Introduction

Industrialization is associated with reduced diversity of the microbiome in the human population [1], which could influence a range of physiological processes including nutrition, metabolism, immunity, neurochemistry, and drug metabolism [2]. Traditional indigenous populations have substantially greater microbial diversity than individuals living in industrialized societies. Nonetheless, our current knowledge of the human gut microbiome [3] is overrepresented by data available from industrialized countries and does not fully address the undersampling of indigenous populations.

Throughout evolution, helminths have coexisted with the gut microbiota in their mutual host [4], and the reduced prevalence of helminth infections from industrialized societies could contribute to the “hygiene hypothesis” [5]. While the effects of helminth colonization on the human gut microbiota have been studied, the results reported have been inconsistent. Some studies found that helminth colonization changes gut microbial diversity and composition and/or a shift in abundance of certain bacterial taxa [6,7,8,9,10,11,12], while others showed no apparent changes in gut microbial profiles [13, 14]. These divergent conclusions could be attributed to different confounders from different geographical locations (e.g., Malaysia [6, 12], Indonesia [9], Liberia [9], Tanzania [11], Western Kenya [10], and Ecuador [13]); different prevalence of helminth species (e.g., Trichuris sp. [13], hookworm [14], Ascaris sp. [10], Strongyloides sp. [8], and Schistosoma spp. [15]), as well as different approaches taken (natural or experimental infection, types of sequencing method, and analysis approaches). Additionally, the direct impact of anthelmintic treatment on the gut microbiome is unclear. While some studies found differences following deworming treatment [10, 12], others have found no impact of treatment on gut microbiota profiles [13]. Other studies that examined anthelmintic albendazole effects on the gut microbiota utilize primarily 16S rRNA sequencing [9, 10, 13, 16, 17]. Hence, a large study incorporating metagenomic sequencing with helminth infection status, albendazole treatment, and additional control groups may provide greater insights into these complex interactions.

Most of the helminth studies mentioned above utilized 16S rRNA sequencing to characterize the gut microbiota, while shotgun metagenomic approaches enable higher taxonomic resolution, at the species or strains level, and can identify not only bacteria but also archaea, fungi, and viruses [18, 19]. However, incomplete reference databases make it a challenge to profile uncharacterized microorganisms. Recently, an approach to assembling sequencing reads into contigs and binning them into putative genomes, known as metagenome-assembled genomes (MAGs), has enabled retrieving semi-complete genomes directly from samples without the need of culturing organisms [20, 21]. The Unified Human Gastrointestinal Genome (UHGG) established an integrated catalog of prokaryotic genomes containing 204,938 nonredundant genomes that represent 4644 prokaryotic species [3] by combining recent studies with large-scale assembly of MAGs from human microbiome data [3, 21, 22] as well as two culture-based studies that sequenced genomes from cultivated human gut bacteria [23, 24]. The Human Reference Gut Microbiome (HRGM) catalog expanded on UHGG to include underrepresented Asian metagenomes from Korea, India, and Japan [25] and added 780 new species from the newly assembled genomes [25]. However, Southeast-Asian countries remain underrepresented.

In this study, we generated shotgun metagenomics data from 650 Malaysian stool samples to investigate helminth-gut microbiome interactions by both cross-sectional and longitudinal analyses. The large sample size allowed us to examine these interactions in five different villages from different locations with different lifestyles. Examination of anthelmintic-treated uninfected individuals in the longitudinal phase enabled assessment of albendazole effects on the gut microbiome independent of helminths. Metagenomic data enabled investigation on the replication rates of individual bacterial species under different conditions. Since a substantial quantity of the microbiome remains undescribed, this large dataset from indigenous populations with traditional lifestyles from the underrepresented South East Asian region provides new insights into helminth-gut microbiome interactions and more comprehensive metagenomic sequences for future human gut microbiome studies.

Results

Gut microbiome analysis of indigenous Malaysians and urban controls

To identify and characterize helminth-associated microbiome effects, this study consisted of a cross-sectional component that compares urban individuals (n = 56) living in Kuala Lumpur (KL) with indigenous Orang Asli (OA) (n = 351) from five different villages (Figs. S1 and S2), as well as a longitudinal component to examine changes to the microbiome after anthelmintic (albendazole) treatment. A total of 650 fecal samples (including longitudinal samples) were processed for metagenomic sequencing, resulting in 11,480,206,516 paired reads after quality control and contamination removal (Supplementary Fig. S3). We compared different OA villages, which have different prevalence of soil-transmitted helminth infections (Supplementary Figs. S1 and S2). In the longitudinal phase of the study, consented OA subjects were treated with 400 mg albendazole for 3 consecutive days after collection of the first fecal sample. At 21 and 42 days following treatment, additional fecal samples were collected; however, this phase of the study was disrupted by the COVID-19 pandemic, reducing the number of paired samples available. KL subjects were not treated with albendazole, and they provided only one sample. Questionnaire data were collected and analyzed for some of the study subjects (n = 340).

When we first mapped the metagenomic sequences to RefSeq (i.e., bacteria, protozoa, fungi, viral, archaea genomes), we observed a very low percentage of mapped reads (median: 41.6%). However, when we mapped the sequences to databases that incorporate MAGs (i.e., HRGM [25] and UHGG [3], the percentage of sequencing reads mapped to HRGM (median: 91.5%) and UHGG (median: 87.9%) was much higher than RefSeq (Fig. 1A). Additionally, the percentage of mapped reads to all three databases was higher in KL subjects than the OA population (HRGM: p = 2.6e−11; UHGG: p = 1.2e−07; RefSeq: p = 2e−07) (Fig. 1A), indicating that there are more unknown microbial genomes in the OA population.

