Journal List > J Korean Med Sci > v.38(15) > 1516082546

Zhang, Zhou, Ma, Chen, Tang, Yang, Li, Liang, Xue, Liu, Zhang, and Wang: Gut Microbiota Dysbiosis Correlates With Long COVID-19 at One-Year After Discharge

Abstract

Background

Long coronavirus disease 2019 (COVID-19) in recovered patients (RPs) is gradually recognized by more people. However, how long it will last and the underlining mechanism remains unclear.

Methods

We conducted a prospective follow-up study to evaluate the long-term symptoms and clinical indices of RPs at one-year after discharge from Union Hospital, Wuhan, China between December 2020 to May 2021. We also performed the 16S rRNA sequencing of stool samples from RPs and healthy controls (HCs) and analyzed the correlation between the gut microbiota and long COVID-19.

Results

In total, 187 RPs were enrolled, among them, 84 (44.9%) RPs reported long COVID-19 symptoms at one-year after discharge. The most common long-term symptoms were cardiopulmonary symptoms, including chest tightness after activity (39/187, 20.9%), palpitations on exercise (27/187, 14.4%), sputum (21/187, 11.2%), cough (15/187, 8.0%) and chest pain (13/187, 7.0%), followed by systemic symptoms including fatigue (34/187, 18.2%) and myalgia (20/187, 10.7%), and digestive symptoms including constipation (14/187, 7.5%), anorexia (13/187, 7.0%), and diarrhea (8/187, 4.3%). Sixty-six (35.9%) RPs presented either anxiety or depression (42/187 [22.8%] and 53/187 [28.8%] respectively), and the proportion of anxiety or depression in the long symptomatic group was significantly higher than that in the asymptomatic group (41/187 [50.6%] vs. 25/187 [24.3%]). Compared with the asymptomatic group, scores of all nine 36-Item Short Form General Health Survey domains were lower in the symptomatic group (all P < 0.05). One hundred thirty RPs and 32 HCs (non-severe acute respiratory syndrome coronavirus 2 infected subjects) performed fecal sample sequencing. Compared with HCs, symptomatic RPs had obvious gut microbiota dysbiosis including significantly reduced bacterial diversities and lower relative abundance of short-chain fatty acids (SCFAs)-producing salutary symbionts such as Eubacterium_hallii_group, Subdoligranulum, Ruminococcus, Dorea, Coprococcus, and Eubacterium_ventriosum_group. Meanwhile, the relative abundance of Eubacterium_hallii_group, Subdoligranulum, and Ruminococcus showed decreasing tendencies between HCs, the asymptomatic group, and the symptomatic group.

Conclusion

This study demonstrated the presence of long COVID-19 which correlates with gut microbiota dysbiosis in RPs at one-year after discharge, indicating gut microbiota may play an important role in long COVID-19.

Graphical Abstract

jkms-38-e120-abf001.jpg

INTRODUCTION

The global pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been ongoing for almost three years, resulting in tremendous numbers of infections. However, for infectors, recovering from the acute disease is not the end, many of them experience ongoing symptoms. As reported by previous follow-up studies of COVID-19,123 a portion of recovered patients (RPs) continue to complain of a multitude of symptoms for months, and these post COVID-19 symptoms can lead to long-term effects on the quality of life and mental health of RPs, which we call long COVID-19.456 However, how long and why the symptoms persist in long COVID-19 patients are largely unclear.
The gut microbiota which has been widely known to regulate immunity and inflammation has a profound impact on human health and disease.78 Microbiota dysbiosis may be involved in pathogenesis and lead to susceptibility to some diseases.7 Several studies have reported gut microbiota alteration in acute and early recovery stage patients with COVID-19.91011 Our previous studies also demonstrated gut microbiota dysbiosis including the increase of opportunistic pathogens and loss of commensal bacteria that was correlated with the symptoms of acute stage and convalescent stage at three-month after discharge.1213 To investigate whether it returns to normal at one-year after discharge and how it acts on long COVID-19, in this study, we enrolled a large number of RPs with different disease severities in the acute phase and did a comprehensive evaluation of the potential effect of gut microbiota on long COVID-19 at one-year after discharge through a series of tests including symptoms, health status and psychiatric questionnaires, laboratory tests, pulmonary function tests (PFTs), chest computed tomography (CT), 6-minute walk tests (6MWT) and 16S rRNA sequencing of stool samples from RPs and healthy controls (HCs).

METHODS

Study design and participants

In this prospective observational follow-up study, we included RPs with COVID-19 at one-year ± one month after discharge from Wuhan Union Hospital, China, between December 2020 and May 2021. In the early stage of the pandemic outbreak, Wuhan Union hospital was designated as the first-line therapy hospital, and the patients hospitalized all had discomfort symptoms and CT abnormalities, so there were no mild patients according to the classification of disease severity of World Health Organization COVID-19 guidelines.14 Study inclusion and exclusion criteria were described in our previous study.1215 The SARS-CoV-2 strain which infected our patients was originally found in the early stage of the pandemic outbreak in Wuhan, Hubei, China, and we had sequenced full genomes of the virus which we showed in our previous article published in European Respiratory Journal.16 Additionally, all patients were unvaccinated.

Evaluation of conditions of RPs at one-year after discharge

RPs were interviewed with symptom questionnaires, 36-Item Short Form General Health Survey (SF-36), Self-Rating Anxiety Scale (SAS), and Self-Rating Depression Scale (SDS), and underwent laboratory tests including complete blood counts, blood biochemistry, C-reactive protein (CRP), lymphocyte subset counts, and serum immunoglobulin (Ig) M and IgG antibodies, PFTs, chest CT scans, and 6MWT. The specific assessment methods of clinical parameters were displayed in Supplementary Data 1.

Stool samples collection and 16s rRNA sequencing

Fecal samples were collected from RPs and age-, sex-, body mass index (BMI)-, and comorbidities-matched HCs. Under the strict epidemic prevention and control measures in China including timely closure of cities, nationwide nucleic acid and IgM/IgG antibodies tests for SARS-CoV-2, combined with examination of epidemiological histories and symptoms, all residents in Wuhan were screened for infection. Subjects with no epidemiological histories, negative SARS-CoV-2 nucleic acid and IgM/IgG antibodies were defined to be non-COVID-19 subjects, and HCs of our study were all non-COVID-19 subjects. Besides, the enrolled RPs and HCs were all residents of Wuhan city, and subjects who received antibiotics, probiotics, or both within 12 weeks before enrollment were excluded from collecting stool samples. Stool specimens (5–10 g) were collected in standard sterile sample pots with an integral spoon after defecation, and were processed in the laboratory within 30 minutes after collection and stored at −80°C until analysis. Detailed 16s rRNA sequencing process was displayed in Supplementary Data 1.

