li AY, Chung AC, Cheung CP, Tso EY, Fung KS, Chan V, Ling L, Joynt G, Hui DS, Chow KM, Ng SSS, Li TC, Ng RW, Yip TC, Wong GL, Chan FK, Wong CK, Chan PK, Ng SC. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021;70:698-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 521]  [Cited by in F6Publishing: 656]  [Article Influence: 218.7]  [Reference Citation Analysis (0)]
39.  Shi J, Sun J, Hu Y. Enteric involvement of SARS-CoV-2: Implications for the COVID-19 management, transmission, and infection control. Virulence. 2020;11:941-944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
40.  Villapol S. Gastrointestinal symptoms associated with COVID-19: impact on the gut microbiome. Transl Res. 2020;226:57-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 186]  [Article Influence: 46.5]  [Reference Citation Analysis (0)]
41.  Kaźmierczak-Siedlecka K, Vitale E, Makarewicz W. COVID-19 -gastrointestinal and gut microbiota-related aspects. Eur Rev Med Pharmacol Sci. 2020;24:10853-10859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
42.  Zhang T, Cui X, Zhao X, Wang J, Zheng J, Zheng G, Guo W, Cai C, He S, Xu Y. Detectable SARS-CoV-2 viral RNA in feces of three children during recovery period of COVID-19 pneumonia. J Med Virol. 2020;92:909-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 152]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
43.  Chen Y, Chen L, Deng Q, Zhang G, Wu K, Ni L, Yang Y, Liu B, Wang W, Wei C, Yang J, Ye G, Cheng Z. The presence of SARS-CoV-2 RNA in the feces of COVID-19 patients. J Med Virol. 2020;92:833-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 505]  [Cited by in F6Publishing: 535]  [Article Influence: 133.8]  [Reference Citation Analysis (0)]
44.  Wölfel R, Corman VM, Guggemos W, Seilmaier M, Zange S, Müller MA, Niemeyer D, Jones TC, Vollmar P, Rothe C, Hoelscher M, Bleicker T, Brünink S, Schneider J, Ehmann R, Zwirglmaier K, Drosten C, Wendtner C. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020;581:465-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4682]  [Cited by in F6Publishing: 4769]  [Article Influence: 1192.3]  [Reference Citation Analysis (0)]
45.  Kevadiya BD, Machhi J, Herskovitz J, Oleynikov MD, Blomberg WR, Bajwa N, Soni D, Das S, Hasan M, Patel M, Senan AM, Gorantla S, McMillan J, Edagwa B, Eisenberg R, Gurumurthy CB, Reid SPM, Punyadeera C, Chang L, Gendelman HE. Diagnostics for SARS-CoV-2 infections. Nat Mater. 2021;20:593-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 511]  [Cited by in F6Publishing: 417]  [Article Influence: 139.0]  [Reference Citation Analysis (0)]
46.  Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32663]  [Cited by in F6Publishing: 28382]  [Article Influence: 7095.5]  [Reference Citation Analysis (3)]
47.  Zuo T, Liu Q, Zhang F, Yeoh YK, Wan Y, Zhan H, Lui GCY, Chen Z, Li AYL, Cheung CP, Chen N, Lv W, Ng RWY, Tso EYK, Fung KSC, Chan V, Ling L, Joynt G, Hui DSC, Chan FKL, Chan PKS, Ng SC. Temporal landscape of human gut RNA and DNA virome in SARS-CoV-2 infection and severity. Microbiome. 2021;9:91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
48.  Zuo T, Zhan H, Zhang F, Liu Q, Tso EYK, Lui GCY, Chen N, Li A, Lu W, Chan FKL, Chan PKS, Ng SC. Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge. Gastroenterology. 2020;159:1302-1310.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 199]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
49.  Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, Wan Y, Chung ACK, Cheung CP, Chen N, Lai CKC, Chen Z, Tso EYK, Fung KSC, Chan V, Ling L, Joynt G, Hui DSC, Chan FKL, Chan PKS, Ng SC. Alterations in Gut Microbiota of Patients With COVID-19 During Time of Hospitalization. Gastroenterology. 2020;159:944-955.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 877]  [Article Influence: 219.3]  [Reference Citation Analysis (0)]
50.  Newsome RC, Gauthier J, Hernandez MC, Abraham GE, Robinson TO, Williams HB, Sloan M, Owings A, Laird H, Christian T, Pride Y, Wilson KJ, Hasan M, Parker A, Senitko M, Glover SC, Gharaibeh RZ, Jobin C. The gut microbiome of COVID-19 recovered patients returns to uninfected status in a minority-dominated United States cohort. Gut Microbes. 2021;13:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