Fig. 1
figure 1

Variation in the gut microbiome of 650 Malaysians from Orang Asli (OA) villages and Kuala Lumpur (KL). A Violin plots illustrating the percentage of mapped reads with RefSeq (i.e., bacteria, protozoa, fungi, viral, archaea), Unified Human Gastrointestinal Genome (UHGG), and human reference gut microbiome (HRGM) databases between OA (green) and KL (purple) samples. B Relative abundance of phyla from the 237 species of the core gut microbiota of OA and KL populations (left). The relative abundance of the main species from Firmicute A (right). C Bar plot shows the bacterial species that are differentially abundant between Orang Asli and Urban cohort from Kuala Lumpur based on the output of the microbiome multivariable association with linear models 2 (MaAsLin2). The length of the bar corresponds to the value of the significant association. Red color represents the bacterial species associated with the Orang Asli subjects, whereas blue color represents the bacterial species associated with the urban cohort. D The percentage of mapped reads to the HRGM database for samples from different OA villages and KL. E Comparison of pairwise beta diversity at species level within group to the KL cohort, assessed by Jaccard distance based on the distance of nucleotide k-mer sketches k = 21 (top) and genus-level classification (bottom). F Principal coordinates analysis (PCoA) of Jaccard distance based on the gut metagenomic profiles (species levels) in all samples, with individuals from different geographical locations denoted by specific color (ADONIS: p = 0.001, R2 = 0.073; ANOSIM: p = 0.001, R = 0.215). The p-values for A, D, and E are computed using Wilcoxon rank-sum test

Utilizing HRGM, we determined the core microbiota for the Malaysian population and found that 237 core bacterial species were 100% shared among the subjects (Fig. 1B; Supplementary Figs. S4 A–E and S5 A–C). The most abundant phylum was Firmicutes A, the majority of which were uncultured species [3] (Fig. 1B). Agathobacter rectalis, Blautia_A wexlerae, and Agathobacter faecis were the main species from Firmicutes A (Fig. 1B). Using a cross-validated random forest model to identify core microbiota species driving the variation between OA vs KL subjects, we achieved a mean prediction accuracy of 98.05% at a kappa of 96.06% (out-of-bag error = 1.8%). Megamonas funiformis, Phocaeicola plebeius A, Bacteroides stercoris, Phocaeicola massiliensis, and HRGM Genome 3145 were the top five predictors between OA and KL subjects (Supplementary Fig. S6 A–C). Of these, HRGM Genome 3145, Gemmiger sp900539695, and Blautia A sp000436615 were more abundant in OA subjects, while Megamonas funiformi, Phocaeicola plebeius A, and Bacteroides stercoris were more abundant in KL subjects (Supplementary Fig. S6 A–C). The bacterial species with the largest variation (cutoff 6.0 for the coefficient of variation) among the core gut microbiota is shown in Supplementary Fig. S7A. To control for covariates, we utilized (MaAsLin2) to identify bacterial taxa differentially abundant between OA and KL subjects that are independent of village, age, and sex. Fourteen bacterial species, of which many are uncharacterized, including HRGM_Genome_2427 (p = 0.009), CAG 964.sp000435335 (p = 0.009), and Ruminococcus_E sp003438075 (p = 0.009), are more abundant in OA subjects, whereas HRGM_Genome_0171 (p = 2e−4) and HRGM_Genome_3486 (p = 0.009) are more abundant in KL subjects (Fig. 1C and Supplementary Table S1).

The Orang Asli live in different geographical settings and have distinctive cultures and lifestyles. We found that KL subjects have higher mapped reads than all OA villages (Fig. 1D; Supplementary Fig. S7 B and C), and the percentage of mapped reads from both villages Rasau (p = 2e−16) and Legong (p = 2e−16) was markedly lower compared to KL (Fig. 1D; Supplementary Fig. S7 B and C). To compare pairwise beta diversity at the species level within each village group to the KL cohort and to use a reference independent strategy as an alternative approach, we assessed Jaccard distances using 21 nucleotide k-mers and genus-level annotations from HRGM, which showed similar results (Fig. 1E). In addition, we observed that Rasau and Legong had the highest beta diversity and nucleotide dissimilarity compared to KL (Fig. 1E). Moreover, comparison of bacterial communities at species level across geographical locations using Jaccard distance revealed substantial differences between villages (ADONIS: p = 0.001, R2 = −0.073; analysis of similarity [ANOSIM]: p = 0.001, R = 0.215) (Fig. 1F, Supplementary Table S2). From the principal coordinate analysis (PCoA) plot (Fig. 1F), we observed clustering of the samples from Rasau and Legong. Conversely, the samples from Bangkong and Sepat were clustered together with KL, while Judah exhibited a more dispersed distribution. Hence, OA subjects in Rasau and Legong were more similar in gut microbial composition and were different from KL and other villages. Equivalent beta-diversity results were observed with other k-mers sketches (31 and 51) and at the species level (Fig. S8 A–G).

Village-dependent effects of helminth infection on the gut microbiome

We determined the infection intensity and the prevalence of intestinal helminth infection among the 351 OA subjects and found that Trichuris infection (61.8%, n = 217) was the most predominant, followed by hookworm (20.8%, 73) and Ascaris (17.9%, 63) infections (Fig. 2A). The distribution of age and gender of these subjects is shown in Fig. S9 A and B. The overall prevalence of helminth infection was 67.2% (n = 236) (Fig. 2A), and infection intensity was summarized in Fig. S9C. For beta diversity at species level, based on PCoA, there were differences in gut microbiome between infected and uninfected individuals; however, statistically, the effect size was small (ADONIS: p = 0.001, R2 = 0.024; ANOSIM: p = 0.001, R = 0.145) (Fig. 2B, Supplementary Table S2), which was also the case for Bray-Curtis distance and nonmultidimensional scaling (NMDS) ordination (Supplementary Fig. S10 A–C).

Fig. 2
figure 2

Effects of intestinal helminth infection status on gut microbial diversity and composition for the 351 Orang Asli individuals. A The prevalence of intestinal helminth infection in the OA cohort based on overall infection status, as well as specific intestinal helminth infection (i.e., trichuriasis, ascariasis, and hookworm infection). B Principal coordinates analysis (PCoA) of Jaccard distances based on gut microbiota profiles (species levels) of the OA cohort. The individuals infected and uninfected with intestinal helminths are denoted by blue and red, respectively (ADONIS: p = 0.001, R2 = 0.024; ANOSIM: p = 0.001, R = 0.145). C Alpha-diversity box plot of species richness based on different status of intestinal helminth infection, number of intestinal helminth infection, Trichuris infection, and intensity of Trichuris infection. Wilcoxon rank-sum test is used for two independent variables, while the Kruskal-Wallis test is used for more than two comparison groups. D The prevalence of intestinal helminth infection (top) and Trichuris infection (bottom) by different geographical locations. E Comparison of alpha diversity (species richness) between individuals from KL and specific OA villages. F Spearman correlation between the intensity of Trichuris infection and percentage of unmapped reads to the HRGM database (p = 3.200e−6, R = 0.250). The blue line represents the linear regression between intensity of Trichuris infection and percentage of unmapped reads. G Bar plot of the F statistic values from ADONIS analysis of variables that contribute to the gut microbiota composition. Colored bars indicate the variables that show significant effects on gut microbiota variation (p < 0.05). H Bar plot of effect size of the variables (village [p = 2.610e−08, pseudo R2 = 0.027], helminth infection [p = 0.029, pseudo R2 = 0.004], and interactions between helminth village [p = 0.002, pseudo R2 = 0.016]) that contribute significantly to the variance of the microbiota based on MDMR analysis. I Heatmap of bacterial species associated with village, helminth infections, and interactions from MaAsLin2. Blue for positive association and red for negative association

