3,033
Views
5
CrossRef citations to date
0
Altmetric
Research Paper

Shotgun Metagenomics of Gastric Biopsies Reveals Compositional and Functional Microbiome Shifts in High- and Low-Gastric-Cancer-Risk Populations from Colombia, South America

, , , , , , & show all
Article: 2186677 | Received 30 Aug 2022, Accepted 27 Feb 2023, Published online: 12 Mar 2023

ABSTRACT

Along with Helicobacter pylori infection, the gastric microbiota is hypothesized to modulate stomach cancer risk in susceptible individuals. Whole metagenomic shotgun sequencing (WMS) is a sequencing approach to characterize the microbiome with advantages over traditional culture and 16S rRNA sequencing including identification of bacterial and non-bacterial taxa, species/strain resolution, and functional characterization of the microbiota. In this study, we used WMS to survey the microbiome in extracted DNA from antral gastric biopsy samples from Colombian patients residing in the high-risk gastric cancer town Túquerres (n = 10, H. pylori-positive = 7) and low-risk town of Tumaco (n = 10, H. pylori-positive = 6). Kraken2/Bracken was used for taxonomic classification and abundance. Functional gene profiles were inferred by InterProScan and KEGG analysis of assembled contigs and gene annotation. The most abundant taxa represented bacteria, non-human eukaryota, and viral genera found in skin, oral, food, and plant/soil environments including Staphylococus, Streptococcus, Bacillus, Aspergillus, and Siphoviridae. H. pylori was the predominant taxa present in H. pylori-positive samples. Beta diversity was significantly different based on H. pylori-status, risk group, and sex. WMS detected more bacterial taxa than 16S rRNA sequencing and aerobic, anaerobic, and microaerobic culture performed on the same gastric biopsy samples. WMS identified significant differences in functional profiles found between H. pylori-status, but not risk or sex groups. H. pylori-positive samples were significantly enriched for H. pylori-specific genes including virulence factors such as vacA, cagA, and urease, while carbohydrate and amino acid metabolism genes were enriched in H. pylori-negative samples. This study shows WMS has the potential to characterize the taxonomy and function of the gastric microbiome as risk factors for H. pylori-associated gastric disease. Future studies will be needed to compare and validate WMS versus traditional culture and 16S rRNA sequencing approaches for characterization of the gastric microbiome.

Introduction

Although Helicobacter pylori infection is the strongest factor associated with developing gastritis and stomach cancer, increasing evidence suggests the gastric microbiota may also have a key role in modulating disease risk in susceptible individualsCitation1–3. Culture and 16S rRNA sequencing have been the primary means to study the gastric microbiome, and while these approaches have revealed important insights into the potential influence of the microbiome on gastric disease risk and progression, these methods have limited scope including low-throughput or nonspecific taxonomic identification. Unlike 16S rRNA sequencing, metagenomic shotgun sequencing (WMS) is an untargeted, microbiome sequencing approach that enables 1) identification of bacterial, eukaryota, viral, and archaea microbes, 2) species and strain level taxonomic resolution, and 3) functional characterization of the microbiotaCitation4,Citation5. As a result, WMS is being increasingly used with 16S rRNA profiling to study the gastrointestinal microbiome, especially to identify risk factors associated with intestinal inflammatory diseases and cancersCitation6–12. While numerous studies have characterized the gastric microbiome using 16S rRNA profilingCitation1, WMS provides an opportunity to further define how the microbiome taxonomic composition and function may contribute as factors associated with disease risk.

In this study, we utilized WMS to survey the microbiome in gastric biopsy samples collected from Colombian patients residing in the high gastric cancer risk (HGCR) town of Túquerres located in the Andes Mountains and low gastric cancer risk (LGCR) coastal town of Tumaco. While both populations have high prevalence of H. pylori infection (>80%), additional factors have been extensively investigated over the last four decades to explain the differences in gastric cancer risk. Geographic location has been hypothesized to represent multifactorial influences of host and H. pylori genotypes as well as dietary and environmental variables including helminth infection and host microbiome composition/functionCitation13. Previous investigations of these Colombian cohorts by traditional culture and 16S rRNA profiling have revealed significant differences in the gastric microbiome composition between gastric cancer populationsCitation13–15. Thus, in the current study, the gastric microbiome was further evaluated using WMS to assess its association as a risk factor for disease in these patient cohorts.

A significant challenge for next-generation sequenced-based profiling of the microbiome in tissue biopsy samples is the low abundance of microbe to host cells, especially in the stomach where microbial density is 5–10 log-folds lower compared to the lower intestine and fecesCitation16. 16S rRNA profiling attempts to overcome this challenge via PCR amplification of 16S rRNA genes regions prior to sequencing. However, this method is prone to technical artifacts due to amplification bias and sensitivity to contaminationCitation17,Citation18. Conversely, WMS does not require PCR and therefore potentially provides a more accurate reflection of the gut microbiota compared to 16S rRNA profilingCitation11. Despite this advantage, WMS is substantially more expensive and resource intensive than 16S rRNA profiling because magnitudes of deeper sequencing are required to detect microbiota genes in tissue samples with low microbe-to-human DNA ratios, even with methods to deplete host DNA prior to sequencingCitation19. Thus, an objective of the current study was to compare gastric microbiota profiles between HGCR and LGCR tissue samples using WMS versus 16S rRNA approaches as well as by traditional bacterial culture. To our knowledge, this study is the first to use WMS, 16S rRNA and culturing profiling methodologies to evaluate and compare the gastric microbiome. We hypothesize WMS is a feasible approach to characterize the composition and functional potential of the gastric microbiota from human stomach biopsy tissue as well as identify significant features that differentiate patients from HGCR and LGCR populations.

Results

Study population

Twenty gastric biopsy tissue samples for microbiome analysis were selected to match for H. pylori status, risk group, histopathology scores, age, and sex from a total set of 163 adult patients undergoing upper gastrointestinal endoscopy from the HGCR and LGCR in Colombia (). There were no statistical differences in age between H. pylori status, risk, or sex groups. Histopathology scores were not statistically different based on H. pylori status or risk group, but samples from males had significantly higher pathology scores compared to those from females (P = 0.008, Mann – Whitney U-test).

Table 1. H. pylori status, risk group, histopathology scores, age, and sex of patient gastric biopsy samples from low and high gastric cancer risk populations in Colombia.

WMS taxonomic profiling of the gastric microbiome

WMS yielded 35,062,050 to 168,230,930 total reads per sample after quality control. Of these reads, WMS profiling classified ~96–99% reads as host (i.e., Homo sapiens), and ~1–4% reads were unclassified taxa (supplemental figure S1). The remaining reads belonged to bacteria (~0.03–0.2%), archaea (~0.0007–0.002%), virus (~0.002–0.006%), and non-human eukaryota (~0.03–0.1%) taxa (supplemental figure S1). Thus, 42,319 to 304,298 total reads per sample classified as bacteria, non-human eukaryota, viral, and archaea operational taxonomic units (OTUs) were further evaluated in this study (supplemental figure S1). There were no statistical differences in the number of reads in total or per taxa between H. pylori status, risk group, or sexes, except that males had significantly higher bacterial reads than females (86,045.0 ± 38,847.2 vs. 51,290.7 ± 26,999.1, P = 0.03). Of these, bacteria and non-human eukaryotes predominated. Virus followed by archaea were the next most abundant. At the bacterial genus level, WMS identified significantly more OTUs compare to 16S rRNA sequencing (P < 0.001) (supplemental figure S2).

When all taxa (i.e., bacteria, archaea, virus, and non-human eukaryota) were evaluated, the top 10 most abundant OTUs represented bacteria, non-human eukaryota, and viral genera that have been described in skin, oral, small/large intestine, food, plant, and soil environments (). Beta diversity was significantly different based on H. pylori status (P = 0.031), risk group (P = 0.022), and sex (P = 0.004) (). Observed OTUs, Chao1, and Shannon alpha diversity metrics were not significant between these groups. The Simpson alpha diversity metric was significantly lower in H. pylori-positive samples (P = 0.024) and males (P = 0.019) (). When Helicobacter reads were excluded from these analyses, beta diversity was no longer statistically different between H. pylori-positive versus H. pylori-negative samples, but remained significantly different between risk groups and sexes (supplemental figure S3). There was no significance difference in alpha diversity metrics when Helicobacter reads were excluded from analyses (supplemental figure S3). Differential abundance analysis by GLM and T-test identified that males were significantly enriched in Keratinibaculum spp., regardless if H. pylori reads were included or excluded (supplemental figure S5A). No OTUs were statistically abundant between the H. pylori status or risk groups. While sample F3 appeared to be an outlier in the PCA plots, statistical significance for beta diversity did not change when omitted, suggesting this sample did not skew results (results not shown).

