2024-06-24 06:44:08
Microbiome-Targeted Interventions (MBTIs) majorpublished
Did you know?
Microbiome Targeted Interventions (MBTIs) are revolutionizing modern medicine. These interventions can precisely modulate the microbiome, offering unprecedented precision in targeting pathogens while preserving beneficial microbes.
Microbiome Targeted Interventions (MBTIs) are cutting-edge treatments that utilize information from Microbiome Signatures to modulate the microbiome, revolutionizing medicine with unparalleled precision and impact.
Karen Pendergrass is a microbiome researcher specializing in microbiome-targeted interventions (MBTIs). She systematically analyzes scientific literature to identify microbial patterns, develop hypotheses, and validate interventions. As the founder of the Microbiome Signatures Database, she bridges microbiome research with clinical practice. In 2012, based on her own investigative research, she became the first documented case of FMT for Celiac Disease—four years before the first published case study.
Microbiome Signatures identifies and validates condition-specific microbiome shifts and interventions to accelerate clinical translation. Our multidisciplinary team supports clinicians, researchers, and innovators in turning microbiome science into actionable medicine.
Karen Pendergrass is a microbiome researcher specializing in microbiome-targeted interventions (MBTIs). She systematically analyzes scientific literature to identify microbial patterns, develop hypotheses, and validate interventions. As the founder of the Microbiome Signatures Database, she bridges microbiome research with clinical practice. In 2012, based on her own investigative research, she became the first documented case of FMT for Celiac Disease—four years before the first published case study.
A Microbiome-Targeted Intervention refers to a deliberate and purposeful manipulation of the gut microbiota’s composition and function to restore health and prevent disease.[1] MBTIs encompass a diverse range of therapeutic approaches that vary in application, goal, and mechanisms of action. These interventions can be broadly categorized into two approaches: those that modify the entire microbial community, such as dietary modulations or fecal microbiota transplantation (FMT), and those targeting specific taxa or strains, including phage therapy, probiotics, and prebiotics.[2] The fundamental principle underlying MBTI is that by restoring a healthy microbiota composition and optimizing microbial function through various mechanisms of action, we can modulate host responses to disease-relevant exposures and stimuli, ultimately leading to improved health outcomes.[3] Thus, MBTIs are a key component of precision microbiome medicine, revolutionizing treatments with unparalleled precision and impact.
The scope of conditions benefiting from MBTI has expanded dramatically as research has accumulated evidence for microbiome-driven pathogenesis across nearly every major organ system. Metabolic and Gastrointestinal Conditions represent the most robustly validated therapeutic area. Obesity and metabolic syndrome have shown particular responsiveness to MBTI through multiple mechanisms: targeting the microbiome by diet modulation can increase short-chain fatty acid (SCFA)-producing bacteria and reduce pathogenic taxa, leading to improved glucose metabolism and lipid profiles.[4] Nonalcoholic fatty liver disease (NAFLD) shows significant improvement with probiotics and synbiotics, with interventions reducing liver inflammation markers and hepatic steatosis as measured by ultrasound and elastography.[5] Irritable bowel disease, encompassing both Crohn’s disease and ulcerative colitis, represents perhaps the most clinically advanced application of MBTI. Probiotics, prebiotics, synbiotics, and particularly FMT have demonstrated efficacy in inducing remission in IBD patients, with one clinical study reporting 95% of ulcerative colitis patients achieved clinical remission eight weeks after FMT. [6][7] Type 1 and type 2 diabetes also benefit from microbiome-targeted approaches, as specific microbial taxa influence glucose homeostasis through SCFA production and immune regulation.[8]
The future of Microbiome-Targeted Interventions (MBTIs) is rapidly evolving, moving beyond traditional probiotics and prebiotics to encompass highly sophisticated, condition-specific, and personalized strategies. This evolution is driven by advances in understanding the intricate mechanisms of host-microbe interactions, including novel approaches such as phage therapy, targeting functional shielding, developing new antimicrobials, leveraging photobiomodulation, exploring drug repurposing, supporting nutritional immunity, and new advancements and insights from the field of microbial metallomics.
