Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that displays great variability in clinical phenotype. Many factors have been described to be correlated with its severity, and microbiota could play a key role in the infection, progression, and outcome of the disease. SARS-CoV-2 infection has been associated with nasopharyngeal and gut dysbiosis and higher abundance of opportunistic pathogens. To identify new prognostic markers for the disease, a multicenter prospective observational cohort study was carried out in COVID-19 patients divided into three cohorts based on symptomatology: mild (n=24), moderate (n=51), and severe/critical (n=31). Faecal and nasopharyngeal samples were taken, and the microbiota was analyzed. Linear discriminant analysis identified M. salivarium, P. dentalis, and H. parainfluenzae as biomarkers of severe COVID-19 in nasopharyngeal microbiota, while P. bivia and P. timonensis were defined in faecal microbiota. Additionally, a connection between faecal and nasopharyngeal microbiota was identified, with a significant ratio between P. timonensis (faeces) and P. dentalis and M. salivarium (nasopharyngeal) abundances found in critically ill patients. This ratio could serve as a novel prognostic tool for identifying severe COVID-19 cases.
Introduction
Coronavirus disease 2019 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The data reported in November 2023 revealed that over 700 million people have been infected with the virus [1]. The appearance of mutations and variants of concern has caused several additional waves of infection and compromise the effectiveness of existing vaccines and antiviral drugs [2]. Moreover, SARS-CoV-2 infection has shown to cause long-term effects on human health, although the mechanisms are still poorly described [3]. Even though most COVID-19 cases are mild, disease can be severe, resulting in hospitalisation, respiratory failure, or even death [1]. Therefore, a remarkable feature of SARS-CoV-2 infection is the great variability in clinical phenotype among infected people. Many factors can correlate with COVID-19 disease severity, including age, gender, body mass index, previous comorbidities, immune responses, and genetics [4–6]. Of note, and unfortunately, the determinants of infection outcome and the pathogenic mechanisms are not completely understood yet.
SARS-CoV-2 primarily infects the respiratory tract by binding to angiotensin-converting enzyme 2 (ACE2) receptor [7]. However, a growing body of evidence suggests that SARS-CoV-2 can also infect other organs since viral particles and nucleic acids have been found in different biological samples, like sputum, bronchoalveolar lavage fluid, faeces, blood, and urine [8, 9]. Thus, studies employing single-cell RNA sequencing have demonstrated that ACE2 is present in various organs and tissues, including the gastrointestinal tract, where ACE2 receptors have been reported to be highly expressed [10]. This supports several lines of evidence suggesting a substantial involvement of the gastrointestinal tract in the pathogenesis of the disease, including the ability of SARS-CoV-2 to infect and replicate in intestinal enterocytes [11], or to increase expression of the viral entry receptor (ACE2 receptor) and several membrane-bound serine proteases (such as transmembrane protease serine 2 (TMPRSS2) and TMPRSS4) in intestinal epithelial cells [12].
In addition, reports have shown that SARS-CoV-2 infection can disrupt nasopharyngeal and intestinal microbiota composition and promote dysbiosis, reducing diversity and increasing the presence of opportunistic pathogens, including Staphylococcus, Corynebacterium and Acinetobacter bacteria [13, 14], which can make patients more susceptible to secondary infections, rising morbidity and mortality [15]. Besides local changes in the respiratory tract, alterations in the distal gut microbiota have also been observed in SARS-CoV-2 infection [16–19]. Previous studies have evidenced a relevant connection between the microbiome in the nasopharynx and the gut, and preliminary data have suggested a bidirectional interaction that could play a role in the development of the immune response both in healthy and pathological conditions, including SARS-CoV-2 infection [20]. In this sense, dysbiosis has been associated not only with the severity of the disease but also with the recovery processes [13, 19]; however, little is understood of the link with the establishment of different symptomatic profiles in this condition, and to date, few studies have focused on the association between COVID-19 severity and the nasopharyngeal and faecal microbiota, being examined simultaneously. Considering that the efficacy of the COVID-19 vaccines and antiviral drugs against SARS-CoV-2 is compromised with the emergence of mutations and new variants of the virus [2], new therapeutic approaches and prognostic tools are still necessary. Therefore, the characterization of the nasopharyngeal and intestinal microbiome will allow to identify predictive biomarkers for the diagnosis and prognosis of the disease, as well as possible therapeutic targets in the management of SARS-CoV-2.
Results
Study patients characteristics
A total of 106 patients who had laboratory confirmation of SARS-CoV-2 infection were included in the present study. Of note, none of these patients reported another viral infection at the time of enrolment. Based on the clinical spectrum criteria reported in the COVID-19 treatment guidelines, patients were categorised into 3 groups: mild (24 patients), moderate illness that needed hospitalisation in Respiratory Unit (51 patients), and severe symptomatology and admitted in the ICU (31 patients) (Table 1). The age of the patients significantly increased with the severity of the symptoms. Patients included in the severe group were significantly older than those with mild or moderate symptoms (p < 0.05; ANOVA). Patient inclusion was carried out evenly in terms of gender as 52 women and 54 men were recruited; nevertheless, a gender-related impact on the clinical course of these patients was observed since males were predominantly in the group of patients with severe symptoms when compared with the mild illness group (p < 0.05; Fisher’s test). Regarding the symptomatology, mild patients showed symptoms of a mild respiratory infection, including fever, cough and headache. 33% of mild patients displayed dyspnoea and low oxygen saturation, 26% reported gastrointestinal complaints (including stomach ache, digestive discomfort and diarrhoea); and 4% reported high respiratory and heart rates. In contrast, moderate and severe patients showed higher frequencies of dyspnoea, low oxygen saturation and increased respiratory or heart rates (p < 0.05; Fisher’s test). When the different comorbidities were considered, patients in the group with severe symptoms showed a higher percentage of cardiomyopathy compared to those from the mild or moderate cohorts (42% vs 3% and 6%, respectively; p < 0.05; Fisher’s test). However, no significant differences were found in the prevalence of the other pathologies among groups. Regarding blood-based biomarkers, platelets (p < 0.05; ANOVA), D-dimer, ferritin and C reactive protein (p < 0.05; Kruskal-Wallis) correlated with the severity of the symptoms, being the severe group significantly different.
