Skip to main content

Network analysis of gut microbial communities reveal key genera for a multiple sclerosis cohort with Mycobacterium avium subspecies paratuberculosis infection

Abstract

Background

In gut ecosystems, there is a complex interplay of biotic and abiotic interactions that decide the overall fitness of an individual. Divulging the microbe-microbe and microbe-host interactions may lead to better strategies in disease management, as microbes rarely act in isolation. Network inference for microbial communities is often a challenging task limited by both analytical assumptions as well as experimental approaches. Even after the network topologies are obtained, identification of important nodes within the context of underlying disease aetiology remains a convoluted task. We therefore present a network perspective on complex interactions in gut microbial profiles of individuals who have multiple sclerosis with and without Mycobacterium avium subspecies paratuberculosis (MAP) infection. Our exposé is guided by recent advancements in network-wide statistical measures that identify the keystone nodes. We have utilised several centrality measures, including a recently published metric, Integrated View of Influence (IVI), that is robust against biases.

Results

The ecological networks were generated on microbial abundance data (n = 69 samples) utilising 16 S rRNA amplification. Using SPIEC-EASI, a sparse inverse covariance estimation approach, we have obtained networks separately for MAP positive (+), MAP negative (-) and healthy controls (as a baseline). Using IVI metric, we identified top 20 keystone nodes and regressed them against covariates of interest using a generalised linear latent variable model. Our analyses suggest Eisenbergiella to be of pivotal importance in MS irrespective of MAP infection. For MAP + cohort, Pyarmidobacter, and Peptoclostridium were predominately the most influential genera, also hinting at an infection model similar to those observed in Inflammatory Bowel Diseases (IBDs). In MAP- cohort, on the other hand, Coprostanoligenes group was the most influential genera that reduces cholesterol and supports the intestinal barrier.

Conclusions

The identification of keystone nodes, their co-occurrences, and associations with the exposome (meta data) advances our understanding of biological interactions through which MAP infection shapes the microbiome in MS individuals, suggesting the link to the inflammatory process of IBDs. The associations presented in this study may lead to development of improved diagnostics and effective vaccines for the management of the disease.

Background

Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disorder [1] that affects brain and spinal cord impacting around 2.5 million people worldwide [2]. The origin of demyelination and inflammation is not clear yet however interplay between environmental and genetic factors are known to develop MS [3, 4]. Growing literature on this topic highlights the role of gut microbiota as a strong environmental influencer in the MS context [5]. It is proposed that perturbations in the gut microbiota could stimulate proinflammatory responses that serve as additional mechanism in the pathogenesis of MS [6, 7]. On the other hand, several studies have linked Mycobacterium avium subspecies paratuberculosis (MAP) infections with MS [8, 9]. MAP is a versatile intracellular parasite that colonizes intraepithelial macrophages in the mucosa-associated lymphoid tissue of the small intestine. It can induce chronic granulomatous gastroenteritis, known as John’s disease or paratuberculosis, in animals, especially ruminants [10]. Various molecular and serological tests have reported the presence of MAP in the blood of individuals with multifactorial diseases, including type 1 diabetes (T1D) [11, 12], Crohn’s disease (CD) [13], multiple sclerosis (MS) [8, 9] and Parkinson’s disease (PD) [14]. Molecular mimicry is known to be one of the potential mechanisms by which MAP triggers autoimmune diseases due to the structural similarity of MAP antigens to self-antigens [15]. Despite the extensive research conducted on the gut microbiota and MS there is hardly any study that identified microbes or their functions linked to MS specially when infected by MAP.

In a complex microbial ecosystem, species rarely act alone. Either they strive for resources following a competitive exclusion principle with one species outcompeting another, or they live in symbiosis, or there are predator-prey interactions. Recovery of both biotic and abiotic interactions then leads to understanding how stable the ecosystem is [16], with the network of interactions able to reveal the important species functioning in the ecosystem. Typically, in microbial networks, through in situ analytical approaches, highly interacting nodes called hubs are identified [17]. In a previous study, identified hubs or keystone nodes were then later confirmed experimentally [18], and were deemed to be important. Furthermore, these inferential network-based approaches have found use in clinical applications. For example, in [19], highly connected hub species associated better with the clinical changes (as compared to highly abundant and prevalent species) in cystic fibrosis patients with chronic lung infections.

