Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Acknowledgements
Author’s response
Authors’ reply
Book Review
Book Reviews
Classics In Indian Medicine
Clinical Case Report
Clinical Case Reports
Clinical Research Methods
Clinico-pathological Conference
Clinicopathological Conference
Conferences
Correspondence
Corrigendum
Editorial
Eminent Indians in Medicine
Errata
Erratum
Everyday Practice
Film Review
History of Medicine
HOW TO DO IT
Images In Medicine
Indian Medical Institutions
Letter from Bristol
Letter from Chennai
Letter From Ganiyari
Letter from Glasgow
Letter from London
Letter from Mangalore
Letter From Mumbai
Letter From Nepal
Masala
Medical Education
Medical Ethics
Medicine and Society
News From Here And There
Notice of Retraction
Notices
Obituaries
Obituary
Original Article
Original Articles
Review Article
Selected Summaries
Selected Summary
Short Report
Short Reports
Speaking for Myself
Speaking for Ourselve
Speaking for Ourselves
Students@nmji
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Acknowledgements
Author’s response
Authors’ reply
Book Review
Book Reviews
Classics In Indian Medicine
Clinical Case Report
Clinical Case Reports
Clinical Research Methods
Clinico-pathological Conference
Clinicopathological Conference
Conferences
Correspondence
Corrigendum
Editorial
Eminent Indians in Medicine
Errata
Erratum
Everyday Practice
Film Review
History of Medicine
HOW TO DO IT
Images In Medicine
Indian Medical Institutions
Letter from Bristol
Letter from Chennai
Letter From Ganiyari
Letter from Glasgow
Letter from London
Letter from Mangalore
Letter From Mumbai
Letter From Nepal
Masala
Medical Education
Medical Ethics
Medicine and Society
News From Here And There
Notice of Retraction
Notices
Obituaries
Obituary
Original Article
Original Articles
Review Article
Selected Summaries
Selected Summary
Short Report
Short Reports
Speaking for Myself
Speaking for Ourselve
Speaking for Ourselves
Students@nmji
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Acknowledgements
Author’s response
Authors’ reply
Book Review
Book Reviews
Classics In Indian Medicine
Clinical Case Report
Clinical Case Reports
Clinical Research Methods
Clinico-pathological Conference
Clinicopathological Conference
Conferences
Correspondence
Corrigendum
Editorial
Eminent Indians in Medicine
Errata
Erratum
Everyday Practice
Film Review
History of Medicine
HOW TO DO IT
Images In Medicine
Indian Medical Institutions
Letter from Bristol
Letter from Chennai
Letter From Ganiyari
Letter from Glasgow
Letter from London
Letter from Mangalore
Letter From Mumbai
Letter From Nepal
Masala
Medical Education
Medical Ethics
Medicine and Society
News From Here And There
Notice of Retraction
Notices
Obituaries
Obituary
Original Article
Original Articles
Review Article
Selected Summaries
Selected Summary
Short Report
Short Reports
Speaking for Myself
Speaking for Ourselve
Speaking for Ourselves
Students@nmji
View/Download PDF

Translate this page into:

Editorial
38 (
5
); 257-263
doi:
10.25259/NMJI_1737_2025

‘Mining’ the Gut: The Microbiome ‘Lassonde curve’

Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

[To cite: Narang H, Ahuja V. ‘Mining’ the gut: The microbiome ‘Lassonde curve’. Natl Med J India 2025;38:257–63. DOI: 10.25259/NMJI_1737_2025]

The prevalent view is that the current excitement and progress in gut microbiome research are primarily a result of recent high-throughput sequencing breakthroughs. While it is true that technology provided the necessary acceleration for this revolution, it is quite possible that it merely acted as a trigger for a far more fundamental change. In the history of human progress, key foundational thinkers and major contributors are often overshadowed by those who follow, particularly those associated with later, more visible technological breakthroughs. A classic example of foundational, theoretical work being eclipsed by visible, practical application is the concept of the digital computer. Alan Turing provided the theoretical blueprint with the Turing machine, while John von Neumann formalised the stored-programme architecture. This architecture became the practical design for the first working digital computers, and in the process, von Neumann became the more celebrated scientist of his time.1

