Skip to main content

Alterations in gut microbiota and host transcriptome of patients with coronary artery disease

Abstract

Background

Coronary artery disease (CAD) is a widespread heart condition caused by atherosclerosis and influences millions of people worldwide. Early detection of CAD is challenging due to the lack of specific biomarkers. The gut microbiota and host-microbiota interactions have been well documented to affect human health. However, investigation that reveals the role of gut microbes in CAD is still limited. This study aims to uncover the synergistic effects of host genes and gut microbes associated with CAD through integrative genomic analyses.

Results

Herein, we collected 52 fecal and 50 blood samples from CAD patients and matched controls, and performed amplicon and transcriptomic sequencing on these samples, respectively. By comparing CAD patients with health controls, we found that dysregulated gut microbes were significantly associated with CAD. By leveraging the Random Forest method, we found that combining 20 bacteria and 30 gene biomarkers could distinguish CAD patients from health controls with a high performance (AUC = 0.92). We observed that there existed prominent associations of gut microbes with several clinical indices relevant to heart functions. Integration analysis revealed that CAD-relevant gut microbe genus Fusicatenibacter was associated with expression of CAD-risk genes, such as GBP2, MLKL, and CPR65, which is in line with previous evidence (Tang et al., Nat Rev Cardiol 16:137-154, 2019; Kummen et al., J Am Coll Cardiol 71:1184-1186, 2018). In addition, the upregulation of immune-related pathways in CAD patients were identified to be primarily associated with higher abundance of genus Blautia, Eubacterium, Fusicatenibacter, and Monoglobus.

Conclusions

Our results highlight that dysregulated gut microbes contribute risk to CAD by interacting with host genes. These identified microbes and interacted risk genes may have high potentials as biomarkers for CAD.

Peer Review reports

Background

Coronary artery disease (CAD) mostly arising from atherosclerosis is the most common type of heart disease, which affects millions of individuals worldwide [1]. The cause of CAD is multifactorial that involve complex environmental and genetic factors [2,3,4]. CAD process can be effectively prevented by drug therapy, percutaneous coronary intervention (PCI), or coronary artery bypass graft (CABG) surgery [5, 6]. However, the early diagnosis of CAD remains substantially difficult due to lack of adequate biomarkers. Although several biomarkers, such as Fibrinogen and C-reactive protein, have been reported to be associated with CAD [7, 8], these biomarkers generally lack sufficient specificity and significantly detectable changes appear mainly in the advanced stages of CAD [9]. Thus, the early diagnosis and intervention of CAD pose a significant public health challenge with enormous medical and societal consequences. There is an urgent requirement for new and effective biomarkers to assist in the early diagnosis, monitoring, and management of CAD.

Gut microbiota comprises the tens of thousands of intestinal bacteria and the biological activity in the human intestine [10,11,12,13], which is essential to the development of human health [14,15,16,17,18,19,20,21]. Host-microbiota interactions, including inflammatory and metabolic processes, have been well-documented to be involved in the etiology of multiple complex diseases [22, 23]. Multiple lines of evidence have shown that the gut microbiota can influence distant cells and organs via a variety of biochemical signals or metabolites [24]. In particular, recent studies have indicated that gut microbiome potentially impact the cardiovascular system [25, 26]. An increased potential for several intestinal flora metabolites, including lipopolysaccharides [27], grammabutyrobetaine [28], and trimethyllysine [29] biosynthesis in the microbiome, has been identified among CAD patients. Most notably, a growing number of studies have linked increased levels of the gut microbe-derived trimethylamine-N-oxide (TMAO) to cardiovascular diseases [30, 31]. It has also recently been demonstrated that the composition of gut microbiota can induce the alterations in various serum metabolite concentrations that are significantly relevant to the severity of CAD [32]. Hence, integrating metagenomics with other omics approaches including transcriptomics to investigate the host-microbiome interactions will allow for distinguishing effective biomarkers and developing precision medicine for the treatment of atherosclerotic cardiovascular diseases [33, 34].

To date, investigating the contribution of gut microbiome changes to CAD is insufficient, and the mechanism underlying synergistic interactions between host genes and gastrointestinal microbes remains unclear. Thus, we performed an integrative genomic analysis to uncover the synergy between host genes and gut microbes associated with CAD. Furthermore, we also conducted a series of bioinformatics analyses to explore the molecular functions and potential mechanisms of microbe-associated host genes for the development of CAD, and give a clue of the potential biomarkers for diagnosing CAD.

Results

Overview of gut microbiota

A total of 52 fecal and 50 blood samples from CAD patients and matched controls were sequenced in the current study (Supplementary Tables S1 and S2). Clinical information, including baseline characteristics, medications, and laboratory data of the CAD and control group, is shown in Table 1. There was no significant difference in age, gender, background medication, or other baseline characteristics between the two groups of subjects (P > 0.05). The degree of coronary artery stenosis was significantly different between the two groups (P < 0.05). At the same time, according to the results of coronary angiography, we judged that patients with CAD were in the same disease stage, that is, at least one main coronary artery stenosis ≥ 70%, but not AMI.

Table 1 Clinical characteristics of patients with CAD and non-CAD

Through 16S rRNA sequencing, we obtained 964 Operational Taxonomic Units (OTUs) from all 52 fecal samples using Qiime2 software. Among them, there were 654 (67.84%) shared OTUs between CAD and control subjects, and 248 (25.73%) and 62 (6.43%) unique OTUs for CAD and control groups, respectively (Fig. 1a). Although no significant difference was observed in Shannon (Fig. 1b) and Phylogenetic Diversity (PD, Fig. 1c) indexes between CAD and control groups, beta-diversity analysis revealed that there was a significant difference in overall bacterial composition between two groups (Fig. 1d).

Fig. 1
figure 1

Overview of intestinal flora composition in control and CAD patients. A OTU Difference Venn diagram between Con and CAD; The Operational Taxonomic Units (OTUs) were generated by using the Uparse. B and C The Shannon and PD indexes; D beta-diversity analysis; The blue circle indicates the PCoA of CAD patients, and red circle indicates that of controls. E and F the major gut microbes at phylum level; G the major gut microbes at genus level

To provide a clearer understanding of the gut microbial features in CAD patients, we summarized the relative abundance of gut microbes at different taxonomic levels. At phylum level, the major gut microbes belong to Firmicutes, Bacteroidota, Fusobacteriota, and Proteobacteria (Fig. 1e and g). At genus level, the top 10 abundant bacteria were Bacteroides, Prevotella_9, Lactobacillus, Limosilactobacillus, Fusobacterium, Ligilactobacillus, Escherichia-Shigella, Faecalibacterium, Klebsiella, and Alloprevotella (Fig. 1f).