For alpha diversity at species level, we observed higher species richness in the samples from infected subjects (p = 2.50e−5) (Fig. 2C). This relationship was confirmed by a linear mixed model analyses controlling for village as a random effect (p = 1.18 × 10−6). Individuals infected with either single (p = 0.005) or multiple species of helminths (p = 0.033) had higher species richness (Fig. 2C). Trichuris-infected OA (p = 9.70e−06) had higher species richness than uninfected (Fig. 2C), including those infected at light (eggs per gram [epg] < 999; p = 0.045) and moderate (epg < 9,999; p = 8.34e−07) intensities (Fig. 2C). Other alpha-diversity indices (i.e., Shannon and Simpson, at species level as well) are shown in Supplementary Fig. S11 A–H, and results for each village are shown in Supplementary Fig. S12 A–E. The prevalence of helminth infection varied according to village and was highest in Rasau (89.6%, n = 43 of 48), followed by Legong (81.0%, 81 of 100), Judah (71.6%, 83 of 116), Sepat (55.0%, 22 of 40), and Bangkong (14.9%, 7 of 47) (Fig. 2D). As Trichuris was the predominant helminth, the prevalence of Trichuris was similar for Rasau (81.3%, 39 of 48), Legong (77.0%, 77 of 100), Judah (65.6%, 76 of 116), Sepat (50.0%, 20 of 40), and Bangkong (10.6%, 5 of 47) (Fig. 2D). There was no infected individual with helminths in KL. The two villages with the highest prevalence, Rasau (p = 2.0e−4) and Legong (p = 8.1e−07), showed higher species richness compared to KL (Fig. 2E). Also, we observed that species richness appeared to be greater when helminth infections in the villages were more prevalent, which was similar to the order of villages for unmapped reads shown in Fig. 1D. To determine if Trichuris infection intensity was associated with unmapped reads, we performed a Spearman correlation test and found that the intensity of Trichuris infection was positively correlated (p = 3.2e−06, R = 0.25) with the percentage of unmapped reads to the HRGM database (Fig. 2F). These results indicated that helminth infections were associated with underrepresentation in the catalog of bacterial genomes.

We next determined the relative contribution of village and helminth infection status on the gut microbiome in relation to other factors (e.g., whether they had probiotic food, diarrhea, or antibiotics in the past 3 months, different age groups, and protozoa infection). ADONIS analysis at species level indicated that only village (p =1.000e−4, F-value = 1.672, R2 = 0.025) and helminth status (p = 0.028, F-value = 1.387, R2 = 0.010) had significant effects on the gut microbiome composition (Fig. 2G). Since village has the largest effect size on gut microbiome composition, we next used MaAsLin2 [26] to identify bacterial species that were differentially abundant between Trichuris infected and uninfected individuals from specific villages. Importantly, we found that the bacterial species that were most differentially abundant between infected and uninfected subjects were unique to each village (Supplementary Fig. S13A). For example, Haemophilus_A.parahaemolyticus and Corynebacterium provencense were different in Bangkong and Rasau, whereas Prevotella.sp900316565 was different in Sepat, C941.sp004557565 and UBA10281.HRGM_Genome_2392 in Judah, Prevotella.sp900546575 and Prevotella.HRGM_Genome_3676 in Legong, and UBA1829.sp900549045 and F082.HRGM_Genome_5331 in Rasau (Supplementary Fig. S13A). Similar patterns of results were obtained with ANCOM-BC (Supplementary Fig. S13B). These results indicated that helminth infections may have different effects on the gut microbiome in different villages.

To specifically test the hypothesis that effects of helminth infection on the microbiome are highly dependent on village, we used multivariate distance matrix regression (MDMR) [27] to test for statistical interactions between helminth infection and village and to calculate relative effect sizes on microbiota variation at species level. We find that there was a significant interaction between village and helminth infection, and that the effect of village is greater than helminth infection status after accounting for the effects of this interaction (Fig. 2H).

To identify bacterial features that are significant in helminth-village interactions, as well as independent of these covariates, we used MaAsLin2 with helminth and village as fixed effects, to identify bacteria that are independent and associated with the interaction between helminth and village. Of the 230 helminth-associated bacteria, more than 55% (n = 135) were associated with village (Fig. 2I). Hence, most of the effects of helminth-associated bacteria are village dependent, and in different villages, there are different bacteria associated with helminth infections. Several Lactobacillus species, including Lactobacillus gasseri and Lactobacillus crispatus, were associated with helminth infection independent of village (Supplementary Fig. S14 and Table S3).

Dynamic changes to the gut microbiome after anthelmintic treatment

Longitudinal interventional approaches provide stronger assessment of cause-and-effect relationships. Fecal samples analyzed at pre- and post-anthelmintic treatment provided insights into the effects of deworming on the gut microbiome. Individual subjects were grouped into four categories (i.e., full responders [n = 43 paired; from 26–33,099 to 0 epg], partial responders [n = 23 paired; from 281–119,875 to 26–71,579 epg], nonresponders [n = 5 paired; from 204–1097 to 281–1632 epg], and uninfected [n = 58 paired]), based on the Trichuris infection intensity before and after deworming (Fig. 3A). While mixed infection was present in some individuals, hookworm and Ascaris infection were always cured after deworming (Supplementary Fig. S15A).

