1
|
Li C, Chen X, Zha W, Fang S, Shen J, Li L, Jiang H, Tian P. Impact of gut microbiota in chronic kidney disease: natural polyphenols as beneficial regulators. Ren Fail 2025; 47:2506810. [PMID: 40441674 PMCID: PMC12123969 DOI: 10.1080/0886022x.2025.2506810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/04/2025] [Accepted: 05/10/2025] [Indexed: 06/02/2025] Open
Abstract
Chronic kidney disease (CKD) poses a severe health risk with high morbidity and mortality, profoundly affecting patient quality of life and survival. Despite advancements in research, the pathophysiology of CKD remains incompletely understood. Growing evidence links CKD with shifts in gut microbiota function and composition. Natural compounds, particularly polyphenols, have shown promise in CKD treatment due to their antioxidant and anti-inflammatory properties and their ability to modulate gut microbiota. This review discusses recent progress in uncovering the connections between gut microbiota and CKD, including microbiota changes across different kidney diseases. We also examine metabolite alterations,such as trimethylamine-N-oxide, tryptophan derivatives, branched-chain amino acids, short-chain fatty acids, and bile acids,which contribute to CKD progression. Further, we outline the mechanisms through which polyphenols exert therapeutic effects on CKD, focusing on signaling pathways like nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), mammalian target of rapamycin (mTOR), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), phosphatidylin-ositol-3-kinase (PI3K)/protein kinase B (Akt), and toll like receptors (TLR), as well as their impact on gut microbiota. Lastly, we consider how dietary polyphenols could be harnessed as bioactive drugs to slow CKD progression. Future research should prioritize multi-omics approaches to identify patients who would benefit from polyphenolic interventions, enabling personalized treatment strategies to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Cheng Li
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Department of Nephrology, Jiujiang University affiliated Hospital, Jiu’jiang, Jiang’xi, China
| | - Xulong Chen
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Weiwei Zha
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Sitian Fang
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nan’chang, Jiangxi, China
| | - Jiangwen Shen
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Lin Li
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Hongli Jiang
- Department of Blood Purification, Kidney Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
| | - PuXun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an, Shaan’xi, China
| |
Collapse
|
2
|
Zheng J, Zhou C, Li Z, Jin X, Zou Y, Bai S, Zheng H, Ling W, Zhao Y, Wang Y, Zhang R, Liu Z, Lu L. Alcaligenes faecalis promotes colitis to colorectal cancer transition through IgA+ B cell suppression and vinculin acetylation. Gut Microbes 2025; 17:2473511. [PMID: 40047249 PMCID: PMC11901412 DOI: 10.1080/19490976.2025.2473511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/24/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Lymphoid tissue-resident commensal bacteria (LRC), a subtype of gut microbiota essential for inflammation-associated carcinogenesis, predominantly attribute to colorectal cancer(CRC), whereas its role was largely unknown. Herein, we found Alcaligenes faecalis (A. faecalis), the main LRC embedded in Peyer's patches, was abundantly enriched in colitis, adenoma, and stage-dependently observed in CRC tissues. Interestingly, A. faecalis alone can not affect intestinal homeostasis, while during colitis, A. faecalis significantly translocated from Peyer's patches to colon, remarkably attenuated immune response abilities of B cells, T cells, and DC cells in PPs, consequently impeded IgA+ B cells homing. Meanwhile, during colitis, the ectopia of A. faecalis in colon tissues, promoted vinculin acetylation by A. faecalis-derived metabolite acetic acid, which impeded intestinal barrier via hindering the binding of vinculin to β-catenin. Our study revealed A. faecalis not only suppress mucosal immune responses via reducing IgA+ B cells in Peyer's patches but also disrupt intestinal barrier via increasing vinculin acetylation, ultimately promoting inflammation-to-cancer transition in CRC.
Collapse
Affiliation(s)
- Jing Zheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chishun Zhou
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zizheng Li
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Jin
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yihua Zou
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huanjin Zheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weichao Ling
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiru Zhao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Wang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linlin Lu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Baba Y, Tajima K, Yoshimura K. Intestinal and esophageal microbiota in esophageal cancer development and treatment. Gut Microbes 2025; 17:2505118. [PMID: 40376843 PMCID: PMC12087659 DOI: 10.1080/19490976.2025.2505118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 03/03/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
Esophageal cancer (EC) is the eleventh most commonly diagnosed cancer, and its prognosis remains poor. Several challenges remain for improving the clinical outcomes of EC, and improving technologies for early detection, diversifying treatment options, and advancing personalized treatment are essential. Alterations in the intestinal and esophageal microbiota are associated with the pathogenesis and progression of EC; for instance, Fusobacterium nucleatum is important in the pathogenesis and progression of esophageal squamous cell carcinoma. Therefore, a novel diagnostic biomarker may be identified using the intestinal microbiota. Furthermore, targeting the intestinal and esophageal microbiota may help in the early detection of EC, use of a novel prognostic biomarker, and even the detection of a therapeutic target, resulting in a more individualized therapeutic approach for EC. In this review, we summarize the clinical research focused on the intestinal and esophageal microbiota in EC development and its treatment, and discuss the challenges in the clinical application of intestinal and esophageal microbiota.