Statistical analysis

Continuous variables were expressed as median (interquartile range) and compared using independent t-test, Mann-Whitney U test, one-way analysis of variance, or Kruskal-Wallis test when appropriate; categorical variables were expressed as number (%) and compared using χ2 test or Fisher’s exact test when appropriate. For correlation analysis, Spearman rank test was performed. Statistical analysis was performed using SPSS version 25.0 (IBM Corp., Armonk, NY, USA). A two-sided P value of < 0.05 was considered statistically significant.

Ethics statement

The study was approved by the Ethics Committee of the Wuhan Union Hospital (2020-0149-02). All participants provided written informed consent prior to participation.

RESULTS

Demographics and clinical characteristics

One hundred eighty-seven RPs were recruited in our study (a detailed flowchart was presented in Supplementary Fig. 1). There were 91 (48.7%) men and the median age was 58.00 (48.00–66.00) years.
The common comorbidities were hypertension (34.8%), followed by chronic pulmonary diseases (18.2%), chronic digestive system diseases (18.2%), hyperlipidemia (16.0%) and diabetes (14.4%). There were 105 (56.1%) moderate, 62 (33.2%) severe, and 20 (10.7%) critical patients. During hospitalization, 3.7% RPs were admitted to intensive care unit, 78.6% RPs were given antibiotic therapy, and the median hospital stay days were 27.00 (18.00–39.00) days.

Long-term symptoms, physical and psychiatric functions, laboratory findings, lung functions and lung CT of RPs at one-year after discharge

At one-year after discharge, 84 (44.9%) RPs reported one or more long COVID-19 symptoms. The most common long-term symptoms were cardiopulmonary symptoms, including chest tightness after activity (39/187, 20.9%), palpitations on exercise (27/187, 14.4%), sputum (21/187, 11.2%), cough (15/187, 8.0%) and chest pain (13/187, 7.0%), followed by systemic symptoms including fatigue (34/187, 18.2%) and myalgia (20/187, 10.7%), and digestive symptoms including constipation (14/187, 7.5%), anorexia (13/187, 7.0%), and diarrhea (8/187, 4.3%) (Table 1). There were no differences between the symptomatic group and the asymptomatic group in terms of sex, age, BMI, comorbidities, and diseases severity in acute infection stage (Table 1). 66 (35.9%) RPs presented either anxiety or depression (42/187 [22.8%] and 53/187 [28.8%], respectively), and the proportion of anxiety or depression in the symptomatic group was significantly higher than that in the asymptomatic group (41/187 [50.6%] vs. 25/187 [24.3%], P < 0.001). Scores of all the SF-36 domains except for the role-emotional domain of RPs were lower than the norm, especially that of general health and healthy transition domains, and the scores of all domains in the symptomatic group were lower compared to the asymptomatic group (all P < 0.05). The medians of all the laboratory findings including lymphocyte subset counts of RPs were within the normal range at one-year after discharge, while in the symptomatic group, the medians of CD3+ T cells and CD4+ T cells were lower than that in the asymptomatic group (Table 2). The 99.4% RPs had positive SARS-CoV-2 IgG antibodies at one-year after discharge, while 11.8% still had positive SARS-CoV-2 IgM (Supplementary Table 1). One hundred seventy-eight participants finished PFTs. Most patients had anomalies of diffusion capacity, with a reduced carbon monoxide diffusing capacity (DLCO)%pred in 43.2% RPs. Forced expiratory volume in 1 second (FEV1)%pred, forced vital capacity (FVC)%pred, FEV1/FVC%pred, vital capacity (VC)%pred, total lung capacity (TLC)%pred, residual volume (RV)%pred, and functional residual capacity (FRC)%pred were not within normal ranges in 9.6%, 4.5%, 16.3%, 5.6%, 19.5%, 39.6% and 30.2% RPs, respectively. However, there were no significant differences in all parameters of lung functions between the symptomatic group and the asymptomatic group. The median of 6-minute walk distance (6MWD) of RPs was 477.00 (434.25–537.00) m with no statistical difference between the symptomatic group and the asymptomatic group. 185 subjects completed lung CT. The proportions of normal, ground-glass opacity (GGO), fibrosis and GGO + fibrosis were 48.1%, 12.4%, 19.5%, and 20.0% respectively, and the median of CT score was 1.00 (0.00–6.00). The symptomatic group and the asymptomatic group had similar CT image proportions and CT scores (Supplementary Table 2).
Table 1