Fig. 3
figure 3

Dynamic changes to the gut microbiota of 129 Orang Asli after albendazole treatment. A Line plots show changes of the infection intensity of Trichuris pre and post response to anthelmintic drugs stratified by full responders (n = 43), partial responders (n = 23), nonresponders (n = 5), and uninfected individuals (n = 58). B Principal coordinates analysis (PCoA) plot of Jaccard distances based on gut microbiota profiles (species levels) of responders (ADONIS: p = 0.001, R2 = 0.014; ANOSIM: p = 0.001, R = 0.072), with pre-anthelmintic treatment (blue) and post-anthelmintic treatment (red). C Principal coordinates analysis (PCoA) plot of Jaccard distances based on gut microbiota profiles (species levels) of uninfected subjects (ADONIS: p = 0.006, R2 = 0.012; ANOSIM: p = 0.001, R = 0.069) (Fig. 3C, Supplementary Table S2), with pre-anthelmintic treatment (blue) and post-anthelmintic treatment (red). D Venn diagram depicting the number of shared and exclusive bacteria species that are found to be differentially abundant (pre and post) between responders and uninfected individuals. The blue area includes 253 bacteria that are altered only in responders, while the yellow and mixed color area indicates the 873 bacteria that are altered in uninfected individuals. E Box plots show the bacterial taxa that are altered by deworming treatment in responders but not in nonresponders (host response). The differences in the abundance of (i) Sutterella HRGM Genome 4418 and (ii) Muricomes contorta_B in helminthic infections (left) and different groups of response (right), namely uninfected and responders in pre and post. F Box plots and line plots show the bacterial taxa that are associated with deworming treatment in both responders and nonresponders (drug response). The differences in the abundance of (i) Collinsella sp900540485 and (ii) Collinsella stercoris in helminthic infections (left) and different groups of response (right), namely uninfected and responders in pre and post. The p-values for E and F are computed using Wilcoxon signed-rank test (responders vs nonresponders) and Wilcoxon rank-sum test (helminthic infections)

First, we compared pre and post samples for responders, which include both full and partial responders. PCoA based on Jaccard distances showed that there are differences in gut microbiota composition at species level between pre and post treatment, but the effect size was small (ADONIS: p = 0.001, R2 = 0.014; ANOSIM: p = 0.001, R = 0.072) (Fig. 3B, Supplementary Table S4). Since albendazole may have a direct effect on the microbiota, we next compared the gut microbiota profile pre and post treatment for uninfected individuals. Similar to the responders, PCoA based on Jaccard distances also indicated differences in gut microbiota composition at species level between pre and post samples, with a small effect size (ADONIS: p = 0.006, R2 = 0.012; ANOSIM: p = 0.001, R = 0.069) (Fig. 3C, Supplementary Table S4). NMDS ordination, Bray-Curtis distance matrix, and beta-dispersion analysis showed similar results (Supplementary Figs. S16 A–E and S17 A–E, Table S4), and there were no significant changes to alpha diversity at species level between pre and post treatment (i.e., Richness, Shannon, Simpson) (Supplementary Fig. S15 B and C).

Using MaAsLin2 for differential abundance testing, we found changes of 911 bacterial species at pre and post treatment among responders. However, there was substantial overlap with changes found in pre and post treatment samples for uninfected individuals (658 species, 72.2%) (Fig. 3D and Supplementary Fig. S18A), with only 253 taxa which were specific to the responders. For example, in both responders and uninfected individuals, the relative abundance of Collinsella sp003466125 (p = 1.52e−08; p = 3.66e−07, respectively) and RUG013.sp001486445 (p = 2.53e−07; p = 3.80e−06) was reduced after deworming, while the relative abundance of Bilophila sp900550745 increased (p = 1.33e−08; p = 5.81e−05) (Supplementary Fig. S18 B and C). To assess the longitudinal effects of albendazole treatment, we also used MaAsLin2 to identify taxa altered by treatment response, controlling for infection status and village as fixed effects. Of the 576 species that were identified to be associated with these covariates, the majority were associated with village (n = 305) and with infection status (n = 200), and only 69 species were associated with treatment response, of which only four species were independent of village and infection status (Supplementary Fig. S19A). Of the four, only one (CAG.245.sp900552135) showed a statistically significant (Supplementary Fig. S19B) association with treatment response but independent of village or infection status (Supplementary Table S5).

Next, we used MaAsLin2 to identify bacterial taxa that were associated with drug response and helminth status, correcting for village as a covariate. There were a total of 293 bacteria species that were associated with drug response, only six of which were associated with host response (Supplementary Fig. S19C). There were only two taxa (Sutterella HRGM Genome 4418 and Muricomes contorta B) associated with deworming treatment in responders but not in nonresponders in this model. The Sutterella HRGM Genome 4418 (p = 8.10e−06) was more abundant in helminth-infected individuals, whereas Muricomes contorta B (p = 0.024) was more enriched in nonhelminth-infected individuals (Fig. 3E). In contrast, there were many more taxa (n = 295) that are associated with deworming treatment in both responders and nonresponders like Collinsella sp900540845 and Collinsella stercoris (Fig. 3F), which indicates that the effects of albendazole were greater on the bacterial communities than helminth infection itself (Supplementary Table S6). Hence, albendazole may have a substantial effect on the microbiota that may be an important confounding factor for deworming studies.

In some individuals, we conducted a follow-up study 42 days post-anthelmintic treatment. There were no differences in alpha diversity on day 42 (Supplementary Fig. S20A), and although beta-diversity analysis at species level showed significant differences between three timepoints (i.e., pre, 21 days, and 42 days) (Supplementary Fig. S20 B–C, Supplementary Table S7), these differences are driven by the pre-treatment samples (Supplementary Fig. S20D). Therefore, the changes in the gut microbiome in both responders and uninfected individuals after albendazole treatment remain stable by day 42.

Bacterial replication in the context of helminth infection

Actively replicating bacteria can be identified by calculating the index of replication based on coverage trends of bidirectional genome replication from a single origin of replication. We used the algorithm growth rate index (GRiD) to estimate the growth rate of gut bacteria in relation to helminth infection status. Spearman correlation analysis on Trichuris egg burden with the growth rate of the bacteria identified 350 bacterial species with growth rate associated with Trichuris egg burden (Fig. 4 A and B, Supplementary Fig. S21, and Supplementary Table S8). Prevotella stercorea replication was most positively associated (p = 1.58e−14, R = 0.39) with egg burden, while Bifidobacterium longum (p = 1.30e-11, R = −0.35) and Phocaeicola vulgatus (p = 3.45e−9, R = −0.31) were negatively associated with egg burden. Using a linear mixed model of Trichuris egg burden while controlling for village, Prevotella stercorea and Bifidobacterium longum were significantly associated with egg burden (p = 4.89e−06 and p = 7.99e−08, respectively). The predicted replication rate of Prevotella sterorea was higher in Trichuris-infected individuals (p = 1.30e−09), while the predicted replication rate of Bifidobacterium longum and Phocaeicola vulgatus was notably lower in Trichuris-infected individuals (p = 4.50e−09and p = 9.80e−09, respectively) (Fig. 4C).