Difference in gut microbiota between CAD and control group

By performing differential abundant analysis, we identified 18 differential abundant genera between CAD patients and health controls (Fig. 2a and Supplementary Table S3). Genus Blautia, Fusicatenibacter, Monoglobus, and Eubacterium were depleted in CAD patients, while Sutterella, Lachnospiraceae_NK4A136_group, UCG-002, UCG-005, [Eubacterium]_hallii_grou, Collinsella, Colidextribacter, NK4A214_group, Negativibacillus, Faecalitalea, Family_XIII_AD3011_group, Peptoniphilus, Fructilactobacillus, and Solobacterium were enriched in CAD patients. Among them, eight of these identified microbes, including Blautia, Fusicatenibacter, Monoglobus, Eubacterium, UCG-002, UCG-005, Collinsella, and NK4A214_group, have been reported to be associated with CAD, acute myocardial infarction, or CAD complicated with non-alcoholic fatty liver disease in previous studies [27, 35,36,37,38,39,40,41,42]. For example, Hu et al. [37] reported that the decreased abundances of Fusicatenibacter was identified in CAD patients, and Ahrens et al. [42] documented that Monoglobus was significantly decreased in CAD patients, which are in line with our findings as shown in Fig. 2a (P = 0.049 for Fusicatenibacter, and P = 0.021 for Monoglobus). We found that Blautia exhibited significantly low-abundances in CAD patients, whereas the enrichment of Blautia was observed in CAD patients with in-stenosis in a previous study [38]. Notably, several microbes, including Sutterella, Colidextribacter, Negativibacillus, Faecalitalea, Peptoniphilus, Fructilactobacillus, and Solobacterium, were identified to be associated with CAD in the current investigation (Fig. 2a). Detailed taxonomic abundance differences of gut microbiota at all taxonomic levels are shown in the Supplementary Fig. 1a-d.

Fig. 2
figure 2

Difference of gut microbiota between CAD and Control group. A The differential abundant genera between CAD and control group. The abundance of microbe was log-transformed. The significant level of each microbe was calculated by using Student’s t-test. B The AUC of genus-level abundance. C The top 10 differential genera between CAD and control group

By applying the Random Forest procedure, we found that 20 bacteria biomarkers can distinguish CAD patients from health controls with a good performance. The overall AUC was 0.81 using genus-level abundance as input (Fig. 2b). By overlapping genera identified from two independent methods of Mean Decrease Accuracy (Fig. 2c) and Mean Decrease Gini (Supplementary Fig. 1e), we emphasized the importance of Rikenellaceae_RC9_gut_group, Coprobacter, Blautia, and Faecalibaculum on CAD, of which several have been reported to be associated with CAD or its comorbidities [38, 43, 44]. Of note, the biomarker genus Blautia was also identified to be significantly differential genera.

Functional implications of gut microbiota dysbiosis in CAD

To further explore gut microbial functional dysbiosis of CAD patients, we performed functional annotation and differential analyses based on bacterial profiles. We observed a depletion of three biological pathways, including beta − Lactam resistance, cationic antimicrobial peptide (CAMP) resistance and viral proteins functions, and an up-regulation of 10 biological pathways, including nitrogen and methane metabolism, microRNAs in cancer and tropane, piperidine and pyridine alkaloid biosynthesis in CAD patients comparing with health controls (P < 0.05, Fig. 3a and Supplementary Table S4).

Fig. 3
figure 3

Function annotation and differential analyses of gut microbiota dysbiosis. A Functional implications of gut microbial dysfunction in patients with CAD; The x axis indicates the log2-transformed fold change of each functional annotation. P value for each functional annotation was showed in the plot. The functional information of bacterial genomes from the KEGG database, annotated using the UProC and PAUDA methods, was mapped to the SILVA database. B The association of differential abundant bacteria with clinical indices. Significances tested by Pearson correlation analysis. * FDR < 0.05; ** FDR < 0.01; *** FDR < 0.001. Color bar indicates the level of correlation coefficients (corr)

Furthermore, we measured the associations of differential abundant bacteria between CAD and health groups with 23 clinical indices, including blood routine, heart, liver, and kidney functions. There were a series of significant associations between gut microbes and clinical indices relevant to heart functions. Genus Sutterella, Fructilactobacillus, and Peptoniphilus were positively associated with cardiac troponin I (cTNI), the cardiac regulatory proteins that control the calcium mediated interaction between actin and myosin. Brain natriuretic peptide (BNP) was significantly associated with genus Negativibacillus. Creatine kinase (CK) showed significantly positive correlations with genus Fusicatenibacter, Blautia, and [Eubacterium]_hallii_group. In addition, C-reactive protein (CRP) is a widely used marker of inflammation [45], and elevated CRP levels are involved in the development and progression of thrombosis and CAD [46]. Our results indicate that CRP exhibit a significantly positive association with genus Peptoniphilus and Fructilactobacillus (Fig. 3b).

Transcriptional analysis revealed genes associated with gut microbes

To reveal transcriptome difference between CAD patients and health controls, we further performed differential gene expression analysis on PBMC transcriptomic profiles. Comparing with health controls, there were 409 up-regulated and 762 down-regulated genes in CAD patients (P < 0.05; Fig. 4a and Supplementary Table S5), including APOE and OLFML3, which have been well-documented to be associated with CAD in previous studies [47, 48]. GSEA analysis showed that CAD group is characterized by depletion in interferon signaling-related pathways, including interferon gamma signaling pathway and interferon alpha/beta signaling pathway, and is enriched in several immune-regulated and inflammation-related pathways, including TNFs bind their physiological receptors, Rap1 signaling, and TNFR2 non-canonical NF-kB pathway (Fig. 4b). Random Forest procedure identified that differentially expressed genes distinguished CAD patients from health controls with an AUC value of 0.84. Moreover, combining gene and microbial biomarkers increased performance of the Random Forest model, with an AUC value of 0.92 (Fig. 4c).

Fig. 4
figure 4

The transcriptome difference between CAD and control group. A The volcano map of differential genes between CAD patients and controls; Red color indicates the significantly up-regulated genes, blue color indicates the significantly down-regulated genes, and gray indicates non-significant genes. B The pathway enrichment analysis of differential genes by using GSEA. The x axis indicates the normalized enrichment score. Dark blue or dark orange indicates FDR-adjusted P value ≤ 0.05, and light blue or light orange indicates FDR-adjusted P value > 0.05. C The ROC plot showing the performance of the combination of gene and microbial biomarkers based on Random Forest model. The blue line indicates the performance of using gene expression biomarker alone, and the red line indicates the performance of combining gene and microbial biomarker. D Heatmap showing the correlation analysis on differentially abundant gut microbes and differentially expressed genes. * indicates the significant correlation (P < 0.05). Color bar in indicate the correlation coefficient (corr). E Representative example demonstrating the correlation of g__Fusicatenibacter (log-transformed abundance) with the log-transformed expression of seven genes. There were one negatively correlated gene of CD70 and six positively correlated genes (i.e., AFF1, GPR65, MLKL, GBP2, SLK, and ETV6)

Considering that the significantly dis-regulated gut microbes might be associated with host gene abnormal expression and biological functions, we performed a correlation analysis on differentially abundant gut microbes (n = 18) and differentially expressed genes (P < 0.01, n = 347). We obtained a total of 697 suggestive or significant correlations (P < 0.05, Fig. 4d). Among them, there were seven significant correlations with g_Fusicatenibacter with FDR-adjusted P < 0.05, including six positively correlated genes of GBP2, MLKL, GPR65, SLK, ETV6, and AFF1, and one negatively correlated gene of CD70 (Fig. 4e). Among these genes, several have been documented to be implicated in the development of CAD [49, 50]. For example, a network-based prioritization analysis identified the interferon-induced guanylate-binding protein 2 (GBP2) as a key regulator orchestrating biological processes relevant to CAD [49]. MLKL, which can directly induce necroptosis [51], has been reported to be involved in different inflammatory diseases, including tumor necrosis factor-induced shock [52], ischemia–reperfusion injuries [53], and obesity [54]. Recently, Kamal and coworkers have found that MLKL is associated to hallmarks of atherosclerosis with and without type II diabetes mellitus, which could be a potential drug target for treating atherosclerotic patients [50]. Thus, these g_Fusicatenibacter-host gene interactions may play an important role in CAD.