Demographics and clinical characteristics of RPs at one-year after discharge

jkms-38-e120-i001
Characteristics Total (n = 187) Symptomatic group (n = 84) Asymptomatic group (n = 103) P value
Sex 0.084
Male 91 (48.7) 35 (41.7) 56 (54.4)
Female 96 (51.3) 49 (58.3) 47 (45.6)
Age, yr 58.00 (48.00–66.00) 57.00 (45.00–67.00) 59.00 (48.00–66.00) 0.729
BMI, kg/m2 25.39 (23.31–27.59) 25.27 (23.33–28.00) 25.82 (23.31–27.47) 0.699
Smokers 34 (18.2) 12 (14.3) 22 (21.4) 0.212
Comorbidities
Hypertension 65 (34.8) 24 (28.6) 41 (39.8) 0.109
Diabetes 27 (14.4) 15 (17.9) 12 (11.7) 0.230
Coronary heart disease 16 (8.6) 6 (7.1) 10 (9.7) 0.533
Chronic pulmonary diseasesa 34 (18.2) 13 (15.5) 21 (20.4) 0.386
Chronic digestive system diseasesb 34 (18.2) 18 (21.4) 16 (15.5) 0.299
Hyperlipidemia 30 (16.0) 12 (14.3) 18 (17.5) 0.554
Hypothyroidism 5 (2.7) 3 (3.6) 2 (1.9) 0.659
Chronic hepatitis B 3 (1.6) 1 (1.2) 1 (1.9) 1.000
Cancer 2 (1.1) 0 (0.0) 2 (1.9) 0.503
Chronic kidney disease 2 (1.1) 2 (2.4) 0 (0.0) 0.200
Information during hospitalization
Disease severity status 0.350
Moderate 105 (56.1) 45 (53.6) 60 (58.3)
Severe 62 (33.2) 32 (38.1) 30 (29.1)
Critical 20 (10.7) 7 (8.3) 13 (12.6)
Highest oxygen support mode 0.783
No oxygen 36 (19.3) 18 (21.4) 18 (17.5)
Nasal catheter or mask 123 (65.8) 54 (64.3) 69 (67.0)
HFNC or NIV or IMV or ECMO 28 (15.0) 12 (14.3) 16 (15.5)
ICU admission 7 (3.7) 4 (4.8) 3 (2.9) 0.703
Antibiotic therapy 147 (78.6) 71 (84.5) 76 (73.8) 0.075
Length of hospital stay, days 27.00 (18.00–39.00) 25.00 (17.00–40.00) 27.00 (20.00–38.50) 0.410
Symptoms after discharge
Any one of the following 84 (44.9) - - -
Cardiopulmonary symptoms
Chest tightness after activity 39 (20.9) - - -
Palpitation on exercise 27 (14.4) - - -
Sputum 21 (11.2) - - -
Cough 15 (8.0) - - -
Chest pain 13 (7.0) - - -
Digestive symptoms
Constipation 14 (7.5) - - -
Anorexia 13 (7.0) - - -
Diarrhea 8 (4.3) - - -
Systemic symptoms
Fatigue 34 (18.2) - - -
Myalgia 20 (10.7) - - -
Data are given as median (interquartile range) or number (%).
RPs = recovered patients, BMI = body mass index, HFNC = high-flow nasal cannula for oxygen therapy, NIV = non-invasive ventilation, IMV = invasive mechanical ventilation, ECMO = extracorporeal membrane oxygenation, ICU = intensive care unit.
aChronic pulmonary diseases include chronic bronchitis, emphysema, chronic obstructive pulmonary disease, tuberculosis, and bronchiectasis, etc.
bChronic digestive system diseases include chronic gastritis and chronic ulcer, etc.
Table 2

Anxiety or depression, SF-36, and laboratory findings of RPs at one-year after discharge

jkms-38-e120-i002
Variables Total Symptomatic group Asymptomatic group P value
Anxiety or depression status (n = 184, 81/103)
Either anxiety or depression 66 (35.9) 41 (50.6) 25 (24.3) < 0.001
Anxiety 42 (22.8) 30 (37.0) 12 (11.7) < 0.001
Depression 53 (28.8) 32 (39.5) 21 (20.4) 0.004
mMRC dyspnea scale 0.001
0 86 (46.0) 26 (31.0) 60 (58.3)
1–2 93 (49.7) 54 (64.3) 39 (37.9)
3–4 8 (4.3) 4 (4.8) 4 (4.8)
SF-36 (n = 183, 80/103)
PF 85.00 (70.00–95.00) 75.00 (51.25–85.00) 90.00 (80.00–95.00) < 0.001
RP 100.00 (50.00–100.00) 75.00 (0.00–100.00) 100.00 (75.00–100.00). < 0.001
BP 84.00 (74.00–100.00) 74.00 (64.50–87.50) 84.00 (74.00–100.00) < 0.001
GH 55.00 (40.00–75.00) 40.00 (30.00–60.00) 70.00 (50.00–85.00) < 0.001
VT 75.00 (60.00–85.00) 65.00 (50.00–75.00) 80.00 (70.00–90.00) < 0.001
SF 87.50 (62.50–100.00) 75.00 (50.00–87.50) 100.00 (75.00–100.00) < 0.001
RE 100.00 (33.30–100.00) 66.70 (0.00–100.00) 100.00 (66.70–100.00) < 0.001
MH 72.00 (60.00–80.00) 68.00 (52.00–75.75) 76.00 (68.00–88.00) < 0.001
HT 50.00 (25.00–75.00) 25.00 (25.00–75.00) 50.00 (25.00–75.00) < 0.001
Laboratory findings (n = 176, 100/76)
WBC, G/L 6.23 (5.25–7.19) 6.09 (5.20–7.40) 6.31 (5.26–7.06) 0.911
Neutrophils, G/L 3.53 (2.84–4.23) 3.63 (2.96–4.17) 3.49 (2.74–4.31) 0.544
Lymphocytes, G/L 2.02 (1.68–2.45) 1.98 (1.66–2.52) 2.05 (1.70–2.44) 0.682
NLR 1.64 (1.32–2.22) 1.63 (1.35–2.26) 1.66 (1.27–2.20) 0.482
Monocyte, G/L 0.38 (0.32–0.43) 0.38 (0.32–0.43) 0.37 (0.31–0.43) 0.670
ALT, U/L 23.50 (16.00–31.00) 25.00 (17.25–40.75) 21.50 (16.00–29.75) 0.035
AST, U/L 25.00 (21.00–29.00) 26.00 (21.00–30.00) 24.00 (21.00–28.00) 0.167
TBIL, µmol/L (n = 102, 43/59) 14.55 (11.00–18.75) 14.40 (10.20–18.20) 14.60 (11.20–19.70) 0.205
Scr, µmol/L 67.85 (55.95–79.98) 66.40 (56.50–82.15) 68.10 (54.26–79.85) 0.979
BUN, mmol/L 5.34 (4.69–6.23) 5.27 (4.67–6.27) 5.36 (4.71–6.21) 0.908
GFR, mL/min/1.73m2 (n = 102, 43/59) 94.72 (79.20–104.96) 94.57 (75.71–104.52) 96.30 (82.02–105.83) 0.730
CRP, mg/L (n= 146, 63/83) 4.93 (2.60–5.32) 4.94 (2.90–5.32) 4.87 (2.55–5.29) 0.498
Percentages of lymphocyte subset count (n = 40, 18/22)
CD3+ T cells 64.00 (55.18–72.88) 57.40 (47.98–71.38) 68.15 (59.80–76.25) 0.036
CD4+ T cells 33.40 (25.75–39.30) 28.20 (24.05–35.60) 35.75 (30.13–40.60) 0.040
CD8+ T cells 21.65 (20.53–28.05) 22.35 (17.35–26.08) 21.65 (20.80–29.40) 0.422
B cells 10.15 (7.43–14.43) 10.35 (8.23–15.30) 9.95 (7.38–12.88) 0.488
NK cells 20.40 (15.73–34.15) 27.85 (16.48–38.05) 19.40 (14.65–25.10) 0.068
Data are given as median (interquartile range) or number (%).
SF-36 = 36-Item Short Form General Health Survey, RPs = recovered patients, COVID-19 = coronavirus disease 2019, mMRC dyspnea scale = modified medical research council dyspnea scale, PF = physical functioning, BP = bodily pain, RP = role physical, GH = general health, VT = vitality, SF = social functioning, RE = role-emotional, MH = mental health, HT = healthy transition, WBC = white blood cells, NLR = neutrophil/lymphocyte ratio, ALT = alanine aminotransferase, AST = aspartate aminotransferase, TBIL = total bilirubin, Scr = serum creatinine, BUN = blood urea nitrogen, GFR = glomerular filtration rate, CRP = C-reactive protein, NK = natural killer.