| Intervention | MBTI Status |
|---|---|
| Phage Therapy | Phage therapy is gaining significant traction as a precise tool for microbiome modulation. Unlike broad-spectrum antibiotics that decimate commensal bacteria along with pathogens, bacteriophages (viruses that infect bacteria) offer highly specific targeting of undesirable bacterial species without disrupting the beneficial microbiota.[9] This precision is particularly valuable in managing infectious diseases and combating antibiotic resistance, where traditional methods fall short.[10] Engineered phages could be designed to eliminate specific pathogens or even deliver therapeutic payloads, offering a novel strategy for infectious disease prevention and treatment. This contrasts with traditional microbiome modulation methods like probiotics and fecal microbiota transplantation (FMT), providing unparalleled specificity and adaptability in microbial targeting.[11] |
| Targeting Functional Shielding and Co-aggregation | A critical area for future MBTIs involves addressing microbial “functional shielding” and co-aggregation, where certain microorganisms create protective environments that enable pathogens to thrive or resist treatments. This phenomenon is observed in cases like Candida in its hyphae stage, providing a niche for Staphylococcus aureus, or co-aggregation between Candida and Porphyromonas gingivalis. [12][13] Understanding these intricate inter-kingdom interactions is key. MBTIs will increasingly focus on disrupting these protective mechanisms. For instance, if Candida facilitates the persistence of other pathogens, interventions could aim to inhibit Candida hyphal formation or its co-aggregation properties. Developing strategies to interfere with these synergistic pathogenic alliances could dramatically improve outcomes in polymicrobial infections and chronic inflammatory conditions. |
| Novel Antimicrobials and Bacteriostatics | The development of novel classes of antimicrobials and bacteriostatics, such as dimethylglyoxime or gallium, represents another promising avenue. [14][15] These agents could offer more selective action against pathogens compared to conventional antibiotics, minimizing off-target effects on the beneficial microbiome. The goal is to inhibit or kill harmful microbes without broadly disrupting the delicate microbial ecosystem, thereby avoiding unintended consequences like secondary infections or the emergence of further resistance. Such targeted antimicrobials are crucial in the fight against multidrug-resistant infections. |
| Photobiomodulation | Photobiomodulation (PBM) is an emerging non-invasive technique that uses light to stimulate cellular function, reduce inflammation, and promote healing. Its application in microbiome medicine is exploratory but holds potential, particularly in modulating microbial communities on accessible surfaces like the skin or oral cavity. PBM could influence microbial growth, biofilm formation, or even host immune responses in a way that shifts the microbial balance towards a healthier state, potentially offering a gentle yet effective method to enhance MBTIs. |
| Supporting Nutritional Immunity Factors | Nutritional immunity represents one of the most ancient and fundamental defense mechanisms by which the host actively restricts the bioavailability of essential nutrients to limit pathogenic microbial growth and virulence.[16] This mechanism extends beyond simple nutrient deprivation to a sophisticated form of chemical warfare where the host manipulates metal availability to simultaneously suppress pathogenic virulence while preserving or enhancing the fitness of commensal microbiota.[17] The distinction between traditional micronutrient supplementation and nutritional immunity- and microbial metallomics-informed MBTIs is critical. Conventional clinical practice often assumes that nutritional deficiencies universally require supplementation (i.e. that iron-deficient patients should supplement ferrous sulfate, that zinc-deficient individuals should supplement zinc oxide or zinc citrate, that copper-depleted hosts benefit from copper gluconate.) However, in the context of dysbiosis and microbial pathogenesis, such blanket supplementation can inadvertently fuel pathogenic growth while simultaneously undermining the host’s natural antimicrobial strategies.[18] Nutritional immunity- and microbial metallomics-informed MBTIs, by contrast, recognize that in many disease states, apparent micronutrient “deficiency” is actually an adaptive host response—a deliberate sequestration strategy designed to starve invading pathogens.[19] Supporting nutritional immunity thus involves a fundamentally different therapeutic paradigm: rather than correcting “deficiency,” the goal becomes enhancing the host’s ability to maintain nutrient restriction while simultaneously ensuring that this restriction selectively disadvantages pathogens without compromising commensal bacteria or essential host physiology.[20] |