51.  Khan M, Mathew BJ, Gupta P, Garg G, Khadanga S, Vyas AK, Singh AK. Gut Dysbiosis and IL-21 Response in Patients with Severe COVID-19. Microorganisms. 2021;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
52.  Magalhães NS, Savino W, Silva PMR, Martins MA, Carvalho VF. Gut Microbiota Dysbiosis Is a Crucial Player for the Poor Outcomes for COVID-19 in Elderly, Diabetic and Hypertensive Patients. Front Med (Lausanne). 2021;8:644751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
53.  Hung YP, Lee CC, Lee JC, Tsai PJ, Ko WC. Gut Dysbiosis during COVID-19 and Potential Effect of Probiotics. Microorganisms. 2021;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
54.  Zuo T, Liu Q, Zhang F, Lui GC, Tso EY, Yeoh YK, Chen Z, Boon SS, Chan FK, Chan PK, Ng SC. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. 2021;70:276-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 238]  [Article Influence: 79.3]  [Reference Citation Analysis (0)]
55.  Sokol H, Contreras V, Maisonnasse P, Desmons A, Delache B, Sencio V, Machelart A, Brisebarre A, Humbert L, Deryuter L, Gauliard E, Heumel S, Rainteau D, Dereuddre-Bosquet N, Menu E, Ho Tsong Fang R, Lamaziere A, Brot L, Wahl C, Oeuvray C, Rolhion N, Van Der Werf S, Ferreira S, Le Grand R, Trottein F. SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota. Gut Microbes. 2021;13:1-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
56.  Zhang F, Wan Y, Zuo T, Yeoh YK, Liu Q, Zhang L, Zhan H, Lu W, Xu W, Lui GCY, Li AYL, Cheung CP, Wong CK, Chan PKS, Chan FKL, Ng SC. Prolonged Impairment of Short-Chain Fatty Acid and L-Isoleucine Biosynthesis in Gut Microbiome in Patients With COVID-19. Gastroenterology. 2022;162:548-561.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 105]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
57.  Shang J, Ye G, Shi K, Wan Y, Luo C, Aihara H, Geng Q, Auerbach A, Li F. Structural basis of receptor recognition by SARS-CoV-2. Nature. 2020;581:221-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2950]  [Cited by in F6Publishing: 2536]  [Article Influence: 634.0]  [Reference Citation Analysis (0)]
58.  Chakraborty C, Sharma AR, Mallick B, Bhattacharya M, Sharma G, Lee SS. Evaluation of molecular interaction, physicochemical parameters and conserved pattern of SARS-CoV-2 Spike RBD and hACE2: in silico and molecular dynamics approach. Eur Rev Med Pharmacol Sci. 2021;25:1708-1723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
59.  Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med. 2020;76:14-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 805]  [Cited by in F6Publishing: 823]  [Article Influence: 205.8]  [Reference Citation Analysis (0)]
60.  Sungnak W, Huang N, Bécavin C, Berg M, Queen R, Litvinukova M, Talavera-López C, Maatz H, Reichart D, Sampaziotis F, Worlock KB, Yoshida M, Barnes JL; HCA Lung Biological Network. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26:681-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1985]  [Cited by in F6Publishing: 1821]  [Article Influence: 455.3]  [Reference Citation Analysis (0)]
61.  Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3643]  [Cited by in F6Publishing: 3918]  [Article Influence: 195.9]  [Reference Citation Analysis (0)]
62.  Sharma RK, Oliveira AC, Yang T, Karas MM, Li J, Lobaton GO, Aquino VP, Robles-Vera I, de Kloet AD, Krause EG, Bryant AJ, Verma A, Li Q, Richards EM, Raizada MK. Gut Pathology and Its Rescue by ACE2 (Angiotensin-Converting Enzyme 2) in Hypoxia-Induced Pulmonary Hypertension. Hypertension. 2020;76:206-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
63.  Koester ST, Li N, Lachance DM, Morella NM, Dey N. Variability in digestive and respiratory tract Ace2 expression is associated with the microbiome. PLoS One. 2021;16:e0248730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
64.  Geva-Zatorsky N, Sefik E, Kua L, Pasman L, Tan TG, Ortiz-Lopez A, Yanortsang TB, Yang L, Jupp R, Mathis D, Benoist C, Kasper DL. Mining the Human Gut Microbiota for Immunomodulatory Organisms. Cell. 2017;168:928-943.e11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 469]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