Bacterial composition differs between sample type and severity index in SARS-CoV-2 infected patients
Nasopharyngeal swabs and faeces were obtained from all the patients included in the study within seven days of symptom onset and used for characterization of the microbiota composition. In the nasopharyngeal microbiota, α-diversity was reduced in the moderate and severe groups compared to the mild group (p < 0.05 and p = 0.07 respectively; ANOVA) (Figure 1A). Conversely, when α-diversity was examined in stool samples, no significant modifications were observed among groups (Figure 1B). On the other hand, ß-diversity analysis through the Bray-Curtis index revealed statistical differences between groups for both nasopharyngeal swabs and stools samples (p < 0.001; PERMANOVA) (Figure 1C and 1D). As a result, microbiota is grouped in three separate clusters depending on the severity of the symptoms, which indicates that microbiota differences are based on changes in bacterial composition.
Similarly, the characterization of microbiota abundance composition revealed heterogeneity in the microbiota profile associated with severity and disease progression in these patients. Relative abundance levels for both phylum and genera were evaluated using the median abundance as a threshold. Specifically, genera with a median relative abundance greater than 0.5% were considered for detailed analysis.
At phylum level, in nasopharyngeal microbiota, the abundance of Bacillota was increased while the abundance of Bacteroidota and Actinobacteroidota was reduced in patients with severe symptomatology (Figure 2A). Conversely, the faecal microbiota of the three groups presented a more homogeneous distribution than the nasopharyngeal one, being the most abundant phyla Bacillota and Bacteroidota (Figure 2B). However, patients with severe symptoms, thus with a worse prognosis, showed a lower abundance of Bacteroidota (Figure 2B). Exact values of relative abundance for different phyla are shown in supplementary file 1.
At genus level, the nasopharyngeal microbiome composition revealed that symptom severity was associated with a higher number of detected genera (Figure 2C). Of note, significant differences in genus abundance for each group of patients were found. Thus, mild patients presented a significantly higher abundance of Alistipes, Muribaculaceae, Lactobacillus and Lachnospiraceae (p < 0.05; Kruskal-Wallis), whereas the group with moderate symptoms showed a significant increase in Alcaligenes, Pseudorobacter and Pseudomonas (p < 0.05; Kruskall-Wallis). Additionally, patients with more severe disease had significantly higher relative abundance of Acinetobacter, Actinomyces, Anaerococcus, Atopobium, Campylobacter, Dolosigranulum, Enterobacter, Enterococcus, Finegoldia, Fusobacterium, Gemella, Haemophilus, Lawsonella, Leptotrichia, Megasphaera, Neisseria, Serratia, Rotia and Veillonella (p < 0.05; Kruskal-Wallis). Unlike nasopharyngeal swabs, stool samples showed a reduction in the number of detected genera as symptom severity increased (Figure 2D). Precisely, mild patients showed more presence of Blautia, Muribaculaceae and different members of the Clostridia class (Clostridia, Coprococcus and Ruminococcus) than the other groups (p < 0.05; Kruskal-Wallis). In moderate ill patients Bacteroides, Barneseilla, Faecalibacterium, Parabacteroides and Streptococcus were significantly increased (p < 0.05; Kruskal-Wallis). Remarkably, Alistipes, Anaerococcus, Dialister, Finelgoldia, Lachnocostridium, Prevotella or Peptoniphilus were more abundant in patients with severe symptoms (p < 0.05; Kruskal-Wallis).
Relative abundance levels for genera identified bacteria and exact p-values can be found in supplementary files 2 and 3.
Differences in bacteria composition could be used as biomarkers to predict disease severity and outcome in SARS-CoV-2 infection
We investigated whether some specific taxa could be associated with the severity of the symptoms. ASVs were evaluated to determine core taxa along with the specific bacteria of each group of patients and samples (Figure 3A,B). Core taxa were identified using a detection threshold of 0.1 and a prevalence threshold of 0.1. In nasopharyngeal swabs, Venn diagram revealed that the three cohorts shared 51 core taxa. 60 specific bacteria were identified in patients with mild symptoms while 32 were observed in patients with moderate symptoms and 8 in patients with severe symptoms. When stools were considered, the three cohorts shared 159 core taxa. 27 were specific of mild patients, another 33 of patients with moderate symptoms and 27 of the severe patients (For more details see supplementary file 4).