Correlations between long COVID-19 symptoms and laboratory findings, lung functions, total CT scores as well as 6MWD

The white blood cells and neutrophils were positively correlated with diarrhea (R = 0.205, P = 0.006; R = 0.183, P = 0.015). The lymphocytes were negatively correlated with palpitation (R = −0.158, P = 0.036) and fatigue (R = −0.194, P = 0.010). Alanine aminotransferase (ALT) was positively correlated with palpitation (R = 0.174, P = 0.021). Total bilirubin (TBIL) was negatively correlated with constipation (R = −0.200, P = 0.030). CRP was positively correlated with chest tightness (R = 0.217, P = 0.008), chest pain (R = 0.163, P = 0.049) and myalgia (R = 0.247, P = 0.003). CD3+ T cells and CD4+ T cells were negatively correlated with “any symptoms” (R = 0.335, P = 0.034; R = 0.329, P = 0.038). CD8+ T cells were negatively correlated with palpitation (R = −0.341, P = 0.031). FEV1%pred was negatively correlated with sputum (R = −0.153, P = 0.042). DLCO%pred was negatively correlated with chest tightness (R = −0.218, P = 0.004), palpitation (R = −0.188, P = 0.015), anorexia (R = −0.196, P = 0.011), and fatigue (R = −0.262, P = 0.001). TLC%pred was negatively correlated with chest tightness (R = −0.152, P = 0.049), chest pain (R = −0.176, P = 0.022), anorexia (R = -0.229, P = 0.003), fatigue (R = −0.178, P = 0.020) and myalgia (R = −0.178, P = 0.021). 6MWD was negatively correlated with myalgia (R = −0.210, P = 0.006). Correlations analysis results are shown in Fig. 1 and Supplementary Table 3.
Fig. 1

Correlations between long-term symptoms and laboratory findings, lung functions, total CT scores as well as 6MWD of RPs with COVID-19 at one-year after discharge. Spearman correlation coefficient is indicated using a color gradient: yellow indicates positive correlation; purple indicates negative correlation.

CT = computed tomography, 6MWD = 6-minute walk distance, RPs = recovered patients, COVID-19 = coronavirus disease 2019, WBC = white blood cells, ALT = alanine aminotransferase, AST = aspartate aminotransferase, TBIL = total bilirubin, BUN = blood urea nitrogen, Scr = serum creatinine, CRP = C-reactive protein, NK = natural killer, FEV1 = forced expiratory volume in 1 second, FVC = forced vital capacity, DLCO = carbon monoxide diffusing capacity, TLC = total lung capacity, VC = vital capacity.
*P < 0.05, **P < 0.01.
jkms-38-e120-g001

Gut microbiota composition and alteration among the symptomatic group, the asymptomatic group of RPs and HCs

Fifty-five symptomatic RPs, 75 asymptomatic RPs, and 32 age, sex, BMI and comorbidities matched HCs performed gut microbiota analysis (Table 3). After merging and filtering, 8,023,050 high-quality sequences were acquired from 165 fecal samples by 16s rRNA sequencing with an average sequence number of 49,524 per sample. As estimated by the Sobs and Shannon index, the community richness and diversity of gut microbiota in the symptomatic RPs were significant decreased than that in HCs (P = 0.002 and P = 0.003, respectively) with increasing tendencies between the symptomatic group, the asymptomatic group, and HCs (Fig. 2A and B). The number of operational taxonomic units (OTUs) in the symptomatic group, the asymptomatic group, and HCs were 881, 943, and 855 respectively, and 638 OTUs overlapped among the three groups (Fig. 2C). Principal coordinate analysis (PCoA) of Bray-Curtis distances was performed to display microbiome space between samples, and the result showed that there were significant differences in gut microbial communities among the three groups (P = 0.005), especially between the symptomatic group and HCs (P = 0.004) (Fig. 2D). To investigate changes in the gut microbiota of RPs, we subsequently assessed relative abundance at the phylum, class, family, and genus levels. At the phylum level, Firmicutes, Actinobacteriota, Proteobacteria, and Bacteroidota were the most dominant phyla in all three groups. Firmicutes was less abundant, but Actinobacteriota and Proteobacteria were more abundant in the symptomatic RPs compared to HCs although the differences were not significant (Fig. 3A). At the class level, the relative abundance of Clostridia and Coriobacteriia were significantly decreased in the symptomatic group of RPs than HCs (P = 0.008 and P = 0.002, respectively), while Bacilli, Actinobacteria, and Gammaproteobacteria were enriched in the symptomatic group than HCs although significant differences were not detected (Fig. 3B). In the class Clostridia of the phylum Firmicutes, seven genera including Eubacterium_hallii_group, Agathobacter, Subdoligranulum belonging to Oscillospiraceae family, Ruminococcus belonging to Ruminococcaceae family, Dorea, Coprococcus, and Eubacterium_ventriosum_group, which were almost short-chain fatty acids (SCFAs)-producing symbionts, significantly depleted in the symptomatic group than HCs, and the relative abundance of Eubacterium_hallii_group, Subdoligranulum, and Ruminococcus showed decreasing tendencies between HCs, the asymptomatic group, and the symptomatic group. In the class Bacilli of the phylum Firmicutes, Veillonella genus belonging to Veillonellaceae family was enriched in the symptomatic RPs (Fig. 4A and B).
Table 3