Initial exploration: The compass and its scalability

Similarly, while high-throughput sequencing is the technological engine of the current microbiome revolution, the compelling revolutionary concept was laid down by Carl Woese in 1977.2 His fundamental and game-changing contribution was the establishment of the Three-Domain System of life, which was based on the phylogenetic analysis of 16S ribosomal RNA (rRNA) gene sequences. The true conceptual breakthrough was his pioneering use of the 16S rRNA gene as a ‘molecular chronometer’ to determine evolutionary relationships. Woese’s work, which is often overshadowed by subsequent technological advances, laid the vital foundation upon which all modern molecular microbiology—and thus, the entire microbiome field—is built. In essence, Woese provided the map and the compass (the 16S rRNA taxonomy), which high-throughput sequencing later made it possible to read at scale and speed.

Microbiome discovery phase: Basic concepts

With the introduction of DNA sequencing technologies, commonly referred to as next-generation sequencing (NGS), microbiological advances have been in focus. From culturing and isolating single microbes, we have progressed to sequencing entire microbial genomes and identifying and characterizing unculturable microbes. This has led to the identification of distinct microbiomes in the oral cavity, respiratory tract, skin and the gastrointestinal (GI) tract, often collectively termed the ‘second human genome.’ These microbial cells in our bodies are estimated to outnumber our own human cells by 30% and contain 100 times more genes than the human genome, thus making us ‘more microbial than human’.35 Around 95% of these microbial cells reside in our gut, thus making the gut microbiome a key organ of the body.6

The gut microbiome in healthy adults is characterized by high diversity and richness, comprising an estimated 500 to 1000 bacterial species, predominantly of the phyla Firmicutes (60%), Bacteroidetes (20%), Actinobacteria (10%), Proteobacteria (5%) and Verrucomicrobia (<1%).7 Additionally, methanogenic archaea (primarily Methanobrevibacter smithii), fungi (Candida, Saccharomyces, Aspergillus) and viruses (bacteriophages, Adenoviruses, Noroviruses) are also present, though constituting only a minor fraction (<2%) by mass of the gut microbiota (Fig. 1). Factors affecting the gut microbiome composition include age, sex, genetics, lifestyle factors such as stress, exercise and sleep, diet, antibiotics and drug exposures, environmental factors and geographical location.810

Alterations in gut microbiota and metabolites across various stages of human life cycle SCFA short chain fatty acids
FIG 1.
Alterations in gut microbiota and metabolites across various stages of human life cycle SCFA short chain fatty acids

The gut microbiome of the Indian population shows considerable differences from the western population. While Prevotella and Megasphaera have been found predominant in the Indian population, the gut microbiome of the western population is dominated by Blautia and Fecalibacterium.1013 These differences are largely attributable to dietary patterns; Indian diets are high in carbohydrates and fibres, whereas western diets contain higher amounts of animal proteins and fats.14 Functionally, the Indian gut microbiome shows enrichment in pathways related to glycan biosynthesis, xenobiotic metabolism and branched-chain amino acid biosynthesis. In contrast, the western microbiomes are more associated with short-chain fatty acids (SCFAs) production and other metabolic pathways.15,16 Similarly, differences in the composition of gut microbiota have also been found between individuals residing in rural and urban areas at sea level and high altitude in India.10

The gut microbiome is involved in various physiological processes, including nutrient metabolism, maintaining gut barrier integrity, immunomodulation, and the gut–brain axis. The gut microbes metabolize dietary fibres, proteins and bile acids into SCFA, trimethylamine-N-oxide (TMAO) and secondary bile acids, respectively, which are involved in the regulation of metabolism, inflammation, cardiovascular and hepatic functions. SCFAs are also involved in the regulation of gut epithelial barrier integrity. The gut microbiome modulates the immune system via several mechanisms, including promotion of immune tolerance via microbial-associated molecular patterns (MAMPs); promoting regulatory T-cell (Treg) differentiation and maintaining Th17/Treg balance; production of immunomodulatory metabolites, including SCFAs; and exerting colonization resistance by stimulating production of antimicrobial peptides and secretory IgA.17