Besides, the linear discriminant analysis (LEfSe) was performed to identify differential microorganisms for each group of patients (Figure 3C,D). In nasopharyngeal samples from mild patients, Burkholderia sp., Paraburkholderia sp. and Massilia sp. were determined; in moderate patients, Pseudomonas veronii, Stenotrophomonas rhizophila and Azotobacter chroococcum; and in severe patients, Mycoplasma salivarium, Prevotella dentalis, Leptotrichia and Haemophilus parainfluenzae. On the other hand, in stool samples, Bacteroides coprocola, Veillonella sp., Ruminococcus bicirculans and Sutterella stercoricanis were identified as predictors of mild condition; Prevotella stercorea, Bacteroides cellulosilyficus, Streptococcus salivarus, Bacteroides stercoris and Prevotella copri as predictors of moderate status; and Escherichia, Enterococcus durans, Alistipes onderdonkii, Prevotella timonensis and Prevotella bivia as markers of severe condition. Unlike the Venn diagram, the presence of a species in the LEfSe analysis does not imply its absence in the other groups, but rather indicates significant differences in abundance. Hence, detailed abundance of these bacteria can be found in supplementary files 5 and 6.
To further assess the role of these potential biomarkers in the prediction of COVID-19 severity, a correlation analysis was performed between these species and symptomatology. In summary, biomarkers identified in nasopharyngeal swabs in patients with severe condition, M. salivarium and Leptotrichia, showed a positive correlation with D dimer and cardiomyopathy, respectively (R > 0.3). In addition, the other two biomarkers related to the highest severity, H. parainfluenzae and P. dentalis, also showed a positive association with CRP, D dimer and cardiomyopathy, even though it was not statistically significant (R = 0.2). More precisely, M. salivarium and Leptotrichia along with P. dentalis showed a significantly negative correlation towards lymphocytes count (p<0.05; R<-0.25) (Figure 4A). In the case of stool samples, biomarkers for mild symptomatology (B. coprocola, R. bicirculans and S. stercoricanis) presented a negative correlation with CRP levels and respiratory rate (p<0.05; R < −0.25). In contrast, the severe biomarkers identified, presented important correlations towards dyspnoea, cardiomyopathy or respiratory rate. Concretely, P. bivia and P. timonensis, revealed a positive correlation with D dimer, ferritin levels and respiratory rate, as well as a negative correlation with lymphocyte count (R > 0.6) (Figure 4B). Additionally, these two species showed a significant positive correlation against CRP levels and a negative correlation towards lymphocyte count (p<0.05). Exact correlation and p-values are shown in supplementary file 7.
Identification of a novel microbiome-based COVID-19 prognosis approach
Considering the possible identified biomarkers and their relationship towards clinical variables, we proposed a new approach to predict disease severity in patients suffering SARS-CoV-2 infection by evaluating correlation among specific bacteria abundance from nasopharyngeal and stool samples The analysis revealed no important associations between nasopharyngeal and faecal microbiota in mild and moderate patients (Figure 5A,B). Nonetheless, in patients with severe symptoms the Spearman’s rho coefficient showed a significant positive correlation between P. timonensis towards P. dentalis and M. salivarium (Figure 5C). Consequently, the ratios P. timonensis / M. salivarium and P. timonensis / P. dentalis were calculated to determine whether they could be used as a predictor of COVID-19 severity. Both seemed to be significantly increased in patients with severe symptoms compared to those with mild or moderate symptomatology (p < 0.05; Kruskal-Walli) (Figure 5D,E). As a result, the ratio between the abundance of these bacteria could serve as reliable predictors of severity of COVID-19.
Discussion
Recent findings have evidenced the prominent role of the microbiome in several diseases, including those caused by viruses, proving that commensal microbiota could either enhance or hinder viral infections [30, 31]. A large body of evidence indicates that respiratory virus infections can induce a microbial imbalance in the airways, increasing the risk of secondary bacterial infections and worsening the prognosis [32]. Besides, alterations in the gut microbiota have also been noted during respiratory virus infections, likely influenced by the “gut-lung axis” [33]. In this regard, different studies have tried to establish relationships between microbiota composition and SARS-CoV-2 infection [34, 35] but just a few have explored microbiota compositional variations among patients with different symptomatology [36]. However, only a few studies have explored the nasopharyngeal-faecal axis as a potential biomarker of severity in patients infected with SARS-CoV-2. Hence, the present study provides some biomarkers located in nasopharyngeal and gut microbiota that could help to predict COVID-19 severity.
In this context, 106 patients with SARS-CoV-2 infection were included in this study and classified into 3 severity groups according to their symptoms. To ensure results were derived only from SARS-CoV-2 infection, included patients were not positive for other viral infections. Considering clinical parameters, the current study confirmed that age and gender are positively associated with more severe symptoms like dyspnoea, increased heart and respiratory rates together with lower oxygen saturation. Results that are in line with other COVID-19 studies conducted globally, which have shown that men and women are disproportionately affected. Data revealed that males suffered from more severe disease than females, including higher ICU admission rates, dyspnoea, increased heart rate, being all clinical signs for higher severity of the COVID-19 disease [37, 38]. Regarding some biochemical parameters those groups of patients with moderate or severe symptoms showed significantly increased levels of D-dimer, ferritin and CRP, which are typically described as biomarkers for COVID-19 severity [39–41]. Interestingly, previous studies have reported that alterations in the gut microbiota composition in COVID-19 patients could be also associated with disease severity, measured with the elevated concentration of the blood markers described above. [42–44]. In this sense, we have confirmed these results in our study as specific bacteria from severe condition positively correlates with the levels of ferritin, CRP, D-dimer or lymphocytes. Consequently, the microbiota could be key in the clinical phenotype of COVID-19 patients, although its specific contribution to the progression of the infection and a poor prognosis needs to be better understood.