Figure 1. Relative percent of reads classified as bacteria, archaea, virus, and non-human eukaryota OTUs at the kingdom (A), phylum (B), and genus (C) level using WMS. Absolute reads classified as bacteria, archaea, virus, and non-human eukaryota OTUs at the kingdom (D), phylum (E), and genus (F) level using WMS. The top 10 most abundant OTUs are shown.

Figure 1. Relative percent of reads classified as bacteria, archaea, virus, and non-human eukaryota OTUs at the kingdom (A), phylum (B), and genus (C) level using WMS. Absolute reads classified as bacteria, archaea, virus, and non-human eukaryota OTUs at the kingdom (D), phylum (E), and genus (F) level using WMS. The top 10 most abundant OTUs are shown.

Figure 2. PCA plots of Aitchison distance for beta diversity comparisons of bacteria, archaea, virus, and non-human eukaryota OTUs at the genus level detected using WMS for H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) the between groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics on bacteria, archaea, virus, and non-human eukaryota OTUs at the genus level detected using WMS for H. pylori status (D), risk group (E), and sex (F) samples. Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

Figure 2. PCA plots of Aitchison distance for beta diversity comparisons of bacteria, archaea, virus, and non-human eukaryota OTUs at the genus level detected using WMS for H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) the between groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics on bacteria, archaea, virus, and non-human eukaryota OTUs at the genus level detected using WMS for H. pylori status (D), risk group (E), and sex (F) samples. Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

Analyses repeated on only bacteria taxa (i.e., archaea, virus, and non-human eukaryote taxa excluded) identified that genera associated with the skin, oral, small/large intestine, and soil were the most abundant in samples () and predominately belonged to the Proteobacteria and Firmicutes phyla. Helicobacter and Klebsiella spp. were the most abundant Proteobacteria detected. Staphylococcus, Streptococcus, Bacillus, and Lactococcus, and Clostridium spp., all of which were Firmicutes, were within the 10 most abundant bacterial taxa detected. The soil-associated genera Arthrobacter and Thermatoga spp. were also present. A significant difference in beta diversity was again present based on H. pylori status (P = 0.012), risk group (P = 0.012), and sex (P = 0.005) (). There were no significant differences in alpha diversity metrics based on H. pylori status when Helicobacter reads were included or excluded from analyses (, supplemental figure S4). If Helicobacter reads were excluded, beta diversity remained significantly different between risk groups (P = 0.011) and sex (P = 0.049) (supplemental figure S4). Keratinibaculum spp. remained significantly enriched in males when only bacterial taxa were evaluated (supplemental figure S5B). Statistical significance for beta diversity did not change when sample F3 was omitted from PCA plots (results not shown).

Figure 3. Relative percent of reads classified as bacteria OTUs at the phylum(A), family(B), and genus (C) level using WMS. Absolute reads classified as bacteria OTUs at the phylum(E), family(F), and genus (G) level using WMS. The top 10 most abundant OTUs are shown.

Figure 3. Relative percent of reads classified as bacteria OTUs at the phylum(A), family(B), and genus (C) level using WMS. Absolute reads classified as bacteria OTUs at the phylum(E), family(F), and genus (G) level using WMS. The top 10 most abundant OTUs are shown.

Figure 4. PCA plots of Aitchison distance for beta diversity comparisons of bacteria OTUs at the genus level detected using WMS between H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics on bacteria OTUs at the genus level detected using WMS for H. pylori status (D), risk group (E), and sex (F) samples. Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

Figure 4. PCA plots of Aitchison distance for beta diversity comparisons of bacteria OTUs at the genus level detected using WMS between H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics on bacteria OTUs at the genus level detected using WMS for H. pylori status (D), risk group (E), and sex (F) samples. Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

There was no significant difference in alpha or beta diversity between groups when non-human eukaryote (supplemental figure S6), archaea (supplemental figure S7), or viral (supplemental figure S8) taxa were individually analyzed. Aspergillus spp., a fungal genus, was the predominant non-human eukaryote OTU noted (supplemental figure S9). Siphoviridae spp., which target bacterial and archaea hosts, were the most enriched viral OTU in all samples (supplemental figure S10). Of archaea, Pyrolobus species were the most common (supplemental figure S11). Together, these results indicate that H. pylori alters the structure and diversity of the gastric microbiome and primarily affects bacterial communities. Non-bacterial taxa were in general uniform across samples, regardless of H. pylori infection status. Since significant differences in diversity were not present for other taxonomic kingdoms, bacteria appear to be the primary constituents of gastric microbiota diversity.

16S rRNA versus WMS for bacterial microbiome characterization

Unlike WMS analysis, 16S rRNA profiling only identified significant differences in beta diversity based on H. pylori-status (). Additionally, observed OTUs, Chao1, Shannon, and Simpson indices were significantly lower in H. pylori-positive samples (). These alpha diversity metrics suggests H. pylori outcompetes other microbes for colonization in the gastric niche, and when H. pylori is absent, the gastric niche may be occupied by a higher diversity of microbes. When Helicobacter reads were excluded, beta diversity as well as Shannon and Simpson indices were no longer statistically different between H. pylori-status groups (supplemental figure 12). Only Helicobacter, Streptococcus, and Lactococcus were shared between the top 10 most abundant bacterial genera between 16S rRNA versus WMS profiling (). Linear regression of relative abundance at the genus level was performed to evaluate how similar the microbiome profiles were between 16S rRNA and WMS per sample. Seven of thirteen H. pylori-positive samples had strong coefficient of determinations (R2>0.75), suggesting these 16S rRNA and WMS methods yielded similar gastric microbiome profiles (supplemental figure 13A). Interestingly, when Helicobacter reads were removed from the linear regression analysis, these R2 values became less than 0.1 (supplemental figure 13B). For the remaining 5 H. pylori-positive and 7 H. pylori-negative samples, microbiome profiles at the genus level were poorly correlated between 16S rRNA and WMS methods, as shown by low R2 values (supplemental figure 13A). These findings indicate that gastric microbiome profiles drastically differ based on the sequencing methodology utilized.

Figure 5. PCA plots of Aitchison distance for beta diversity comparisons of bacteria OTUs at the genus level detected using 16S rRNA profiling for H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics of bacteria OTUs at the genus level detected using 16S rRNA profiling for H. pylori status (D), risk group (E), and sex (F). Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

Figure 5. PCA plots of Aitchison distance for beta diversity comparisons of bacteria OTUs at the genus level detected using 16S rRNA profiling for H. pylori status (A), risk group (B), and sex (C) samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. Observed OTUs, Chao1, Shannon and Simpson alpha diversity metrics of bacteria OTUs at the genus level detected using 16S rRNA profiling for H. pylori status (D), risk group (E), and sex (F). Statistical comparisons of alpha diversity metrics between groups was performed using the Wilcoxon rank sum test with P-value adjustment using the Holm method.

Figure 6. Relative percent of reads classified as bacterial OTUs at the phylum (A), family (B), and genus (C) level detected using 16S rRNA profiling. Absolute reads classified as bacterial OTUs at the phylum (D), family (E), and genus (F) level detected using 16S rRNA profiling. The top 10 most abundant OTUs are shown.

Figure 6. Relative percent of reads classified as bacterial OTUs at the phylum (A), family (B), and genus (C) level detected using 16S rRNA profiling. Absolute reads classified as bacterial OTUs at the phylum (D), family (E), and genus (F) level detected using 16S rRNA profiling. The top 10 most abundant OTUs are shown.