The gut-organ axes refer to the bidirectional communication networks between the gastrointestinal tract and other organs, including the brain, liver, lung, kidney and others (Fig. 2). The gut microbiome plays a central role in the functioning of these axes. These axes are mediated by microbial metabolites (e.g. SCFAs, bile acids, neurotransmitters), immune signalling, neural pathways (including the vagus nerve) and endocrine factors. The gut microbiota regulate host metabolism, immune responses and gut barrier integrity, thereby influencing the functions and disease susceptibility of extra-intestinal organs.18

Various gut-organ axes and diseases associated with gut microbiome dysbiosis
FIG 2.
Various gut-organ axes and diseases associated with gut microbiome dysbiosis

During the first three years of life, the gut microbiome undergoes structured changes that parallel and influence the development of metabolism, immunity and cognition.19 By 3 years of age, the gut microbiome achieves diversity and functional capacity resembling that of healthy adults.

Perturbation of the gut microbiome in disease

In adults, the microbiome is relatively stable and resilient, meaning it can return to its baseline state after perturbations like dietary changes or a short course of antibiotics.20 However, prolonged use of antibiotics and other drugs, immune dysfunction, environmental exposures and other factors lead to long-term alterations in the microbiome, referred to as dysbiosis. Dysbiosis has been associated with multiple diseases, including gastrointestinal disorders such as Clostridioides difficile infection (CDI), inflammatory bowel disease (IBD), and irritable bowel syndrome, as well as extra-intestinal conditions like metabolic syndrome, obesity, atherosclerosis, lung cancer, asthma, Parkinson’s disease and chronic kidney disease.2123 However, despite extensive data linking dysbiosis to various clinical diseases, a causal relationship is yet to be established in human studies. Dysbiosis can disrupt the gut-organ axes, contributing to the pathogenesis of conditions such as metabolic dysfunction-associated steatotic liver disease, neuropsychiatric disorders and dysfunction of other organs. For example, altered gut microbial metabolites and bile acid metabolism contribute to hepatic inflammation and steatosis; altered gut metabolites, endocrine signalling, and immune dysregulation can influence neuroinflammation and central nervous system homeostasis; and disruption of gut barrier integrity leads to increased exposure of microbial products to the pancreas, contributing to metabolic dysregulation and islet dysfunction.

The promise of microbiome manipulation therapies

In light of the microbiome–disease associations, even in the absence of causality, it appears reasonable to consider microbiome manipulation as a therapeutic strategy against these diseases.24 These microbiome manipulation strategies include diet, antibiotics, prebiotics, probiotics and faecal microbiota transplantation. Amongst these, the intervention having the most robust data supporting its efficacy and safety is faecal microbiota transplantation (FMT).

FMT, which involves the infusion of a faecal suspension from a healthy individual into the gastrointestinal tract of an individual with GI disease, has been evaluated in randomized controlled trials and cohort studies, demonstrating clear benefits for the management of recurrent CDI and, to a lesser extent, ulcerative colitis (UC). Additionally, studies suggest FMT may offer therapeutic potential in a variety of other conditions, including Crohn’s disease, irritable bowel syndrome, hepatic encephalopathy, metabolic syndrome and in the eradication of multidrug-resistant organisms from the gut.25 Response to FMT is also affected by donor factors, recipient factors and procedural variables. Certain donors, sometimes termed ‘super-donors,’ have microbiota profiles associated with higher rates of clinical response, particularly those enriched with butyrate-producing taxa (e.g. Lachnospiraceae, Oscillospiraceae) and higher overall microbial diversity. Baseline recipient microbiome composition, immune status, genetics, age and underlying disease state also impact microbiota engraftment and clinical efficacy. The route of administration (colonoscopy versus upper GI endoscopy and oral capsules), frequency and number of infusions, the amount infused and use of antibiotics before FMT can all affect engraftment and efficacy.26