In general, once a network topology is obtained, network-wide statistical measures such as centralities are calculated which, ascertain how central a node is within the expanse of a network. For example, Degree Centrality, Cluster Rank, Betweenness, Collective Influence, Network Neighborhood, and Local H-index are some centrality measures that can quantify the importance of nodes. Whilst each one of these on their own can serve to highlight a particular nuance, a more sophisticated approach that can simultaneously consider a set of centrality measures covering local and global features of network can offer better synergy by reducing biases inherent with some of the measures. For this purpose, a more sophisticated network-wide statistical measure called Integrated Value of Influence (IVI) [20] is proposed that uses six important centrality measures (as above) as building blocks to derive Hubness and Spreading scores eventually combining them to a single IVI measure. This single measure then enables recovery of the most important topological characteristics of the network identifying keystone nodes that have biological relevance with the measure robust against biases. The aim of this paper is to then to consolidate these recent advancements in network statistics to identify keystone nodes in multiple sclerosis patients with and without MAP infection. We then associate these keystone nodes with the anthropometric and sociodemographic information. For association, we utilize the Generalized Linear Latent Variable Model (GLLVM) [21] approach where the abundance of individual microbes is regressed against covariates of interest by also incorporating a small number of latent variables. The fitted beta coefficients through the GLLVM approach then gives directionality (positive or negative association) against the covariates of interest consolidating the clinical or environmental context under which the data is generated. However, fitting GLLVM against sources of variability when there are thousands of taxa, significantly more than the number of samples, is computationally challenging and impractical for larger datasets. IVI leverages this by ranking taxa in terms of their influence, thus allowing exploration of the top most influential nodes, enabling better convergence of the likelihood in the reduced space of the variables.

Methods

Bioinformatics

Our previous study [22] provides a comprehensive overview of the study design, stool sampling, their processing, and bioinformatic analysis. In brief, this study involved comparative analysis of three distinct study groups; MS patients who tested positive for MAP infection (MAP+), MS patients who tested negative for MAP infection (MAP-) and a control group consisting of healthy individuals. Each participant provided two samples, labelled as T1 and T2 typically collected a month apart. A total of 97 individuals were screened for participation in this study at the Multiple Sclerosis Center of the University of Cagliari, Italy. The collected samples underwent 16 S rRNA amplicon sequencing using V3-V4 primer set on an Illumina MiSeq instrument. Out of the 97 stool samples collected from the participants, only 74 had provided a sufficient DNA yield for microbiome analyses. An additional 5 samples were excluded due to low read numbers (< 5000 reads), resulting in a total of 69 samples included in final analyses.

Network inference

We have used an OTU table of n = 69 x P = 16,787 OTUs (see [22]) where VSEARCH pipeline [23] was used to construct OTUs at 99% similarity threshold. The summary statistics of reads mapping to these OTUs for samples as follows: [Minimum: 5,074; 1st Quartile: 14,380; Median: 18,060; Mean: 20,634; 3rd Quartile: 22,651; Maximum: 96,572]. After obtaining the taxonomy of OTUs using SILVA SSU Ref NR database release v.138 [24], the abundances of OTUs belonging to the same genus were collated together giving an n = 69 x P = 128 dimensional genera table. Note that where the OTUs were not resolved at genus level, they were put in the “__Unknown__” category. From the 69 samples, we inferred the network separately for Healthy Control (n = 24), MAP+ (n = 27), and MAP- (n = 18) cohorts. We have used the SPIEC-EASI [25] approach using the standard parameters in the function spiec.easi(abundance_table, method=’mb’, lambda.min.ratio = 1e-2, nlambda = 20, pulsar.params = list(rep.num = 50)), where abundance_table is the table extracted separately for Healthy Control (HC), MAP + and MAP- individuals.

Network wide statistics

Having obtained the network topology for all three cohorts (Healthy Control, MAP+, and MAP- MS patients), we have calculated several network wide statistics using the R packages igraph [26], influential [20], and centiserve [27]. We have used the statistics given in Supplementary Table S1, with comparative analyses of these statistics for different cohort given in Supplementary Figure S1 (Supplementary_Materials.docx).

Generalised Linear Latent Variable Model (GLLVM)

To find the relationship between most influential nodes [top 20 selected based on Integrated View of Influence (IVI) metric] and the sources of variation (Sex, Age, BMI, Time Points, Weight Change, Having Children, Having Pets, Smoker, Work Routine, Sports, Leisure Time, Sleep Duration, Antibiotics, Sweet Consumption, Drinking Water, Alcohol Consumption, Probiotics Consumption, Stool Consistency, and Disease Duration [only available for MAP+, and MAP- cohort]), we have used Generalised Linear Latent Variable Model (GLLVM) [21] which extends the basic generalized linear model that regresses the mean abundances \({\mu }_{ij}\) (for \(i\)-th sample and \(j\)-th microbe) of individual microbes against environmental covariates \({x}_{i}\) as above by incorporating latent variables (confounders) \({u}_{i}\) as \(g\left({\mu }_{ij}\right)={\eta }_{ij}={\alpha }_{i}+{\beta }_{0j}+{\varvec{x}}_{i}^{T}{\varvec{\beta }}_{j}+{\varvec{u}}_{i}^{T}{\varvec{\theta }}_{j}\), where \({\varvec{\beta }}_{j}\) are the microbe specific coefficients associated with individual covariate. A 95% confidence interval of \({\varvec{\beta }}_{j}\) whether positive (increasing the abundance of microbe) or negative (decreasing the abundance of microbe), and not crossing 0 boundary gives directionality with respect to a particular covariate. \({\varvec{\theta }}_{j}\) are the corresponding coefficients associated with latent variable. \({\beta }_{0j}\) are microbes’ specific intercepts, whilst \({\alpha }_{i}\) are optional sample effects which can either be chosen as fixed effects or random effects (not used in this study). To model the distribution of individual microbes, we have used Negative Binomial distribution. Additionally, the approximation to the log-likelihood is done through Variational Approximation (VA) with final sets of parameters in glvmm() function being family = ‘negative.binomial’, method="VA”, control.start=list(n.init = 7, jitter.var = 0.1) that converged the optimization algorithm associated with GLLVM for HC, MAP+ and MAP- cohort.