Culture versus 16S rRNA and WMS for bacterial microbiome characterization

Bacterial genera detected by 16S rRNA and WMS analysis were compared with those cultured from gastric biopsies. 117 non-Helicobacter bacteria isolates representing 20 genera were cultured from 17 gastric biopsy samples under aerobic, anaerobic, and microaerobic conditions (). No non-Helicobacter bacteria were cultured in three samples (B4, E1, and G1; ). Non-Helicobacter bacterial genera identified by 16S rRNA, WMS, and culture profiling were plotted on a binary presence/absence heatmap to determine the concurrence of detection between methods per sample (). For culture-based detection, there were 49 instances in which a specific genus was present in a specific gastric biopsy sample (hashed boxes in ). WMS had more of these genera in common with culture than 16S rRNA sequencing (41/49 versus 35/49, respectively) (), but these proportions were not statistically different using the z-score test. Thirty-one of the isolates detected by culture were identified by both WMS and 16S rRNA profiling (). Interestingly, culture detected four isolates not identified by 16S rRNA or WMS profiling (Gordonia in sample H4, Atopobium in samples C4 and E4, and Aerococcus in sample A4). While Gordonia and Aerococcus were identified by 16S rRNA or WMS profiling in other samples (Gordonia in samples A4 and B4, Aerococcus G1, G2, A5, C4, D3, B4, C5), Atopobium was not detected by either sequencing approach in any of the 20 biopsies (). Under the aerobic, anaerobic, and microaerobic conditions, growth for fungi or archaea were not observed after 2–3 weeks of culture from any gastric biopsy sample.

Figure 7. A) Heatmap showing absolute number non-Helicobacter bacterial isolates cultured per genera from each gastric biopsy sample. Culture conditions in which genera were isolated are indicated in parenthesis next to taxa names. Ae, aerobic; An, anaerobic; M, microaerobic. B) The concurrence of genera detected by culture, 16s rRNA and/or WMS profiling per gastric biopsy sample using a heatmap showing the binary presence/absence of genera per method. Note hashed boxes represent the 49 total instances in which at least one isolate from specific genus was cultured from a gastric biopsy sample.

Figure 7. A) Heatmap showing absolute number non-Helicobacter bacterial isolates cultured per genera from each gastric biopsy sample. Culture conditions in which genera were isolated are indicated in parenthesis next to taxa names. Ae, aerobic; An, anaerobic; M, microaerobic. B) The concurrence of genera detected by culture, 16s rRNA and/or WMS profiling per gastric biopsy sample using a heatmap showing the binary presence/absence of genera per method. Note hashed boxes represent the 49 total instances in which at least one isolate from specific genus was cultured from a gastric biopsy sample.

Functional prediction of bacterial gastric microbiome from WMS

WMS reads classified as bacterial were assembled into contigs and annotated for protein coding genes. Bacterial reads were assembled into 1,361 to 7,628 contigs comprising 289,174 to 2,366,126 bp of total metagenomics sequence and included 1,120 to 5,513 annotated protein coding genes (supplemental table S1). DIAMOND analysis of the annotated protein gene sequences against the nr database was performed to determine the closest taxonomic homolog for each gene. Beta diversity in the taxonomic profiles of annotated protein genes was statistically different between H. pylori-positive versus H. pylori-negative samples (supplemental figure S14). Only genes corresponding to Helicobacter spp. were statistically more abundant in H. pylori-positive compared to H. pylori-negative samples based on differential abundance analysis by GLM and T-test (FDR<0.001). There were no statistical differences in beta diversity or differentially abundant genes between risk groups or sexes (supplemental figure 14). H. pylori-positive compared to H. pylori-negative samples were assembled into more contigs (3,565.4 ± 1,983.1 vs. 3,941.0 ± 1,474.6, P = 0.635) with larger overall metagenomics sequence (740,570.4 ± 379,153.0 vs. 1,590,043.4 ± 536,639.7, P = 0.002) as well as had more genes annotated (2,435.6 ± 1,038.4 vs. 3,812.1 ± 885.3, P = 0.006).

Gene annotations were analyzed using InterProScan, Gene Ontology (GO), and KEGG to evaluate and compare the functional potential of the H. pylori-positive versus H. pylori-negative gastric microbiomes. The beta diversity for these functional features was significantly different based on H. pylori status (, supplemental figure S15), but not risk groups or sexes (supplemental figures 16, 17, 18). Eleven InterProScan protein domains and 11 GO terms were significantly enriched in H. pylori-positive compared to H. pylori-negative samples based on differential abundance analysis by GLM and T-test (FDR<0.01; , supplemental figure S15C). The protein annotations corresponding to these enriched InterProScan protein domains and GO terms features were attributed to H. pylori. Two KEGG pathways (carbohydrate and amino acid metabolism) were statistically higher in H. pylori-negative samples (FDR<0.05; ) and were attributed to over 15 different microbial taxa present at different abundances in each sample. The most commonly shared genera attributed to carbohydrate metabolism annotations in H. pylori-negative samples was Neisseria (n = 4/7; samples F3, A5, G2, and H2) followed by Corynebacterium (n = 2/7; samples E1 and G1). For amino acid metabolism genes, the most commonly shared genera in these samples was represented by Bacillus (n = 2/7; samples F3 and A5), Corynebacterium (n = 2/7; samples E1 and G1), Propionibacterium/Cutibacterium (n = 2/7; samples G1 and H2), and Ralstonia (n = 2/7; samples F3 and G2). No significant enrichments for InterProScan protein domains, GO terms, or KEGG pathways were detected between risk groups or sexes.

Figure 8. Comparison of InterProScan protein domain abundance in annotated bacterial genes by WMS after contig assembly with metaSpades using a heatmap of the 50 most abundant features (A) and PCA plot of Aitchison distance for beta diversity comparisons (B) between H. pylori-positive versus -negative samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. C) Differential abundance analysis of InterProScan protein domains in annotated bacterial genes between H. pylori status determined using Welch’s t-test and generalized linear model (GLM) on centered log-ratio (clr) transformed abundances with a false discovery rate (FDR) correction for P-values using the Benjamini-Hochberg (BH) method. The abundance of differentially expressed features with a FDR≤0.01 for both tests is shown.

Figure 8. Comparison of InterProScan protein domain abundance in annotated bacterial genes by WMS after contig assembly with metaSpades using a heatmap of the 50 most abundant features (A) and PCA plot of Aitchison distance for beta diversity comparisons (B) between H. pylori-positive versus -negative samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. C) Differential abundance analysis of InterProScan protein domains in annotated bacterial genes between H. pylori status determined using Welch’s t-test and generalized linear model (GLM) on centered log-ratio (clr) transformed abundances with a false discovery rate (FDR) correction for P-values using the Benjamini-Hochberg (BH) method. The abundance of differentially expressed features with a FDR≤0.01 for both tests is shown.

Figure 9. Comparison of KEGG metabolism pathway abundance in annotated bacterial genes by WMS after contig assembly with metaSpades using a heatmap of the 50 most abundant features (A) and PCA plot of Aitchison distance for beta diversity comparisons (B) between H. pylori-positive versus -negative samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. C) Differential abundance analysis of KEGG pathways in annotated bacterial genes between H. pylori status determined using Welch’s t-test and generalized linear model (GLM) on centered log-ratio (clr) transformed abundances with a false discovery rate (FDR) correction for P-values using the Benjamini-Hochberg (BH) method. The abundance of differentially expressed features with a FDR≤0.05 for both tests is shown.

Figure 9. Comparison of KEGG metabolism pathway abundance in annotated bacterial genes by WMS after contig assembly with metaSpades using a heatmap of the 50 most abundant features (A) and PCA plot of Aitchison distance for beta diversity comparisons (B) between H. pylori-positive versus -negative samples. Statistical analysis for beta diversity was performed using PERMANOVA to determine significance differences (P-value) and percentage of the variance explained (R2) between the groups. C) Differential abundance analysis of KEGG pathways in annotated bacterial genes between H. pylori status determined using Welch’s t-test and generalized linear model (GLM) on centered log-ratio (clr) transformed abundances with a false discovery rate (FDR) correction for P-values using the Benjamini-Hochberg (BH) method. The abundance of differentially expressed features with a FDR≤0.05 for both tests is shown.

Detection of H. pylori virulence factors gene sequences using WMS

Strain-specific H. pylori virulence factor genes detected by WMS were further evaluated. Across all 13 H. pylori-positive samples, more than 80% of the genes in the reference H. pylori strain 26695 genome were detected by WMS. Per sample, strain-specific virulence factor profiles were apparent, including differences in cag pathogenicity-associated island (PAI) and vacuolating cytotoxin (vacA) status as well as a repertoire of outer membrane protein including those belonging to Hop and Hor families. (). While vacA sequences were detected in the majority of H. pylori-positive samples by WMS, the sequences were not long enough to identify subtypes.

Figure 10. H. pylori virulence factor genes detected in assembled contigs and mapped to H. pylori reference strain 26695. Rings (outer to inner): 1) Forward protein coding genes (green), 2) Reverse protein coding genes (purple), 3) Virulence factor genes from H. pylori reference strain 26695 (orange), and 4) histogram showing number of genes detected by WMS per H. pylori-positive sample (n = 13).