| Advancements in Microbial Metallomics | The field of microbial metallomics, studying the role of metals in microbial life, is crucial for advancing MBTIs. Environmental heavy metals, even at trace levels, can act as cofactors for pathogens and their virlence factors, or cause mismetallation. Future MBTIs will increasingly consider these elemental interactions to develop targeted interventions. [21] Advancements in the field will inevitably lead to therapies that precisely target the metal-dependent pathways of pathogens or enhance host mechanisms to sequester potentially harmful metals, thereby influencing microbial virulence and host susceptibility. These interventions extend beyond simple nutrient supplementation to a sophisticated understanding of metal bioavailability and utilization within the microbial ecosystem. |
| Drug Repurposing | Drug repurposing represents an emerging and highly promising frontier for MBTI advancement. Rather than developing novel compounds from scratch, drug repurposing leverages the extensive safety, pharmacokinetic, and clinical data already available for FDA-approved or clinically validated drugs, dramatically accelerating the timeline from bench discovery to clinical application. [22] This approach is particularly valuable for MBTI development because it enables rapid translation of microbiome research findings into clinically accessible interventions while bypassing the lengthy and expensive drug development process. Many approved pharmaceuticals were originally developed for entirely different indications but possess incidental antimicrobial properties that, when applied strategically to dysbiosis, can shift microbial communities favorably. For instance, certain cardiac medications, antihistamines, and psychiatric drugs demonstrate selective antimicrobial effects against dysbiosis-driving pathogens while sparing beneficial commensals.[23] Metformin, the first-line diabetes medication used by hundreds of millions of patients, exemplifies this principle: originally developed as an antidiabetic agent, metformin’s effects on glucose metabolism are now understood to work partly through direct modulation of gut dysbiosis, selectively promoting growth of SCFA-producing commensals like Akkermansia muciniphila and Faecalibacterium prausnitzii.[24] Recognition of this mechanism has led to emerging clinical interest in metformin as an MBTI for obesity and metabolic syndrome independent of its glucose-lowering effects. |
| Advances in Targeted Prebiotics | Prebiotics represent one of the oldest and most naturalistic MBTI approaches. However, traditional prebiotic development has been relatively crude, with most prebiotics being broadly fermented by multiple bacterial taxa without specific selectivity for target species.[25] One of the most profound discoveries emerging from precision prebiotic research involves the relationship between prebiotic molecular weight and microbial selectivity , ensuring prebiotic compabitility with the desired outcomes. [26] This principle fundamentally challenges the traditional “one-size-fits-all” prebiotic approach, which is crucial to the advancement of the proper use of fibers in microbiome medicine applications. |
Neuropsychiatric and Neurodegenerative Conditions represent an emerging frontier where MBTI shows increasing promise. Alzheimer’s disease and mild cognitive impairment have demonstrated associations with specific microbiota alterations, particularly depletion of short-chain fatty acid-producing bacteria and enrichment of pro-inflammatory taxa.[27] Mediterranean-ketogenic dietary interventions that modulate the microbiome to increase beneficial SCFAs and protective taxa while reducing inflammatory metabolites have shown improvements in cognitive function and reduced AD pathological markers.[28]
Autism spectrum disorder (ASD) presents particularly compelling evidence for microbiota-targeted intervention potential: probiotic supplementation with Lactobacillus and Bifidobacterium species improved core ASD features and reduced gastrointestinal dysfunction while microbiota transfer therapy showed promising improvements in both gastrointestinal and behavioral symptoms in open-label trials.[29]
Depression and anxiety, conceptualized through the microbiota-gut-brain axis, benefit from microbiome modulation: probiotics and dietary interventions targeting microbial production of neuroactive metabolites (short-chain fatty acids, tryptophan metabolites) have demonstrated measurable improvements in depressive symptoms and anxiety scores.[30]
Post-traumatic stress disorder similarly shows emerging evidence for benefit from microbiome-targeted approaches focusing on restoring SCFA-producing bacteria.[31]
Immune-Mediated and Inflammatory Diseases beyond IBD have shown responsiveness to MBTI. Rheumatoid arthritis and other autoimmune diseases are characterized by intestinal dysbiosis, and multiple probiotic trials have demonstrated reduced disease activity and inflammatory markers.[32] Systemic lupus erythematosus (SLE) and multiple sclerosis (MS) exhibit microbiota signatures that can be modified through dietary and probiotic interventions to restore immune tolerance.[33] Allergic diseases including atopic dermatitis, eczema, and asthma all show associations with dysbiosis, and microbiome-targeted interventions including probiotics and dietary modifications have demonstrated improvements in allergic symptoms and inflammatory markers.[34]