Collapse
Affiliation(s)
- Yuta Baba
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
- Division of Hematology, Department of Medicine, Showa Medical University Fujigaoka Hospital, Kanagawa, Japan
| | - Kohei Tajima
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Kiyoshi Yoshimura
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| |
Collapse
|
4
|
Steinert RE, Rehman A, Sadabad MS, Milanese A, Wittwer-Schegg J, Burton JP, Spooren A. Microbial micronutrient sharing, gut redox balance and keystone taxa as a basis for a new perspective to solutions targeting health from the gut. Gut Microbes 2025; 17:2477816. [PMID: 40090884 PMCID: PMC11913388 DOI: 10.1080/19490976.2025.2477816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
In health, the gut microbiome functions as a stable ecosystem maintaining overall balance and ensuring its own survival against environmental stressors through complex microbial interaction. This balance and protection from stressors is maintained through interactions both within the bacterial ecosystem as well as with its host. As a consequence, the gut microbiome plays a critical role in various physiological processes including maintaining the structure and function of the gut barrier, educating the gut immune system, and modulating the gut motor, digestive/absorptive, as well as neuroendocrine system all of which are crucial for human health and disease pathogenesis. Pre- and probiotics, widely available and clinically established, offer various health benefits primarily by beneficially modulating the gut microbiome. However, their clinical outcomes can vary significantly due to differences in host physiology, diets, individual microbiome compositions, and other environmental factors. This perspective paper highlights emerging scientific insights into the importance of microbial micronutrient sharing, gut redox balance, keystone species, and the gut barrier in maintaining a diverse and functional microbial ecosystem, and their relevance to human health. We propose a novel approach that targets microbial ecosystems and keystone taxa performance by supplying microbial micronutrients in the form of colon-delivered vitamins, and precision prebiotics [e.g. human milk oligosaccharides (HMOs) or synthetic glycans] as components of precisely tailored ingredient combinations to optimize human health. Such a strategy may effectively support and stabilize microbial ecosystems, providing a more robust and consistent approach across various individuals and environmental conditions, thus, overcoming the limitations of current single biotic solutions.
Collapse
Affiliation(s)
- Robert E. Steinert
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
- Department of Surgery and Transplantation, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| | | | - Alessio Milanese
- Data Science, Science & Research, Dsm-Firmenich, Delft, Netherlands
| | | | - Jeremy P. Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
| | - Anneleen Spooren
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| |
Collapse
|
5
|
Deng F, Yang D, Qing L, Chen Y, Zou J, Jia M, Wang Q, Jiang R, Huang L. Exploring the interaction between the gut microbiota and cyclic adenosine monophosphate-protein kinase A signaling pathway: a potential therapeutic approach for neurodegenerative diseases. Neural Regen Res 2025; 20:3095-3112. [PMID: 39589173 PMCID: PMC11881707 DOI: 10.4103/nrr.nrr-d-24-00607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/07/2024] [Accepted: 09/10/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut microbiota and cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling pathway in the host's central nervous system plays a crucial role in neurological diseases and enhances communication along the gut-brain axis. The gut microbiota influences the cAMP-PKA signaling pathway through its metabolites, which activates the vagus nerve and modulates the immune and neuroendocrine systems. Conversely, alterations in the cAMP-PKA signaling pathway can affect the composition of the gut microbiota, creating a dynamic network of microbial-host interactions. This reciprocal regulation affects neurodevelopment, neurotransmitter control, and behavioral traits, thus playing a role in the modulation of neurological diseases. The coordinated activity of the gut microbiota and the cAMP-PKA signaling pathway regulates processes such as amyloid-β protein aggregation, mitochondrial dysfunction, abnormal energy metabolism, microglial activation, oxidative stress, and neurotransmitter release, which collectively influence the onset and progression of neurological diseases. This study explores the complex interplay between the gut microbiota and cAMP-PKA signaling pathway, along with its implications for potential therapeutic interventions in neurological diseases. Recent pharmacological research has shown that restoring the balance between gut flora and cAMP-PKA signaling pathway may improve outcomes in neurodegenerative diseases and emotional disorders. This can be achieved through various methods such as dietary modifications, probiotic supplements, Chinese herbal extracts, combinations of Chinese herbs, and innovative dosage forms. These findings suggest that regulating the gut microbiota and cAMP-PKA signaling pathway may provide valuable evidence for developing novel therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Fengcheng Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lingxi Qing