Different gene programs associated with gut microbes

We further performed differential gene program analysis to explore whether there exist distinct gene sets with different functions associated with gut microbes. Based on the unsupervised clustering analysis, these differential genes were grouped into two clusters (Fig. 4d). Cluster 1 (highlighted in red) were positively correlated with genera that were depleted in CAD patients (e.g., Blautia, Eubacterium, Fusicatenibacter, and Monoglobus). Cluster 2 (highlighted in blue) were positively correlated with genera that were enriched in CAD patients. Pathway enrichment analysis demonstrated that genes in cluster 1 showed significant enrichments in immune-related pathways (FDR-adjusted P < 0.05), including regulation of RUNX1 expression and activity, interferon gamma signaling, DDX58/IFIH1-mediated induction of interferon-alpha/beta and interferon signaling. This highlighted a set of genes involved in interferon signaling pathway, such as BP2, GBP4, GBP5, GBP6, PML, STAT1, CASP10, IFIH1, TRAF6, UBA7, and UBE2L6 (Fig. 4d). Although no significant enrichment was observed for genes in cluster 2 (FDR-adjusted P < 0.05), we identified that several pathways exhibited suggestive enrichments for genes in cluster 2 (P < 0.05), including GRB7 events in ERBB2 signaling, sodium-coupled sulphate, di- and tri-carboxylate transporters, and PI3K events in ERBB4 signaling. Together, these two distinct gene programs relevant to gut microbes potentially have different biological functions contributing to CAD.

Discussion

The role of gut microbes in CAD is still not fully understood. Deciphering correlation of gut microbes with CAD related gene functions and clinical indices contribute in understanding the underlying pathophysiological mechanism of CAD. In the current study, we performed 16S rRNA sequencing to comprehensively elucidate difference in profiles of gut microbiome in CAD patients and health controls. Our results exhibited that structure of gut microbiome differed in CAD and health group. In the CAD patients, 4 genera such as Blautia, Eubacterium, and Fusicatenibacter were depleted, and 14 genera such as Sutterella, and Collinsella were enriched.

Accumulating evidence has been documented that remarkably disturbed gut microbiota were detected in patients with cardiovascular diseases [25,26,27,28]. Mitra et al. [55] indicated that host microbiome associated families, such as Porphyromonadaceae, Bacteroidaceae, Micrococcacaea, and Streptococcacaea, were increased in asymptomatic atherosclerotic plaques patients than symptomatic atherosclerotic plaques patients. Based on these findings, we particularly focused on the relationship of altered Blautia, Eubacterium, and Fusicatenibacter with CAD. These three microbes were major producers of short chain fatty acids (SCFAs), which are important secondary metabolites capable of regulating cardiometabolic health [56, 57]. Previous studies [58, 59] have reported a reduced abundance of SCFA producers in CAD patients in different population and resolution. A recent study by Guo et al. [60] indicated that SCFA was significantly lower in AMI group than in control group. In addition, bacteria that can produce SCFA, such as Ruminococcus and Bacteroides, were significantly reduced in CAD patients [61].

Butyrate and other SCFAs produced by gut microbes serve as signaling molecules to modulate blood pressure, inflammatory responses and other metabolic functions. There are increasing evidence indicates the importance of SCFAs in regulating cardiac function [61, 62]. Considerable studies have demonstrated the parasympathetic activation effects of the SCFA propionate [61]. Park and coworkers reported that butyrate potentiates lipoteichoic acid-induced inflammasome activation through the inhabitation of histone deacetylase (HDAC) [63]. Kibbie et al. [64] have demonstrated that butyrate is enable to directly reduce CD4 + T cell activation and proliferation via HDAC inhibition and GPR43 signaling. Recently, Zhong et al. [65] uncovered that butyrate attenuates vascular calcification, a common pathological change in cardiovascular tissues, via its dual effects on HDAC inhibition and NFκB activation based on cellular and animal models. Also, they found that knockdown of Gpr41 could attenuated butyrate-induced vascular smooth muscle cells calcification. Zhou et al. [66] demonstrated that oral propionate supplementation improve MI therapy by parasympathetic activation based on the gut-brain axis. Moreover, a study of malonate intervention in the treatment of myocardial infarction in mice has showed that malonate enable to shift the cardiac metabolic pattern of oxidative phosphorylation to glucose metabolic pattern and promote cardiomyocyte proliferation, hemodialysis and cardiac regeneration in adult mice after myocardial infarction [67]. Jiang et al. [68] reported that intramembranous butyrate injection improved cardiac function by promoting macrophage differentiation and inhibiting inflammation and sympathetic remodeling after myocardial infarction. Our results emphasized the importance of gut microbial functions involved in cationic antimicrobial peptide (CAMP) resistance in CAD. CAMPs are hybrid peptides playing an important role in defensing against invasive bacterial infection for host. However, in addition to antimicrobial effect, CAMPs were believed to be an important link between inflammation and atherosclerotic cardiovascular disease [69]. For example, elevated plasma α-defensin is associated with an increased risk for cardiovascular morbidity [70]. PR-39, a proline-arginine-rich cationic antimicrobial peptide, has been reported to play a cardioprotective role in myocardial I/R injury through inhibition of tumor necrosis factor-α (TNF-α)-induced degradation of the NFκB inhibitor IκBα [71].

The correlation of gut microbiome with CAD indicated that it could be early diagnostic marker for the risk of CAD. In this study, the AUC for gut microbiome-based diagnostic RF model was 93.9%. This was supported by a previous study [39], which has reported that predictive model based on gut microbiome is able to provide a robust prediction of CAD (AUC = 87.7%). Moreover, combining gut microbiome with clinical variables increased power of prediction model, generating an AUC of 90.4%. These results reinforced the capacity of gut microbiome as diagnostic biomarkers for CAD.

To investigate the interactional functions of gut microbes and host genes on CAD, we performed correlation analysis of the abundances of gut microbes and the expression levels of host genes. We observed significant reduction of interferon signaling pathways in CAD patients, as well as strong association of depleted bacteria with genes involved in interferon signaling pathways. Our result suggested that depletion of SCFA-producing bacteria (Blautia, Eubacterium, and Fusicatenibacter) may contribute to the decrease of interferon signaling pathways in CAD patients. One mechanism by which microbes can modulate the host disease is through regulation of cytokine signaling [72]. Interferons (IFNs) are classified into three families (type I, type II, and type III) based on sequence homology. Among them, IFN-γ is a key cytokine implicated in both innate and adaptive immunity, and studies on the involvement of IFN-γ in multiple stages of the atherosclerotic process, playing different roles, have been carried out for decades. In vitro and in vivo studies [73] have shown that IFN-γ has both pro- and anti-atherogenic properties and plays a large role in all stages of CAD progression. Meanwhile, the type I interferons have the capacity to regulate the development or function of virtually every immune effector cell, contributing to the anti-inflammatory and anti-tumor responses, which is essential in the process of CAD [74,75,76].