Results

Recovered keystone nodes vary between HC, MAP + and MAP- cohort

The networks of genera inferred for HC, MAP + and MAP- cohorts are shown in Figs. 1, 2 and 3 along with the statistics for top influential nodes. Although we have used several network wide metrics, we mainly emphasized on three strategies for the identification of key stone genera: (a) Spreading Score: which itself a combination of four metrics i.e., Neighbourhood connectivity (NC), Cluster rank (CR), Betweenness centrality (BC) and Collective influence (CI) that are used to identify the potential genera having higher spreading potential within the microbiome network. (b) Hubness Score: It is used to identify genera that have high centrality measures [(Degree centrality (DCi) and Local H-index (LHindex)] and (c) Integrated View of Influence (IVI): It integrates both spreading and hubness score as a single metric and identifies microbial network most influential nodes.

Fig. 1
figure 1

Network inferred for HC samples using SPIEC-EASY algorithm for OTUs collated at genus level. The size of the nodes in the network corresponds to Integrated View of Influence scores whilst the nodes are colored at Phylum level. Left panel shows the top 20 important nodes along with their scores based on composite measure Integrated View of Influence, along with the Spreading Score and the Hubness Score. To improve clarity, bars of nodes that were not in top 20 list for a given metric were not drawn. These influential genera (n = 25) are then annotated on the network figure shown in the middle panel

Fig. 2
figure 2

Network inferred for MAP + samples using SPIEC-EASY algorithm for OTUs collated at genus level. The size of the nodes in the network corresponds to Integrated View of Influence scores whilst the nodes are colored at Phylum level. Left panel shows the top 20 important nodes along with their scores based on composite measure Integrated View of Influence, along with the Spreading Score and the Hubness Score. To improve clarity, bars of nodes that were not in top 20 list for a given metric were not drawn. These influential genera (n = 26) are then annotated on the network figure shown in the middle panel

Fig. 3
figure 3

Network inferred for MAP- samples using SPIEC-EASY algorithm for OTUs collated at genus level. The size of the nodes in the network corresponds to Integrated View of Influence scores whilst the nodes are colored at Phylum level. Left panel shows the top 20 important nodes along with their scores based on composite measure Integrated View of Influence, along with the Spreading Score and the Hubness Score. To improve clarity, bars of nodes that were not in top 20 list for a given metric were not drawn. These 26 influential genera (after combining the results from all metrics) are then annotated on the network figure shown in the middle panel

We have taken a union of the top 20 nodes selected for either of the three metrics, Spreading Score, Hubness Score, and IVI. These are shown in the left panels of Figs. 1, 2 and 3 and selected 25, 26, and 26 genera for HC, MAP+, and MAP- cohorts, respectively. Majority of the influential nodes belonged to the phylum Firmicutes (21 for HC, 18 for MAP+, and 15 for MAP-) with the most influential nodes according to IVI being: Flavonifractor (HC); Pyramidobacter (MAP+); and [Eubacterium]_coprostanoligenes_group (MAP-). Of all the influential nodes, Eggerthella and [Ruminococcus]_gnavus_group were selected for all three cohorts. Amongst other common nodes: GCA_900066575 (Lachnospiraceae) and Ruminococcus are common between HC and MAP+; Erysipelatoclostridium, UCG_010 (Oscillospirales), Megamonas, and Succinovibrio are common between HC and MAP-; and Eisenbergiella and Oscillibacter are common between MAP + and MAP-.

Supplementary Data Table S1 contains all interactions recovered for each cohort where, genera (top 5 for each cohort) that achieved very high IVI scores are highlighted along with their interacting secondary connections. These come out to be 14 unique genera with Eggerthella common between MAP + and HC cohort. Secondary connections of the top 5 IVI nodes that are common between MAP + and MAP- are Aldercreutzia, [Ruminococcus]_gnavus_group, Oscillobacter, and Acidaminococcus. Secondary connections of the top 5 IVI nodes had a high degree of overlap between HC and MAP+, and these include Flavonifractor, [Clostridium]_innocuum_group, Eisenbergiella, Colidextribacter, and Ruminococcus.

Top 20 influential keystone nodes based on IVI and their relationship with the clinical parameters and the exposome

We then employed a GLLVM to regress the top 20 most influential genera against different sources of variation. These associations are shown in Figs. 4 and 5, and 6. The covariates include Sex, Age, BMI, Time Points, Weight Change, Having Children, Having Pets, Smoker, Work Routine, Sports, Leisure Time, Sleep Duration, Antibiotics, Sweet Consumption, Drinking Water, Alcohol Consumption, Probiotics Consumption, Stool Consistency, and Disease Duration, with the information provided by the subjects at the time of sample collection.