65.  Penninger JM, Grant MB, Sung JJY. The Role of Angiotensin Converting Enzyme 2 in Modulating Gut Microbiota, Intestinal Inflammation, and Coronavirus Infection. Gastroenterology. 2021;160:39-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 74]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
66.  Viana SD, Nunes S, Reis F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities -Role of gut microbiota dysbiosis. Ageing Res Rev. 2020;62:101123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 88]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
67.  Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, Hansbro PM. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 622]  [Cited by in F6Publishing: 792]  [Article Influence: 99.0]  [Reference Citation Analysis (0)]
68.  Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O. The role of the lung microbiota and the gut-lung axis in respiratory infectious diseases. Cell Microbiol. 2018;20:e12966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 233]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
69.  Enaud R, Prevel R, Ciarlo E, Beaufils F, Wieërs G, Guery B, Delhaes L. The Gut-Lung Axis in Health and Respiratory Diseases: A Place for Inter-Organ and Inter-Kingdom Crosstalks. Front Cell Infect Microbiol. 2020;10:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 338]  [Article Influence: 84.5]  [Reference Citation Analysis (0)]
70.  S A, K G, A AM. Intermodulation of gut-lung axis microbiome and the implications of biotics to combat COVID-19. J Biomol Struct Dyn. 2021;1-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
71.  Aktas B, Aslim B. Gut-lung axis and dysbiosis in COVID-19. Turk J Biol. 2020;44:265-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 81]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
72.  Hernández-Terán A, Mejía-Nepomuceno F, Herrera MT, Barreto O, García E, Castillejos M, Boukadida C, Matias-Florentino M, Rincón-Rubio A, Avila-Rios S, Mújica-Sánchez M, Serna-Muñoz R, Becerril-Vargas E, Guadarrama-Pérez C, Ahumada-Topete VH, Rodríguez-Llamazares S, Martínez-Orozco JA, Salas-Hernández J, Pérez-Padilla R, Vázquez-Pérez JA. Dysbiosis and structural disruption of the respiratory microbiota in COVID-19 patients with severe and fatal outcomes. Sci Rep. 2021;11:21297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
73.  Zhu X, Han Y, Du J, Liu R, Jin K, Yi W. Microbiota-gut-brain axis and the central nervous system. Oncotarget. 2017;8:53829-53838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 152]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
74.  Morais LH, Schreiber HL 4th, Mazmanian SK. The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19:241-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 704]  [Article Influence: 176.0]  [Reference Citation Analysis (0)]
75.  Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci. 2011;12:453-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 925]  [Article Influence: 71.2]  [Reference Citation Analysis (0)]
76.  Taché Y, Saavedra JM. Introduction to the Special Issue "The Brain-Gut Axis". Cell Mol Neurobiol. 2022;42:311-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
77.  Dogra N, Mani RJ, Katare DP. The Gut-Brain Axis: Two Ways Signaling in Parkinson's Disease. Cell Mol Neurobiol. 2022;42:315-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
78.  Manosso LM, Arent CO, Borba LA, Ceretta LB, Quevedo J, Réus GZ. Microbiota-Gut-Brain Communication in the SARS-CoV-2 Infection. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
79.  Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018;1693:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 438]  [Cited by in F6Publishing: 629]  [Article Influence: 125.8]  [Reference Citation Analysis (0)]
80.  O'Donnell MP, Fox BW, Chao PH, Schroeder FC, Sengupta P. A neurotransmitter produced by gut bacteria modulates host sensory behaviour. Nature. 2020;583:415-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 116]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
81.  Mittal R, Debs LH, Patel AP, Nguyen D, Patel K, O'Connor G, Grati M, Mittal J, Yan D, Eshraghi AA, Deo SK, Daunert S, Liu XZ. Neurotransmitters: The Critical Modulators Regulating Gut-Brain Axis. J Cell Physiol. 2017;232:2359-2372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 283]  [Article Influence: 40.4]  [Reference Citation Analysis (0)]