Fig. 4
figure 4

Gut bacterial replication in the context of intestinal helminth infection. A Heatmap of the growth rate index (GRiD) score, which infers an index of replication for top 20 gut bacteria in relation to helminth infection status of individuals based on Spearman correlation test followed by false discovery rate (FDR) correction. Samples are shown in rows, by village, whereas the GRiD score of each bacterium is shown in columns. The first vertical side bar color codes the intestinal helminth infection status, while the second side bar indicates the infection intensity of Trichuris. B GRiD score correlation between bacterial species with the infection intensity of Trichuris. The bar chart shows the Spearman’s rank correlation coefficient. Blue and gray colors represent the positive and negative correlations respectively. C Box plots of GRiD score for Prevotella stercorea (left), Bifidobacterium longum (middle), and Phocaeicola vulgatus (right) in Trichuris infected and uninfected individuals. The GRiD scores of these species between Trichuris infected and uninfected individuals were tested using Wilcoxon rank-sum test

For the longitudinal deworming component of the study, we observed that the growth rate of 93 bacterial species was different between pre and post treatment samples among the responders. Among these bacterial species, slightly more than one-third of them (n = 33) were also identified from the cross-sectional analysis (Supplementary Fig. S22A). Spearman correlation analysis demonstrated that the growth rate of uncultured Oscillibacter sp. (p = 6.000e−04) and Phocaeicola vulgatus (p = 0.006) was significantly correlated with Trichuris burden (Supplementary Fig. S22B and Supplementary Table S9). After we verified the results by building a linear mixed model controlling for village, both uncultured Oscillibacter sp. (p = 0.033) and Phocaeicola_vulgatus (p = 0.028) were negatively associated with Trichuris burden, indicating more replication in responders after anti-helminthic treatment (Supplementary Fig. S22C). However, considerable portions of responder-associated taxa (32 out of 93) were also observed in the noninfected individuals (n = 67) (Supplementary Fig. S22D). Therefore, it could be difficult to disentangle the effects of Trichuris infection and direct effects of albendazole treatment on the dynamics of the microbiome. Hence, we conducted MaAsLin2 analyses to identify bacterial replication (based on GRiD score) that were associated with Trichuris infection while controlling for treatment group and village. Collinsella_sp._TF06.26 (adjusted p = 0.004) was positively associated with Trichuris infection, while Phocaiecola vulgatus (adjusted p = 0.04), Burkholderia sp. K4410.MGS.135 (p = 0.04), Bacteroides stercoris (adjusted p = 0.04), and Phocaeicola massiliensis (adjusted p = 0.04) were negatively associated with Trichuris infection (Supplementary Table S10).

Functional gene profiles of the Orang Asli microbiota and the effects of albendazole treatment

We used the HUMAnN tool to investigate pathway inference and gene families with Pfam domains (Figs. S23–S25). To adjust for covariates, we used MaAsLin2 to identify the pathways and gene families that were differentially abundant between the following: (1) Orang Asli and Urban cohorts, while controlling for age and sex; (2) Helminth positive and negative individuals, while including village as a covariate; and (3) Treatment response groups while controlling for village. Using these models, we find that the L-tryptophan biosynthesis superpathway was enriched in the Orang Asli microbiome compared to urban controls from KL (Supplementary Figs. S23 and S25A). Tryptophan, an essential amino acid, and its catabolites have been suggested to affect intestinal homeostasis through the aryl hydrocarbon receptor and may be important in inflammatory bowel diseases [28]. Hence, future work on microbial metabolism in the Orang Asli may focus on this pathway.

After controlling for village, there were no significant pathways differentiating helminth positive and negative individuals. Between different villages, we found the strongest significance for the peptidoglycan biosynthesis II (Staphylococci) pathway (Supplementary Figs. S23 and S25B). Villages with high helminth prevalence have individuals enriched in this pathway, but we did not find a significant relationship between helminth infection and Staphylococcus aureus abundance. This pathway may be important in the generation of peptidoglycan in other gram-positive bacteria, and the significance of the geographical difference in the abundance of this pathway is still unclear. We also found that the microbiome after albendazole treatment is enriched for the L-glutamate degradation V (via hydroxyglutarate) pathway (Supplementary Figs. S23 and S25C), which is an indication that albendazole may affect the fermentation of amino acids in an anoxic environment. Additionally, in our gene family enrichment analysis, our only substantial observation is that the phosphoenolpyruvate carboxylase gene family was decreased after albendazole treatment (Supplementary Figs. S2 and S25D). This also indicates how albendazole can affect metabolic processes of the microbiome; however, the implications of these results remain unclear.

Discussion

In this study, we examined 650 stool metagenomes from a cohort of 351 indigenous Malaysians from five villages with different prevalence rates (14.9–89.6%) of helminth infections, along with 56 urban citizens (uninfected) living in Kuala Lumpur City. To our knowledge, this is the largest study utilizing shotgun metagenomics to investigate the interactions between helminth infection and the human gut microbiome.

We found that mapping reads to the HRGM database, which incorporates MAGs, increase the quality and quantity of taxonomic classifications, compared to using from the NCBI database alone, especially for the indigenous Orang Asli. We also found that the microbiota is dominated by Firmicutes A, which is represented by mostly uncultured bacteria, highlighting the underrepresentation of cultured bacteria from indigenous groups. This could be an important caveat for most of the previous studies on helminths and the gut microbiota, which were conducted using 16s rRNA sequencing [10,11,12, 16, 29] with the taxonomic classification based on mapping to the reference databases Greengenes, SILVA, and Ribosomal Database Project. In a recent shotgun metagenomic study on 175 Cameroonian samples, the data was also mapped onto a reference database from NCBI [30], and the investigators noted that the classification of the relative abundance of bacteria did not correspond to data from 16S Greengenes classifications for the V4 region [30]. A different study also indicated that 16s rRNA gene sequencing only provided a portion of the gut microbiota profile compared to shotgun metagenomics [31]. Hence, we suggest that assembling a Malaysian gut microbiome reference catalog will provide substantial benefit for future microbiome studies, especially from underrepresented geographic regions, and for rural and indigenous populations.