Fig. 4
figure 4

𝜷− coefficients returned from GLLVM procedure for intrinsic covariates considered in this study, and the top 20 influential nodes returned for HC samples in Fig. 1 using IVI metric, with the complete results including extrinsic parameters shown in Supplementary Figure S2. Those coefficients which are positively associated with the microbial abundance of a particular genera are represented in red color whilst those that are negatively associated are represented with blue color, respectively. Non-significant associations, if any, are represented with the black color. For categorical variables, one level acts as a reference and is annotated with REF. Genera also found influential for MAP + and MAP- cohort are represented with red color, whilst those found in either of MAP- and MAP + cohort are represented with green and blue colors, respectively

Fig. 5
figure 5

𝜷− coefficients returned from GLLVM procedure for intrinsic covariates considered in this study, and for the top 20 most influential nodes returned for MAP + samples in Fig. 2 using IVI metric, with the complete results including extrinsic parameters shown in Supplementary Figure S3. Those coefficients which are positively associated with the microbial abundance of a particular genera are represented in red color whilst those that are negatively associated are represented with blue color, respectively. Non-significant associations, if any, are represented with the black color. For categorical variables, one level acts as a reference and is annotated with REF. Genera also found influential for HC and MAP- cohort are represented with red color, whilst those found in either of MAP- and HC cohort are represented with green and blue colors, respectively

Fig. 6
figure 6

𝜷− coefficients returned from GLLVM procedure for intrinsic covariates considered in this study, and for the top 20 most influential nodes returned for MAP- samples in Fig. 3 using IVI metric, with the complete results including extrinsic parameters shown in Supplementary Figure S4. Those coefficients which are positively associated with the microbial abundance of a particular genera are represented in red color whilst those that are negatively associated are represented with blue color, respectively. Non-significant associations, if any, are represented with the black color. For categorical variables, one level acts as a reference and is is annotated with REF. Genera also found influential for HC and MAP + cohort are represented with red color, whilst those found in either of HC and MAP + cohort are represented with blue color

There are some keystone nodes that are common across all cohorts. These include Ruminococcus genera in general, and also include [Ruminococcus]_gnavus_group and Erysipelatoclostridium. Also, Eisenbergiella is the only genera which is influential in MS cohort, irrespective of MAP infection. For Eisenbergiella, using GLLVM, some of the covariates differ in their association between MAP + and MAP- cohort. These include Age, Probiotics consumptions (> 1 year,3–6 months), Sex(male), Sleep duration, Smoker, Stool consistency, Weight change (Loss), and Work routine (sitting) which are all positively associated with Eisenbergiella in MAP- individuals whilst they are negatively associated with Eisenbergiella in MAP + individuals. Also, drinking water > 2 L, Leisure time(normal), probiotics consumption(currently), sweet consumption 5–6 per week, and work routine (sedentary, standing) are negatively associated with Eisenbergiella for MAP- individuals and positively associated with Eisenbergiella for MAP + individuals.

To identify the important genera in the networks associated with MAP+, MAP- and HCs, we have employed network-based statistics based on centrality, connectivity and hubness scores/strategies. Supplementary Figure S1 shows comparison of network-wide statistics among all the study cohorts. The statistics revealed that MAP- individuals have the highest IVI scores than MAP + and HCs. The trend is same in terms of Hubness score, Local H-index, Laplacian centrality, Spreading score and Geodesic K-path centrality. However, for the MAP + group, Neighborhood connectivity, Lin centrality and Latora closeness is highest as compared to the other cohorts. These measures suggest that in MAP + networks, microbial genera are closer, central and have more connections to neighborhood genera as compared to the MAP- and HC cohorts.

Discussion

The human microbiome exhibits significant diversity in its composition, interconnections, and resilience both within and across individuals. Based on the interconnectedness of microbes, our aim is to identify keystone microbes associated with diifferent clinical parameters. The “keystone” concept has its roots in microbial ecology that designate a species with a significant role on community relative to its abundance. The concept has been extended to microbial abundance data where certain microbial species may play a crucial role in shaping the community structure and function. Although these species are few and far between, they have a markedly increased influence. Rahman and Schomberg et al., [28] elaborates on this further and utilized this approach in identifying important enzymes in microbial networks. Furthermore, their results highlighted high “between centrality” values relative to node degree as a means to identify “choke points” that play a significant role in carrying out fundamental metabolic conversions in bacteria and act as central hubs in metabolic networks influencing various interaction and pathways.

Using IVI, a composite approach built from several centrality measures, we identified the top 3 most influential nodes for MAP + cohort which are Pyramidobacter, Peptoclostridium, and Eggerthella. Eggerthella, a microbe associated with cysteine degradation [29] has been associated with multiple sclerosis although its causal role is not fully established yet [30]. The genus Pyramidobacter comprises strains that are anaerobic, non-motile, asaccharolytic bacilli producing acetic and isovaleric acids along with small quantities of propionic and isobutyric acids [31]. It is thought to enhance fiber degradation and may depend on thiamine to grow [32]. Though the direct link of Pyramidobacter with MS remains unclear, it was previously associated with 17-Hamilton Depression Rating Scale (HAMD-17) assessment [33].