Figure 10. H. pylori virulence factor genes detected in assembled contigs and mapped to H. pylori reference strain 26695. Rings (outer to inner): 1) Forward protein coding genes (green), 2) Reverse protein coding genes (purple), 3) Virulence factor genes from H. pylori reference strain 26695 (orange), and 4) histogram showing number of genes detected by WMS per H. pylori-positive sample (n = 13).

Complete urease operons were detected in H. pylori protein annotations (). Urease activity is essential for H. pylori to neutralize stomach acid and colonize the gut. Interestingly, other urease-like genes were not detected in non-H. pylori contigs or protein annotation (see urease activity GO: 0009039 in supplemental figure 15C). This suggests that these bacteria may utilize alternative mechanisms to neutralize stomach acid and enable colonization in the gastric environment. Alternatively, increased pH of the stomach acid as a result of gastric disease progression (i.e., NAG and MAG-IM) may have facilitated increased colonization of non-H. pylori species. Gastric pH was not measured in patients to evaluate this potential association.

Discussion

Gastric cancer is the 5th most common cancer and the 4th most common cause of cancer death globally, especially in emerging countries like Colombia, South America where H. pylori infection can exceed 80%Citation20,Citation21. While H. pylori infection is the strongest risk factor associated with gastric disease, age, sex, smoking, diet, and hygiene may also modify riskCitation20. Recently, non-H. pylori microbes present in the gastric microbiome have been appreciated as important factors that may modulate gastric disease statesCitation1,Citation2,Citation22. Previous studies have shown that non-H. pylori microbes can be readily cultured from the gastric tissue samplesCitation14. Furthermore, non-H. pylori microbes residing in the gastric niche have been appreciated to interfere with urease-breath test resultsCitation23, implying sufficient colonization loads exist in the stomach to yield physiological effects on the host.

Eradication of H. pylori infection with antibiotics effectively reduces gastric disease and cancer risk. For example, INS-GAS mice, a transgenic mouse line that overexpresses human gastrin and has increased indices of gastric disease, have reduced gastric disease progression and severity when maintained under germfree conditions or when treated with antibiotics under conventional housingCitation24–26. Additionally, antibiotic treatment that failed to completely clear H. pylori in human patients still significantly reduced their risk for gastric cancerCitation27, implying that other microbes present in the gastric niche of the host may contribute to disease progression.

With the advent of next-generation sequencing technology, targeted 16S rRNA profiling has facilitated culture-free approaches to evaluate the gastric microbiome. Numerous studies have found that while H. pylori is the predominant bacterial taxa present in the stomach in infected individuals, there is a high diversity of non-H. pylori taxa also detectableCitation2,Citation22. In H. pylori-negative patients, the gastric microbiome remains highly diverse including a variety of bacteria taxa from all major phyla. Additionally, the magnitude of gastric microbiome diversity is also associated with the host states, such as increases in gastric pH due to acid suppressant drugs, antibiotic use, and during progression of disease noted in H. pylori-associated cancerCitation2,Citation22,Citation28–30. While culture and 16S rRNA sequencing have provided insights into the diversity of bacterial microbes inhabiting the stomach, these approaches fail to capture the functional diversity of the gastric microbiome and how this may impact the host. In general, it is also largely unknown if non-bacterial taxa are present in the gastric niche and contribute to normal or pathological states in the host. To this end, we utilized non-targeted WMS to characterize the composition and function of the gastric microbiome in human patients from high- and low-risk gastric cancer populations residing in Colombia, South America. In addition to capturing the bacterial diversity and strain-specific genetic signatures that included annotation of microbial protein coding gene sequences, WMS allowed us to identify eukaryota, viral, and archaea microbes present in the gastric microbiome. We did not identify any statistical differences in eukaryota, viral, or archaea populations between H. pylori status, risk group, or sex. In our study, Candida spp. were detected in all gastric biopsy tissues samples by WMS, but were not isolated by culture, even though media we used for culture will support the growth of fungi. The lack of fungal growth possibly is due in part to the polymicrobial nature of the gastric biopsies where other bacteria colonizing this niche can inhibit slower growing fungi, as well as reducing fungal viability. Further, 16S-based bacterial analysis would not detect fungal presence, given 18S sequencing is used to identify fungi in tissue samples. Future studies investigating the role of fungi in gastric cancer progression are warranted. Interestingly, Candida spp. have been recently detected in colorectal cancer tumor DNA from human patients and associated with metastatic progressionCitation31.

Our current WMS analysis confirms previous culture-based and 16S rRNA microbiome studies that non-H. pylori bacteria are present in stomach tissues of Colombian patients from high- and low-gastric-cancer-risk cohortsCitation13. While H. pylori is the predominant taxa in the stomach when present, significant differences exist the gastric microbial population between gastric cancer risk groups and sex, even when H. pylori is excluded from analyses (see supplemental figures S3 and S4). This finding reinforces the significance non-Helicobacter microbes may have in modulating gastric cancer risk and progression. Additional studies are needed to understand how the gastric microbiota contributes to cancer risk as well as interplays with other potential multifactorial attributes between Túquerres and Tumaco populations, for example, diet and sex, parasitic infections, and genetic characterization of H. pylori.

Several soil-associated bacterial taxa were abundant in the gastric microbiome. Bacillus, Actinomyces, and Arthrobacter spp. were detected by both WMS and culture in our study. Perturbations in presences of these taxa have been associated with gastric cancer riskCitation32–35. Interestingly, the abundance of Keratinibaculum spp., thermophilic anaerobes originally isolated from soilCitation36, was significantly higher in males versus females. Future studies will be needed to validate the presence and enrichment of Keratinibaculum spp. in the gastric niche as well as its association with sex and/or gastric disease progression, especially since males had significantly higher pathology scores compared to females. The significant differences in the gastric microbial beta diversity as well as enrichment of Keratinibaculum spp. between males versus females is particularly interesting considering males have a greater than 2-fold higher incidence and death rate of gastric cancer compared to femalesCitation20. Keratinibaculum spp. have not been previously described in the gastric microbiome. It is plausible these soil-associated microbes were introduced from consumption of food or water sources. The role of these taxa in gastric carcinogenesis requires further study.

Proteobacteria and Firmicutes phyla predominated in the gastric microbiome. Klebsiella spp., belonging to the Proteobacteria phylum, were abundant in the gastric microbiome and increased levels have been previously associated with gastric cancerCitation37. Several taxa from the Firmicutes phylum were enriched in the gastric microbiome and have been associated with gastric or intestinal cancer, including Clostridium Citation38–41, Lactococcus Citation42, Staphylococcus Citation14, and Streptococcus Citation14 spp.

Staphylococcus and Streptococcus species are considered commensals of the skin and oral sites. However, Staphylococcus epidermidis and Streptococcus salivarius isolated from stomach biopsy samples from the same Colombian cohort of the current study were capable of gastric colonization as well as modulated H. pylori-associated gastric pathology and host immune responses in a germfree INS-GAS mouse model of gastric cancerCitation14. Staphylococcus epidermidis was cultured from both high- and low-risk gastric cancer patients, but Staphylococcus epidermidis was more often present in the stomach biopsies of low-risk gastric cancer patientsCitation14. Both strains exhibited urease activity and were able to maintain persistent colonization up to 5 months in the germfree mouse gastric niche alone or during co-infection with H. pylori. Neither strain affected H. pylori gastric colonization, but Streptococcus salivarius caused significantly higher gastric pathology than H. pylori only or H. pylori with Staphylococcus epidermidis. In a different study from our lab, we showed germfree INS-GAS colonized with a restricted intestinal microbiota that includes Clostridium spp. exacerbated gastric pathology and carcinogenesis due to H. pylori infectionCitation43. This study emphasizes how non-H. pylori stomach microflora play a role in the pathology of H. pylori-induced gastric cancer. In particular, this study reinforces previous investigations observing that oral commensal and opportunistic pathogenic microbes may have the potential to colonize the stomach and act as risk factors alongside H. pylori to influence chronic inflammation and carcinogenesisCitation44. While future research is required to ascertain if taxa detected by next-generation sequencing approaches represent actively colonizing versus swallowed/transient microbes, we hypothesize some of these taxa have the potential to adhere to the mucosal surface and sustain active colonization in the gastric niche, as suggested by the previously published studies described above.