In this metagenomic study, we found that intestinal helminth infection status was associated with higher species richness, which was consistent with our previous findings and others conducted using 16s rRNA sequencing [9, 11, 12, 16, 30, 32]. However, we did not find a significant difference at pre and post deworming, which could be because of smaller sample size and could also be confounded by the effects of albendazole. Additionally, other studies have not observed an effect of helminths on microbial diversity [7, 10, 13, 16, 17, 33]. It is important to note that each study cohort has different prevalence rates for different helminth species, as well as distinct genetics, lifestyles, and living conditions. This study has a larger sample size than our previous studies [12, 34] and has enabled us to examine the interactions of helminth infection and the gut microbiome in different villages. Indeed, village has the largest effect size on gut microbiome variation, followed by helminth infection status. Notably, villages with higher helminth prevalence rates also have higher microbial diversity, but in different villages, helminth infection is associated with differential abundances of distinct bacterial taxa. It is important to note that the different villages represent diverse environments, practicing unique lifestyles, and have different hygiene practices. Compared to other villages, Rasau and Legong villages (with higher helminth prevalence) are located near the forest with high exposure to the soil environment, which may harbor more microbes [35], and mouse experiments have shown that exposure to soil increases gut microbiota diversity [36]. From our questionnaire, a higher percentage of villagers from Rasau and Legong are plantation agricultural workers (Rasau: 30.4%; Legong: 14.8%; others: < 6.7%), lack of toilet facility (52.5%; 20.8%; < 13.0%), and practice open defecation (46.5%; 32.0%; < 7.4%) more than other villages. As Trichuris eggs become infective in the soil, this may increase exposure to Trichuris, as well as other microbes in the contaminated soil, resulting in higher microbial diversity in different settings.

We also found that deworming helminth-negative individuals can influence the gut microbiome that overlaps substantially with changes in individuals responding to drug treatment by having reduced worm burdens. This indicates that albendazole may directly affect the microbiome, or that there are population effects that can influence uninfected people. There are four previous studies on albendazole [9, 10, 13, 16]. The first study conducted among Ecuador school children did not find any difference in bacterial composition among both Trichuris infected and uninfected groups after a combination of albendazole and ivermectin treatment [13]. In contrast, the second study in Indonesia found an increase of Actinobacteria and decrease in Bacteroidetes with albendazole treatment versus placebo in individuals that remained helminth-infected post treatment, but not in uninfected individuals [16]. In addition, Rosa et al. demonstrated that the gut bacterial composition was altered in a helminth-uninfected group in Indonesia after 2 years [9]. Another study in Kenya found significantly reduced Chao richness in uninfected individuals after deworming treatment, suggesting an effect of albendazole [10]. Albendazole is a prodrug that metabolizes rapidly to albendazole sulfoxide (the active anthelmintic compound) and albendazole sulphone (the inactive compound). Some bacterial species (Enterobacter aerogenes NCIM 2695, Klebsiella aerogenes NCIM 2258, Pseudomonas aeruginosa NCIM 2074, and Streptomyces griseus NCIM 2622) could be involved in metabolizing albendazole to albendazole sulfoxide and albendazole sulphone [37]. Albendazole can also be metabolized by the resident microbiota in gut rumens in sheep and cattle [38]. Hence, the gut microbiota could play a crucial role in metabolizing albendazole and influence drug bioavailability and efficacy on infected individuals. Why albendazole has lower efficacy against Trichuris infection than hookworm and Ascaris warrants further investigation [39]. Future studies could apply metabolomics profiling to investigate metabolite differences between response groups to better understand the underlying mechanisms. The observation that the L-glutamate degradation V (via hydroxyglutarate) pathway was enriched after albendazole treatment and that the phosphoenolpyruvate carboxylase gene family was depleted after albendazole treatment indicates that albendazole can affect metabolic processes, and future work should focus on the effects of albendazole on microbial metabolism.

Conclusions

We find that this metagenomic study of rural indigenous populations required reference databases that included MAGs to improve taxonomic and functional classification of sequencing reads. However, unmapped reads remain a challenge as villages with higher prevalence of helminth infections have more unmapped reads. Hence, this large metagenomic dataset from five different villages in Malaysia with different helminth infection prevalences should facilitate further characterization of microbiome-parasite associations in other nonindustrialized populations. Helminth effects on the microbiome were village dependent, and albendazole treatment had a substantial effect on the microbiome. These results may explain some of the discrepancies from previous studies on helminth-microbiota interactions.

Methods

Study design and sample collection

This study consists of both cross-sectional and longitudinal phases. Cross-sectional comparisons were made on the OA and between OA and urban cohorts (KL) living in the capital city of Malaysia, Kuala Lumpur. Within the Orang Asli community, we studied five Orang Asli villages: (1) Rasau village (Perak state); (2) Judah village (Selangor state); (3) Sepat village (Selangor state); (4) Bangkong village (Selangor state); and (5) Legong village (Kedah state). The locations of each village are displayed on a map using ArcGIS (version 10.7.1) together with other information including states, tribes, and subtribes (Supplementary Fig. S1). A total number of 351 samples were collected from Orang Asli subjects and 56 samples from KL subjects in this cross-sectional component (aged 4 years and older) (Supplementary Fig. S2).

For the longitudinal phase, Orang Asli subjects who provided consent were treated with 400 mg albendazole for 3 consecutive days after the first stool sample collection. Stool samples were collected from the treated subjects at 21 days and 42 days following anthelmintic treatment. However, due to the restriction during the COVID-19 pandemic, only four Orang Asli villages were included in this phase, excluding Legong village. There was no follow-up for urban controls after the cross-sectional phase because they were not treated with albendazole. Sample selection for analysis was based on a complete set of paired stool samples (pre [pre-anthelmintic treatment] and post [21-day post-anthelmintic treatment]) (n = 129) and three timepoints stool samples collection (pre, 21 days and 42 days; n = 110). Four subject samples were removed from the longitudinal analysis due to incomplete data collection. Then, subjects were categorized into three groups for comparison: responders, nonresponders, and uninfected, based on their infection status before and after the albendazole treatment. Responders (n = 66 paired samples) refer to individuals who were positive at baseline and became negative or showed reduction of infection intensity after deworming. Nonresponders (n = 5 paired samples) refer to individuals who were positive at baseline and showed increment or maintain of egg counts after deworming. Uninfected (n = 58 paired samples) refers to negative individuals before and after the treatment. Nonresponders were not included in the gut metagenome analysis due to insufficient sample size. The detailed number of samples collected at each timepoints was shown in Supplementary Fig. S2.