IBD, which includes ulcerative colitis and Crohn’s disease, has an unknown aetiology and a multifactorial pathogenesis. IBD arises from a complex interplay of genetic susceptibility, environmental exposures, immune dysregulation and alterations in gut microbiome. Manipulation of the gut microbiome targets the pathophysiology of IBD at a stage upstream of the dysregulated immune response by addressing the microbial dysbiosis that contributes to immune activation and barrier dysfunction.27,28 Alteration of the microbiome with a combination of FMT as well as dietary changes has been explored in this disease. A novel study combining both these approaches showed that FMT with an anti-inflammatory diet (AID) in mild-to-moderate UC not only led to induction of remission rates as high as 50%, but also AID was able to maintain clinical remission in 50% of patients at 48 weeks.29 Furthermore, FMT was found to refurbish the crypt-associated microbiota (CAM), and FMT-restored CAM taxa correlated negatively with disease activity.30 Despite successful trials of FMT in IBD, it has not gained mainstream acceptance as an established therapeutic modality, and this leads us to analyze the possible reasons for the lack of confidence in microbiome manipulation therapies.

Microbiome manipulation therapy: Struggle to meet broader transformative expectations The field has delivered on a narrow, high-impact application (treatment of recurrent CDI) but is still struggling to meet the broader, transformative expectations for chronic disease treatment. The main hurdles preventing widespread success are the immense inter-individual variability of the microbiome, the complexity of microbial interactions, and the ‘chicken-or-egg’ dilemma (unclear if dysbiosis causes or is merely a result in most diseases). The challenge starts with defining a ‘healthy’ human gut microbiome due to individual and geographical variability. To address this, various indices have been developed. The HACK index (Health-Associated Core Keystone index) is a ranked metric for gut microbiome taxa that quantifies their association with host health and microbiome resilience. It was developed by analysing the prevalence and health associations of 201 gut microbial taxa across 45 000 adult gut microbiome samples from 42 countries and 28 disease categories. It provides a reproducible, standardized way to compare microbiomes, regardless of profiling strategy.31 Other indices similar to the HACK index have also been developed, including Gut Microbiome Health Index (GMHI), Gut Microbiome Index (GMI), Metagenomic Aerotolerant Predominance Index (MAPI) and Keystone Species Score.3236 Longitudinal microbiome studies have shown that intra-individual variability in gut microbiome is generally lower than inter-individual variability, with the majority of microbial community structure and genetic profiles remaining stable over extended periods of time (months to years). The individual-specific microbial signatures enable ‘microbial fingerprinting’ to accurately group samples from the same person over many years. One study reported an accuracy of 95% at 1 year and 85% at 4 years in detecting samples from the same individual.37

Translation of microbiome sciences into routine clinical practice is not without obstacles. Regulatory barriers stem from methodological heterogeneity in the studies and the absence of clear regulatory pathways for microbiome-based diagnostics and therapeutics. Ethical issues involve concerns about data sharing and privacy, informed consent and responsible communication of results. Variability in sample collection, sequencing platforms, bioinformatic pipelines and interpretation of results leads to difficulty in reproducing the findings. Table 1 lists the remaining knowledge gaps in microbiome therapeutics.