While only~0.03–0.2% of the total WMS reads were mapped to known bacterial sequences, we determined that cultured bacteria taxa from the gastric biopsies were more often detected by WMS than 16S rRNA analysis, suggesting that WMS may have stronger taxonomic resolution. Interestingly, Atopobium was the only genera detected by culture but not 16S rRNA or WMS profiling. We speculate that Atopobium strains may have low genetic abundance in the gastric biopsy samples that prevented detection by 16S rRNA or WMS profiling methods, but were enriched for growth by the culturing conditions/media. Sample preparation methods that deplete host DNA in human colonic biopsy samples before WMS have been shown to increase the bacterial-to-human DNA ratio without distorting microbiome profiles, thereby increasing sequencing coverage of the microbial genomes in tissues samplesCitation19. Application of similar bacterial DNA enrichment methods for gastric biopsy tissues may improve detection of underrepresented species as well as the taxonomic and functional analysis by WMS. Follow-up studies in larger cohorts of patients will be needed to validate that WMS provides superior accuracy and depth of microbiome analysis compared to traditional 16S rRNA approaches.

In our study, we found a poor correlation between the relative abundance at the genus level detected for 16S rRNA profiling versus WMS despite using DNA from the same extraction. This finding suggests these approaches may have different sensitivities or biases for taxonomic detection since 16S rRNA profiling relies on sequencing PCR amplicons while WMS targets bacterial genomic fragments. It has been well described in the literature that 16S rRNA profiling is susceptible to technical artifacts due to contamination as well as PCR amplification biasCitation17,Citation18. Conversely, WMS may be less prone to these caveats. Furthermore, the 16S rRNA gene is also susceptible to misidentification since some species (and genera like Escherichia and Shigella) can have nearly identical 16S rRNA sequences, but are considered different species based on the gold standard for microbial speciation, whole-genome sequence analysis (e.g., core gene phylogeny, average nucleotide identity, digital DNA-DNA hybridization)Citation45,Citation46. WMS may also identify genera differently since this method can use any genomic sequence to identify a taxon, instead of a single gene (i.e., 16S rRNA sequence).

A potential limitation of our 16S rRNA profiling is a negative extraction control or negative PCR control to monitor for potential background noise was not included. However, since 16S rRNA and WMS profiling used the same DNA extracts, overlapping contamination would have been expected using both sequencing approaches which is not supported by the poor correlation in OTU abundance between these methods. Our culture results suggest the predominant isolates cultured from these gastric biopsy samples were also detected by 16S rRNA and WMS. Additionally, the OTUs we have detected by both 16S rRNA and WMS have been described in previous studies characterizing the gastric microbiota from these Colombian cohorts as well as other populationsCitation1,Citation2,Citation13.

WMS enabled identification of statistically significant differences in functional and metabolic genes harbored by the gastric microbiome between H. pylori-positive versus -negative patients. Interestingly, carbohydrate and amino acid metabolism pathways were enriched in H. pylori-negative patients. Comparative genome analysis and metabolic experiments have shown H. pylori lacks the traditional glycolysis pathway used by bacteria to metabolize glucose into pyruvate and instead relies on alternative pathways to process carbohydratesCitation47. Additionally, H. pylori has a limited gene content for de novo biosynthesis of the amino acids histidine, leucine, methionine, phenylalanine, and valine. As demonstrated experimentally, H. pylori instead requires media supplemented with these amino acids to grow in the absence of serum in vitro; presumably serum acts as transporters for these nutrientsCitation47. Enrichment of carbohydrate and amino acid metabolism genes in H. pylori-negative samples were mainly represented by Bacillus, Corynebacterium, Cutibacterium, Neisseria, and Ralstonia spp. The biological significance of enrichment in carbohydrate and amino acid metabolism genes in the gastric microbiota requires further investigation, especially how it may relate to the pathogenesis of gastric disease and cancers. Continued optimization of WMS methods for gastric tissues is needed to increase sequencing coverage for non-Helicobacter species occupying the gastric microbiota as well as further elucidate their genetic features.

As expected, H. pylori represented the predominant taxa in H. pylori-positive patients and significant enrichment for H. pylori-specific genes were detected according to InterProScan domains and GO terms. These included genes for DNA methylation for epigenetic modificationCitation48 (IPR002941, GO: 0008170, GO 00063006), outer membrane proteinsCitation49 (IPR002718, IPR040838), and urease activity (GO: 0009039). In particular, WMS was able to detect strain-specific H. pylori virulence factors genes that facilitate host colonization (urease, outer membrane proteins), immune evasion (outer membrane proteins), gastric epithelial injury (vacuolating cytotoxin, cag PAI), and promote oncogenesis (cag PAI)Citation50.

Previously, our group performed whole genome sequencing on H. pylori isolates cultured from gastric biopsy tissues collected from the same high- and low-risk Colombian populationsCitation15, which included five samples also analyzed by WMS in the current study (samples B4, C2, C5, D3, and D4). These five H. pylori isolates were cag-positive and vacA-positive and harbored a variety of outer membrane proteins. WMS was able to detect vacA (3/5 samples), cag PAI (4/5 samples), and outer membrane proteins (4/5 samples) (data not shown). The gold standard for detecting and characterizing disease-associated H. pylori strains has been culture to isolate the organism from the stomach and enable subsequent biochemical, molecular and whole genome sequence characterization. However, given the fastidious nature of this species, this has been a challenging bottleneck. Additionally, culture and passage of H. pylori may promote genotypic drifts given the high genomic plasticity of this species, which may confound associations between H. pylori genetics and host disease progressionCitation51. Therefore, WMS may augment traditional culture and genome sequencing approaches to identify, monitor, and characterize risk factors attributed to H. pylori colonization in patients, including detection of numerous strain-specific genes including disease-promoting virulence factors like vacA and cag PAICitation52. While contig sizes and protein gene annotations from non-H. pylori taxa were too low to appreciably characterize their strain-specific features and describe the microbiome beyond the genus level, continued optimization of WMS methods for gastric tissues, such as depletion of host DNA, may improve microbial genome sequencing coverage and therefore contig assembly for H. pylori and other taxa.

While numerous 16S rRNA profiling studies have been published, to our knowledge the gastric microbiome has been characterized using WMS in only a single report in the literature by Hu et al.Citation53 In this study, the authors evaluated gastric wash samples from 6 patients with advanced gastric adenocarcinoma and 5 patients with superficial gastritis and found significant differences in the composition and functional pathways related to bacterial OTUs. The authors did not report characterization of non-bacterial taxa. Unlike the study by Hu et al., we surveyed the gastric microbiome using gastric biopsy samples, which represented mucosal-associated microbes. Host-microbe interactions at the mucosal surface in the stomach and other sites of the gastrointestinal tract are hypothesized to be more influential for host homeostasis and disease progression since microbes are in closer proximity to the epithelial barrier and immune sites compared to organisms primarily residing in the luminal contentCitation54. In the study by Hu et al., the authors did not perform 16S rRNA or culture profiling of the gastric microbiota for comparison with WMS.

In a different published report by Thorell et al., metatranscriptomic RNA sequencing of stomach biopsy tissues from Nicaragua patients with different H. pylori infection statuses and premalignant tissue changes was performed to evaluate the composition of the transcriptionally active microbial community in the gastric microbiotaCitation55. Similar to our findings, these authors determined that RNA transcripts from H. pylori predominated in H. pylori-positive samples, while fungi, Bacteroidetes, Firmicutes, and Actinobacteria transcripts were also detected in biopsy tissues as well. Additionally, the authors noted highly expressed H. pylori nickel transport genes which parallels our findings of genes enriched for nickel cation binding (GO:0016151) in H. pylori-positive samples. Significant differences in stomach microbiota at different stages of gastric disease progression were not noted in this study. While these authors performed 16S rRNA profiling to support taxa identified by metatranscriptomic RNA sequencing, the authors did not statistically analyze the agreement of taxa identified between sequencing methods.

In conclusion, our study demonstrates that WMS is a feasible approach to characterize the gastric microbiome. By continuing to study the gastric microbiome with WMS, novel insights into the structure, function, and interactions of H. pylori with other microorganisms and host disease risk may be elucidated. Future studies comparing WMS versus culture and 16S rRNA profiling in larger sample populations are warranted to optimize WMS as a novel approach to detect and characterize non-H. pylori taxa present in the stomach. By integrating next-generation sequencing approaches with traditional culture techniques, future studies will have the potential to identify specific microbes associated with augmented or suppressed risk for stomach disease in human patients.