Cardiovascular and Metabolic Conditions have revealed intimate mechanistic links to the microbiome through the production of metabolites like trimethylamine-N-oxide (TMAO) that promote atherosclerosis. Microbiome-targeted interventions reducing TMAO-producing bacteria while increasing SCFA producers have been associated with improved cardiovascular markers and reduced inflammatory signaling.[35] Hypertension similarly responds to microbiome modulation focusing on increasing diversity and SCFA production.[36]
Liver Diseases including alcohol-associated liver disease show significant potential for MBTI. The gut-liver axis enables dysbiosis to promote hepatic inflammation and progression to cirrhosis; probiotics, antibiotics (such as rifaxomicin), and FMT have each demonstrated clinical benefits in reducing liver inflammation and improving hepatic function.[37] Hepatobiliary cancers present emerging opportunities for microbiome-targeted therapy: specific microbial taxa are enriched in responders to anti-PD-1 immunotherapy, suggesting that microbiome modulation may enhance cancer immunotherapy efficacy.[38]
Gastrointestinal and Colorectal Conditions beyond IBD include irritable bowel syndrome (IBS), where microbiome-targeted dietary interventions demonstrate particular promise.[39] Chronic constipation responds to probiotics and dietary modifications that increase SCFA-producing bacteria and restore colonic motility through neuroendocrine signaling.[40] Colorectal cancer prevention and treatment represent particularly important applications, as dysbiosis is associated with increased carcinogenic pathways; dietary fiber interventions and FMT have potential to restore protective bacterial communities and enhance immunotherapy responses.[41]
Pulmonary and Respiratory Conditions increasingly show responsiveness to microbiome modulation. Chronic obstructive pulmonary disease, asthma, and bronchiectasis—all characterized by dysbiosis-driven inflammation—benefit from probiotics and dietary interventions that restore barrier function and immune regulation.[42] COVID-19 severity correlates with distinct respiratory microbiome signatures, and interventions targeting dysbiosis show promise in mitigating disease severity and improving immune responses.[43]
Reproductive Health and Gynecologic Conditions represent an important frontier where Microbiome-targeted interventions (MBTIs) have demonstrated clinical value. Polycystic ovary syndrome (PCOS), endometriosis, bacterial vaginosis (BV), and recurrent miscarriage all exhibit microbiota dysbiosis amenable to intervention. Probiotics and dietary modifications targeting the vaginal and gut microbiomes have shown improvements in clinical outcomes, including reduced pain severity and improved fertility.[44]
Oral Health and Periodontal Disease show remarkable responsiveness to microbiome-targeted approaches. Dysbiosis in the oral microbiome contributes to both periodontal disease and systemic conditions through bacterial translocation and inflammatory signaling; probiotics, antimicrobial peptides, and dietary interventions have demonstrated clinical benefits.[45]
The emergence of Microbiome-targeted interventions (MBTIs) has catalyzed a paradigm shift in medical practice from conventional, symptom-focused treatment to mechanistically informed, causally targeted interventions. [46] Microbiome medicine represents precision medicine at the molecular level, where therapeutic strategies are guided by understanding specific host-microbe interactions rather than generic disease categories. This advancement is evident across multiple therapeutic domains. In metabolic diseases, MBTIs have demonstrated the ability to modify fundamental pathways: probiotics and synbiotics significantly reduce fasting glucose, hemoglobin A1c, and lipid parameters in patients with metabolic syndrome and diabetes.[47]
For inflammatory conditions like inflammatory bowel disease, microbiome-targeted interventions including dietary interventions, probiotics, and fecal transplants (FMT) represent emerging treatment prospects with particular success when tailored to individual microbial signatures.[48] The integration of multi-omics technologies—metagenomics, metabolomics, microbial metallomics, and metatranscriptomics—has enabled microbiome medicine to move beyond descriptive associations to predictive and mechanistic understanding.[49] Rather than assuming all dysbiosis is equivalent, researchers can now identify which specific taxa alterations drive disease in particular patients and which interventions are most likely to restore functionality. This precision approach optimizes therapeutic efficacy while potentially reducing unnecessary interventions.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Did you know?