The results of alpha diversity revealed only substantial changes in richness in the nasopharyngeal microbiome associated with moderate and severe symptoms. Although most of the studies have proposed that SARS-CoV-2 infection is associated with lower microbial diversity in nasopharyngeal samples [13, 45, 46], others did not find differences in alpha diversity composition among groups with different symptomatology [47, 48]. This could be explained by the diverse methods of sample collection, and also the SARS-CoV-2 variant and the treatment with antibiotics [49].
Furthermore, in the present study, beta diversity analysis showed that in both types of samples, every group of patients presented distinctly differentiated clusters. As previously reported, COVID-19 disease severity is more dependent on the presence or absence of certain bacteria rather than alterations in bacterial diversity and richness [ 50]. This argument was supported by the characterization of bacterial microbiota composition at phyla and genera levels. In nasopharyngeal swabs, the abundance of Bacteroidota and Actinobacteriota was reduced compared to mild patients, which goes in accordance with previous reports where these bacteria have been previously linked to a better prognosis of SARS-CoV-2 infection, since can exert beneficial effects by preventing respiratory diseases, including COVID-19 [51–55]. Moreover, the abundance of Bacillota and Pesudomonadota was increased in patients with severe symptomatology, thus supporting previous studies in which higher counts of Bacillota (Staphylococcus sp. and Streptococcus sp.) and Pesudomonadota (Pseudomonas sp.) were associated with moderate and severe symptoms of COVID-19 [56].
Regarding genera composition, Alistipes and Muribaculaceae were highly abundant in patients with mild symptoms. These bacteria have been well characterised in gut microbiota, but little is known about their presence in nasopharyngeal microbiota. Different experimental studies in mice have suggested their role in viral infections. Thus, Muribaculaceae was found in the lung microbiota in SARS-CoV-2 infected mice that were treated with a selective inhibitor of the main protease (Mpro) [57]. In the case of Alistipes, in a study conducted in children infected with respiratory syncytial virus (RVS), its abundance was reduced in the nasopharyngeal microbiota in comparison with non infected subjects [58]. Overall, these genera could be associated with a protective role against viral infection, and their higher presence in nasopharyngeal samples from mild COVID-19 patients could prevent the progression to severe disease. In contrast, the increased presence of other genera, such as Corynebacterium, Acinetobacter, Staphylococcus and Veillonella, was associated with the severity of SARS-CoV-2 infection. This is supported by previous studies in which these genera were associated with both disease severity and systemic inflammation [59]. In addition, higher abundance of Enterococcus was observed in severe patients, thus confirming other studies in critically ill patients [60].
On the contrary, findings at phylum level in stool samples did not reveal notable modifications among patient groups. However, in mild ill patients, different genera from Clostridia class (Clostridia, Coprococcus, Dorea, Lachnospiraceae, Roseburia and Ruminococcus), and Barnesiella were identified as highly abundant. While the class Clostridia was associated with a reduced production of proinflammatory cytokines in COVID-19 patients and in those who recovered from the infection [61, 62], Barnesiella prevents colonisation by antibiotic-resistant bacteria such as Enterococcus, which has been reported as a frequent cause of systemic infection in critically ill COVID-19 patients [63, 64]. Thus, the reduction of gut abundance of Barnesiella and Clostridia members could be associated with more severe symptoms in COVID-19 patients. Moreover, the specific genera detected in the severe illness group, Lachnocostridum, Anaerococcus and Peptoniphilus, have been previously recognised as opportunistic pathogens, which could induce gut inflammation and contribute to a poor prognosis [65].
Considering the variations observed in both nasopharyngeal and gut microbiota composition and their miscellaneous effects, the identification of specific bacteria could be used as a biomarker to predict COVID-19 severity. Accordingly, the present study has identified unique ASVs for the different severities of COVID-19. For nasopharyngeal samples, species belonging to the genus Lactobacillus (L. fermentum or L. reuteri) or Prevotella (P. pallens, P. ori and P. shahii) have been pinpointed. Interestingly, mild patients have also shown a higher content of Lactobacillus, which could also contribute to the host defence against viral infection at early stages, as reported in asymptomatic COVID-19 patients [66]. In the case of Prevotella sp., its role in COVID-19 infection has not been clearly elucidated, but previously published microbiome analysis has revealed that its abundance was higher in mild patients [67]. Nevertheless others have suggested that it could be a biomarker of critical phenotype in COVID-19 patients [68, 69]. It is interesting to highlight that Anaerococcus prevotii was one of the species exclusively found in stools in mild patients. This microorganism has already been linked to lower inflammation in COVID-19 patients [70]. Conversely, Coprobacillus cateniformis was only found in severe patients, and could be involved in the development of a worse condition in these patients through ACE2 upregulation [17]. Even though these bacteria were found to be unique for each group, LEfSe was performed to obtain specific severity prognostic biomarkers depending on the abundance threshold. Therefore, for mild patients, Burkholderia and Paraburkholderia were identified in nasopharyngeal swabs and B. coprocola and R. bicirculans in stool samples. Although the information regarding the first two species in humans is limited, a few studies have reported their presence in the commensal human microbiota [71, 72]. Contrarily, B. coprocola and R. bicirculans have been found in both healthy and COVID-19 patients, being the abundance of R. bicirculans reduced in infected subjects [73].