Fecal sample preparation and analysis

All the stool samples collected were divided into two portions: (i) preserved in 2.5% potassium dichromate and stored at 4 °C for intestinal helminth infection screening and (ii) aliquoted in 1.5 ml cryovial tube, frozen immediately in dry ice, and kept at −80 °C for shotgun metagenomic analysis (Supplementary Fig. S26). To detect and quantify helminth infections, Kato-Katz was performed. A thick smear was prepared from the fresh stool according to the manufacturer’s instructions (Kato-Katz kit, Mahidol University, Thailand) [40]. Infection intensity was stratified into light, moderate, or heavy according to WHO cutoffs [41]. Formalin ether sedimentation was performed according to Chin et al. (2016) [42]. Stool samples were considered positive if any soil-transmitted helminths were detected from any of these two methods. DNA was extracted from stool samples using Qiagen DNeasy PowerSoil Pro Kit (Qiagen, Hilden, Germany). DNA library was prepared using Illumina TruSeq DNA Nano Library kit (Illumina, USA). Paired-end metagenomic sequencing was performed on the NovaSeq 6000 S4 platform to generate an average of 20 million paired-end reads per sample (range 13–35 million paired-end reads), with a read length of 150 bp and insert size of 350 bp.

Sequencing analysis pipeline

The overall bioinformatic analysis workflow from preprocessing to downstream analysis is shown in Supplementary Fig. S3. In brief, the whole process of quality filtering and trimming of the raw sequence reads was performed by using KneadData (version 0.7.4) integrated with Trimmomatic [42], Bowtie [43], and FastQC [44] tools. Sequence reads were trimmed by using Trimmomatic with default settings, based on a sliding window trimming approach (SLIDINGWINDOW:4:20) when average base Phred quality score over four reads dropped below 33 (PHRED 33). Next, sequence reads were mapped against reads mapping to the reference genome (hg37) using Bowtie2 with default parameters (very sensitive end-to-end alignment) to remove human host genome. The filtered reads were then used for the downstream analyses. Additionally, FastQC was used to perform quality checks on the raw metagenomic reads before preprocessing and after preprocessing to ensure high-quality metagenomic reads for downstream analysis.

For taxonomic classification, Kraken2 (version 2.1.0) [45], a k-mer matching algorithm classifier, was used for assigning taxonomic labels to the trimmed reads. The trimmed reads were mapped using Kraken2 against (1) RefSeq database (bacterial, protozoa, fungi, viral, and archaeal) and two MAGs integrated databases: (2) HRGM database, with 232,098 reference genomes [25], and the UHGG database, with 204,938 reference genomes [3] using default settings. After taxonomic classification by Kraken2, Bayesian Re-estimation of Abundance with KrakEN (Bracken2) (version 2.6.0) [46] was used to compute the relative abundance of bacteria for each taxa (from phylum to species level) using default settings with a read length parameter of 150. The mapped reads of the OA and KL cohorts were then plotted into a violin plot using ggplot2 package [47] to access which databases provide better taxonomic classification. The distribution of the mapped reads was determined using the Shapiro test from the rstatix package [48]. Then, the difference between the mapped reads of OA and KL was determined using the Wilcoxon rank-sum test from ggplot2 package [47]. The data generated from Bracken2 were exported in the form of BIOM (Biological Observation Matrix) table and analyzed using R programming language (version 4.0.5, R Studio, Inc., Boston, MA, USA). The BIOM table was imported and filtered using the phyloseq package [49]. Only those taxa with a minimum prevalence of 20% across all the samples and a minimum coefficient of variation of 3.0 were included in the following analysis (Supplementary Fig. S27). In general, ggplot2 [47] and ggpubr package [50] were used to create visualization plots.

In order to confirm our findings, we performed reference independent strategy by using Sourmash (version 4.0.0) [51] to compute k-mer sketches. To discard erroneous k-mers, the low abundance of k-mers was trimmed using “trim-low-abun” from k-mer project, with a k-mer abundance cutoff of 3.0 and trimming coverage of 18. Signatures were generated for each sample using “sourmash compute” with a compression ratio of 10,000 (−scaled 10,000) and k-mer lengths of 21, 31, and 51 (−k21, −k31, −k51). A signature output was generated for Jaccard distance comparisons. Before the k-mer comparison, “sourmash index” was used to create a Sequence Bloom Tree database from a collection of signatures. Lastly, “sourmash compare” was used with default settings to compare the signatures at each length of k.

The core microbiota was determined by including taxa present across all samples (i.e., prevalence of 100% across all the samples). Then, the core microbiota was visualized using bar chart to compare the heterogeneity between OA and KL cohorts as well as the heterogeneity across different villages. Alpha diversity, in terms of species richness [52], Shannon [53], and Simpson index [54], was analyzed using the microbiomeSeq package [55]. Beta-diversity analysis was performed on both the Jaccard and Bray-Curtis dissimilarity matrix calculated from the taxon abundance data standardized using Hellinger. Differences in beta diversity between groups (i.e., different OA villages and different helminth infection status) or between different timepoints (pre vs post) were displayed with principal coordinates analysis (PCoA) plots and NMDS plots.

Metagenomes were annotated for functional genes and pathways using HUMAnN v3.0 and its UniRef 50, Pfam, and MetaCyc pathway databases using read sequences that were trimmed and quality filtered using KneadData [56]. Read counts were normalized for gene length (reads per kilobase), transformed by centered log ratio, and filtered to remove very low prevalent features before statistical analyses were carried out with MaAsLin2 v1.10 using the same linear-mixed effects models as for the taxonomic comparisons [26].

Growth rate index (GRiD) (version 1.3) was used to evaluate the growth rate of microbial species in metagenomic samples [57]. Samples were mapped to a GRiD database (ftp://ftp.jax.org/ohlab/GriD_environ_specific_database/stool_microbes.tar.gz), a stool-specific database created based on microbes mostly found in stool. GRiD score > 1.02 indicates bacteria are in growth phase, whereas GRiD score < 1.02 indicates that bacteria are in stationary or lag phase. The downstream analysis was conducted as described in Supplementary Fig. S28.