Some limitations in the present study are warranted to comment. First, it’s worth noting that the functional profiling of the gut microbiome was computationally inferred from amplicon sequencing data, and it could differ from that measured by transcriptomic sequencing techniques. According to a previous study [77], there can be significant genomic and phenotypic diversity among strains of the same species. This diversity has important implications for the ecological function of an organism, as well as the conditions required to culture it. Thus, it is impossible to reliably assess whether a microbe is “culturable” based on 16S rRNA gene sequence. Second, current study only curated a moderate sample size to establish the link between microbiome and host transcriptomic profiles for uncovering the pathology of CAD. The identified links does not imply causality but may reflect indirect discovery of causal associations. Future studies with more large-scale samples are needed to conduct for validating the identified relationships and exploring the underlying causality. Additionally, in light of multiple lines of evidence [78,79,80,81,82,83] have documented that integrating multi-omic datasets including GWAS, single-cell sequencing data, and epigenetic data contribute to uncover the molecular etiology of complex diseases. More integrative genomic analyses of microbiome with other omics are warranted for distinguishing the pathology of CAD.

Conclusions

In summary, our results provide evidence to support that gut microbes play critical roles in CAD mediated by host genes. Reduction of SCFA-producing bacteria and interferon signaling genes are associated with CAD. Depletion of SCFA-producing bacteria, e.g. Blautia, Eubacterium, and Fusicatenibacter, may contribute to dysfunction of interferon signaling and cardiovascular functions. These identified microbes may have potential for diagnosis or therapy of CAD.

Methods

Study participants and sample collection

In the current cross-sectional study, 31 patients with CAD and 21 healthy controls were recruited from the Department of Cardiology, Hangzhou First People's Hospital, Zhejiang University School of Medicine. This study was reviewed and approved by the Ethics Committee of Zhejiang University. The enrolled patients signed a written informed consent form. All patients’ medical history and baseline data were obtained from the electronic medical records system. To obtain high-quality samples, patients with CAD included in this study were required to meet the following criteria: 1) age > 25 years and < 80 years; 2) show ≥ 70% stenosis in at least one major branch of the coronary artery. The healthy control group population should meet the following criteria: 1) age > 45 years and < 70 years; 2) all three coronary vessels ≤ 20% stenosis. Besides, individuals without coronary angiography testing or with one of follow disorders, including cancer, AMI, antibiotic exposure, and any other cardiac-related disease or systemic disease were excluded.

During the hospitalization period, these 52 subjects were educated how to properly collect midcourse fecal samples to reduce the possibility of environmental bacterial contamination. One morning fasting fecal sample was taken for the first time within 24 h of admission for each patient. All fresh fecal samples were stored in − 80 °C for subsequent processing and sequencing. Pre-operative blood samples for 50 of these participants were drawn and stored in PAXgene blood RNA tubes at -80 °C before further processing [84].

16S rRNA sequencing

Total genomic DNA was extracted from fecal samples. The V3-V4 region of bacterial 16S rRNA gene was amplified using primers (341F: 5′-CCTACGGGNGGCWGCAG-3′, 806R: 5′-GGACTACHVGGGTWTCTAAT-3′) with barcodes. The mixed PCR products were purified using the Qiagen Gel Extraction Kit (Qiagen, Germany). Sequencing libraries were generated using the TruSeq® DNA PCR-Free Sample Preparation Kit (Illumina, USA). The library quality was assessed using the Qubit@ 2.0 Fluorometer (Thermo Scientific) and the Agilent Bioanalyzer 2100 system. Finally, the library was sequenced on an Illumina NovaSeq 6000 platform, generating 250 bp paired-end reads.

Raw sequences were demultiplexed based on barcodes. After trimming off the barcodes and primers, paired-end sequences were assembled with FLASH (v1.2.7). The assembled raw tags were quality controlled with Fastp (v0.23.1) [85] to obtain high-quality clean tags (Supplementary Table S1). Chimeric sequences were further removed from clean tags using vsearch (v2.22.1) [86]. The filtered sequences were clustered based on 97% identity to generate Operational Taxonomic Units (OTUs). The Uparse (v7.0.1001, http://www.drive5.com/uparse/) method was applied to cluster the filtered sequences into OTUs at 97% identity threshold. Representative sequences were extracted and annotated with Mothur (v1.48.0) [87] and SILVA (v138.1) database [88]. The MUSCLE (v3.8.31) [89] tool was leveraged to perform sequence alignment for obtaining phylogenetic relationships of all OTU representative sequences. Finally, the data for each sample were normalized based on the sample with the lowest number of sequences by rrarefy() function in the vegan (v2.6–4) R package [90], which generates on randomly rarefied community data frame based on the given sequencing depth to remove the effects of different sample sizes. Subsequently, alpha and beta diversity analyses were based on the normalized data.

Transcriptomic sequencing

Total RNA was extracted from the isolated PBMCs using the QIAzol and miRNeasy Mini Kit (Qiagen, CA, USA). The RNA integrity (average RIN: 7.05; standard deviation (SD): 1.27, Supplementary Table S2) was tested using the Bioanalyzer 2100 system with the RNA Nano 6000 Assay Kit (Agilent Technologies, CA, USA). Poly-T oligo-attached magnetic beads were used to purify mRNA from the total RNA, which was subsequently used to establish cDNA libraries for RNA sequencing. Quality-controlled cDNA library was sequenced on Illumina Novaseq platform (Beijing, China), producing paired-end sequences (150 bp).

Raw transcriptomic sequencing data were qualified using the FastQC (https://www.bioinformatics.babraham.ac.uk/projects/fastqc/). Ensembl human reference genome (http://asia.ensembl.org/info/data/ftp/index.html, file name: Homo_sapiens.GRCh37.75.cdna.all.fa) was used for alignment and annotation. The Hisat2 (v2.0.5) tool [91] was used to establish index for reference genome, and align reads to the reference genome. The FeatureCounts (v1.5.0-p3) method [92] was used to summarize read counts for each gene.

Functional enrichment analysis

We performed differential gene expression (DGE) analysis using the DESeq2 (v1.36.0) [93] (Supplementary Table S5). P-values were evaluated by the Student’s t-test [94], and multiple testing corrections were carried out using the Benjamini & Hochberg false discovery rate (FDR) method [95]. Gene set enrichment analysis (GSEA) and over representative analysis (ORA) were performed by using the WEB-based Gene Set AnaLysis Toolkit (WebGestalt, https://www.webgestalt.org/) [96]. Pathways in the Reactome database [97] were used as a reference.

Bioinformatics and statistical analyses

Qiime (v1.9.1) [98] was used to calculate alpha diversity indexes and Unifrac distance and sample clustering tree. Unifrac distance measures the evolutionary branch weight of different OTUs in the sample OTU table to assess the differences between OTUs. After obtaining the Unifrac distance matrix, the UPGMA algorithm was used to construct a sample clustering tree. PCoA analyses was conducted with ade4 package in R (v2.15.3). Differences in alpha diversity indexes between groups were analyzed by the Wilcoxon rank-sum test [99] in R (v2.15.3).

Tax4Fun [100] was applied to predict functional profiles. Specifically, it extracted the full-length 16S rRNA gene sequences of prokaryotic genomes from the KEGG database [101] and used the BLASTN algorithm to align them to the SILVA SSU Ref NR database [102] (with a BLAST bitscore > 1500) to establish a relevant matrix. The functional information of bacterial genomes from the KEGG database, annotated using the UProC and PAUDA methods, was then mapped to the SILVA database [88] for functional annotation. OTUs were clustered based on the SILVA database reference sequences.