Characteristics of matched RPs and HCs of whom the stool samples performed gut microbiota analysis

jkms-38-e120-i003
Characteristics RPs HCs (n = 32) P value
Symptomatic group (n = 55) Asymptomatic group (n = 75)
Sex 0.089
Male 23 (41.8) 44 (58.7) 13 (40.6)
Female 32 (58.2) 31 (41.3) 19 (59.4)
Age, yr 57.00 (53.00–68.00) 62.00 (51.00–67.00) 57.5 (51.25–64.80) 0.599
BMI, kg/m2 25.10 (23.37–26.87) 25.39 (23.18–27.14) 24.05 (22.86–26.00) 0.143
Comorbidities
Hypertension 16 (29.1) 31 (41.3) 7 (21.9) 0.106
Chronic pulmonary diseasesa 10 (18.2) 16 (21.3) 3 (9.4) 0.335
Chronic digestive system diseasesb 12 (21.8) 13 (17.3) 4 (12.5) 0.542
Hyperlipidemia 8 (14.5) 15 (20.0) 4 (12.5) 0.555
Diabetes 10 (18.2) 11 (14.7) 4 (12.5) 0.780
Coronary heart disease 4 (7.3) 9 (12.0) 2 (6.3) 0.635
Hypothyroidism 2 (3.6) 3 (4.0) 3 (9.4) 0.559
Chronic hepatitis B 0 (0.0) 1 (1.3) 1 (3.1) 0.471
Cancer 0 (0.0) 1 (1.3) 0 (0.0) 1.000
Chronic kidney disease 0 (0.0) 0 (0.0) 0 (0.0) -
Data are given as number (%) or median (interquartile range).
RPs = recovered patients, HCs = healthy controls, BMI = body mass index.
aChronic pulmonary diseases include chronic bronchitis, emphysema, COPD, tuberculosis, and bronchiectasis, etc.
bChronic digestive system diseases include chronic gastritis and chronic ulcer, etc.
Fig. 2

Differences of alpha diversity (A, B), Venn diagram of OTUs (C) and beta diversity (D) in symptomatic RPs, asymptomatic RPs, and HCs. P value < 0.05 is considered to be statistically significant.

RPs = recovered patients, HCs = healthy controls, OTU = operational taxonomic unit.
**P < 0.01.
jkms-38-e120-g002
Fig. 3

The relative abundance of gut microbiota at the phylum (A) and class (B) levels in symptomatic RPs, asymptomatic RPs, and HCs. The category “Other” covers all other phyla or classes with a low taxa abundance.

RPs = recovered patients, HCs = healthy controls.
jkms-38-e120-g003
Fig. 4

Taxonomic differences of gut microbiota between symptomatic RPs, asymptomatic RPs, and HCs at family (A) and genus (B) levels. Bars indicate mean ± standard deviation. P value < 0.05 is considered to be statistically significant.

RPs = recovered patients, HCs = healthy controls.
*P < 0.05, **P < 0.01, ***P < 0.001.
jkms-38-e120-g004

Correlations between altered gut microbiota and laboratory findings

We found that the relative abundance of Eubacterium_hallii_group was positively correlated with lymphocytes (R = 0.224, P = 0.006), Agathobacter was positively correlated with TBIL (R = 0.210, P = 0.048), and negatively correlated with CRP (R = −0.238, P = 0.008). Subdoligranulum was positively correlated with lymphocytes (R = 0.169, P = 0.040), TBIL (R = 0.219, P = 0.039), and B cells (R = 0.357, P = 0.036). Ruminococcus was positively correlated with lymphocytes (R = 0.202, P = 0.014), and negatively correlated with CRP (R = −0.215, P = 0.017) and natural killer (NK) cells (R = −0.348, P = 0.040). Dorea was negatively correlated with neutrophils (R = −0.188, P = 0.022). Eubacterium_ventriosum_group was positively correlated with lymphocytes (R = 0.207, P = 0.012), and negatively correlated with ALT (R = −0.182, P = 0.027) and NK cells (R = −0.411, P = 0.014). Erysipelatoclostridium was positively correlated with CRP (R = 0.259, P = 0.004). Veillonella was negatively correlated with lymphocytes (R = −0.184, P = 0.025) (Fig. 5, Supplementary Table 4).
Fig. 5

Correlation analysis of altered gut bacteria and laboratory findings. Spearman correlation coefficient is indicated using a color gradient: yellow indicates positive correlation; purple indicates negative correlation.

WBC = white blood cell, ALT = alanine aminotransferase, AST = aspartate aminotransferase, TBIL = total bilirubin, BUN = blood urea nitrogen, Scr = serum creatinine, CRP = C-reactive protein, NK = natural killer.
*P < 0.05, **P < 0.01.
jkms-38-e120-g005