Staphylococcus aureus is often carried without causing disease. About 20–30% of people harbor it in their noses or on their skin as a harmless commensal. However, as a pathobiont, it can pivot to a formidable pathogen in the right circumstances.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Lorem ipsum dolor sit amet, consectetur adipiscing elit. Proin ut laoreet tortor. Donec euismod fermentum pharetra. Nullam at tristique enim. In sit amet molestie
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
Alias iure reprehenderit aut accusantium. Molestiae dolore suscipit. Necessitatibus eum quaerat. Repudiandae suscipit quo necessitatibus. Voluptatibus ullam nulla temporibus nobis. Atque eaque sed totam est assumenda. Porro modi soluta consequuntur veritatis excepturi minus delectus reprehenderit est. Eveniet labore ut quas minima aliquid quibusdam. Vitae possimus fuga praesentium eveniet debitis exercitationem deleniti.
2024-06-24 06:44:08
Microbiome-Targeted Interventions (MBTIs) majorpublished
Fecal Microbiota Transplantation (FMT) involves transferring fecal bacteria from a healthy donor to a patient to restore microbiome balance.
Phage therapy uses viruses to target and kill specific bacteria, offering a precise alternative to antibiotics, especially for resistant infections.
Short-chain fatty acids are microbially derived metabolites that regulate epithelial integrity, immune signaling, and microbial ecology. Their production patterns and mechanistic roles provide essential functional markers within microbiome signatures and support the interpretation of MBTIs, MMAs, and systems-level microbial shifts across clinical conditions.
Crohn's disease is a chronic inflammatory condition of the gastrointestinal tract that can cause a wide range of symptoms, including abdominal pain, diarrhea, and fatigue. The exact cause of the disease remains unclear, but it is believed to result from a combination of genetic predisposition and environmental factors. Although there is no cure, ongoing advancements in medical research continue to improve management strategies and quality of life for those affected by Crohn's disease.
Phage therapy uses viruses to target and kill specific bacteria, offering a precise alternative to antibiotics, especially for resistant infections.
Drug repurposing involves identifying new therapeutic uses for existing drugs, offering a cost-effective and time-efficient pathway to enhance treatment options and address unmet medical needs.
Nutritional immunity restricts metal access to pathogens, leveraging sequestration, transport, and toxicity to control infections and immunity.
Microbial Metallomics is the study of how microorganisms acquire, use, regulate, and transform metals in any biological or environmental context.
Dimethylglyoxime represents a novel therapeutic paradigm that exploits a fundamental metabolic difference between pathogenic bacteria and their mammalian hosts. By selectively depleting bacterial access to nickel, a cofactor essential for multiple pathogenic enzymes but unnecessary for human physiology, DMG offers a theoretically host-sparing antimicrobial approach.
Gallium is studied for its unique antimicrobial and anticancer properties. It inhibits metalloproteinases, disrupts bacterial iron metabolism, and may enhance antibiotic efficacy, particularly against resistant strains. Gallium compounds show potential as non-traditional therapeutic agents in treating infections and inhibiting cancer cell invasion and metastasis.