Differences in log-transformed bacterial abundance and predicted functional profiles between groups were analyzed by the Student’s t-test in R (v2.15.3), analogue to previous studies [103, 104]. Additionally, we applied the Random Forest method [105] to recognize bacterial biomarkers that classify CAD patients from health controls. R package “randomForest” (RF) was used to perform random forest classification with default “mtry” parameters. Variable importance was measured by permuting each predictor variable and measuring the decrease in model accuracy. We adopted a nested cross-validation procedure [106] in which the least important variables were sequentially removed until the mean error rate of the model reached two standard deviations above the lowest error rate. We calculated the area under the ROC curve (AUC) with the “ROCR” package [107] in R (v2.15.3). To select biomarkers, we sorted gut microbes according to their RF importance and calculated the AUC using different numbers of top gut microbes. The gut microbes with the highest AUC were selected as biomarkers.

To assess the correlations of bacterial abundances with level blood indicators and expression levels of genes in paired peripheral blood, the Pearson correlation test was applied, and FDR-adjusted P < 0.05 was considered to be significant. All the plots were visualized in R (v2.15.3).

Availability of data and materials

Original contributions presented in the study are included in the manuscript. Detailed clinical baseline data is available at BIG Submission (BIG SUB, https://ngdc.cncb.ac.cn/omix/preview/FQcgNvTM, OMIX ID: OMIX004322). The raw and processed blood RNA sequencing data and fecal 16S sequencing data have been documented in the NCBI Gene Expression Omnibus (GEO) database (Accession Nos. GSE242046 and GSE242047). The codes to reproduce the analyzed results are available in a dedicated GitHub repository (https://github.com/mayunlong8989/microbiome_CAD).

Abbreviations

CAD:

Coronary artery disease

TMAO:

Trimethylamine-N-oxide

CAMP:

Cationic antimicrobial peptide

BNP:

Brain natriuretic peptide

cTNI:

Cardiac troponin I

CK:

Creatine kinase

CRP:

C-reactive protein

SCFA:

Short chain fatty acids

PBMCs:

Peripheral blood mononuclear cells

DGE:

Differential gene expression

FDR:

False discovery rate

GSEA:

Gene set enrichment analysis

ORA:

Over representative analysis

KEGG:

The Kyoto Encyclopedia of Genes and Genomes

AUC:

Area under the ROC curve

References

  1. Gheorghiade M, Sopko G, De Luca L, Velazquez EJ, Parker JD, Binkley PF, Sadowski Z, Golba KS, Prior DL, Rouleau JL, et al. Navigating the crossroads of coronary artery disease and heart failure. Circulation. 2006;114(11):1202–13.

    Article  PubMed  Google Scholar 

  2. Sing CF, Stengârd JH, Kardia SL. Genes, environment, and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2003;23(7):1190–6.

    Article  CAS  PubMed  Google Scholar 

  3. Van Der Harst P, Verweij N. Identification of 64 novel genetic loci provides an expanded view on the genetic architecture of coronary artery disease. Circ Res. 2018;122(3):433–43.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Zhong Y, Chen L, Li J, Yao Y, Liu Q, Niu K, Ma Y, Xu Y. Integration of summary data from GWAS and eQTL studies identified novel risk genes for coronary artery disease. Medicine. 2021;100(11):e24769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Crea F, Liuzzo G. Pathogenesis of acute coronary syndromes. J Am Coll Cardiol. 2013;61(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  6. Beerkens FJ, Claessen BE, Mahan M, Gaudino MFL, Tam DY, Henriques JPS, Mehran R, Dangas GD. Contemporary coronary artery bypass graft surgery and subsequent percutaneous revascularization. Nat Rev Cardiol. 2022;19(3):195–208.

    Article  CAS  PubMed  Google Scholar 

  7. Behague I, Poirier O, Nicaud V, Evans A, Arveiler D, Luc Gr, Cambou JP, Scarabin PY, Bara L, Green F. β Fibrinogen gene polymorphisms are associated with plasma fibrinogen and coronary artery disease in patients with myocardial infarction: the ECTIM study. Circulation. 1996;93(3):440–9.

    Article  CAS  PubMed  Google Scholar 

  8. Chen L, Yao Y, Jin C, Wu S, Liu Q, Li J, Ma Y, Xu Y, Zhong Y. Integrative genomic analysis identified common regulatory networks underlying the correlation between coronary artery disease and plasma lipid levels. BMC Cardiovasc Disord. 2019;19(1):1–10.

    Article  Google Scholar 

  9. Laaksonen R, Ekroos K, Sysi-Aho M, Hilvo M, Vihervaara T, Kauhanen D, Suoniemi M, Hurme R, März W, Scharnagl H, et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J. 2016;37(25):1967–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li J, Ma Y, Bao Z, Gui X, Li AN, Yang Z, Li MD. Clostridiales are predominant microbes that mediate psychiatric disorders. J Psychiatr Res. 2020;130:48–56.

    Article  PubMed  Google Scholar 

  11. Ma Y, Li MD. Establishment of a strong link between smoking and cancer pathogenesis through DNA methylation analysis. Sci Rep. 2017;7(1):1–13.

    Google Scholar 

  12. Cani PD. Human gut microbiome: hopes, threats and promises. Gut. 2018;67(9):1716–25.

    Article  CAS  PubMed  Google Scholar 

  13. Tomofuji Y, Kishikawa T, Maeda Y, Ogawa K, Otake-Kasamoto Y, Kawabata S, Nii T, Okuno T, Oguro-Igashira E, Kinoshita M. Prokaryotic and viral genomes recovered from 787 Japanese gut metagenomes revealed microbial features linked to diets, populations, and diseases. Cell Genomics. 2022;2(12): 100219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shreiner AB, Kao JY, Young VB. The gut microbiome in health and in disease. Curr Opin Gastroenterol. 2015;31(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63.

    Article  CAS  PubMed  Google Scholar 

  17. Schmidt TSB, Raes J, Bork P. The human gut microbiome: from association to modulation. Cell. 2018;172(6):1198–215.

    Article  CAS  PubMed  Google Scholar 

  18. Yang Z, Li J, Gui X, Shi X, Bao Z, Han H, Li MD. Updated review of research on the gut microbiota and their relation to depression in animals and human beings. Mol Psychiatry. 2020;25(11):2759–72.

    Article  PubMed  Google Scholar 

  19. Xu L, Ma Y, Yuan J, Zhang Y, Wang H, Zhang G, Tu C, Lu X, Li J, Xiong Y. COVID-19 quarantine reveals that behavioral changes have an effect on myopia progression. Ophthalmology. 2021;128(11):1652–4.

    Article  PubMed  Google Scholar 

  20. Bonfiglio F, Liu X, Smillie C, Pandit A, Kurilshikov A, Bacigalupe R, Zheng T, Nim H, Garcia-Etxebarria K, Bujanda L. GWAS of stool frequency provides insights into gastrointestinal motility and irritable bowel syndrome. Cell Genom. 2021;1:3.

    Google Scholar 

  21. Villacampa EG, Larsson L, Mirzazadeh R, Kvastad L, Andersson A, Mollbrink A, Kokaraki G, Monteil V, Schultz N, Appelberg KS. Genome-wide spatial expression profiling in formalin-fixed tissues. Cell Genom. 2021;1(3):100065.

    Article  Google Scholar 

  22. Sonnenburg JL, Bäckhed F. Diet–microbiota interactions as moderators of human metabolism. Nature. 2016;535(7610):56–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chaguza C, Smith JT, Bruce SA, Gibson R, Martin IW, Andam CP. Prophage-encoded immune evasion factors are critical for Staphylococcus aureus host infection, switching, and adaptation. Cell Genom. 2022;2(11):100094.