82.  Konturek PC, Brzozowski T, Konturek SJ. Stress and the gut: pathophysiology, clinical consequences, diagnostic approach and treatment options. J Physiol Pharmacol. 2011;62:591-599.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Vodička M, Ergang P, Hrnčíř T, Mikulecká A, Kvapilová P, Vagnerová K, Šestáková B, Fajstová A, Hermanová P, Hudcovic T, Kozáková H, Pácha J. Microbiota affects the expression of genes involved in HPA axis regulation and local metabolism of glucocorticoids in chronic psychosocial stress. Brain Behav Immun. 2018;73:615-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
84.  Ojeda J, Ávila A, Vidal PM. Gut Microbiota Interaction with the Central Nervous System throughout Life. J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 41]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
85.  Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30:492-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 789]  [Cited by in F6Publishing: 1352]  [Article Influence: 338.0]  [Reference Citation Analysis (0)]
86.  Cerf-Bensussan N, Gaboriau-Routhiau V. The immune system and the gut microbiota: friends or foes? Nat Rev Immunol. 2010;10:735-744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 453]  [Cited by in F6Publishing: 450]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
87.  Xu K, Cai H, Shen Y, Ni Q, Chen Y, Hu S, Li J, Wang H, Yu L, Huang H, Qiu Y, Wei G, Fang Q, Zhou J, Sheng J, Liang T, Li L. [Management of COVID-19: the Zhejiang experience]. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2020;49:147-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 165]  [Reference Citation Analysis (0)]
88.  Jahani M, Dokaneheifard S, Mansouri K. Hypoxia: A key feature of COVID-19 launching activation of HIF-1 and cytokine storm. J Inflamm (Lond). 2020;17:33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 82]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
89.  Rahman A, Tabassum T, Araf Y, Al Nahid A, Ullah MA, Hosen MJ. Silent hypoxia in COVID-19: pathomechanism and possible management strategy. Mol Biol Rep. 2021;48:3863-3869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 76]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
90.  Tian M, Liu W, Li X, Zhao P, Shereen MA, Zhu C, Huang S, Liu S, Yu X, Yue M, Pan P, Wang W, Li Y, Chen X, Wu K, Luo Z, Zhang Q, Wu J. HIF-1α promotes SARS-CoV-2 infection and aggravates inflammatory responses to COVID-19. Signal Transduct Target Ther. 2021;6:308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 106]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
91.  Gopal AB, Chakraborty S, Padhan PK, Barik A, Dixit P, Chakraborty D, Poirah I, Samal S, Sarkar A, Bhattacharyya A. Silent hypoxia in COVID-19: a gut microbiota connection. Curr Opin Physiol. 2021;23:100456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
92.  Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2488]  [Cited by in F6Publishing: 2792]  [Article Influence: 279.2]  [Reference Citation Analysis (0)]
93.  von Rosenvinge EC, O'May GA, Macfarlane S, Macfarlane GT, Shirtliff ME. Microbial biofilms and gastrointestinal diseases. Pathog Dis. 2013;67:25-38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
94.  Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG, Salazar N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front Microbiol. 2016;7:185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1002]  [Cited by in F6Publishing: 1125]  [Article Influence: 140.6]  [Reference Citation Analysis (0)]
95.  Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol. 2019;10:277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 970]  [Cited by in F6Publishing: 1596]  [Article Influence: 319.2]  [Reference Citation Analysis (0)]
96.  Odenwald MA, Turner JR. The intestinal epithelial barrier: a therapeutic target? Nat Rev Gastroenterol Hepatol. 2017;14:9-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 732]  [Cited by in F6Publishing: 674]  [Article Influence: 96.3]  [Reference Citation Analysis (0)]