Correspondingly, in patients with moderate symptoms, P. veronii was detected in nasopharyngeal samples whereas P. stercorea, B. cellulosilyficus, B. stercoris and P. copri were identified in stool samples. In general, these findings agree with previous studies [74]. Xu et al. found that infected patients showed higher abundance of B. cellulosilyficus [75], whereas B. stercoris and P. copri were associated with ACE2 upregulation and increased proinflammatory cytokine production, respectively, in COVID-19 patients [73, 76].
Likewise, in critically ill patients, the biomarkers detected for nasopharyngeal microbiota were M. salivarium, P. dentalis, Leptotrichia and H. parainfluenzae, while, in stool samples, Escherichia sp., E. durans, A. Onderdonkii, P. timonensis and P. bivia were the species recognised as biomarkers. Both P. bivia and P. timonensis have been defined as unique microorganisms in COVID-19 patients microbiota [73, 77], whilst a higher abundance of M. salivarium, H. parainfluenzae and E. durans were related to poor prognosis [78–80]. When considering A. onderdonkii, it is a short-chain fatty acid-producing bacteria that has been reported to be inversely associated with COVID-19 severity [17, 50]; however, there are conflicting evidences regarding its pathogenicity that indicate that A. onderdonkii may have protective effects against some diseases, including COVID-19 [17]. Nonetheless, considering that Zuo T et al. employed a different methodology to analyse microbiota composition from stool samples, A. onderdonkii could be considered as a biomarker of severe condition in SARS-CoV-2 infected subjects.
Of note, the use of these bacteria as biomarkers of severity in SARS-CoV-2 infection is further supported by the fact that these species exhibited positive correlations with critical clinical variables mentioned above. Specifically, possible severe biomarkers from nasopharyngeal samples such as M. salivarium, H. parainfluenzae and P. dentalis showed a negative correlation towards lymphocytes count. This also happens for stool severe biomarkers P. bivia and P. timonensis. As it is well established that microbiota can modify blood cells count [81], it would not be a surprise that these bacteria could promote COVID-19 severity by lowering lymphocyte count. In addition, P. bivia and P. timonensis showed positive correlation with ferritin, CRP and D-dimer levels, as well as cardiomyopathy and respiratory rates. Several studies have revealed both the relationship between CRP levels, gut microbiota and COVID-19 severity [82], as well as the positive correlation of specific bacteria with D-dimer, CRP and the levels of pro-inflammatory mediators in plasma [83].
Finally, although the composition of the nasal and faecal microbiota in COVID-19 patients has been previously studied [32], the interaction between these microbiotas has not been extensively evaluated. In this study, we analyzed the relationship between specific bacteria from nasopharyngeal and stool samples, confirming a correlation between the abundances of species in these different sample types. Concretely, a strong positive correlation between P. timonensis (in stool samples) towards P. dentalis and M. salivarium (in nasopharyngeal samples) was found in severe patients. To maximise the potential use of these biomarkers, the ratio of the abundance of these species was also significantly increased within the highest severity of this condition. As a result, the ratio proposed in this study could be used as a novel predictor to identify critically ill COVID-19 patients as the ratio Bacillota and Bacteroidetes has been used as a marker of dysbiosis [84]. In this case, the ratio P. timonensis/P. dentalis and P. timonensis /M. Salivarium could be a prognostic tool for severe SARS-CoV-2, and its increase could be associated with a higher risk COVID-19 severity development.
Materials and methods
Ethics approval
The study was conducted in accordance with the declaration of Helsinki and the protocol approved by the Clinical Research Ethics Committee of Granada (CEIC) (ID of the approval omicovid-19 1133-N-20). All patients provided written informed consent before being included in the study. The samples were managed by the ibs.GRANADA Biobank following the protocols approved by the Andalusian Biomedical Research Ethics Coordinating Committee.
Subject recruitment and sample collection
A multicenter prospective observational cohort study was carried out between September 2020 and July 2021. Patients with SARS-CoV-2 infection were recruited from the University Hospital San Cecilio, the University Hospital Virgen de las Nieves, and the Primary Care centres, Salvador Caballero and Las Gabias in Granada (Spain). These patients were Caucasian and laboratory-confirmed SARS-CoV-2 positive by quantitative reverse transcription polymerase chain reaction (RT-qPCR) performed on nasopharyngeal swabs collected by healthcare practitioners. No confounding factors such as other viral infections were present. Patients were classified in three groups based on severity profile following the described guidelines [21]; mild (n=24), moderate (n=51) and severe/critical (n=31). Mild illness includes individuals who have any of the various signs and symptoms of COVID-19 (e.g., fever, cough, sore throat, malaise, headache, muscle pain, nausea, vomiting, diarrhoea, loss of taste and smell) but do not have shortness of breath, dyspnoea, or abnormal chest imaging. Moderate illness includes patients that show fever and respiratory symptoms with radiological findings of pneumonia. Severe/critical illness includes patients with any of the following criteria: respiratory distress (≥30 breaths/min), oxygen saturation ≤93% at rest, arterial partial pressure of oxygen (PaO2)/fraction of inspired oxygen (FiO2) ≤300 mmHg, respiratory failure and requiring mechanical ventilation, shock, and with other organ failures that required intensive care unit (ICU) care.