DISCUSSION

In this work, we evaluated the long-term symptoms and clinical indices of RPs at one-year after discharge and compared the differences between RPs with long-term symptoms and those without. Simultaneously, we explored the differences in the gut microbiota between the symptomatic RPs and the asymptomatic RPs, as well as HCs, and analyzed the associations of altered gut microbiota with clinical parameters. We found that long-term symptoms, physical function impairment, psychiatric disorders, lung functions reduction, and radiographic abnormalities persisted to 12 months in a proportion of individuals. The differences between the symptomatic RPs and asymptomatic RPs were mainly in anxiety or depression, SF-36 domains and CD3+ T cells and CD4+ T cells. The gut microbiota of RPs still did not restore at one-year after discharge, mainly manifested as lower bacterial diversity and significantly reduced SCFAs-producing symbionts, especially in the symptomatic group, and altered gut microbiota was significantly correlated with clinical indices of the recovery stage.
In our study, the participants enrolled were inpatients with relatively serious conditions at acute phase during the early stage of the pandemic. According to our results, 44.9% RPs still had one or more symptoms including chest tightness after activity, chronic fatigue, palpitation on exercise, sputum, myalgia, and so on at one-year after discharge, and many of the symptoms tended to become chronic which reduced quality of daily life and had a psychological impact on RPs. The largest follow-up study of COVID-19 published in the Lancet suggested that 4.5% and 10.8% of people, who were infected with delta and omicron variants respectively, experienced long COVID-19,17 but our patients were infected with the earliest SARS-CoV-2 which was more virulent and with relatively serious conditions in acute stage, thus, there were more patients experiencing long COVID-19 in our study. We found that there was persistent impairment of the health-related quality of life of RPs and 35.9% RPs were assessed as having anxiety or depression. The results were similar with previous follow-up research on SARS that reported decreased health-related quality of life and reduced mental health at one-year discharge.181920 A study of SARS had observed that even after 2–4 years of discharge, over 39% of the respondents had depression issues, 40.3% had chronic fatigue problems, and 27.1% met the modified 1994 Centers for Disease Control and Prevention criteria for chronic fatigue syndrome.21 These results indicate that long COVID-19 deserves constant attention.
We explored the composition and alteration of gut microbiota in RPs with long-term symptoms and those without, as well as HCs. The results suggested that RPs, especially the symptomatic group, had lower bacterial diversity, and depleted SCFAs-producing symbiotes than HCs. Gu et al.22 found that acutely infected COVID-19 patients had significantly reduced bacterial diversity, and the abundance of the Ruminococcaceae family and several genera from the Lachnospiraceae family (such as Agathobacter, unclassified Lachnospiraceae, and Eubacterium_hallii_group) was dramatically reduced compared with HCs. Zuo et al.23 showed that Eubacterium rectale, Ruminococcus obeum, and Dorea formicigenerans were significantly depleted in COVID-19 patients. These data were consistent with our results, but Gu et al. also suggested that COVID-19 patients had an increased relative abundance of opportunistic pathogens, including Streptococcus, Rothia, Veillonella, Erysipelatoclostridium, and Actinomyces.22 Our previous work also indicated an increase in opportunistic pathogens and inflammation-related pathogens, including Escherichia, Intestinibacter, Clostridium, and Flavonifractor in RPs at three-month after discharge.12 However, in this study, we only found Veillonella was enriched in RPs, and the enrichment of other opportunistic pathogens was not presented in our data, indicating the relative abundance of these taxa gradually reduced or returned to normal at a long-term time after discharge.
In fact, we had also analyzed the gut microbiota of healthcare workers with COVID-19 in the acute phase and three-month discharge, and the results had been issued in other journals.1213 Results showed that the alpha diversities were all lower, and the relative abundance of beneficial commensals mainly SCFAs-producing strains were decreased compared with HCs, which were consistent with the results in this study. However, due to the strict prevention and control of the epidemic situation and policies, we were unable to collect stool samples from a large number of relatively severe patients during the acute phase. In this study, we enrolled patients with different disease severities in the acute phase, and the number of subjects was larger, so it was more representative. Our work indicated that the gut microbiota dysbiosis persisted even at one-year after discharge.
The gut microbiota is vital for the development of the human immune system and homeostasis, and it has a profound impact on human health and disease.2425 A healthy intestinal microbiota maintains homeostatic immune responses through the exposure of structural ligands (for example, lipopolysaccharide or peptidoglycan) and secreted metabolites (for example, SCFAs). Dysbiosis of the gut microbiota can cause systemic inflammation and an outgrowth of opportunistic pathogens, which can lead to chronic inflammation at distal sites. The gut microbiota can implicate in the pathogenesis of lung injury via several potential mechanisms, including direct translocation of bacteria from the gut to the lung, immune modulation effects of microbes-related metabolites, and so on, which we call the link as gut-lung axis.72627 Although COVID-19 mainly involves the lungs, the gut microbiota may play an important role in the rehabilitation process through the gut-lung axis. In addition, a lot of investigations have reported gut microbiota dysbiosis in subjects with mental disorders including anxiety and depression,2829 and our results showed that the symptomatic RPs had increased proportions of anxiety or depression which indicated the gut microbiota may have an influence on anxiety or depression of long COVID-19 patients through the gut-brain axis.30
In our study, the SCFAs-producing salutary commensal bacteria were dramatically reduced in RPs, including Eubacterium_hallii_group, Subdoligranulum, Ruminococcus, Dorea, Coprococcus, and Eubacterium_ventriosum_group.31323334 SCFAs, as paramount microbiota metabolites of the intestine lumen, play an important role in the modulation of the immune system not only for the intestine but also for distal sites or organs,3536 including influencing pulmonary diseases or psychiatric illness through the gut-lung axis or gut-brain axis. Many studies have demonstrated that SCFAs are involved in the lung inflammation of influenza, bacterial pneumonia, chronic obstructive pulmonary disease, and asthma.3738394041 It was reported that SCFAs enhanced antiviral CD8+ T-cells and macrophages to protect against respiratory infection.42434445 Tian et al.39 demonstrated that increased production of propionate is associated with reduced lung inflammation through animal experiments. In addition, one study suggested that propionate can attenuate the lipopolysaccharide-induced epithelial-mesenchymal transition and even fibrosis.46 There were also studies reported that SCFAs especially butyrate in the central nervous system can affect the function of hippocampus and promote the expression of brain-derived neurotrophic factor, which has been shown to have antidepressant-like effects in animal models.4748 Taken together, we posit that significantly reduced levels of SCFAs and SCFAs-producing commensal bacteria may delay rehabilitation of pulmonary and psychiatric symptoms and cause long COVID-19.
Our study has several limitations. First, this was a single-center study, but the number of participants was enough and the sample quality was high. To our knowledge, our study has the largest number of participants and was the first study investigating the gut microbiota of RPs with COVID-19 at one-year after discharge. Second, this was a cross-sectional study. We did not conduct dynamic monitoring of RPs. Next, we will continue paying attention to the convalescence and gut microbiota of COVID-19 RPs for a longer time. Third, lifestyle and diet may affect gut microbiota, and we did not have detailed information on them, but all participants in the study came from Hubei province and shared similar diet habits and lifestyles (mainly wheat flour, rice, pork, eggs, vegetables), which can largely reduce the impact of diet on outcomes. Fourth, while we discussed the potential roles of aberrant bacteria, the deep mechanism of association between gut microbiota alteration and SARS-CoV-2 infection and convalescence was not identified. In the future study, multi-omics analysis beyond 16S rRNA sequencing can be a good way to investigate the deep-seated mechanism of the gut-lung axis.
In conclusion, this study provides evidence for the presence of long COVID-19 in RPs at one-year after discharge, and gut microbiota may play an important role in long COVID-19.