Nutritional immunity restricts metal access to pathogens, leveraging sequestration, transport, and toxicity to control infections and immunity.
Microbial Metallomics is the study of how microorganisms acquire, use, regulate, and transform metals in any biological or environmental context.
Microbial Metallomics is the study of how microorganisms acquire, use, regulate, and transform metals in any biological or environmental context.
Microbial Metallomics is the study of how microorganisms acquire, use, regulate, and transform metals in any biological or environmental context.
Drug repurposing involves identifying new therapeutic uses for existing drugs, offering a cost-effective and time-efficient pathway to enhance treatment options and address unmet medical needs.
Metformin is a synthetic derivative of guanidine derived from the guanidine alkaloid of the plant Galega officinalis L. with significant hypoglycemic effects. It is a first-line antihyperglycemic agent due to its efficacy, low cost, and favorable safety profile.
Short-chain fatty acids are microbially derived metabolites that regulate epithelial integrity, immune signaling, and microbial ecology. Their production patterns and mechanistic roles provide essential functional markers within microbiome signatures and support the interpretation of MBTIs, MMAs, and systems-level microbial shifts across clinical conditions.
Autism Spectrum Disorder (ASD) is a neurodevelopmental condition characterized by social, communication, and behavioral challenges. It involves genetic and environmental factors, including microbiome imbalances which influence symptom severity and overall health.
TMAO is a metabolite formed when gut bacteria convert dietary nutrients like choline and L-carnitine into trimethylamine (TMA), which is then oxidized in the liver to TMAO. This compound is linked to cardiovascular disease, as it promotes atherosclerosis, thrombosis, and inflammation, highlighting the crucial role of gut microbiota in influencing heart health.
Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.
Fecal Microbiota Transplantation (FMT) involves transferring fecal bacteria from a healthy donor to a patient to restore microbiome balance.
Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.
Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.
Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.
Fecal Microbiota Transplantation (FMT) involves transferring fecal bacteria from a healthy donor to a patient to restore microbiome balance.
Microbial Metallomics is the study of how microorganisms acquire, use, regulate, and transform metals in any biological or environmental context.
Staphylococcus aureus is a versatile skin and mucosal commensal that can transition into a highly virulent pathobiont. Known for its immune-evasive strategies, toxin production, and antibiotic resistance, it plays a significant role in chronic infections and microbiome imbalance.
Pathobionts are native microbes with the capacity to cause disease under disrupted host or microbiome conditions.
Heavy metals play a significant and multifaceted role in the pathogenicity of microbial species.
Heavy metals play a significant and multifaceted role in the pathogenicity of microbial species.
Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T.
Microbiome-based interventions to modulate gut ecology and the immune system.Mucosal Immunology. 2022;15(6):1095-1113.
Read ReviewHitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T.
Microbiome-based interventions to modulate gut ecology and the immune system.Mucosal Immunology. 2022;15(6):1095-1113.
Read ReviewHitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T.
Microbiome-based interventions to modulate gut ecology and the immune system.Mucosal Immunology. 2022;15(6):1095-1113.
Read ReviewA. Mederle, M. Dima et al.
Impact of Gut Microbiome Interventions on Glucose and Lipid Metabolism in Metabolic Diseases: A Systematic Review and Meta-Analysis.Life. 2024.
S. Sharpton, Bharat Maraj et al.
Gut microbiome–targeted therapies in nonalcoholic fatty liver disease: a systematic review, meta-analysis, and meta-regression.The American Journal of Clinical Nutrition. 2019.
Despoina Gyriki, Christos G Nikolaidis et al.
Exploring the Gut Microbiome’s Role in Inflammatory Bowel Disease: Insights and Interventions.Journal of Personalized Medicine. 2024.
A. Tikunov, V. A. Fedorets et al.
Intestinal Microbiome Changes and Clinical Outcomes of Patients with Ulcerative Colitis after Fecal Microbiota Transplantation.Journal of Clinical Medicine. 2023.