97.  Kim HS. Do an Altered Gut Microbiota and an Associated Leaky Gut Affect COVID-19 Severity? mBio. 2021;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
98.  Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C. The luminal short-chain fatty acid butyrate modulates NF-kappaB activity in a human colonic epithelial cell line. Gastroenterology. 2000;118:724-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 320]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
99.  Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, Xavier RJ, Teixeira MM, Mackay CR. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282-1286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2007]  [Cited by in F6Publishing: 2154]  [Article Influence: 143.6]  [Reference Citation Analysis (0)]
100.  Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M, Gordon JI. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci U S A. 2008;105:16767-16772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1035]  [Cited by in F6Publishing: 1070]  [Article Influence: 66.9]  [Reference Citation Analysis (0)]
101.  Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B, Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1875]  [Cited by in F6Publishing: 1929]  [Article Influence: 175.4]  [Reference Citation Analysis (2)]
102.  Chhibber-Goel J, Gopinathan S, Sharma A. Interplay between severities of COVID-19 and the gut microbiome: implications of bacterial co-infections? Gut Pathog. 2021;13:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
103.  Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. Int J Mol Sci. 2019;20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 536]  [Cited by in F6Publishing: 790]  [Article Influence: 158.0]  [Reference Citation Analysis (0)]
104.  Chen Y, Gu S, Chen Y, Lu H, Shi D, Guo J, Wu WR, Yang Y, Li Y, Xu KJ, Ding C, Luo R, Huang C, Yu L, Xu M, Yi P, Liu J, Tao JJ, Zhang H, Lv L, Wang B, Sheng J, Li L. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut. 2022;71:222-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 98]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
105.  Ceccarelli G, Borrazzo C, Pinacchio C, Santinelli L, Innocenti GP, Cavallari EN, Celani L, Marazzato M, Alessandri F, Ruberto F, Pugliese F, Venditti M, Mastroianni CM, d'Ettorre G. Oral Bacteriotherapy in Patients With COVID-19: A Retrospective Cohort Study. Front Nutr. 2020;7:613928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
106.  Rao KS, Suryaprakash V, Senthilkumar R, Preethy S, Katoh S, Ikewaki N, Abraham SJK. Role of Immune Dysregulation in Increased Mortality Among a Specific Subset of COVID-19 Patients and Immune-Enhancement Strategies for Combatting Through Nutritional Supplements. Front Immunol. 2020;11:1548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
107.  Al-Hadidi SH, Alhussain H, Abdel Hadi H, Johar A, Yassine HM, Al Thani AA, Eltai NO. The Spectrum of Antibiotic Prescribing During COVID-19 Pandemic: A Systematic Literature Review. Microb Drug Resist. 2021;27:1705-1725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
108.  Ramirez J, Guarner F, Bustos Fernandez L, Maruy A, Sdepanian VL, Cohen H. Antibiotics as Major Disruptors of Gut Microbiota. Front Cell Infect Microbiol. 2020;10:572912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 268]  [Article Influence: 67.0]  [Reference Citation Analysis (0)]
109.  Chattopadhyay I, Shankar EM. SARS-CoV-2-Indigenous Microbiota Nexus: Does Gut Microbiota Contribute to Inflammation and Disease Severity in COVID-19? Front Cell Infect Microbiol. 2021;11:590874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
110.  Rafiqul Islam SM, Foysal MJ, Hoque MN, Mehedi HMH, Rob MA, Salauddin A, Tanzina AY, Biswas S, Noyon SH, Siddiki AMAMZ, Tay A, Mannan A. Dysbiosis of Oral and Gut Microbiomes in SARS-CoV-2 Infected Patients in Bangladesh: Elucidating the Role of Opportunistic Gut Microbes. Front Med (Lausanne). 2022;9:821777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
111.  Baud D, Dimopoulou Agri V, Gibson GR, Reid G, Giannoni E. Using Probiotics to Flatten the Curve of Coronavirus Disease COVID-2019 Pandemic. Front Public Health. 2020;8:186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 136]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