The genera that are typically affected by thiamine supplementation including those that were found important for MAP + cohort include Erysipelotrichaceae, Lachnospiraceae, Selenomonas, Pyramidobacter, Christensenellaceae R7 and, Ruminococcaceae NK4A214. Pyramidobacter species are vital cellulolytic bacteria and produce acetate as the main fermentation product [34], the enhanced Pyramidobacter by thiamine could support the fiber degradation. Our findings of acetate producers implicated in MAP + cohort is in line with literature [35] where based on metabolomics, higher concentration of acetate is observed for MAP infection predominantly in males. It is worth noting that Peptoclostridium, a causative agent of diarrhea and colitis [36] adds another dimension to the complex interplay of microbial species in the gut. Its potential implications in MS individuals with MAP infection further adds credence to the association of Multiple Sclerosis (MS) with Inflammatory Bowel Disease (IBD) [37, 38] particularly Crohn’s disease and Ulcerative colitis. Additionally, genome wide association studies have revealed a shared risk locus between IBD and MS indicating a common underlying pathological mechanism affecting both conditions [39]. Therefore, one would expect the microbial signature between MS and IBD to be similar. In individuals without MAP infection, there is abundance of Eubacterium coprostanoligenes, Megamonas Ruminococcus gnavus and, Alistipes. Eubacterium coprostanoligenes is known for its capability of transforming cholesterol to coprostanol [40]. It plays a major role in stimulating lactic acid metabolism toward the production of SCFAs, that support the intestinal barrier [41]. This process also leads to secondary bile acids secretions which play a role in the balance between health and disease particularly in association with inflammatory bowel disease [42]. Megamonas found to be increased in MS [43], which is also implicated in Amyotrophic lateral sclerosis (ALS) [44]. Alistipes are differentially abundant in RRMS (Relapse Remitting Multiple Sclerosis) [45] that signifies its potential in immune related responses. Ruminococcus gnavus is known to produce a polysaccharide that induces TNFα production emphasizing its role in immune modulation [46].

In our study, we have also identified interaction within each cohort focusing on those genera with very high IVI scores. In MAP + cohort, a noticeable cooccurrence relationship occurs between the genera Murdochiella and Pyramidobacter. This observation is in line with a study conducted by Caudet et al. [47], which confirmed the higher relative abundance of these two species in a similar context. Similarly, Eubacterium eligens and Intestinimonas are identified as cooccurring species. Both are regarded as butyriciproducens that produce SCFA, especially butyric acid [48]. Interestingly, these species were stimulated in fermentations from patients with IBD [49]. Faecalibacterium and Intestinimonas has been considered as potential probiotics for treating and alleviating inflammatory bowel disease [50], although Intestinimonas could potentially increase in abundance in animal models of inflammation [51]. Nonetheless, it’s important to consider potential complexities, as indicated by a study that found an inverse relationship between Rikenellaceae and Pyramidobacter [52]. In MAP- individuals, Bifidobacterium and Alistipes were identified as cooccurring species, with this behavior also observed in individuals with Parkinson’s Disease [53]. The overlap of our observed co-occurrence patterns with other neurological conditions gives credence to network-based approaches to understanding disease etiology.

Furthermore, we have also implemented GLLVM regression model to investigate the association of the keystone nodes with the covariates of interest. Notably, Eisenbergiella, a gram negative, non-motile, non-spore producing bacteria demonstrated distinct association with individuals characterized as MAP + and MAP- considering various covariates of interest. These covariates include Age, Probiotics consumptions (> 1 year,3–6 months), Sex(male), Sleep duration, Smoker, Stool consistency, Weight change(Loss), and Work routine (sitting), Drinking water > 2 L, Leisure time(normal), probiotics consumption(currently), sweet consumption 5–6 per week, and work routine (sedentary, standing). Our findings suggests that modulation of Eisebergiella abundance as a biocontrol agent can potentially be useful in clinical settings.

In conclusion, our research explores the complex microbial landscape for microbe-microbe interactions within individuals affected by multiple sclerosis (MS), with a specific focus on those with Mycobacterium avium subspecies paratuberculosis (MAP) infection. Incorporating genera identified through the IVI statistic led to interesting associations for both MAP + and MAP- groups. Particularly, Pyramidobacter, Peptoclostridium, and Eggerthella in the MAP + cohort, and their potential metabolic nuances may pave the way for development of therapeutic agents. The downstream GLLVM regression model further elucidated the association of Eisenbergiella with various covariates of interests, suggesting a potential link between its abundance and factors i.e., age, probiotic consumption, and lifestyle. However, our conclusions are drawn based on a limited sample size. Further research work including a larger cohort, with temporal sampling will unravel further associations between microbes that may have been missed in this study, and will lead to development of microbial modulation strategies that might have beneficial effects.

Data availability

Sequence data that support the findings of this study have been deposited in the European Nucleotide Archive with the primary accession code PRJEB67783.