ACKNOWLEDGMENTS

We thank all study participants and all staff who provided help for our program in our hospital.

Notes

Funding: This work was supported by the Fundamental Research Funds for the Central Universities (No. 2020kfyXGYJ009) and (No. 2020kfyXGYJ034), the Natural Science Foundation of Hubei Province (No. 2020CFB809) and Scientific Research Fund of Health Commission of Hubei Province (No. WJ2021M234).

Disclosure: The authors have no potential conflicts of interest to disclose.

Data Availability Statement: The sequences from our study were deposited in the NCBI Sequence Read Archive (PRJNA810742).

Author Contributions:

  • Conceptualization: Zhang JC, Wang XR.

  • Data curation: Zhang DM, Zhou YY, Ma YL, Chen P, Tang J, Yang BH, Li H, Liang MY, Xue YE, Liu Y.

  • Formal analysis: Zhang DM, Zhou YY, Ma YL, Chen P, Tang J.

  • Funding acquisition: Zhang JC, Wang XR, Ma YL.

  • Methodology: Zhang DM, Zhou YY, Ma YL, Chen P.

  • Writing - original draft: Zhang DM, Zhou YY.

  • Writing - review & editing: Zhang JC, Wang XR.

References

1. Lerum TV, Aaløkken TM, Brønstad E, Aarli B, Ikdahl E, Lund KM, et al. Dyspnoea, lung function and CT findings 3 months after hospital admission for COVID-19. Eur Respir J. 2021; 57(4):2003448. PMID: 33303540.
2. Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet. 2021; 397(10270):220–232. PMID: 33428867.
3. Wu X, Liu X, Zhou Y, Yu H, Li R, Zhan Q, et al. 3-month, 6-month, 9-month, and 12-month respiratory outcomes in patients following COVID-19-related hospitalisation: a prospective study. Lancet Respir Med. 2021; 9(7):747–754. PMID: 33964245.
4. Yelin D, Margalit I, Yahav D, Runold M, Bruchfeld J. Long COVID-19-it’s not over until? Clin Microbiol Infect. 2021; 27(4):506–508. PMID: 33316400.
5. Shah W, Hillman T, Playford ED, Hishmeh L. Managing the long term effects of covid-19: summary of NICE, SIGN, and RCGP rapid guideline. BMJ. 2021; 372:n136. PMID: 33483331.
6. World Health Organization. A clinical case definition of post COVID-19 condition by a Delphi consensus, 6 October 2021. Updated 2021. Accessed October 6, 2021. https://www.who.int/publications/i/item/WHO-2019-nCoV-Post_COVID-19_condition-Clinical_case_definition-2021.1 .
7. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, et al. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017; 15(1):55–63. PMID: 27694885.
8. Pflughoeft KJ, Versalovic J. Human microbiome in health and disease. Annu Rev Pathol. 2012; 7(1):99–122. PMID: 21910623.
9. Yeoh YK, Zuo T, Lui GC, Zhang F, Liu Q, Li AY, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021; 70(4):698–706. PMID: 33431578.
10. Chen Y, Gu S, Chen Y, Lu H, Shi D, Guo J, et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut. 2022; 71(1):222–225. PMID: 33833065.
11. Ren Z, Wang H, Cui G, Lu H, Wang L, Luo H, et al. Alterations in the human oral and gut microbiomes and lipidomics in COVID-19. Gut. 2021; 70(7):1253–1265. PMID: 33789966.
12. Zhou Y, Zhang J, Zhang D, Ma WL, Wang X. Linking the gut microbiota to persistent symptoms in survivors of COVID-19 after discharge. J Microbiol. 2021; 59(10):941–948. PMID: 34382150.
13. Zhou Y, Shi X, Fu W, Xiang F, He X, Yang B, et al. Gut microbiota dysbiosis correlates with abnormal immune response in moderate COVID-19 patients with fever. J Inflamm Res. 2021; 14:2619–2631. PMID: 34168484.
14. World Health Organization. COVID-19 clinical management: living guidance, 25 January 2021. Updated 2021. Accessed January 25, 2021. https://apps.who.int/iris/handle/10665/338882 .
15. Liang L, Yang B, Jiang N, Fu W, He X, Zhou Y, et al. Three-month follow-up study of survivors of coronavirus disease 2019 after discharge. J Korean Med Sci. 2020; 35(47):e418. PMID: 33289374.
16. Wang X, Zhou Q, He Y, Liu L, Ma X, Wei X, et al. Nosocomial outbreak of COVID-19 pneumonia in Wuhan, China. Eur Respir J. 2020; 55(6):2000544. PMID: 32366488.
17. Antonelli M, Pujol JC, Spector TD, Ourselin S, Steves CJ. Risk of long COVID associated with delta versus omicron variants of SARS-CoV-2. Lancet. 2022; 399(10343):2263–2264. PMID: 35717982.
18. Tansey CM, Louie M, Loeb M, Gold WL, Muller MP, de Jager J, et al. One-year outcomes and health care utilization in survivors of severe acute respiratory syndrome. Arch Intern Med. 2007; 167(12):1312–1320. PMID: 17592106.
19. Ong KC, Ng AW, Lee LS, Kaw G, Kwek SK, Leow MK, et al. 1-year pulmonary function and health status in survivors of severe acute respiratory syndrome. Chest. 2005; 128(3):1393–1400. PMID: 16162734.
20. Hui DS, Wong KT, Ko FW, Tam LS, Chan DP, Woo J, et al. The 1-year impact of severe acute respiratory syndrome on pulmonary function, exercise capacity, and quality of life in a cohort of survivors. Chest. 2005; 128(4):2247–2261. PMID: 16236881.
21. Lam MH, Wing YK, Yu MW, Leung CM, Ma RC, Kong AP, et al. Mental morbidities and chronic fatigue in severe acute respiratory syndrome survivors: long-term follow-up. Arch Intern Med. 2009; 169(22):2142–2147. PMID: 20008700.
22. Gu S, Chen Y, Wu Z, Chen Y, Gao H, Lv L, et al. Alterations of the gut microbiota in patients with coronavirus disease 2019 or H1N1 influenza. Clin Infect Dis. 2020; 71(10):2669–2678. PMID: 32497191.