Methods

Study population, samples, and histopathology

As previously cited, subjects between 40 and 60 years of age with dyspeptic symptoms that warranted upper gastrointestinal tract endoscopy were recruited in Tumaco (LGCR) and Túquerres (HGCR) in 2010Citation14,Citation15. Subjects that had received proton pump inhibitors, H2-receptor antagonists, or antimicrobials during the 30-day period previous to the endoscopic procedure were excluded from this study. Other exclusion criteria were major diseases or previous gastrectomy. Participation was voluntary and informed consent was obtained from all participants. The Ethics Committees of the participating hospitals in Nariño and the Universidad del Valle in Cali, Colombia, and the Institutional Review Board of Vanderbilt University approved all study protocols, and all experiments were performed in accordance with the relevant guidelines and regulations. A total of 163 participants were recruited (81 from the HGCR and 82 from the LGCR) and a sample population of 20 patients (10 from HGCR, 10 from LGCR) consisting of 10 males and 10 females was selected for this study. Four biopsy samples (2 antrum, 1 incisura, and 1 corpus) were used for histopathology, one frozen antral biopsy was used for H. pylori culture, and another frozen antral biopsy for both DNA extraction and culture. By histologic diagnosis, 17 patients had non-atrophic gastritis (NAG) and 3 had multifocal atrophic gastritis with intestinal metaplasia (MAG-IM) (, supplemental table S19). Thirteen of the patients were H. pylori-positive by a modified Steiner stain and antral culture ().

16S rRNA sequencing and WMS

DNA was extracted from antral gastric biopsy samples using the DNeasy PowerLyzer PowerSoil Kit (Qiagen, Germantown, MD) following the manufacture’s protocol. DNA was stored at −20°C until use. 16S rRNA library preparations and sequencing were performed as previously describedCitation56. WMS libraries were prepared from>5 ug of DNA per sample using NexteraFlex and sequenced by 2 × 250 bp paired reads on an Illumina Novaseq SP500.

Bioinformatic analysis

All scripts for the bioinformatic workflow were performed using default parameters unless stated otherwise and have also been deposited in GitHub (https://github.com/TonyMannion/WMS_Gastric_Biopsies). 16S rRNA and WMS reads were decontaminated for low-quality base pairs and adapter sequence using BBDuk version 38.90 (sourceforge.net/projects/bbmap/) before taxonomic classification. Decontaminated 16S rRNA reads were analyzed using Kraken2Citation57 against the SILVA 138 database, a highly curated and comprehensive database of 16S rRNA gene sequences (available at https://benlangmead.github.io/aws-indexes/k2). Decontaminated WMS reads analyzed by Kraken2Citation58 followed by BrackenCitation59 for taxonomic classification and abundance against more than 34,000 RefSeq genomes from archaea, bacteria, fungi, protozoa, eukaryota, human, and viral species (PlusPF: standard plus protozoa & fungi and EuPathDB46: eukaryotic pathogen genomes with contaminants removed; available at https://benlangmead.github.io/aws-indexes/k2). This database includes more than 60 different strains of H. pylori. Bacterial reads were extracted using Kraken2 from decontaminated WMS reads and assembled into contigs using metaSPAdesCitation60. BBMap version 38.90 (sourceforge.net/projects/bbmap/) was used to estimate contig coverage by mapping bacterial reads to assembled contig sequences, and no contigs were excluded based on coverage prior to gene annotation with RAST hosted by PATRICCitation61. DIAMONDCitation62 analysis against the nr database was used to predict taxonomy for protein gene annotations. InterProScan version 5.52–86.0Citation63 and KAASCitation64 (program: GHOSTX; method: BBG; GENES data set: hsa, dme, ath, sce, pfa, eco, sty, hin, pae, nme, hpy, rpr, mlo, bsu, sau, lla, spn, cac, mge, mtu, ctr, bbu, syn, aae, mja, afu, pho, ape) were used to analyze gene annotations for protein domains and KEGG pathways, respectively. GO terms were included in the outputs from InterProScan domain analysis. Protein gene annotations with ≥90% percent identity to the Helicobacter pylori 26695 genome (Genbank: AE000511) based DIAMOND analysis were mapped and visualized on the circular chromosome using PATRIC.

Bacterial culture methods for gastric biopsies from patients

Antral biopsies were frozen at −80°C in thioglycolate containing 20% glycerol. The biopsies were thawed in an anaerobic atmosphere (10% CO2, 10% H2, 80% N2), and were homogenized in Brain Heart Infusion (BHI) with 20% glycerol with tissue grinders. The homogenate was divided into aliquots to isolate bacteria under diverse culture conditions. For aerobic culture, the homogenates were plated onto chocolate agar, blood agar, MacConkey agar, and Brucella broth medium containing 10% FCS. The plates were incubated at 37°C in 5% CO2 for 24–48 hours. For anaerobic culture, the homogenates were plated onto pre-reduced Brucella blood agar plates (BBL) and inoculated into thioglycolate broth. The cultures were incubated at 37°C in an anaerobic chamber (Coy Lab Products) with mixed gas (10% CO2, 10% H2, 80% N2) for 48 hours. For microaerobic culture to detect the growth of H. pylori, the homogenates were plated onto H. pylori-selective plates and Brucella blood agar plates after passing through 0.65 µm syringe filter. The plates were placed into a vented jar filled with mixed gas (10% CO2, 10% H2, 80% N2) and incubated at 37°C for up to 3 weeks. The plates were checked every 2–3 days for growth. All bacterial strains isolated from the different culture conditions were identified by 16S rRNA sequencing.

Statistical analysis

The Phyloseq, Vegan, and Microbiome packages in R were used to plot and statistically analyze 16S rRNA and shotgun metagenomic data, as described previouslyCitation56. Briefly, alpha diversity was analyzed by the Observed and Chao1 metrics to describe the species richness (i.e., “how many microbes in a sample?”) and the Shannon and Simpson metrics to describe species evenness (i.e., “how are microbes balanced to each other in a sample?”). For beta diversity analyses, the Aitchison distance (i.e., the Euclian distance between samples after centered log-ratio (clr) transformation of abundances)Citation65 was plotted on PCA plots and permutational ANOVA (PERMANOVA) using the adonis function in Vegan. To identify differentially abundant features, the Welch’s t-test and generalized linear model (GLM) on clr transformed abundances with a false discovery rate (FDR) correction for P-values using the Benjamini-Hochberg (BH) method. A P-value or FDR≤0.05 was considered statistically significant. All scripts for these analyses were performed using default parameters unless stated otherwise and have also been deposited in GitHub (https://github.com/TonyMannion/WMS_Gastric_Biopsies).

Supplemental material

Disclosure statement

No potential conflict of interest was reported by the authors.

Data availability statement

The authors confirm that the data supporting the findings of this study are available within

the article and the raw sequencing results can be accessed with the accession number

PRJNA861967. https://www.ncbi.nlm.nih.gov/bioproject/PRJNA861967

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2023.2186677.

Additional information

Funding

This work was supported by the NIH under Grant R01DK58587, R01CA77955, P01CA116087, P30DK058404 to RP; R35CA210088, P30ES002109, P01CA28842 to JGF.