TABLE 1. Ten grand challenges in microbiome research
1 What is a healthy microbiome?–still undefined
2 Role of virome, mycobiome, and archaeome in health and disease
3 Standardization of methodologies for sampling, microbiome sequencing, and data analysis
4 Annotation of the functional dark matter for mechanistic insight into microbe-host interactions
5 Translating microbiota modulation therapies in routine clinical practice
6 Integration multi-omics approaches (genomics, transcriptomics, proteomics, metabolomics)
for functional insights
7 Establishing causality between microbiome alterations and diseases
8 Determining population-specific differences in microbiome composition and function
9 Development of validated biomarkers for diagnosis, prognosis, and therapeutic monitoring
10 Ethical, regulatory, and data-sharing challenges in microbiome research

Microbiome ‘Lassonde curve’

The Lassonde curve describes the lifecycle of value creation in the mining industry. It is a speculative model for junior mining companies, detailing the rise and fall of stock value as they move from exploration to production.38 The Lassonde curve provides an appropriate, if metaphorical, framework to understand the hype and consolidation phases. The core concept of a hype cycle with distinct stages can be used as an analogy to explain the journey of microbiome-related technologies from discovery to practical application. Gut microbiome researchers are “mining the metagenome” to extract the genetic and functional gold that can be leveraged for health, and hence, possibly the example of the Lassonde curve can be used, where investigators in this field are shifting from surface-level enthusiasm to the difficult, deep-digging work required to reach sustained productivity. The key stages are: Initial exploration, Discovery, Orphan period (speculative interest wanes), and Production (Table 2).

TABLE 2. The ‘Lassonde curve’ of microbiome manipulation therapies
Stage in microbiome development
Initial exploration This corresponds to the foundational microbial ecology research conducted for decades before the recent ‘omics’ revolutions.
Microbiome discovery ‘Dysbiosis’ and the orphan period Limited tools, largely culture-based methods. High-throughput sequencing technologies like 16S rRNA gene sequencing and metagenomics enabled a wave of new research, creating unprecedented excitement and attracting major interest. The realization that the microbiome profoundly influences human health led to a ‘gold rush’ for microbiome-based therapies, diagnostics, and consumer products. As initial excitement gives way to reality, the limitations and complexity of the microbiome become apparent. The field enters an ‘orphan period’ characterized by: Dysbiosis confusion: An altered microbiome (‘dysbiosis’) is correlated with numerous diseases, but establishing direct causation is often difficult.
Therapeutic production Technical challenges: Issues with data normalization, reproducibility, and linking sequence data to actual microbial function hinder progress. Worker fatigue: Some initial workers pull back as promised breakthroughs fail to materialize rapidly. The field matures by moving beyond simple observational studies to more rigorous, mechanistic research. Value creation comes from successful, targeted interventions that move through clinical trials, such as:
Faecal microbiota transplantation (FMT) for specific conditions like C. difficile infection.
Live biotherapeutic products (LBPs) and other defined microbial ecosystem therapeutics (MET). Precise interventions: The development of small molecules, prebiotics, and engineered microbes that act predictably on the microbiome to restore a healthy state.

The success precedent is strong

The unmet expectation is not that the science is flawed, but that it has not yet been successfully translated to more complex, chronic diseases like IBD, diabetes or other diseases. The treatment of CDI with FMT is an undeniable success story. Reinforcing this success are seminal studies that have shown that the gut microbiome may not only be a marker of current health but also a potential predictor of future health and overall longevity. People with advanced age display higher gut microbial diversity and enrichment of beneficial taxa, including Akkermansia and Lactobacillus, which are associated with increased SCFA production, improved amino-acid metabolism and reduced systemic inflammation— factors that contribute to healthy aging. Conversely, age-related dysbiosis, characterized by reduced diversity and increased proinflammatory taxa, has been linked to frailty, chronic inflammation and age-related diseases.39

The field of microbiota sciences has made major strides in the past 20 years. Looking ahead, the next 20 years are expected to bring in the development of novel biotics (including engineered probiotics, prebiotics and synthetic communities), which will allow targeted or personalized modulation of the microbiome. Discovery of microbiome-derived novel antibiotics and anti-inflammatory compounds is expected to accelerate. Precision gene-editing tools (e.g. CRISPR-Cas9) may enable modification of specific organisms in the gut, unlocking new ways to treat disease or maintain health. Microbiome composition may serve as both a diagnostic marker and a predictor of future disease risk, improving early detection and preventive healthcare. Machine learning and advanced computational tools will interpret complex microbiome data, improve diagnostics, and facilitate large-scale, multidisciplinary research collaborations. Research focus will expand from broad taxonomic surveys to detailed strain-level characterization and integrated “multi-omics” (combining genomics, metabolomics, proteomics) to offer a comprehensive view of the human microbiome. Collectively, these advances will drive the field towards precision medicine and a mechanistic understanding of microbiome-related health and disease.