23. Zuo T, Zhang F, Lui GC, Yeoh YK, Li AY, Zhan H, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020; 159(3):944–955.e8. PMID: 32442562.
24. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012; 336(6086):1268–1273. PMID: 22674334.
25. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021; 19(1):55–71. PMID: 32887946.
26. McAleer JP, Kolls JK. Contributions of the intestinal microbiome in lung immunity. Eur J Immunol. 2018; 48(1):39–49. PMID: 28776643.
27. Enaud R, Prevel R, Ciarlo E, Beaufils F, Wieërs G, Guery B, et al. 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. PMID: 32140452.
28. Li S, Hua D, Wang Q, Yang L, Wang X, Luo A, et al. The role of bacteria and its derived metabolites in chronic pain and depression: recent findings and research progress. Int J Neuropsychopharmacol. 2020; 23(1):26–41. PMID: 31760425.
29. Hu S, Li A, Huang T, Lai J, Li J, Sublette ME, et al. Gut microbiota changes in patients with bipolar depression. Adv Sci (Weinh). 2019; 6(14):1900752. PMID: 31380217.
30. Järbrink-Sehgal E, Andreasson A. The gut microbiota and mental health in adults. Curr Opin Neurobiol. 2020; 62:102–114. PMID: 32163822.
31. Zhang J, Guo Z, Xue Z, Sun Z, Zhang M, Wang L, et al. A phylo-functional core of gut microbiota in healthy young Chinese cohorts across lifestyles, geography and ethnicities. ISME J. 2015; 9(9):1979–1990. PMID: 25647347.
32. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014; 12(10):661–672. PMID: 25198138.
33. Holmstrøm K, Collins MD, Møller T, Falsen E, Lawson PA. Subdoligranulum variabile gen. nov., sp. nov. from human feces. Anaerobe. 2004; 10(3):197–203. PMID: 16701519.
34. Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016; 167(4):1125–1136.e8. PMID: 27814509.
35. Ratajczak W, Rył A, Mizerski A, Walczakiewicz K, Sipak O, Laszczyńska M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim Pol. 2019; 66(1):1–12. PMID: 30831575.
36. Sublette ME, Cheung S, Lieberman E, Hu S, Mann JJ, Uhlemann AC, et al. Bipolar disorder and the gut microbiome: a systematic review. Bipolar Disord. 2021; 23(6):544–564. PMID: 33512753.
37. Qin N, Zheng B, Yao J, Guo L, Zuo J, Wu L, et al. Influence of H7N9 virus infection and associated treatment on human gut microbiota. Sci Rep. 2015; 5(1):14771. PMID: 26490635.
38. Sencio V, Barthelemy A, Tavares LP, Machado MG, Soulard D, Cuinat C, et al. Gut dysbiosis during influenza contributes to pulmonary pneumococcal superinfection through altered short-chain fatty acid production. Cell Reports. 2020; 30(9):2934–2947.e6. PMID: 32130898.
39. Tian X, Hellman J, Horswill AR, Crosby HA, Francis KP, Prakash A. Elevated gut microbiome-derived propionate levels are associated with reduced sterile lung inflammation and bacterial immunity in mice. Front Microbiol. 2019; 10:159. PMID: 30891007.
40. Vaughan A, Frazer ZA, Hansbro PM, Yang IA. COPD and the gut-lung axis: the therapeutic potential of fibre. J Thorac Dis. 2019; 11(Suppl 17):S2173–S2180. PMID: 31737344.
41. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014; 20(2):159–166. PMID: 24390308.
42. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A. 2011; 108(13):5354–5359. PMID: 21402903.
43. Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, Pernot J, et al. Dietary fiber confers protection against flu by shaping Ly6c- patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity. 2018; 48(5):992–1005.e8. PMID: 29768180.
44. Wu T, Li H, Su C, Xu F, Yang G, Sun K, et al. Microbiota-derived short-chain fatty acids promote LAMTOR2-mediated immune responses in macrophages. mSystems. 2020; 5(6):e00587-20.
45. Wu T, Xu F, Su C, Li H, Lv N, Liu Y, et al. Alterations in the gut microbiome and cecal metabolome during Klebsiella pneumoniae-induced pneumosepsis. Front Immunol. 2020; 11:1331. PMID: 32849494.
46. Chen D, Qiu YB, Gao ZQ, Wu YX, Wan BB, Liu G, et al. Sodium propionate attenuates the lipopolysaccharide-induced epithelial-mesenchymal transition via the PI3K/Akt/mTOR signaling pathway. J Agric Food Chem. 2020; 68(24):6554–6563. PMID: 32452677.
47. Yamawaki Y, Fuchikami M, Morinobu S, Segawa M, Matsumoto T, Yamawaki S. Antidepressant-like effect of sodium butyrate (HDAC inhibitor) and its molecular mechanism of action in the rat hippocampus. World J Biol Psychiatry. 2012; 13(6):458–467. PMID: 21812623.
48. Wei Y, Melas PA, Wegener G, Mathé AA, Lavebratt C. Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the Bdnf gene. Int J Neuropsychopharmacol. 2014; 18(2):pyu032. PMID: 25618518.

SUPPLEMENTARY MATERIALS

Supplementary Data 1

Methods supplement
jkms-38-e120-s001.doc

Supplementary Table 1

Antibody tests of the RPs at one-year after discharge
jkms-38-e120-s002.xls

Supplementary Table 2

Pulmonary functions, 6MWD and chest CT images of the RPs at one-year after discharge
jkms-38-e120-s003.xls

Supplementary Table 3

Correlation analysis of altered gut bacteria and laboratory findings
jkms-38-e120-s004.xls

Supplementary Table 4

Correlation analysis of symptoms and laboratory findings, lung functions, CT scores and 6MWD of RPs with COVID-19 at one-year after discharge
jkms-38-e120-s005.xls

Supplementary Fig. 1

Study design and flow chart.
jkms-38-e120-s006.doc
TOOLS
Similar articles