Abbreviations

16SrRNA:

16 S ribosomal RNA

MS:

Multiple sclerosis

PD:

Parkinson’s disease

MAP:

Mycobacterium avium subspecies paratuberculosis

OTUs:

Operational Taxonomic Units

T1D:

Type 1 diabetes

CD:

Crohn’s disease

IVI:

Integrated Value of Influence

GLLVM:

Generalised Linear Latent Variable Model

NC:

Neighbourhood connectivity

CR:

Cluster rank

BC:

Between centrality

DCi :

Degree centrality

LHindex :

Local H-index

CI:

Collective influence

References

  1. Thompson AJ, Baranzini SE, Geurts J, Hemmer B, Ciccarelli O. Multiple sclerosis. Lancet. 2018;391(10130):1622–36.

    Article  PubMed  Google Scholar 

  2. Tullman MJ. Overview of the epidemiology, diagnosis, and disease progression associated with multiple sclerosis. Am J Manag Care. 2013;19(2 Suppl):S15–20.

    PubMed  Google Scholar 

  3. Goodin DS, Khankhanian P, Gourraud P-A, Vince N. The nature of genetic and environmental susceptibility to multiple sclerosis. PLoS ONE. 2021;16(3):e0246157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Marrie RA. Environmental risk factors in multiple sclerosis aetiology. Lancet Neurol. 2004;3(12):709–18.

    Article  PubMed  Google Scholar 

  5. Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Paz Soldan MM, et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci Rep. 2016;6(1):28484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Durack J, Lynch SV. The gut microbiome: relationships with disease and opportunities for therapy. J Exp Med. 2019;216(1):20–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kadowaki A, Quintana FJ. The gut–CNS axis in multiple sclerosis. Trends Neurosci. 2020;43(8):622–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ekundayo TC, Olasehinde TA, Falade AO, Adewoyin MA, Iwu CD, Igere BE, Ijabadeniyi OA. Systematic review and meta-analysis of Mycobacterium avium subsp. paratuberculosis as environmental trigger of multiple sclerosis. Multiple Scler Relat Disorders. 2022;59:103671.

    Article  CAS  Google Scholar 

  9. Hayashi F, Isobe N, Cossu D, Yokoyama K, Sakoda A, Matsushita T, et al. Elevated mycobacterium avium subsp. paratuberculosis (MAP) antibody titer in Japanese multiple sclerosis. J Neuroimmunol. 2021;360:577701.

    Article  CAS  PubMed  Google Scholar 

  10. Eslami M, Shafiei M, Ghasemian A, Valizadeh S, Al-Marzoqi AH, Shokouhi Mostafavi SK, et al. Mycobacterium avium paratuberculosis and Mycobacterium avium complex and related subspecies as causative agents of zoonotic and occupational diseases. J Cell Physiol. 2019;234(8):12415–21.

    Article  CAS  PubMed  Google Scholar 

  11. Sechi LA, Paccagnini D, Salza S, Pacifico A, Ahmed N, Zanetti S. Mycobacterium avium subspecies paratuberculosis bacteremia in type 1 diabetes mellitus: an infectious trigger? Clin Infect Dis. 2008;46(1):148–9.

    Article  PubMed  Google Scholar 

  12. Manca Bitti ML, Masala S, Capasso F, Rapini N, Piccinini S, Angelini F et al. Mycobacterium avium subsp. paratuberculosis in an Italian cohort of type 1 diabetes pediatric patients. Journal of Immunology Research. 2012;2012.

  13. Naser SA, Ghobrial G, Romero C, Valentine JF. Culture of Mycobacterium avium subspecies paratuberculosis from the blood of patients with Crohn’s disease. Lancet. 2004;364(9439):1039–44.

    Article  PubMed  Google Scholar 

  14. Arru G, Caggiu E, Paulus K, Sechi GP, Mameli G, Sechi LA. Is there a role for Mycobacterium avium subspecies paratuberculosis in Parkinson’s disease? J Neuroimmunol. 2016;293:86–90.

    Article  CAS  PubMed  Google Scholar 

  15. Cossu D, Cocco E, Paccagnini D, Masala S, Ahmed N, Frau J, et al. Association of Mycobacterium avium subsp. paratuberculosis with multiple sclerosis in sardinian patients. PLoS ONE. 2011;6(4):e18482.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Allesina S, Tang S. Stability criteria for complex ecosystems. Nature. 2012;483(7388):205–8.

    Article  CAS  PubMed  Google Scholar 

  17. Paine RT. Food web complexity and species diversity. Am Nat. 1966;100(910):65–75.

    Article  Google Scholar 

  18. Carlström CI, Field CM, Bortfeld-Miller M, Müller B, Sunagawa S, Vorholt JA. Synthetic microbiota reveal priority effects and keystone strains in the Arabidopsis phyllosphere. Nat Ecol Evol. 2019;3(10):1445–54.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Layeghifard M, Li H, Wang PW, Donaldson SL, Coburn B, Clark ST, et al. Microbiome networks and change-point analysis reveal key community changes associated with cystic fibrosis pulmonary exacerbations. Npj Biofilms Microbiomes. 2019;5(1):4.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Salavaty A, Ramialison M, Currie PD. Integrated value of influence: an integrative method for the identification of the most influential nodes within networks. Patterns. 2020;1(5).