References

  1. , . Turing and Von Neumann: From logic to the computer. philosophies. 2023;8:22.
    [CrossRef] [Google Scholar]
  2. , . Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc Natl Acad Sci USA. 1977;74:5088-90.
    [CrossRef] [PubMed] [Google Scholar]
  3. . Inventory of a reservoir: friends & foes. Indian J Med Res. 2015;142:4.
    [CrossRef] [PubMed] [Google Scholar]
  4. , , , , , , et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65.
    [CrossRef] [PubMed] [Google Scholar]
  5. , , . Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164:337-40.
    [CrossRef] [PubMed] [Google Scholar]
  6. , , . Gut microbiome: a biomedical revolution. Nat Rev Gastroenterol Hepatol. 2024;21:830-3.
    [CrossRef] [PubMed] [Google Scholar]
  7. , , , , , , et al. Species-level analysis of human gut microbiota with metataxonomics. Front Microbiol. 2020;11:2029.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , , , , et al. Gut microbiota development across the lifespan: disease links and health-promoting interventions. J Intern Med. 2025;297:560-83.
    [CrossRef] [PubMed] [Google Scholar]
  9. , . Human gut microbiome: a primer for the clinician. JGH Open. 2023;7:337-50.
    [CrossRef] [PubMed] [Google Scholar]
  10. , , , , , , et al. Analysis of the gut microbiome of rural and urban healthy Indians living in sea level and high altitude areas. Sci Rep. 2018;8:10104.
    [CrossRef] [PubMed] [Google Scholar]
  11. , , , , , , et al. A cross-sectional comparative study of gut bacterial community of Indian and Finnish children. Sci Rep. 2017;7:10555.
    [CrossRef] [PubMed] [Google Scholar]
  12. , , , , . Metagenomics analysis reveals features unique to Indian distal gut microbiota. PLoS ONE. 2020;15:e0231197.
    [CrossRef] [PubMed] [Google Scholar]
  13. , , , , , , et al. Molecular characterization and meta-analysis of gut microbial communities illustrate enrichment of Prevotella and Megasphaera in Indian subjects. Front Microbiol. 2016;7:660.
    [CrossRef] [PubMed] [Google Scholar]
  14. , , , , , . Role of diet-microbiome interaction in gastrointestinal disorders and strategies to modulate them with microbiome-targeted therapies. Annu Rev Nutr. 2023;43:355-83.
    [CrossRef] [PubMed] [Google Scholar]
  15. , , , , , , et al. Combined analysis of cross-population healthy adult human microbiome reveals consistent differences in gut microbial characteristics between western and non-western countries. Comput Struct Biotechnol J. 2024;23:87-95.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , , , , , et al. The unique composition of Indian gut microbiome, gene catalogue, and associated fecal metabolome deciphered using multi-omics approaches. GigaScience. 2019;8:giz004.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , , , . Gut microbiota contributes to the intestinal and extraintestinal immune homeostasis by balancing Th17/Treg cells. Int Immunopharmacol. 2024;143:113570.
    [CrossRef] [PubMed] [Google Scholar]
  18. , , , , , . From gut to brain: The roles of intestinal microbiota, immune system, and hormones in intestinal physiology and gut-brain-axis. Mol Cell Endocrinol. 2025;607:112599.
    [CrossRef] [PubMed] [Google Scholar]
  19. , , , . The progression of microbiome therapeutics for the management of gastrointestinal diseases and beyond. Gastroenterology. 2024;167:885-902.
    [CrossRef] [PubMed] [Google Scholar]