    Google Scholar 

  24. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Buglioni A, Burnett JC Jr. A gut-heart connection in cardiometabolic regulation. Nat Med. 2013;19(5):534–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Trøseid M, Andersen GØ, Broch K, Hov JR. The gut microbiome in coronary artery disease and heart failure: Current knowledge and future directions. EBioMedicine. 2020;52: 102649.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Zhu Q, Gao R, Zhang Y, Pan D, Zhu Y, Zhang X, Yang R, Jiang R, Xu Y, Qin H. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol Genomics. 2018;50(10):893–903.

    Article  CAS  PubMed  Google Scholar 

  28. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Li XS, Obeid S, Wang Z, Hazen BJ, Li L, Wu Y, Hurd AG, Gu X, Pratt A, Levison BS. Trimethyllysine, a trimethylamine N-oxide precursor, provides near-and long-term prognostic value in patients presenting with acute coronary syndromes. Eur Heart J. 2019;40(32):2700–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Senthong V, Li XS, Hudec T, Coughlin J, Wu Y, Levison B, Wang Z, Hazen SL, Tang WH. Plasma trimethylamine N-Oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol. 2016;67(22):2620–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xue H, Chen X, Yu C, Deng Y, Zhang Y, Chen S, Chen X, Chen K, Yang Y, Ling W. Gut microbially produced indole-3-propionic acid inhibits atherosclerosis by promoting reverse cholesterol transport and its deficiency is causally related to atherosclerotic cardiovascular disease. Circ Res. 2022;131(5):404–20.

    Article  CAS  PubMed  Google Scholar 

  32. Liu H, Chen X, Hu X, Niu H, Tian R, Wang H, Pang H, Jiang L, Qiu B, Chen X. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome. 2019;7(1):1–14.

    Article  Google Scholar 

  33. Asgari S, Luo Y, Huang C-C, Zhang Z, Calderon R, Jimenez J, Yataco R, Contreras C, Galea JT, Lecca L. Higher native Peruvian genetic ancestry proportion is associated with tuberculosis progression risk. Cell Genom. 2022;2(7):100151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sadler MC, Auwerx C, Deelen P, Kutalik Z. Multi-layered genetic approaches to identify approved drug targets. Cell Genom. 2023;3:100341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tang WW, Li DY, Hazen SL. Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol. 2019;16(3):137–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kummen M, Mayerhofer CC, Vestad B, Broch K, Awoyemi A, Storm-Larsen C, Ueland T, Yndestad A, Hov JR, Trøseid M. Gut microbiota signature in heart failure defined from profiling of 2 independent cohorts. J Am Coll Cardiol. 2018;71(10):1184–6.

    Article  PubMed  Google Scholar 

  37. Hu X, Zhou R, Li H, Zhao X, Sun Y, Fan Y, Zhang S. Alterations of gut microbiome and serum metabolome in coronary artery disease patients complicated with non-alcoholic fatty liver disease are associated with adverse cardiovascular outcomes. Front Cardiovasc Med. 1926;2022:8.

    Google Scholar 

  38. Fang C, Zuo K, Fu Y, Li J, Wang H, Xu L, Yang X. Dysbiosis of gut microbiota and metabolite phenylacetylglutamine in coronary artery disease patients with stent stenosis. Front Cardiovasc Med. 2022;9:589.

    Article  Google Scholar 

  39. Zheng Y-Y, Wu T-T, Liu Z-Q, Li A, Guo Q-Q, Ma Y-Y, Zhang Z-L, Xun Y-L, Zhang J-C, Wang W-R. Gut microbiome-based diagnostic model to predict coronary artery disease. J Agric Food Chem. 2020;68(11):3548–57.

    Article  CAS  PubMed  Google Scholar 

  40. Liu Z, Li J, Liu H, Tang Y, Zhan Q, Lai W, Ao L, Meng X, Ren H, Xu D. The intestinal microbiota associated with cardiac valve calcification differs from that of coronary artery disease. Atherosclerosis. 2019;284:121–8.

    Article  CAS  PubMed  Google Scholar 

  41. Han Y, Gong Z, Sun G, Xu J, Qi C, Sun W, Jiang H, Cao P, Ju H. Dysbiosis of gut microbiota in patients with acute myocardial infarction. Front Microbiol. 2021;12: 680101.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Ahrens AP, Culpepper T, Saldivar B, Anton S, Stoll S, Handberg EM, Xu K, Pepine C, Triplett EW, Aggarwal M. A six-day, lifestyle-based immersion program mitigates cardiovascular risk factors and induces shifts in gut microbiota, specifically Lachnospiraceae, Ruminococcaceae, Faecalibacterium prausnitzii: a pilot study. Nutrients. 2021;13(10):3459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chai X, Wen L, Song Y, He X, Yue J, Wu J, Chen X, Cai Z, Qi Z. DEHP exposure elevated cardiovascular risk in obese mice by disturbing the arachidonic acid metabolism of gut microbiota. Sci Total Environ. 2023;875: 162615.

    Article  CAS  PubMed  Google Scholar 

  44. Zhou B, Wang H, Jin H, Wang L. GW29-e1224 The analysis of the diversity of the intestinal flora in patients with acute myocardial infarction. J Am Coll Cardiol. 2018;72(16S):C112–C112.

    Article  Google Scholar 

  45. Yeh ET, Anderson HV, Pasceri V, Willerson JT. C-reactive protein: linking inflammation to cardiovascular complications. Am Heart Assoc. 2001;104:974–5.

    CAS  Google Scholar 

  46. Zheng JL, Lu L, Hu J, Zhang RY, Zhang Q, Chen QJ, Shen WF. Increased serum YKL-40 and C-reactive protein levels are associated with angiographic lesion progression in patients with coronary artery disease. Atherosclerosis. 2010;210(2):590–5.

    Article  CAS  PubMed  Google Scholar 

  47. Kolovou G, Yiannakouris N, Hatzivassiliou M, Malakos J, Daskalova D, Hatzigeorgiou G, Cariolou MA, Cokkinos DV. Association of apolipoprotein E polymorphism with myocardial infarction in Greek patients with coronary artery disease. Curr Med Res Opin. 2002;18(3):118–24.

    Article  CAS  PubMed  Google Scholar 

  48. Bos MM, de Vries L, Rensen PC, van Dijk KW, Blauw GJ, van Heemst D, Noordam R. Apolipoprotein E genotype, lifestyle and coronary artery disease: gene-environment interaction analyses in the UK Biobank population. Atherosclerosis. 2021;328:33–7.

    Article  CAS  PubMed  Google Scholar 

  49. Zhao Y, Chen J, Freudenberg JM, Meng Q, Consortium C, Rajpal DK, Yang X. Network-based identification and prioritization of key regulators of coronary artery disease loci. Arterioscler Thromb Vasc Biol. 2016;36(5):928–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kamal AM, Sebak SA, Sanad EF. Mixed Lineage Kinase Domain-Like Pseudokinase (MLKL) gene expression in human atherosclerosis with and without type 2 diabetes mellitus. Iran Biomed J. 2021;25(4):265.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 2017;18(2):127–36.

    Article  CAS  PubMed  Google Scholar 

  52. Newton K, Dugger DL, Wickliffe KE, Kapoor N, de Almagro MC, Vucic D, Komuves L, Ferrando RE, French DM, Webster J. Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis. Science. 2014;343(6177):1357–60.