112.  Xia Y, Cao J, Wang M, Lu M, Chen G, Gao F, Liu Z, Zhang D, Ke X, Yi M. Effects of Lactococcus lactis subsp. lactis JCM5805 on colonization dynamics of gut microbiota and regulation of immunity in early ontogenetic stages of tilapia. Fish Shellfish Immunol. 2019;86:53-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
113.  Mak JW, Chan FK, Ng SC. Probiotics and COVID-19-Authors’ reply. Lancet Gastroenterol Hepatol. 2020;5 (8):722-723.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Bhattacharya M, Sharma AR, Mallick B, Sharma G, Lee SS, Chakraborty C. Immunoinformatics approach to understand molecular interaction between multi-epitopic regions of SARS-CoV-2 spike-protein with TLR4/MD-2 complex. Infect Genet Evol. 2020;85:104587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 60]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
115.  Petruk G, Puthia M, Petrlova J, Samsudin F, Strömdahl AC, Cerps S, Uller L, Kjellström S, Bond PJ, Schmidtchen AA. SARS-CoV-2 spike protein binds to bacterial lipopolysaccharide and boosts proinflammatory activity. J Mol Cell Biol. 2020;12:916-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 93]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
116.  Chakraborty C, Sharma AR, Bhattacharya M, Sharma G, Lee SS, Agoramoorthy G. Consider TLR5 for new therapeutic development against COVID-19. J Med Virol. 2020;92:2314-2315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 44]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
117.  Haran JP, Pinero JC, Zheng Y, Palma NA, Wingertzahn M. Virtualized clinical studies to assess the natural history and impact of gut microbiome modulation in non-hospitalized patients with mild to moderate COVID-19 a randomized, open-label, prospective study with a parallel group study evaluating the physiologic effects of KB109 on gut microbiota structure and function: a structured summary of a study protocol for a randomized controlled study. Trials. 2021;22:245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
118.  van der Lelie D, Taghavi S. COVID-19 and the Gut Microbiome: More than a Gut Feeling. mSystems. 2020;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
119.  Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nat Rev Immunol. 2013;13:790-801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 917]  [Cited by in F6Publishing: 909]  [Article Influence: 82.6]  [Reference Citation Analysis (0)]
120.  Vignesh R, Swathirajan CR, Tun ZH, Rameshkumar MR, Solomon SS, Balakrishnan P. Could Perturbation of Gut Microbiota Possibly Exacerbate the Severity of COVID-19 via Cytokine Storm? Front Immunol. 2020;11:607734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
121.  Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, Guan L, Wei Y, Li H, Wu X, Xu J, Tu S, Zhang Y, Chen H, Cao B. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17476]  [Cited by in F6Publishing: 17173]  [Article Influence: 4293.3]  [Reference Citation Analysis (0)]
122.  Lu X, Zhang L, Du H, Zhang J, Li YY, Qu J, Zhang W, Wang Y, Bao S, Li Y. SARS-CoV-2 infection in children. New Engl J of Med. 2020;382 (17):1663-1665.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Zhang JJ, Dong X, Cao YY, Yuan YD, Yang YB, Yan YQ, Akdis CA, Gao YD. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy. 2020;75:1730-1741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2139]  [Cited by in F6Publishing: 2239]  [Article Influence: 559.8]  [Reference Citation Analysis (0)]
124.  Xiao F, Tang M, Zheng X, Liu Y, Li X, Shan H. Evidence for Gastrointestinal Infection of SARS-CoV-2. Gastroenterology. 2020;158:1831-1833.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1985]  [Cited by in F6Publishing: 1891]  [Article Influence: 472.8]  [Reference Citation Analysis (1)]
125.  Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, Wu Y, Zhang L, Yu Z, Fang M, Yu T, Wang Y, Pan S, Zou X, Yuan S, Shang Y. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8:475-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6231]  [Cited by in F6Publishing: 6333]  [Article Influence: 1583.3]  [Reference Citation Analysis (0)]