A. Mederle, M. Dima et al.
Impact of Gut Microbiome Interventions on Glucose and Lipid Metabolism in Metabolic Diseases: A Systematic Review and Meta-Analysis.Life. 2024.
T. Hitch, L. Hall et al.
Microbiome-based interventions to modulate gut ecology and the immune system.Mucosal Immunology. 2022.
Iyiola Aanuoluwa Temitayo, Ann Christopher Francis et al.
Microbiome editing in infectious disease prevention and therapy: CRISPR Applications in Host-Microbe Interactions.International Journal of Biological and Pharmaceutical Sciences Archive, May 2025.
Iyiola Aanuoluwa Temitayo, Ann Christopher Francis et al.
Microbiome editing in infectious disease prevention and therapy: CRISPR Applications in Host-Microbe Interactions.International Journal of Biological and Pharmaceutical Sciences Archive, May 2025.
Schlecht LM, Peters BM, Krom BP, Freiberg JA, Hänsch GM, Filler SG, Jabra-Rizk MA, Shirtliff ME.
Systemic Staphylococcus aureus infection mediated by Candida albicans hyphal invasion of mucosal tissue.Microbiology. 2015;161(1):168–181.
Read ReviewBartnicka D., Gonzalez-Gonzalez M. et al.
Candida albicans Shields the Periodontal Killer Porphyromonas gingivalis from Recognition by the Host Immune System and Supports the Bacterial Infection of Gingival Tissue.Int J Mol Sci. 2020 Mar 14;21(6):1984.
Read ReviewBenoit SL, Schmalstig AA, Glushka J, Maier SE, Edison AS, Maier RJ.
Nickel chelation therapy as an approach to combat multi-drug resistant enteric pathogens.Scientific Reports. 2019
Read ReviewLi F, Liu F, Huang K, Yang S.
Advancement of Gallium and Gallium-Based Compounds as Antimicrobial Agents.Front Bioeng Biotechnol. 2022 Feb 4;10:827960.
Read ReviewClaire Healy, Natalia MuozWolf et al.
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease.Respiratory Research. 2021.
F. Malard, J. Dor et al.
Introduction to host microbiome symbiosis in health and disease.Mucosal Immunology. 2021.
Marialaura Marchetti, Omar De Bei et al.
Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development.International Journal of Molecular Sciences. 2020.
Claire Healy, Natalia MuozWolf et al.
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease.Respiratory Research. 2021.
Claire Healy, Natalia MuozWolf et al.
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease.Respiratory Research. 2021.
Benoit SL, Schmalstig AA, Glushka J, Maier SE, Edison AS, Maier RJ.
Nickel chelation therapy as an approach to combat multi-drug resistant enteric pathogens.Scientific Reports. 2019
Read ReviewIyiola Aanuoluwa Temitayo et al.
Microbiome editing in infectious disease prevention and therapy: CRISPR Applications in Host-Microbe Interactions.International Journal of Biological and Pharmaceutical Sciences Archive, May 2025.
Iyiola Aanuoluwa Temitayo et al.
Microbiome editing in infectious disease prevention and therapy: CRISPR Applications in Host-Microbe Interactions.International Journal of Biological and Pharmaceutical Sciences Archive, May 2025.
Maqsood Ali, Navid Iqbal et al.
Potential Therapeutic Agents: New Players of Obesity Treatment.Journal of Microbiology and Biotechnology. 2025.
Bedu-Ferrari. et al.
Prebiotics and the Human Gut Microbiota: From Breakdown Mechanisms to the Impact on Metabolic Health.Nutrients. 2022.
J. She, C. Wong et al.
Targeted prebiotics alter the obese gut microbiome in humans.Signal Transduction and Targeted Therapy. 2021.
Tao Zhang, Guangqi Gao et al.
Gut microbiome-targeted therapies for Alzheimer’s disease.Gut Microbes. 2023.
Emily R Murray, Mylon Kemp et al.
The Microbiota–Gut–Brain Axis in Alzheimer’s Disease: A Review of Taxonomic Alterations and Potential Avenues for Interventions.Archives of Clinical Neuropsychology. 2022.