    Article  CAS  PubMed  Google Scholar 

  53. Newton K, Dugger D, Maltzman A, Greve J, Hedehus M, Martin-McNulty B, Carano R, Cao T, Van Bruggen N, Bernstein L. RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury. Cell Death Differ. 2016;23(9):1565–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xu H, Du X, Liu G, Huang S, Du W, Zou S, Tang D, Fan C, Xie Y, Wei Y. The pseudokinase MLKL regulates hepatic insulin sensitivity independently of inflammation. Mol Metab. 2019;23:14–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mitra S, Drautz-Moses DI, Alhede M, Maw MT, Liu Y, Purbojati RW, Yap ZH, Kushwaha KK, Gheorghe AG, Bjarnsholt T. In silico analyses of metagenomes from human atherosclerotic plaque samples. Microbiome. 2015;3(1):1–14.

    Article  Google Scholar 

  56. Liu X, Mao B, Gu J, Wu J, Cui S, Wang G, Zhao J, Zhang H, Chen W. Blautia—a new functional genus with potential probiotic properties? Gut Microbes. 2021;13(1):1875796.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Ranaivo H, Thirion F, Béra-Maillet C, Guilly S, Simon C, Sothier M, Van Den Berghe L, Feugier-Favier N, Lambert-Porcheron S, Dussous I. Increasing the diversity of dietary fibers in a daily-consumed bread modifies gut microbiota and metabolic profile in subjects at cardiometabolic risk. Gut Microbes. 2022;14(1):2044722.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Pagonas N, Seibert FS, Liebisch G, Seidel M, Giannakopoulos T, Sasko B, Ritter O, Babel N, Westhoff TH. Association of plasma propionate concentration with coronary artery disease in a large cross-sectional study. Front Cardiovasc Med. 2023;10:1063296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jadoon A, Mathew AV, Byun J, Gadegbeku CA, Gipson DS, Afshinnia F, Pennathur S. Group MKTCCI: Gut microbial product predicts cardiovascular risk in chronic kidney disease patients. Am J Nephrol. 2018;48(4):269–77.

    Article  PubMed  Google Scholar 

  60. Dong C, Yang Y, Wang Y, Hu X, Wang Q, Gao F, Sun S, Liu Q, Li L, Liu J. Gut microbiota combined with metabolites reveals unique features of acute myocardial infarction patients different from stable coronary artery disease. J Adv Res. 2023;46:101–12.

    Article  CAS  PubMed  Google Scholar 

  61. Hu T, Wu Q, Yao Q, Jiang K, Yu J, Tang Q. Short-chain fatty acid metabolism and multiple effects on cardiovascular diseases. Ageing Res Rev. 2022;81:101706.

    Article  CAS  PubMed  Google Scholar 

  62. Wang A, Li Z, Sun Z, Zhang D, Ma X. Gut-derived short-chain fatty acids bridge cardiac and systemic metabolism and immunity in heart failure. J Nutr Biochem. 2023;120:109370.

    Article  CAS  PubMed  Google Scholar 

  63. Park O-J, Ha Y-E, Sim J-R, Lee D, Lee E-H, Kim S-Y, Yun C-H, Han SH. Butyrate potentiates Enterococcus faecalis lipoteichoic acid-induced inflammasome activation via histone deacetylase inhibition. Cell Death Discovery. 2023;9(1):107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kibbie JJ, Dillon SM, Thompson TA, Purba CM, McCarter MD, Wilson CC. Butyrate directly decreases human gut lamina propria CD4 T cell function through histone deacetylase (HDAC) inhibition and GPR43 signaling. Immunobiology. 2021;226(5): 152126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhong H, Yu H, Chen J, Mok SWF, Tan X, Zhao B, He S, Lan L, Fu X, Chen G. The short-chain fatty acid butyrate accelerates vascular calcification via regulation of histone deacetylases and NF-κB signaling. Vascul Pharmacol. 2022;146: 107096.

    Article  CAS  PubMed  Google Scholar 

  66. Zhou M, Li D, Xie K, Xu L, Kong B, Wang X, Tang Y, Liu Y, Huang H. The short-chain fatty acid propionate improved ventricular electrical remodeling in a rat model with myocardial infarction. Food Funct. 2021;12(24):12580–93.

    Article  CAS  PubMed  Google Scholar 

  67. Bae J, Salamon RJ, Brandt EB, Paltzer WG, Zhang Z, Britt EC, Hacker TA, Fan J, Mahmoud AI. Malonate promotes adult cardiomyocyte proliferation and heart regeneration. Circulation. 2021;143(20):1973–86.

    Article  CAS  PubMed  Google Scholar 

  68. Jiang X, Huang X, Tong Y, Gao H. Butyrate improves cardiac function and sympathetic neural remodeling following myocardial infarction in rats. Can J Physiol Pharmacol. 2020;98(6):391–9.

    Article  CAS  PubMed  Google Scholar 

  69. Li Y. The role of antimicrobial peptides in cardiovascular physiology and disease. Biochem Biophys Res Commun. 2009;390(3):363–7.

    Article  CAS  PubMed  Google Scholar 

  70. Joseph G, Tarnow L, Astrup AS, Hansen TK, Parving H-H, Flyvbjerg A, Frystyk J. Plasma α-Defensin Is Associated with Cardiovascular Morbidity and Mortality in Type 1 Diabetic Patients. J Clin Endocrinol Metab. 2008;93(4):1470–5.

    Article  CAS  PubMed  Google Scholar 

  71. Bao J, Sato K, Li M, Gao Y, Abid R, Aird W, Simons M, Post MJ. PR-39 and PR-11 peptides inhibit ischemia-reperfusion injury by blocking proteasome-mediated IκBα degradation. Am J Physiol Heart Circ Physiol. 2001;281(6):H2612–8.

    Article  CAS  PubMed  Google Scholar 

  72. Stefan KL, Kim MV, Iwasaki A, Kasper DL. Commensal microbiota modulation of natural resistance to virus infection. Cell. 2020;183(5):1312-1324.e1310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Voloshyna I, Littlefield MJ, Reiss AB. Atherosclerosis and interferon-γ: new insights and therapeutic targets. Trends Cardiovasc Med. 2014;24(1):45–51.

    Article  CAS  PubMed  Google Scholar 

  74. Kartha VK, Duarte FM, Hu Y, Ma S, Chew JG, Lareau CA, Earl A, Burkett ZD, Kohlway AS, Lebofsky R. Functional inference of gene regulation using single-cell multi-omics. Cell Genom. 2022;2(9):100166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Koh KD, Bonser LR, Eckalbar WL, Yizhar-Barnea O, Shen J, Zeng X, Hargett KL, Sun DI, Zlock LT, Finkbeiner WE. Genomic characterization and therapeutic utilization of IL-13-responsive sequences in asthma. Cell Genom. 2023;3(1):100229.

    Article  CAS  PubMed  Google Scholar 

  76. Bossini-Castillo L, Glinos DA, Kunowska N, Golda G, Lamikanra AA, Spitzer M, Soskic B, Cano-Gamez E, Smyth DJ, Cattermole C. Immune disease variants modulate gene expression in regulatory CD4+ T cells. Cell Genom. 2022;2(4):100117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Steen AD, Crits-Christoph A, Carini P, DeAngelis KM, Fierer N, Lloyd KG, Cameron Thrash J. High proportions of bacteria and archaea across most biomes remain uncultured. ISME J. 2019;13(12):3126–30.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Ma Y, Zhou Y, Jiang D, Dai W, Li J, Deng C, Chen C, Zheng G, Zhang Y, Qiu F: Integration of human organoids single‐cell transcriptomic profiles and human genetics repurposes critical cell type‐specific drug targets for severe COVID‐19. Cell Prolif. 2023:e13558.