126.  Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, Wang B, Xiang H, Cheng Z, Xiong Y, Zhao Y, Li Y, Wang X, Peng Z. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA. 2020;323:1061-1069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14113]  [Cited by in F6Publishing: 14083]  [Article Influence: 3520.8]  [Reference Citation Analysis (0)]
127.  Xu XW, Wu XX, Jiang XG, Xu KJ, Ying LJ, Ma CL, Li SB, Wang HY, Zhang S, Gao HN, Sheng JF, Cai HL, Qiu YQ, Li LJ. Clinical findings in a group of patients infected with the 2019 novel coronavirus (SARS-Cov-2) outside of Wuhan, China: retrospective case series. BMJ. 2020;368:m606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1221]  [Cited by in F6Publishing: 1221]  [Article Influence: 305.3]  [Reference Citation Analysis (0)]
128.  Liu K, Fang YY, Deng Y, Liu W, Wang MF, Ma JP, Xiao W, Wang YN, Zhong MH, Li CH, Li GC, Liu HG. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin Med J (Engl). 2020;133:1025-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 823]  [Article Influence: 205.8]  [Reference Citation Analysis (0)]
129.  Shi H, Han X, Jiang N, Cao Y, Alwalid O, Gu J, Fan Y, Zheng C. Radiological findings from 81 patients with COVID-19 pneumonia in Wuhan, China: a descriptive study. Lancet Infect Dis. 2020;20:425-434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2174]  [Cited by in F6Publishing: 2221]  [Article Influence: 555.3]  [Reference Citation Analysis (0)]
130.  Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, Qiu Y, Wang J, Liu Y, Wei Y, Xia J, Yu T, Zhang X, Zhang L. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13871]  [Cited by in F6Publishing: 12323]  [Article Influence: 3080.8]  [Reference Citation Analysis (1)]
131.  Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, Tan W. Detection of SARS-CoV-2 in Different Types of Clinical Specimens. JAMA. 2020;323:1843-1844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1710]  [Cited by in F6Publishing: 2571]  [Article Influence: 642.8]  [Reference Citation Analysis (0)]
132.  Zhang J, Wang S, Xue Y. Fecal specimen diagnosis 2019 novel coronavirus-infected pneumonia. J Med Virol. 2020;92:680-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 288]  [Article Influence: 72.0]  [Reference Citation Analysis (0)]
133.  Ling Y, Xu SB, Lin YX, Tian D, Zhu ZQ, Dai FH, Wu F, Song ZG, Huang W, Chen J, Hu BJ, Wang S, Mao EQ, Zhu L, Zhang WH, Lu HZ. Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin Med J (Engl). 2020;133:1039-1043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 563]  [Cited by in F6Publishing: 572]  [Article Influence: 143.0]  [Reference Citation Analysis (0)]
134.  Young BE, Ong SWX, Kalimuddin S, Low JG, Tan SY, Loh J, Ng OT, Marimuthu K, Ang LW, Mak TM, Lau SK, Anderson DE, Chan KS, Tan TY, Ng TY, Cui L, Said Z, Kurupatham L, Chen MI, Chan M, Vasoo S, Wang LF, Tan BH, Lin RTP, Lee VJM, Leo YS, Lye DC; Singapore 2019 Novel Coronavirus Outbreak Research Team. Epidemiologic Features and Clinical Course of Patients Infected With SARS-CoV-2 in Singapore. JAMA. 2020;323:1488-1494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1346]  [Cited by in F6Publishing: 1317]  [Article Influence: 329.3]  [Reference Citation Analysis (0)]
135.  Wu Y, Guo C, Tang L, Hong Z, Zhou J, Dong X, Yin H, Xiao Q, Tang Y, Qu X, Kuang L, Fang X, Mishra N, Lu J, Shan H, Jiang G, Huang X. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol Hepatol. 2020;5:434-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1050]  [Cited by in F6Publishing: 1096]  [Article Influence: 274.0]  [Reference Citation Analysis (0)]
136.  Peng L, Liu J, Xu W, Luo Q, Chen D, Lei Z, Huang Z, Li X, Deng K, Lin B, Gao Z. SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimens. J Med Virol. 2020;92:1676-1680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 274]  [Article Influence: 68.5]  [Reference Citation Analysis (0)]
137.  Wu Y, Cheng X, Jiang G, Tang H, Ming S, Tang L, Lu J, Guo C, Shan H, Huang X. Altered oral and gut microbiota and its association with SARS-CoV-2 viral load in COVID-19 patients during hospitalization. NPJ Biofilms Microbiomes. 2021;7:61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 95]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]