  21. Niku J, Hui FK, Taskinen S, Warton DI. Gllvm: fast analysis of multivariate abundance data with generalized linear latent variable models in r. Methods Ecol Evol. 2019;10(12):2173–82.

    Article  Google Scholar 

  22. Ashraf H, Dikarlo P, Masia A, Zarbo I, Solla P, Ijaz U, Sechi L. Mycobacterium avium subspecies paratuberculosis (MAP) infection, and its impact on gut microbiome of individuals with multiple sclerosis. 2023.

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kurtz ZD, Müller CL, Miraldi ER, Littman DR, Blaser MJ, Bonneau RA. Sparse and compositionally robust inference of microbial ecological networks. PLoS Comput Biol. 2015;11(5):e1004226.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Csardi G, Nepusz T. The igraph software package for complex network research. InterJournal Complex Syst. 2006;1695(5):1–9.

    Google Scholar 

  27. Jalili M, Salehzadeh-Yazdi A, Asgari Y, Arab SS, Yaghmaie M, Ghavamzadeh A, Alimoghaddam K. CentiServer: a comprehensive resource, web-based application and R package for centrality analysis. PLoS ONE. 2015;10(11):e0143111.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Rahman SA, Schomburg D. Observing local and global properties of metabolic pathways:‘load points’ and ‘choke points’ in the metabolic networks. Bioinformatics. 2006;22(14):1767–74.

    Article  CAS  PubMed  Google Scholar 

  29. Braccia DJ, Jiang X, Pop M, Hall AB. The capacity to produce hydrogen sulfide (H2S) via cysteine degradation is ubiquitous in the human gut microbiome. Front Microbiol. 2021;12:705583.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Cekanaviciute E, Yoo BB, Runia TF, Debelius JW, Singh S, Nelson CA et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proceedings of the National Academy of Sciences. 2017;114(40):10713-8.

  31. Downes J, Vartoukian SR, Dewhirst FE, Izard J, Chen T, Yu W-H, et al. Pyramidobacter piscolens gen. nov., sp. nov., a member of the phylum ‘Synergistetes’ isolated from the human oral cavity. Int J Syst Evol MicroBiol. 2009;59(Pt 5):972.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Pan X, Xue F, Nan X, Tang Z, Wang K, Beckers Y, et al. Illumina sequencing approach to characterize thiamine metabolism related bacteria and the impacts of thiamine supplementation on ruminal microbiota in dairy cows fed high-grain diets. Front Microbiol. 2017;8:1818.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Yao S, Xie H, Wang Y, Shen N, Chen Q, Zhao Y, et al. Predictive microbial feature analysis in patients with depression after acute ischemic stroke. Front Aging Neurosci. 2023;15:1116065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bainbridge ML, Cersosimo LM, Wright A-DG, Kraft J. Rumen bacterial communities shift across a lactation in Holstein, Jersey and Holstein× Jersey dairy cows and correlate to rumen function, bacterial fatty acid composition and production parameters. FEMS Microbiol Ecol. 2016;92(5):fiw059.

    Article  PubMed  Google Scholar 

  35. Karunasena E, McMahon KW, Chang D, Brashears MM. Host responses to the pathogen Mycobacterium avium subsp. paratuberculosis and beneficial microbes exhibit host sex specificity. Appl Environ Microbiol. 2014;80(15):4481–90.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Pereira FL, Oliveira Júnior CA, Silva RO, Dorella FA, Carvalho AF, Almeida GM, et al. Complete genome sequence of Peptoclostridium difficile strain Z31. Gut Pathogens. 2016;8(1):1–7.

    Article  CAS  Google Scholar 

  37. Sadovnick A, Paty D, Yannakoulias G. Concurrence of multiple sclerosis and inflammatory bowel disease. N Engl J Med. 1989;321(11):762–3.

    Article  CAS  PubMed  Google Scholar 

  38. Gupta G, Gelfand JM, Lewis JD. Increased risk for demyelinating diseases in patients with inflammatory bowel disease. Gastroenterology. 2005;129(3):819–26.

    Article  PubMed  Google Scholar 

  39. Yang Y, Musco H, Simpson-Yap S, Zhu Z, Wang Y, Lin X, et al. Investigating the shared genetic architecture between multiple sclerosis and inflammatory bowel diseases. Nat Commun. 2021;12(1):5641.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes. 2020;12(1):1802866.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Petri RM, Neubauer V, Humer E, Kröger I, Reisinger N, Zebeli Q. Feed additives differentially impact the epimural microbiota and host epithelial gene expression of the bovine rumen fed diets rich in concentrates. Front Microbiol. 2020;11:119.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Tomova A, Bukovsky I, Rembert E, Yonas W, Alwarith J, Barnard ND, Kahleova H. The effects of vegetarian and vegan diets on gut microbiota. Front Nutr. 2019;6:47.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Sanchez JMS, DePaula-Silva AB, Libbey JE, Fujinami RS. Role of diet in regulating the gut microbiota and multiple sclerosis. Clin Immunol. 2022;235:108379.