References

  • Rajilic-Stojanovic M, Figueiredo C, Smet A, Hansen R, Kupcinskas J, Rokkas T, Andersen L, Machado JC, Ianiro G, Gasbarrini A, et al. 2020. Systematic review: gastric microbiota in health and disease. Aliment Pharmacol Ther. 51(6):582–21. doi:10.1111/apt.15650.
  • Sheh A, Fox JG. 2013. The role of the gastrointestinal microbiome in Helicobacter pylori pathogenesis. Gut Microbes. 4:505–531. doi:10.4161/gmic.26205.
  • Kwon SK, Park JC, Kim KH, Yoon J, Cho Y, Lee B, Lee JJ, Jeong H, Oh Y, Kim SH, et al. 2022. Human gastric microbiota transplantation recapitulates premalignant lesions in germ-free mice. Gut. 71(7):1266–1276. doi:10.1136/gutjnl-2021-324489.
  • Bharti R, Grimm DG. 2021. Current challenges and best-practice protocols for microbiome analysis. Brief Bioinform. 22(1):178–193. doi:10.1093/bib/bbz155.
  • Wensel CR, Pluznick JL, Salzberg SL, Sears CL. 2022. Next-generation sequencing: insights to advance clinical investigations of the microbiome. J Clin Invest. 132(7). doi:10.1172/JCI154944.
  • Vogtmann E, Hua X, Zeller G, Sunagawa S, Voigt AY, Hercog R, Goedert JJ, Shi J, Bork P, Sinha R. 2016. Colorectal Cancer and the Human Gut Microbiome: Reproducibility with Whole-Genome Shotgun Sequencing. PLoS One. 11(5):e0155362. doi:10.1371/journal.pone.0155362.
  • Jovel J, Patterson J, Wang W, Hotte N, O’Keefe S, Mitchel T, Perry T, Kao D, Mason AL, Madsen KL, Wong GK.2016. Characterization of the gut microbiome using 16S or Shotgun Metagenomics. Front Microbiol, 7:459. doi:10.3389/fmicb.2016.00459.
  • Mas-Lloret J, Obón-Santacana M, Ibáñez-Sanz G, Guinó E, Pato ML, Rodriguez-Moranta F, Mata A, García-Rodríguez A, Moreno V, Pimenoff VN. 2020. Gut microbiome diversity detected by high-coverage 16S and shotgun sequencing of paired stool and colon sample. Sci Data. 7(1):92. doi:10.1038/s41597-020-0427-5.
  • Lloyd-Price J, et al. 2019. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 569(7758):655–662. doi:10.1038/s41586-019-1237-9.
  • Liu Y, et al. Exploring Gut Microbiota in Patients with Colorectal Disease Based on 16S rRNA Gene Amplicon and Shallow Metagenomic Sequencing. Front Mol Biosci, 2021;8:703638. doi:10.3389/fmolb.2021.703638.
  • Durazzi F, Sala C, Castellani G, Manfreda G, Remondini D, De Cesare A. 2021. Comparison between 16S rRNA and shotgun sequencing data for the taxonomic characterization of the gut microbiota. Sci Rep. 11(1):3030. doi:10.1038/s41598-021-82726-y.
  • Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB, Bousvaros A. 2012. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13(9):R79. doi:10.1186/gb-2012-13-9-r79.
  • Yang I, et al. 2016. Different gastric microbiota compositions in two human populations with high and low gastric cancer risk in Colombia. Sci Rep. 6(1):18594. doi:10.1038/srep18594.
  • Shen Z, et al. 2022. Gastric Non-Helicobacter pylori Urease-Positive Staphylococcus epidermidis and Streptococcus salivarius isolated from humans have contrasting effects on H. pylori-Associated Gastric pathology and Host Immune Responses in a Murine model of Gastric cancer. mSphere. 7(1):e0077221. doi:10.1128/msphere.00772-21.
  • Mannion A, Dzink-Fox J, Shen Z, Piazuelo MB, Wilson KT, Correa P, Peek RM, Camargo MC, Fox JG. 2021. Helicobacter pylori antimicrobial resistance andgene variants in High- and Low-Gastric-Cancer-Risk populations. J Clin Microbiol. 59(5). doi:10.1128/JCM.03203-20.
  • Gorkiewicz G, Moschen A. 2018. Gut microbiome: a new player in gastrointestinal disease. Virchows Arch. 472(1):159–172. doi:10.1007/s00428-017-2277-x.
  • Eisenhofer R, Minich JJ, Marotz C, Cooper A, Knight R, Weyrich LS. 2019. Contamination in low microbial biomass microbiome studies: issues and recommendations. Trends Microbiol. 27(2):105–117. doi:10.1016/j.tim.2018.11.003.
  • Brooks JP, et al. 2015. The truth about metagenomics: quantifying and counteracting bias in 16S rRNA studies. BMC Microbiol. 15(1):66. doi:10.1186/s12866-015-0351-6.
  • Bruggeling CE, Garza DR, Achouiti S, Mes W, Dutilh BE, Boleij A. 2021. Optimized bacterial DNA isolation method for microbiome analysis of human tissues. Microbiologyopen. 10(3):e1191. doi:10.1002/mbo3.1191.
  • Etemadi A, Safiri S, Sepanlou SG, Ikuta K, Bisignano C, Shakeri R, Amani M, Fitzmaurice C, Nixon M, Abbasi N, et al. 2020. The global, regional, and national burden of stomach cancer in 195 countries, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol Hepatol. 5(1):42–54. doi:10.1016/S2468-1253(19)30328-0.
  • Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. 2021. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 71(3):209–249. doi:10.3322/caac.21660.
  • Noto JM, Peek RMJ, Leong JM. 2017. The gastric microbiome, its interaction with Helicobacter pylori, and its potential role in the progression to stomach cancer. PLoS Pathog. 13(10):e1006573. doi:10.1371/journal.ppat.1006573.
  • Osaki T, Mabe K, Hanawa T, Kamiya S. 2008. Urease-positive bacteria in the stomach induce a false-positive reaction in a urea breath test for diagnosis of Helicobacter pylori infection. J Med Microbiol. 57(7):814–819. doi:10.1099/jmm.0.47768-0.
  • Lee C-W,Rickman B, Rogers AB, Ge Z, Wang TC, Fox JG. 2008. Helicobacter pylori eradication prevents progression of gastric cancer in hypergastrinemic INS-GAS mice. Cancer Res. 68(9):3540–3548. doi:10.1158/0008-5472.CAN-07-6786.
  • Lofgren JL, Whary MT, Ge Z, Muthupalani S, Taylor NS, Mobley M, Potter A, Varro A, Eibach D, Suerbaum S, et al. 2011. Lack of Commensal Flora in Helicobacter pylori–infected INS-GAS mice reduces Gastritis and delays Intraepithelial Neoplasia. Gastroenterology. 140(1):210–220. doi:10.1053/j.gastro.2010.09.048.
  • Lee CW, Rickman B, Rogers AB, Muthupalani S, Takaishi S, Yang P, Wang TC, Fox JG. 2009. Combination of sulindac and antimicrobial eradication of Helicobacter pylori prevents progression of gastric cancer in hypergastrinemic INS-GAS mice. Cancer Res. 69(20):8166–8174. doi:10.1158/0008-5472.CAN-08-3856.
  • Ma JL, Zhang L, Brown LM, Li JY, Shen L, Pan KF, Liu WD, Hu Y, Han ZX, Crystal-Mansour S, et al. 2012. Fifteen-year effects of Helicobacter pylori, garlic, and vitamin treatments on gastric cancer incidence and mortality. J Natl Cancer Inst. 104(6):488–492. doi:10.1093/jnci/djs003.
  • von Rosenvinge EC, Song Y, White JR, Maddox C, Blanchard T, Fricke WF. 2013. Immune status, antibiotic medication and pH are associated with changes in the stomach fluid microbiota. Isme J. 7(7):1354–1366. doi:10.1038/ismej.2013.33.
  • Freedberg DE, Lebwohl B, Abrams JA. 2014. The impact of proton pump inhibitors on the human gastrointestinal microbiome. Clin Lab Med. 34(4):771–785. doi:10.1016/j.cll.2014.08.008.
  • Alarcón T, Llorca L, Perez-Perez G. 2017. Impact of the Microbiota and Gastric disease development by Helicobacter pylori BT - Molecular Pathogenesis and signal Transduction by Helicobacter pylori. editorsTegtmeyer N Backert S. pp. 253–275. Springer International Publishing. 10.1007/978-3-319-50520-6_11.
  • Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X, Iliev ID. 2022. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 185(20):3807–3822.e12. doi:10.1016/j.cell.2022.09.015.
  • Zhang Y, Shen J, Shi X, Du Y, Niu Y, Jin G, Wang Z, Lyu J. 2021. Gut microbiome analysis as a predictive marker for the gastric cancer patients. Appl Microbiol Biotechnol. 105(2):803–814. doi:10.1007/s00253-020-11043-7.