  79. Lv Y, Xu X, Wang Z, Huang Y, Ma Y, Wu M. Integrated multi-omics data analysis identifies a novel genetics-risk gene of IRF4 associated with prognosis of oral cavity cancer. Curr Bioinform. 2022;17(8):744–58.

    Article  CAS  Google Scholar 

  80. Ma Y, Qiu F, Deng C, Li J, Huang Y, Wu Z, Zhou Y, Zhang Y, Xiong Y, Yao Y. Integrating single-cell sequencing data with GWAS summary statistics reveals CD16+ monocytes and memory CD8+ T cells involved in severe COVID-19. Genome Med. 2022;14(1):16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Xiang B, Deng C, Qiu F, Li J, Li S, Zhang H, Lin X, Huang Y, Zhou Y, Su J. Single cell sequencing analysis identifies genetics-modulated ORMDL3+ cholangiocytes having higher metabolic effects on primary biliary cholangitis. J Nanobiotechnol. 2021;19(1):406.

    Article  CAS  Google Scholar 

  82. Ma Y, Huang Y, Zhao S, Yao Y, Zhang Y, Qu J, Wu N, Su J. Integrative genomics analysis reveals a 21q22. 11 locus contributing risk to COVID-19. Human Mol Genet. 2021;30(13):1247–58.

    Article  CAS  Google Scholar 

  83. Ma Y, Deng C, Zhou Y, Zhang Y, Qiu F, Jiang D, Zheng G, Li J, Shuai J, Zhang Y, et al. Polygenic regression uncovers trait-relevant cellular contexts through pathway activation transformation of single-cell RNA sequencing data. Cell Genom. 2023;3(9): 100383.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Huang Y, Luo J, Zhang Y, Zhang T, Fei X, Chen L, Zhu Y, Li S, Zhou C, Xu K. Identification of MKNK1 and TOP3A as ovarian endometriosis risk-associated genes using integrative genomic analyses and functional experiments. Comput Struct Biotechnol J. 2023;21:1510–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018;34(17):i884–90.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Rognes T, Flouri T, Nichols B, Quince C, Mahé F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4: e2584.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, Lesniewski RA, Oakley BB, Parks DH, Robinson CJ. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75(23):7537–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73(16):5261–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004;32(5):1792–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Dixon P. VEGAN, a package of R functions for community ecology. J Veg Sci. 2003;14(6):927–30.

    Article  Google Scholar 

  91. Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37(8):907–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30.

    Article  CAS  PubMed  Google Scholar 

  93. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):1–21.

    Article  Google Scholar 

  94. Owen DB. The power of Student’s t-test. J Am Stat Assoc. 1965;60(309):320–33.

    Google Scholar 

  95. Green GH, Diggle PJ. On the operational characteristics of the Benjamini and Hochberg False Discovery Rate procedure. Stat Appl Genet Mol Biol. 2007;6(1):Article27.

  96. Liao Y, Wang J, Jaehnig EJ, Shi Z, Zhang B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 2019;47(W1):W199–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Fabregat A, Jupe S, Matthews L, Sidiropoulos K, Gillespie M, Garapati P, Haw R, Jassal B, Korninger F, May B. The reactome pathway knowledgebase. Nucleic Acids Res. 2018;46(D1):D649–55.

    Article  CAS  PubMed  Google Scholar 

  98. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7(5):335–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Lam F, Longnecker M. A modified Wilcoxon rank sum test for paired data. Biometrika. 1983;70(2):510–3.

    Article  Google Scholar 

  100. Wemheuer F, Taylor JA, Daniel R, Johnston E, Meinicke P, Thomas T, Wemheuer B. Tax4Fun2: prediction of habitat-specific functional profiles and functional redundancy based on 16S rRNA gene sequences. Environ Microbiome. 2020;15(1):1–12.

    Article  Google Scholar 

  101. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45(D1):D353–61.

    Article  CAS  PubMed  Google Scholar 

  102. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2012;41(D1):D590–6.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Liu K, Xu Q, Wang L, Wang J, Guo W, Zhou M. The impact of diet on the composition and relative abundance of rumen microbes in goat. Asian Australas J Anim Sci. 2017;30(4):531.

    Article  CAS  PubMed  Google Scholar 

  104. Benson AK, Kelly SA, Legge R, Ma F, Low SJ, Kim J, Zhang M, Oh PL, Nehrenberg D, Hua K. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci. 2010;107(44):18933–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Liaw A, Wiener M. Classification and regression by randomForest. R News. 2002;2(3):18–22.

    Google Scholar 

  106. Wainer J, Cawley G. Nested cross-validation when selecting classifiers is overzealous for most practical applications. Expert Syst Appl. 2021;182:115222.

    Article  Google Scholar 

  107. Sing T, Sander O, Beerenwinkel N, Lengauer T. ROCR: visualizing classifier performance in R. Bioinformatics. 2005;21(20):3940–1.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all participants and investigators who provide a contribution to the work.

Funding

This research was funded by the Hangzhou Medical and Health Technology Project (No. Z20200135), the Construction Fund of Key Medical Disciplines of Hangzhou (No. OO20200121), the National Natural Science Foundation of China (No. 32200535), and the Scientific Research Foundation for Talents of Wenzhou Medical University (No. KYQD20201001).

Author information

Authors and Affiliations

Authors

Contributions

CL, MX, and LJ conducted the experiments and wrote the article. LM, YC, GX, and LX were involved in parts of the experiments. XY, MY, and ZY conceived the hypothesis, designed the experiments, and reviewed the article. ZY and MY helped in guiding the revision. All authors reviewed and approved the final article.

Corresponding authors

Correspondence to Yunlong Ma, Yizhou Xu or Yigang Zhong.

Ethics declarations

Ethics approval and consent to participate

All procedures followed were in accordance with the ethical standards of the responsible committee on human experimentation (institutional and national) and with the Helsinki Declaration of 1975 (in its most recently amended version). This study was reviewed and approved by the Ethics Committee of Zhejiang University. Informed consent was obtained from all patients included in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Supplementary Table S1. Summary of sequnce reads for all samples. Supplementary Table S2. Summary of the basic information of all RNA samples. Supplementary Table S3. Differential abundant analysis of 16S-seq profiles between CAD patients and controls. Supplementary Table S4. The results of functional annotation and differential analyses on bacterial profiles between CAD patients and controls. Supplementary Table S5. Differential gene expression analysis of transcriptomic profiles between CAD patients and controls.

Additional file 2:

Supplementary Figure S1. Differential of gut microbiota between CAD patients and controls at class (A), order (B), family (C), and species (D) level. The abundance of microbe was log-transformed. The significant level of each microbe was calculated by using Student’s t-test. (E) The top 10 differential genera between CAD and control group by means of Mean Decrease Gini.

Additional file 3:

Supplementary Figure S2. The association of differential abundant bacteria with clinical indices accordining to different levels of order (A), family (B), and species (C). Significances tested by Pearson correlation analysis. * FDR < 0.05; ** FDR < 0.01; *** FDR < 0.001. Color bar indicates the level of correlation coefficients (corr).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, L., Mou, X., Li, J. et al. Alterations in gut microbiota and host transcriptome of patients with coronary artery disease. BMC Microbiol 23, 320 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-03071-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-023-03071-w

Keywords