Nasopharyngeal swabs and stools samples from patients were collected by healthcare staff in the moderate and severe/critical cohorts, while the patients of the mild cohort provided stools self-sampled at home. Stools and nasopharyngeal swabs were collected in collection tubes containing preservative media (OMNIgene®•GUT, DNAGENOTEK®, Ottawa, Ontario, Canada) and stored at −80°C until processing.
Microbial DNA extraction, library preparation and next generation sequencing
For all faecal and nasopharyngeal samples, DNA was isolated according to the modified protocol reported by Rodríguez-Nogales et al. [22] using Qiagen Allprep PowerFecal DNA kit (Qiagen, Germany). DNA was quantified using Qubit dsDNA HS assay kit (12640ES60, Yeason Biotechnology, Shanghai, China) and total DNA was amplified by targeting variable regions V3-V4 of the bacterial 16 S rRNA gene. Quality control of amplified products was achieved by running a high-throughput Invitrogen 96-well-E-gel (Thermo Fisher Scientific, Waltham, MA, USA). PCR products from the same samples were pooled in one plate and normalised with the high-throughput Invitrogen SequalPrep 96-well Plate kit. Pool samples were sequenced using an Illumina MiSeq.
Bioinformatic tools
Paired end reads quality was checked with FastQC software [23]. Trimming of adapters and filtering of low quality sequences was performed with Trimmomatic [24]. Filtered reads were further processed with QIIME2 software (open access, Northern Arizona University, Flagstaff, AZ, USA) by employing DADA2 software [25] to carry out denoising steps to obtain amplicon sequence variants (ASVs). SILVA reference database (138 99% full length) was used for taxonomic assignment [26]. Microbiota results were further analyze with the help of different R packages. Alpha and beta diversity as well as relative abundance were appraised with the Phyloseq package [27]. Beta diversity differences were obtained with a Permutational Multivariate Analysis of Variance (PERMANOVA) included in the Vegan package (Eulerr along with microbiomeutilities package [28] was utilised for constructing Venn diagrams [29]. Meanwhile, microbial package was employed to perform linear discriminant analysis (LDA) effect size (LEfSe) with an LDA score of 3 [29].
Statistical analysis
All data presented were analyzed and visualized using R software. For numerical clinical variables, normality and homoscedasticity were assessed using the shapiro.test and leveneTest functions from the stats and car package, respectively. Data were displayed as mean ± standard deviation (SD) for normally distributed variables, whereas median and interquartile range (IQR) were used for variables with non-normal distributions.
Statistical differences were evaluated using ANOVA for parametric data or Kruskal-Wallis test for non-parametric data, both available in the stats package. When significant differences were identified, post-hoc group comparisons were conducted using Tukey’s method through the TukeyHSD function for ANOVA and Dunn’s test via the dunnTest function from the FSA package for Kruskal-Wallis.
Categorical variables were expressed as percentages, and statistical significance was determined using Fisher’s exact test with the fisher.test function from the stats package. Finally, for correlation analysis, Spearman’s correlation coefficient was employed for non-normally distributed variables, while Pearson’s correlation coefficient was used for normally distributed variables. The cor function from the stats package was used to calculate these coefficients, and the corrplot package was employed for visualization. Statistical significance was defined as a p-value of less than 0.05. To illustrate group differences, different letters (a, b or c) were used to denote statistically significant differences among groups. All data, models, and analytical output are on the linked GitHub repository https://github.com/albarodnog/COVID-INMUNOBIOTICS-Group.git.
Conclusion
This inter-individual variability between the COVID-19 patients could contribute to the different symptomatology observed. This study has identified a correlation between changes in the nasopharyngeal and stool microbiota with COVID-19 severity. A novel biomarker linked to severity of COVID-19 infection has been described based on changes in the abundance of bacterial species in nasopharyngeal and faecal samples. This knowledge can support the development of biomarkers to gauge disease severity and, also, the design of novel therapeutic strategies to mitigate adverse outcomes. Further investigations are imperative to explore how the association between nasopharyngeal and faecal microbiota can be modulated to uncover its role in enhancing immune health, preventing or treating SARS-CoV-2 infections, and fostering immunity.
Data availability statement
Participant data cannot be made publicly available due to the sensitive nature of the personal health data and privacy and confidentiality reasons. However, under certain conditions, these data could be accessible for statistical and scientific research. For further information, please contact the corresponding authors.
Acknowledgements
We acknowledge the collaboration of all the participants who voluntarily and selflessly participated in the study.
Additional information
Conflict of interest statement
All authors declare no interest.
Funding information
The research project was sup-ported by Government of Andalucia (Spain) (CV20-99908).
Author contributions
Benita Martín-Castaño, Margarita Martínez-Zaldívar, Emilio Mota, Fernando Cobo, Concepcion Morales-García, Marta Alvarez-Estevez, Federico García, Silvia Merlos, Paula García-Flores, Manuel Colmenero-Ruiz, José Hernández-Quero, María Nuñez were involved in the sample collection. Patricia Diez-Echave, Jorge García-García, Alba Rodríguez-Nogales, Maria Elena Rodríguez-Cabezas, Laura Hidalgo-García, Antonio Jesús Ruiz-Malagon, José Alberto Molina-Tijeras, María Jesús Rodríguez-Sojo and Anaïs Redruello were involved in the processing of simples and obtaining results. Rocio Morón and Emilio Fernández-Varón had access to the data and were involved in the conception and data analysis and interpretation. Alba Rodríguez-Nogales, Javier Martin, Maria Elena Rodriguez-Cabezas, Benita Martín-Castaño, Rocio Morón, Jorge García-García, Ángel Carazo and Julio Gálvez had access to the data and were involved in the conception and design of the work, data analysis and interpretation, critical revision of the article and final approval before submission. All authors reviewed the final manuscript and agreed to be account able for all aspects of the work.