Andreas Petropoulos, E. Stavropoulou et al.
Microbiota Gut–Brain Axis and Autism Spectrum Disorder:Mechanisms and Therapeutic Perspectives. Nutrients. 2025.
I. Pedroso, S. Kumbhare et al.
Mental Health Symptom Reduction Using Digital Therapeutics Care Informed by Genomic SNPs and Gut Microbiome Signatures.Journal of Personalized Medicine. 2022.
P. Petakh, Khrystyna Duve et al.
Molecular mechanisms and therapeutic possibilities of short-chain fatty acids in posttraumatic stress disorder patients: a mini-review.Frontiers in Neuroscience. 2024.
Baskar Balakrishnan, Veena Taneja
Microbial modulation of the gut microbiome for treating autoimmune diseases.Expert Review of Gastroenterology & Hepatology. 2018.
Sudeep Edpuganti, Krupa Sara Thomas et al.
Gut Microbiome Dysbiosis in Autoimmune Diseases: Mechanisms, Biomarkers, and Therapeutic Strategies.International Journal of Progressive Sciences and Technologies. 2025.
K. Frasier, Haley Fonceca et al.
Unraveling the Role of the Skin Microbiome in Immunodermatological Diseases: Implications for Therapeutic Interventions.Clinical Dermatology and Surgery. 2024.
Razan Abdulaal, Yehya Tlaiss et al.
The role of microbiome dysbiosis in cardiovascular disease: Mechanisms and therapeutic implications.Global Cardiology Science and Practice. 2025.
W.H. Wilson Tang, Takeshi Kitai et al.
Gut Microbiota in Cardiovascular Health and Disease.Circulation Research. 2017.
annaz Ranjbarian, B. Schnabl
Gut Microbiome-Centered Therapies for Alcohol-Associated Liver Disease.Seminars in Liver Disease. 2023.
J. Mao, Dongxu Wang et al.
Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers.Journal for ImmunoTherapy of Cancer. 2021.
Sofia D. Shaikh, Natalie Sun et al.
Irritable Bowel Syndrome and the Gut Microbiome: A Comprehensive Review.Journal of Clinical Medicine. 2023.
Ruili Pan, Linlin Wang et al.
Crosstalk between the Gut Microbiome and Colonic Motility in Chronic Constipation: Potential Mechanisms and Microbiota Modulation.Nutrients. 2022.
Amlia Cinthia Meneses Do Rgo, Irami Arajo Filho
Microbiome and Treatment Resistance in Colorectal Cancer: Mechanisms and Solutions.Surgery Research Journal, Dec. 2024.
Michel Mac Aogin, A. Dicker et al.
Infection and the microbiome in bronchiectasis.European Respiratory Review. 2024.
Carter Merenstein, G. Liang et al.
Signatures of COVID-19 Severity and Immune Response in the Respiratory Tract Microbiome.mBio. 2021.
Caitriona Brennan, Kristina Chan et al.
Harnessing the power within: engineering the microbiome for enhanced gynecologic health.Reproduction and Fertility. 2024.
J. D. Corra, P.P.S. Carlos et al.
The Healthy Oral Microbiome: A Changing Ecosystem throughout the Human Lifespan.Journal of Dental Research. 2024.
Walaa K. Mousa, A. Ali
The Gut Microbiome Advances Precision Medicine and Diagnostics for Inflammatory Bowel Diseases.International Journal of Molecular Sciences. 2024.
A. Mederle, M. Dima et al.
Impact of Gut Microbiome Interventions on Glucose and Lipid Metabolism in Metabolic Diseases: A Systematic Review and Meta-Analysis.Life. 2024.
Despoina Gyriki, Christos G Nikolaidis et al.
Exploring the Gut Microbiome’s Role in Inflammatory Bowel Disease: Insights and Interventions.Journal of Personalized Medicine. 2024.
Walaa K. Mousa, A. Ali
The Gut Microbiome Advances Precision Medicine and Diagnostics for Inflammatory Bowel Diseases.International Journal of Molecular Sciences. 2024.