  • Liatsos C, Papaefthymiou A, Kyriakos N, Galanopoulos M, Doulberis M, Giakoumis M, Petridou E, Mavrogiannis C, Rokkas T, Kountouras J. 2022. Helicobacter pylori, gastric microbiota and gastric cancer relationship: unrolling the tangle. World J Gastrointest Oncol. 14(5):959–972. doi:10.4251/wjgo.v14.i5.959.
  • Dong Z, Chen B, Pan H, Wang D, Liu M, Yang Y, Zou M, Yang J, Xiao K, Zhao R, Zheng X, Zhang L, Zhang Y. 2019. Detection of microbial 16S rRNA Gene in the Serum of patients with gastric Cancer. Front Oncol. 9. doi:10.3389/fonc.2019.00608.
  • Liu X, Shao L, Liu X, Ji F, Mei Y, Cheng Y, Liu F, Yan C, Li L, Ling Z. Alterations of gastric mucosal microbiota across different stomach microhabitats in a cohort of 276 patients with gastric cancer. EBioMedicine, 2019;40:336–348. doi:10.1016/j.ebiom.2018.12.034.
  • Huang Y, Sun Y, Ma S, Chen L, Zhang H, Deng Y. 2013. Isolation and characterization of Keratinibaculum paraultunense gen. nov., sp. nov., a novel thermophilic, anaerobic bacterium with keratinolytic activity. FEMS Microbiol Lett. 345(1):56–63. doi:10.1111/1574-6968.12184.
  • Pappas-Gogos G, Tepelenis K, Fousekis F, Katsanos K, Pitiakoudis M, Vlachos K. 2022. The implication of Gastric Microbiome in the treatment of gastric cancer. Cancers (Basel). 14(8):2039. doi:10.3390/cancers14082039.
  • Hsieh YY, Tung SY, Pan HY, Yen CW, Xu HW, Lin YJ, Deng YF, Hsu WT, Wu CS, Li C. 2018. Increased abundance of Clostridium and Fusobacterium in gastric microbiota of patients with gastric cancer in Taiwan. Sci Rep. 8(1):158. doi:10.1038/s41598-017-18596-0.
  • Smet A, Kupcinskas J, Link A, Hold GL, Bornschein J. 2022. THe role of microbiota in gastrointestinal cancer and cancer treatment: chance or curse? Cell Mol Gastroenterol Hepatol. 13(3):857–874. doi:10.1016/j.jcmgh.2021.08.013.
  • Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, Figueiredo C. 2018. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 67(2):226 LP–236. doi:10.1136/gutjnl-2017-314205.
  • Yang J, Zhou X, Liu X, Ling Z, Ji F. 2021;. Role of the Gastric Microbiome in Gastric Cancer: From Carcinogenesis to Treatment. Front Microbiol. 12. 10.3389/fmicb.2021.641322.
  • Kapsetaki SE, Marquez Alcaraz G, Maley CC, Whisner CM, Aktipis AD. 2022. Microbes, and cancer across the tree of life: a Systematic review. Curr Nutr Rep. 11(3):508–525. doi:10.1007/s13668-022-00420-5.
  • Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, Ge Z, Wang TC, Fox JG. 2014. Gastric colonisation with a restricted commensal microbiota replicates the promotion of neoplastic lesions by diverse intestinal microbiota in the Helicobacter pylori INS-GAS mouse model of gastric carcinogenesis. Gut. 63(1):54–63. doi:10.1136/gutjnl-2013-305178.
  • Bakhti SZ, Latifi-Navid S. 2021. Oral microbiota and Helicobacter pylori in gastric carcinogenesis: what do we know and where next? BMC Microbiol. 21(1):71. doi:10.1186/s12866-021-02130-4.
  • Land M, Hauser L, Jun SR, Nookaew I, Leuze MR, Ahn TH, Karpinets T, Lund O, Kora G, Wassenaar T, Poudel S, Ussery DW. 2015. Insights from 20 years of bacterial genome sequencing. Funct Integr Genomics. 15(2):141–161. doi:10.1007/s10142-015-0433-4.
  • Clarridge JE. 2004. Impact of 16S rRNA gene sequence analysis for identification of bacteria on clinical microbiology and infectious diseases. Clin Microbiol Rev. 3rd. 17(4): 840–862. table of contents. 10.1128/CMR.17.4.840-862.2004.
  • Mannion A, Shen Z, Fox JG. 2018. Comparative genomics analysis to differentiate metabolic and virulence gene potential in gastric versus enterohepatic Helicobacter species. BMC Genomics. 19:830. doi:10.1186/s12864-018-5171-2.
  • Krebes J, Morgan RD, Bunk B, Spröer C, Luong K, Parusel R, Anton BP, König C, Josenhans C, Overmann J, et al. 2014. The complex methylome of the human gastric pathogen Helicobacter pylori. Nucleic Acids Res. 42(4):2415–2432. doi:10.1093/nar/gkt1201.
  • Oleastro M, Ménard A. 2013. The role of Helicobacter pylori outer membrane proteins in adherence and Pathogenesis. Biology (Basel). 2:1110–1134. doi:10.3390/biology2031110.
  • Roesler BM, Rabelo-Gonçalves EMA, Zeitune JMR. 2014. Virulence factors of Helicobacter pylori: a review. Clin Med Insights Gastroenterol. 7:9–17. doi:10.4137/CGast.S13760.
  • Ailloud F, Didelot X, Woltemate S, Pfaffinger G, Overmann J, Bader RC, Schulz C, Malfertheiner P, Suerbaum S. 2019. Within-host evolution of Helicobacter pylori shaped by niche-specific adaptation, intragastric migrations and selective sweeps. Nat Commun. 10(1):2273. doi:10.1038/s41467-019-10050-1.
  • Cover TL. 2016. Helicobacter pylori diversity and Gastric cancer risk. MBio. 7(1):e01869. doi:10.1128/mBio.01869-15.
  • Hu Y-L, Pang W, Huang Y, Zhang Y, Zhang C-J. 2018;. The Gastric Microbiome is Perturbed in advanced Gastric Adenocarcinoma identified through Shotgun Metagenomics. Front Cell Infect Microbiol. 8. 10.3389/fcimb.2018.00433.
  • Maynard CL, Elson CO, Hatton RD, Weaver CT. 2012. Reciprocal interactions of the intestinal microbiota and immune system. Nature. 489(7415):231–241. doi:10.1038/nature11551.
  • Thorell K, Bengtsson-Palme J, Liu OHF, Palacios Gonzales RV, Nookaew I, Rabeneck L, Paszat L, Graham DY, Nielsen J, Lundin SB, et al. 2017. In Vivo Analysis of the Viable Microbiota and Helicobacter pylori Transcriptome in Gastric Infection and Early Stages of Carcinogenesis. Infect Immun. 85(10). doi:10.1128/IAI.00031-17.
  • Sheh A, Artim SC, Burns MA, Molina-Mora JA, Lee MA, Dzink-Fox J, Muthupalani S, Fox JG. 2022. Analysis of gut microbiome profiles in common marmosets (Callithrix jacchus) in health and intestinal disease. Sci Rep. 12(1):4430. doi:10.1038/s41598-022-08255-4.
  • Lu J, Salzberg SL. 2020. Ultrafast and accurate 16S rRNA microbial community analysis using Kraken 2. Microbiome. 8(1):124. doi:10.1186/s40168-020-00900-2.
  • Wood DE, Lu J, Langmead B. 2019. Improved metagenomic analysis with Kraken 2. Genome Biol. 20(1):257. doi:10.1186/s13059-019-1891-0.
  • Lu J, Breitwieser FP, Thielen P, Salzberg SL. Bracken: estimating species abundance in metagenomics data. PeerJ Computer Science. 2017;3:e104 https://doi.org/10.7717/peerj-cs.104
  • Nurk S, Meleshko D, Korobeynikov A, Pevzner PA. 2017. metaSpades: a new versatile metagenomic assembler. Genome Res. 27(5):824–834. doi:10.1101/gr.213959.116.
  • Davis JJ, Wattam AR, Aziz RK, Brettin T, Butler R, Butler RM, Chlenski P, Conrad N, Dickerman A, Dietrich EM, et al. The PATRIC Bioinformatics Resource Center: expanding data and analysis capabilities. Nucleic Acids Res. 2020;48(D1):D606–D612. doi:10.1093/nar/gkz943.
  • Buchfink B, Xie C, Huson DH. 2015. Fast and sensitive protein alignment using DIAMOND. Nat Methods. 12(1):59–60. doi:10.1038/nmeth.3176.
  • Jones P, Binns D, Chang HY, Fraser M, Li W, McAnulla C, McWilliam H, Maslen J, Mitchell A, Nuka G, et al. 2014. InterProScan 5: genome-scale protein function classification. Bioinformatics. 30(9):1236–1240. doi:10.1093/bioinformatics/btu031.
  • Moriya Y, Itoh M, Okuda S, Yoshizawa AC, Kanehisa M. 2007. KAAS: an automatic genome annotation and pathway reconstruction server. Nucleic Acids Res. 35(Web Server):W182–5. doi:10.1093/nar/gkm321.
  • Gloor GB, Macklaim JM, Pawlowsky-Glahn V, Egozcue JJ. 2017. Microbiome datasets are compositional: anD this is not optional. Front Microbiol. 8:2224. doi:10.3389/fmicb.2017.02224.