Statistical analysis

Multiple efforts were done for the differential abundance analysis of the bacteria from OA and KL cohorts. Random forest (randomForest package) was used to identify microbiome taxa predictive of OA and KL [52] groups. We generated a “SMOTE” (Synthetic Minority Oversampling Technique) (consisting of 280 OA and 336 KL) dataset using the package DMwR [58] to address the imbalance number of samples between OA (n = 594) and KL (n = 56) samples [59]. SMOTE algorithm is a technique to address the imbalanced datasets by oversampling the minority class. A new data of the minority class was created artificially using the nearest neighbors of these cases and hence leading to a more balanced dataset [60]. Then, the random forest model was built based on this “SMOTEd” data set and tuned with the methods described by Brwonlee (2016) [61], followed by the significant testing using the methods described by Douglas (2020) [62]. Another more detail analysis via MaAsLin2 (Microbiome Multivariable Association with Linear Models2) from MaAsLin2 package of R was conducted to identify microbiome taxa predictive of OA and KL groups while controlled for village, age, and genders.

For alpha diversity, Wilcoxon rank-sum test [63] was performed to compare groups statistically in the cross-sectional study (i.e., helminth-infected vs Uninfected and OA villages versus the KL), whereas Wilcoxon signed-rank test was used for paired samples in longitudinal study (i.e., pre vs post for both responders and uninfected). We also conducted a linear mixed model to examine the impact of helminth on alpha diversity while controlling the village location [64].

As for beta diversity, the comparison on pairwise distance of the samples between OA villages and KL was conducted using Wilcoxon rank-sum test. This same analysis was also applied to the output generated from k-mers sketches. Permutational multivariate analysis of variance (PERMANOVA) under ADONIS function [65] from the vegan package was conducted with 10,000 permutations on both the Jaccard and Bray-Curtis dissimilarity matrix. This analysis was first performed on specific variables of interest (i.e., different geographical locations, helminth status, and pre vs post). ADONIS was used to assess the effect of multiple variables on the gut microbial composition (e.g., if they had probiotic food, diarrhea, and antibiotics drug in the past 3 months, different age groups, subtribes, and protozoa infections), as well as analysis of similarity (ANOSIM) [66]. To test for multivariate dispersions among groups, the permutation multivariate analysis of dispersion (PERMDISP) [65] was performed via the betadisper function and Tukey’s test under the vegan package [67].

The MDMR was used to specifically test for “statistical interactions” between helminth status and village location and to calculate the relative effect sizes on microbiome variation [27]. Differential abundance analysis was performed using MaAsLin2 [26] for both cross-sectional (i.e., helminth positive and negative individuals while including village as a covariate) and longitudinal data (i.e., between host and drug response while controlling for village as well as between responders and uninfected while controlling for treatment and helminthic infections). Analysis of composition microbiomes with bias correction (ANCOM-BC) [68] was also conducted to validate the output generated from MaAsLin2, for cross-sectional data only.

For GRiD analysis, Spearman’s rank correlation test was conducted to examine the association between the bacterial species growth rate with Trichuris infection intensity for cross-sectional study. Then, the results were corrected using Benjamini-Hochberg with a false discovery rate (FDR) of 5% [69]. As for the longitudinal study, comparison of bacterial species growth rate between pre and post treatment of both responders and uninfected was computed using Wilcoxon rank-sum test and corrected using Benjamini-Hochberg with a FDR of 5%. A linear mixed model was built via the lmertest package [64] to determine the impact of the Trichuris intensity on the specific microbial growth rate by controlling for the village location for both the longitudinal and cross-sectional studies. Besides, multivariate association with linear models 2 (MaAsLin2) [26] was used to determine the microbial growth rate with differentially abundance in helminthic infections while controlling for village location and treatments.

Overall, the Wilcoxon rank-sum test and Wilcoxon signed-rank test from the rstatix package [48] were used to determine the p-value between groups for specific taxa in cross-sectional and longitudinal study respectively.

Key resources table

List of the bioinformatic tools and R packages used is displayed in the table below:

Tasks/analysis

Name

Source

Identifier

Bioinformatic tools

   

Quality control, filtering, and trimming of raw sequence

KneadData

No publication

https://github.com/biobakery/kneaddata

Trimmomatic

Bolger et al., 2014 [70]

https://github.com/biobakery/kneaddata

Bowtie2

Langmead et al., 2012 [43]

https://github.com/biobakery/kneaddata

FastQC

Andrew, 2017 [44]

https://www.bioinformatics.babraham.ac.uk/projects/fastqc/

Taxonomy assignment

Kraken2

Wood et al., 2019 [45]

https://github.com/DerrickWood/kraken2/wiki#downloads

Estimate relative abundance of species or genera

Bracken2

Lu et al., 2017 [46]

https://github.com/jenniferlu717/Bracken

Compute hash sketches from DNA sequence

Sourmash

Brown et al., 2016 [51]

https://sourmash.readthedocs.io/en/latest/

Functional analysis

bioBakery 3

Beghini et al., 2021 [56]

https://github.com/biobakery/biobakery/wiki

Replication of bacterial species

GRiD

Emiola et al., 2018 [57]

https://github.com/ohlab/GRiD

Pathoscope 2.0

Hong et al., 2014 [71]

https://github.com/PathoScope/PathoScope

R packages

   

Data import, filtering, and processing

phyloseq

McMurdie et al., 2013 [49]

https://joey711.github.io/phyloseq/index.html

Core microbiota analysis

DMwR

Amunategui, 2014 [58]

http://amunategui.github.io/smote/

 

randomForest

Breiman et al., 2018 [72]

https://cran.r-project.org/web/packages/randomForest/index.html

Alpha diversity

phyloseq

McMurdie et al., 2013 [49]

https://joey711.github.io/phyloseq/index.html

microbiomeSeq

Ssekagiri et al., 2017 [55]

https://github.com/umerijaz/microbiomeSeq

rstatix

Kassambara, 2021 [48]

https://cran.r-project.org/web/packages/rstatix/index.html

Beta diversity

phyloseq

McMurdie et al., 2013 [49]

https://joey711.github.io/phyloseq/index.html

Differential abundance analysis

MaAsLin2

Malick et al., 2021 [26]

https://huttenhower.sph.harvard.edu/maaslin/

ANCOMBC

Lin et al., 2020 [68]

http://www.bioconductor.org/packages/release/bioc/vignettes/ANCOMBC/inst/doc/ANCOMBC.html

ADONIS and ANOSIM

vegan

Oksanen et al., 2020 [67]

https://cran.r-project.org/web/packages/vegan/index.html

Interaction between covariates

MDMR

McArtor, 2018 [27].

https://cran.r-project.org/web/packages/MDMR/index.html

lmerTest

Kuznetsova et al., 2020 [64]

https://cran.r-project.org/web/packages/lmerTest/index.html

Correlation test

psych

Revelle, 2022 [69]

https://cran.r-project.org/web/packages/psych/index.html