References
- 1.WHO COVID-19 dashboardGeneva: World Health Organization
- 2.SARS-CoV-2 variants, spike mutations and immune escapeNat Rev Microbiol 19:409–424
- 3.Long covid-mechanisms, risk factors, and managementBMJ 374
- 4.Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: a systematic review and meta-analysisInt J Infect Dis 94:91–95
- 5.Genomewide Association Study of Severe Covid-19 with Respiratory FailureN Engl J Med 383:1522–1534
- 6.Autoantibodies against type I IFNs in patients with life-threatening COVID-19Science 370
- 7.A pneumonia outbreak associated with a new coronavirus of probable bat originNature 579:270–273
- 8.SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimensJ Med Virol 92:1676–1680
- 9.Isolation of infectious SARS-CoV-2 from urine of a COVID-19 patientEmerg Microbes Infect 9:991–993
- 10.Digestive system is a potential route of COVID-19: an analysis of single-cell coexpression pattern of key proteins in viral entry processGut 69:1010–1018
- 11.SARS-CoV-2 productively infects human gut enterocytesScience 369:50–54
- 12.TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytesSci Immunol 5
- 13.The nasopharyngeal microbiome in COVID-19Emerg Microbes Infect 12
- 14.Gut and airway microbiota dysbiosis and their role in COVID-19 and long-COVIDFront Immunol 14
- 15.Secondary infection in COVID-19 critically ill patients: a retrospective single-center evaluationBMC Infect Dis 22
- 16.Alterations of the Gut Microbiota in Patients With Coronavirus Disease 2019 or H1N1 InfluenzaClin Infect Dis 71:2669–2678
- 17.Alterations in Gut Microbiota of Patients With COVID-19 During Time of HospitalizationGastroenterology 159:944–955
- 18.Altered oral and gut microbiota and its association with SARS-CoV-2 viral load in COVID-19 patients during hospitalizationNPJ Biofilms Microbiomes 7
- 19.Integrated gut virome and bacteriome dynamics in COVID-19 patientsGut Microbes 13:1–21
- 20.Disentangling the interactions between nasopharyngeal and gut microbiome and their involvement in the modulation of COVID-19 infectionMicrobiology Spectrum 11:e02194–23
- 21.NIH COVID-19 Treatment Guidelines
- 22.Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: impact on microRNAs expression and microbiota compositionMol Nutr Food Res 61
- 23.FastQC: A quality control tool for high throughput sequence dataBabraham Bioinformatics
- 24.Trimmomatic: A flexible trimmer for Illumina Sequence DataBioinformatics
- 25.DADA2: High-resolution sample inference from Illumina amplicon dataNat Methods 13:581–3
- 26.Species abundance information improves sequence taxonomy classification accuracyNat Commun 10
- 27.Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2Genome Biology 15
- 28.microbiomeutilities: Utilities for Microbiome AnalyticsThe microbiome R package relies on the independently developed
- 29.R Foundation for Statistical Computing
- 30.A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjectsPLoS Pathog 10
- 31.Abnormal blood microbiota profiles are associated with inflammation and immune restoration in HIV/AIDS individualsmSystems 8
- 32.Assessment of microbiota in the gut and upper respiratory tract associated with SARS-CoV-2 infectionMicrobiome 11
- 33.Emerging pathogenic links between microbiota and the gut-lung axisNat Rev Microbiol 15:55–63
- 34.The human microbiome and COVID-19: A systematic reviewPLoS One 16
- 35.The Microbiota/Host Immune System Interaction in the Nose to Protect from COVID-19Life (Basel 10
- 36.Metagenomic assessment of gut microbial communities and risk of severe COVID-19Genome Med 15
- 37.Temporal trends of sex differences for COVID-19 infection, hospitalisation, severe disease, intensive care unit (ICU) admission and death: a meta-analysis of 229 studies covering over 10M patientsF1000Res 11
- 38.Aging, Male Sex, Obesity, and Metabolic Inflammation Create the Perfect Storm for COVID-19Diabetes 69:1857–1863
- 39.Hyperferritinemia in critically ill COVID-19 patients - Is ferritin the product of inflammation or a pathogenic mediator?Clin Chim Acta 509:249–251
- 40.D-dimer level in COVID-19 infection: a systematic reviewExpert Rev Hematol 13:1265–1275
- 41.Increased Serum Levels of Hepcidin and Ferritin Are Associated with Severity of COVID-19Med Sci Monit 26
- 42.Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndromeGut 71:544–552
- 43.Multi-kingdom gut microbiota analyses define COVID-19 severity and post-acute COVID-19 syndromeNat Commun 13
- 44.Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19Gut 70:698–706
- 45.Characterization of the Human Oropharyngeal Microbiomes in SARS-CoV-2 Infection and Recovery PatientsAdv Sci (Weinh 8
- 46.Nasopharyngeal microbiome reveals the prevalence of opportunistic pathogens in SARS-CoV-2 infected individuals and their association with host typesMicrobes Infect 24
- 47.Nasopharyngeal Microbiota Profiling of SARS-CoV-2 Infected PatientsBiol Proced Online 22
- 48.SARS-CoV-2 does not have a strong effect on the nasopharyngeal microbial compositionSci Rep 11