  20. , , , , , . Gut microbiome stability and resilience: elucidating the response to perturbations in order to modulate gut health. Gut. 2021;70:595-605.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , , . Real-time analysis of mucosal flora in patients with inflammatory bowel disease in India. J Clin Microbiol. 2010;48:4279-82.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , , , et al. Gut microbiome diversity in acute severe colitis is distinct from mild to moderate ulcerative colitis. J Gastroenterol Hepatol. 2021;36:731-9.
    [CrossRef] [PubMed] [Google Scholar]
  23. , , , , . Insights into gut dysbiosis: Inflammatory diseases, obesity, and restoration approaches. Int J Mol Sci. 2024;25:9715.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , . A decade of advances in human gut microbiome-derived biotherapeutics. Nat Microbiol. 2025;10:301-12.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , , , , , et al. Fecal microbiota transplantation: current challenges and future landscapes. Clin Microbiol Rev. 2024;37:2.
    [CrossRef] [PubMed] [Google Scholar]
  26. , , . The practice of fecal microbiota transplantation in inflammatory bowel disease. Intest Res. 2024;22:44-64.
    [CrossRef] [PubMed] [Google Scholar]
  27. , , , , , , et al. Gut microbiota: Sculptors of the intestinal stem cell niche in health and inflammatory bowel disease. Gut Microbes. 2021;13:1990827.
    [CrossRef] [PubMed] [Google Scholar]
  28. , , , . Gut bacteriome in inflammatory bowel disease: An update on recent advances. Indian J Gastroenterol. 2024;43:103-11.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , , , , , et al. Fecal microbiota transplantation with anti-inflammatory diet (FMT-AID) followed by anti-inflammatory diet alone is effective in inducing and maintaining remission over 1 year in mild to moderate ulcerative colitis: A randomized controlled trial. Gut. 2022;71:2401-13.
    [CrossRef] [PubMed] [Google Scholar]
  30. , , , , , , et al. Fecal microbiota transplantation refurbishes the crypt-associated microbiota in ulcerative colitis. iScience. 2023;26:106738.
    [CrossRef] [PubMed] [Google Scholar]
  31. , , , , , , et al. Toward a health-associated core keystone index for the human gut microbiome. Cell Rep. 2025;44:115378.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , , , , , et al. A predictive index for health status using species-level gut microbiome profiling. Nat Commun. 2020;11:1.
    [CrossRef] [PubMed] [Google Scholar]
  33. , , , , , , et al. Proposal of a health gut microbiome index based on a meta-analysis of Korean and global population datasets. J Microbiol. 2022;60:533-49.
    [CrossRef] [PubMed] [Google Scholar]
  34. , , , , , , et al. Gut Microbiome Wellness Index 2 enhances health status prediction from gut microbiome taxonomic profiles. Nat Commun. 2024;15:1.
    [CrossRef] [PubMed] [Google Scholar]
  35. , , , , , , et al. Gut health predictive indices linking gut microbiota dysbiosis with healthy state, mild gut discomfort, and inflammatory bowel disease phenotypes using gut microbiome profiling. Microbiol Spectr 2025 June 25
    [CrossRef] [PubMed] [Google Scholar]
  36. , , . Characteristics of putative keystones in the healthy adult human gut microbiota as determined by correlation network analysis. Front Microbiol. 2024;15:1.
    [CrossRef] [PubMed] [Google Scholar]
  37. , , , , , , et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell. 2021;184:2302-15.e12.
    [CrossRef] [PubMed] [Google Scholar]
  38. , , , . Confirming the Lassonde curve through life cycle analysis and its effect on share price: A case study of three ASX listed gold companies. Resources Policy. 2022;77:102704.
    [CrossRef] [Google Scholar]
  39. , . The human gut microbiome and aging. Gut Microbes. 2024;16:1.
    [CrossRef] [PubMed] [Google Scholar]
Show Sections