    Article  CAS  PubMed  Google Scholar 

  44. Zeng Q, Shen J, Chen K, Zhou J, Liao Q, Lu K, et al. The alteration of gut microbiome and metabolism in amyotrophic lateral sclerosis patients. Sci Rep. 2020;10(1):12998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Reynders T, Devolder L, Valles-Colomer M, Van Remoortel A, Joossens M, De Keyser J, et al. Gut microbiome variation is associated to multiple sclerosis phenotypic subtypes. Ann Clin Transl Neurol. 2020;7(4):406–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Henke MT, Kenny DJ, Cassilly CD, Vlamakis H, Xavier RJ, Clardy J. Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide. Proceedings of the National Academy of Sciences. 2019;116(26):12672-7.

  47. Caudet J, Trelis M, Cifre S, Soriano JM, Rico H, Merino-Torres JF. Interplay between intestinal bacterial communities and unicellular parasites in a morbidly obese population: a neglected trinomial. Nutrients. 2022;14(15):3211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rosés C, Viadel B, Nieto JA, Soriano-Romaní L, Romo-Hualde A, Agudelo A et al. Gut microbiota modulatory capacity of Brassica oleracea italica x alboglabra (Bimi®). 2023.

  49. Calvete-Torre I, Sabater C, Antón MJ, Moreno FJ, Riestra S, Margolles A, Ruiz L. Prebiotic potential of apple pomace and pectins from different apple varieties: modulatory effects on key target commensal microbial populations. Food Hydrocolloids. 2022;133:107958.

    Article  CAS  Google Scholar 

  50. Li A, Ding J, Shen T, Han Z, Zhang J, Abadeen ZU, et al. Environmental hexavalent chromium exposure induces gut microbial dysbiosis in chickens. Ecotoxicol Environ Saf. 2021;227:112871.

    Article  CAS  PubMed  Google Scholar 

  51. Li W, Lu L, Liu B, Qin S. Effects of phycocyanin on pulmonary and gut microbiota in a radiation-induced pulmonary fibrosis model. Biomed Pharmacother. 2020;132:110826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ramos AF, Terry SA, Holman DB, Breves G, Pereira LG, Silva AG, Chaves AV. Tucumã oil shifted ruminal fermentation, reducing methane production and altering the microbiome but decreased substrate digestibility within a RUSITEC fed a mixed hay–concentrate diet. Front Microbiol. 2018;9:1647.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Li Z, Liang H, Hu Y, Lu L, Zheng C, Fan Y, et al. Gut bacterial profiles in Parkinson’s disease: a systematic review. CNS Neurosci Ther. 2023;29(1):140–57.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank the staff particularly Caterina Scodino at Department of Clinical, Surgery and Experimental Sciences, University of Sassari, Italy for helping with acquiring and handling the samples.

Funding

The authors acknowledge the following fundings. HA is supported by an EU funded scholarship (Programma Operativo Nazionale), University of Sassari financed by Region of Sardinia, and Erasmus + Training Mobility to University of Glasgow. UZI is supported by UKRI Engineering and Physical Sciences Research Council (EP/V030515/1). LAS is supported by the Regione Autonoma Sardegna grant: legge regionale 12 22 December 2022 n. 22 and PRIN 2022 n: 2022BP837R.

Author information

Authors and Affiliations

Authors

Contributions

H.A. (Conceptualization, Methodology, Validation, Formal Analysis, Investigation, Data Curation, Writing - Original Draft, Visualization)P.D. (Resources, Writing - Review & Editing)I.R.Z. (Resources, Writing - Review & Editing)P.S. (Resources, Writing - Review & Editing)U.Z.I. (Methodology, Software, Formal Analysis, Writing - Original Draft, Supervision, Visualization, Funding acquisition)L.A.S. (Conceptualization, Methodology, Resources, Writing - Review & Editing, Supervision, Project administration, Funding acquisition)

Corresponding authors

Correspondence to Umer Zeeshan Ijaz or Leonardo A. Sechi.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Ethics Review Board at University of Sassari at Azienda Sanitaria Locale (ASL) 1 (Prot.llo N°2150/CE, 17/02/15).

Consent for publication

All participants provided written informed consent to participate in the study and a self-reported detailed questionnaire recording participant’s medical history, dietary habits, sleeping pattern and routine lifestyle.

Competing interests

P.D. is employed by BIOMES NGS GmbH. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1.

Details on the network-wide statistics used including supplementary figures

Supplementary Material 2.

Co-occurrence network obtained at genera level for HC, MAP+, and MAP− cohorts. Furthermore, top 5 interacting genera are highlighted for each cohort

Supplementary Material 3.

Network-wide statistics for HC network

Supplementary Material 4.

Network-wide statistics for MAP+ network

Supplementary Material 5.

Network-wide statistics for MAP− network

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ashraf, H., Dikarlo, P., Masia, A. et al. Network analysis of gut microbial communities reveal key genera for a multiple sclerosis cohort with Mycobacterium avium subspecies paratuberculosis infection. Gut Pathog 16, 37 (2024). https://doi.org/10.1186/s13099-024-00627-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13099-024-00627-7

Keywords