- 49.A cross-sectional study on the nasopharyngeal microbiota of individuals with SARS-CoV-2 infection across three COVID-19 waves in IndiaFront Microbiol 14
- 50.The relationship between gut microbiota and COVID-19 progression: new insights into immunopathogenesis and treatmentFront Immunol 14
- 51.Nasal Microbiome in COVID-19: A Potential Role of Corynebacterium in AnosmiaCurr Microbiol 80
- 52.Oral booster probiotic bifidobacteria in SARS-COV-2 patientsInt J Immunopathol Pharmacol 35
- 53.Characterization of oral bacterial and fungal microbiome in recovered COVID-19 patientsBMC Microbiol 23
- 54.The nasal cavity microbiota of healthy adultsMicrobiome 2
- 55.Nasal Bacteriomes of Patients with Asthma and Allergic Rhinitis Show Unique Composition, Structure, Function and InteractionsMicroorganisms 11
- 56.Incidence of co-infections and superinfections in hospitalized patients with COVID-19: a retrospective cohort studyClin Microbiol Infect 27:83–88
- 57.Mild and Severe SARS-CoV-2 Infection Induces Respiratory and Intestinal Microbiome Changes in the K18-hACE2 Transgenic Mouse ModelMicrobiol Spectr 9
- 58.Nasal Microbiota in RSV BronchiolitisMicroorganisms 8
- 59.Metagenomic analysis reveals oropharyngeal microbiota alterations in patients with COVID-19Signal Transduct Target Ther 6
- 60.Signatures of COVID-19 Severity and Immune Response in the Respiratory Tract MicrobiomemBio 12
- 61.Correlation Analysis between Gut Microbiota Alterations and the Cytokine Response in Patients with Coronavirus Disease during HospitalizationMicrobiol Spectr 10
- 62.Alterations in Gut Microbiota Composition in Patients with COVID-19: A Pilot Study of Whole Hypervariable 16S rRNA Gene SequencingBiomedicines 11
- 63.Intestinal microbiota containing Barnesiella species cures vancomycin-resistant Enterococcus faecium colonizationInfect Immun 81:965–73
- 64.Enterococcal bloodstream infections in critically ill patients with COVID-19: a case seriesAnn Med 53:1779–1786
- 65.Gram-positive anaerobic cocci--commensals and opportunistic pathogensFEMS Microbiol Rev 37:520–53
- 66.Lactobacillus plantarum induces innate cytokine responses that potentially provide a protective benefit against COVID-19: A single-arm, double-blind, prospective trial combined with an in vitro cytokine response assayExp Ther Med 23
- 67.Comparison of the respiratory tract microbiome in hospitalized COVID-19 patients with different disease severityJ Med Virol 94:5284–5293
- 68.Inflammation-type dysbiosis of the oral microbiome associates with the duration of COVID-19 symptoms and long COVIDJCI Insight 6
- 69.Metatranscriptomic analysis revealed Prevotella as a potential biomarker of oropharyngeal microbiomes in SARS-CoV-2 infectionFront Cell Infect Microbiol 13
- 70.Clinical implications of gut microbiota and cytokine responses in coronavirus disease prognosisFront Immunol 14
- 71.Comparative analysis of the gut microbiota composition between knee osteoarthritis and Kashin-Beck disease in Northwest ChinaArthritis Res Ther 24
- 72.Plasma metagenomics reveals regional variations of emerging and re-emerging pathogens in Chinese blood donors with an emphasis on human parvovirus B19One Health 17
- 73.Microbiome Profiling Using Shotgun Metagenomic Sequencing Identified Unique Microorganisms in COVID-19 Patients With Altered Gut MicrobiotaFront Microbiol 12
- 74.Altered infective competence of the human gut microbiome in COVID-19Microbiome 11
- 75.Integrated analysis of gut microbiome and host immune responses in COVID-19Front Med 16:263–275
- 76.COVID-19 severity is associated with population-level gut microbiome variationsFront Cell Infect Microbiol 12
- 77.Evaluation of co-circulating pathogens and microbiome from COVID-19 infectionsPLoS One 17
- 78.Microbial signatures in the lower airways of mechanically ventilated COVID-19 patients associated with poor clinical outcomeNat Microbiol 6:1245–1258
- 79.Longitudinal study across SARS-CoV-2 variants identifies transcriptionally active microbes (TAMs) associated with Delta severityiScience 26
- 80.Increased rates of secondary bacterial infections, including Enterococcus bacteremia, in patients hospitalized with coronavirus disease 2019 (COVID-19)Infect Control Hosp Epidemiol 43:1416–1423
- 81.Gut microbiota promote hematopoiesis to control bacterial infectionCell Host Microbe 15:374–81
- 82.Gut Microbiota Diversity and C-Reactive Protein Are Predictors of Disease Severity in COVID-19 PatientsFront Microbiol 12
- 83.Gut Microbiota Dysbiosis Correlates with Abnormal Immune Response in Moderate COVID-19 Patients with FeverJ Inflamm Res 14:2619–2631
- 84.Microbial ecology: human gut microbes associated with obesityNature 444:1022–3
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
- Reviewed Preprint version 2:
Copyright
© 2024, Martin-Castaño et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 371
- downloads
- 24
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.