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yifei Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jilian Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Meiling Jia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Qian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Runda Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihua Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
6
|
Lv LX, Yin JN, Sun YL, Wei MY, Jiang WQ, Gu YC, Yang XP, Shao CL. Marine natural products as potential anti-Pseudomonas aeruginosa agents: challenges and advances. Eur J Med Chem 2025; 292:117670. [PMID: 40305937 DOI: 10.1016/j.ejmech.2025.117670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
Antimicrobial resistance (AMR) has become a pressing need to address in the major global public health challenges, posing a serious threat to human health. Pseudomonas aeruginosa (PA) is one of the most concerning Gram-negative pathogens and is typically treated with broad-spectrum antibiotics. PA exhibits resistance to multiple antibiotics, multifactorial virulence, and dynamic hyperadaptation, which results in a particularly formidable challenge in eliminating PA from patients. The problem of drug resistance is becoming increasingly serious, and the development of new antibiotics is extremely lagging behind, resulting in no drug with a new structure and mechanism being approved for the treatment of infections caused by drug-resistant Gram-negative bacteria over the past half-century. Consequently, the development of new antibiotics is of utmost urgency and importance. Marine natural products (MNPs) have become an important source for developing new antibiotics due to their unique properties. So far, 44 potential molecules with significant anti-PA activity have been isolated from marine organisms, of which 19 have been reported as quorum-sensing system inhibitors (QSIs) with potential for further development. In this review, we provide a comprehensive summary of the current status of drug resistance, pathogenic mechanisms, and resistance mechanisms associated with PA infections. We also highlight the challenges and opportunities presented by MNPs in the development of anti-PA drugs, and offer recommendations to accelerate the antibiotic development process, thereby providing valuable insights for the study and exploitation of novel antibiotics.
Collapse
Affiliation(s)
- Liu-Xia Lv
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Jun-Na Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Yi-Lin Sun
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Wen-Qing Jiang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Xiao-Ping Yang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China.
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China; Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, People's Republic of China.
| |
Collapse
|
7
|
Liu W, Yang X, Zhou Y, Huang Z, Huang J. Gut microbiota in melanoma: Effects and pathogeneses. Microbiol Res 2025; 296:128144. [PMID: 40120565 DOI: 10.1016/j.micres.2025.128144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
The gut microbiota exhibits intricate connections with the body's immune system and holds significant implications for various diseases and cancers. Currently, accumulating evidence suggests a correlation between the composition of the gut microbiota and the development, treatment, and prognosis of melanoma. However, the underlying pathogenesis remains incompletely elucidated. In this comprehensive review, we present an in-depth review of the role played by gut microbiota in melanoma tumorigenesis, growth, metastasis, treatment response, and prognosis. Furthermore, we discuss the potential utility of gut microbiota as a promising prognostic marker. Lastly, we summarize three routes through which gut microbiota influences melanoma: immunity, aging, and the endocrine system. By modulating innate and adaptive immunity in patients with melanoma across different age groups and genders, the gut microbiota plays a crucial role in anti-tumor immune regulation from tumorigenesis to prognosis management, thereby impacting tumor growth and metastasis. This review also addresses current study limitations while highlighting future research prospects.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xin Yang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuwei Zhou
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ziru Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jian Huang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Hicks R, Gozal D, Ahmed S, Khalyfa A. Interplay between gut microbiota and exosome dynamics in sleep apnea. Sleep Med 2025; 131:106493. [PMID: 40203611 DOI: 10.1016/j.sleep.2025.106493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
Sleep-disordered breathing (SDB) is characterized by recurrent reductions or interruptions in airflow during sleep, termed hypopneas and apneas, respectively. SDB impairs sleep quality and is linked to substantive health issues including cardiovascular and metabolic disorders, as well as cognitive decline. Recent evidence suggests a link between gut microbiota (GM) composition and sleep apnea. Indeed, GM, a community of microorganisms residing in the gut, has emerged as a potential player in various diseases, and several studies have identified associations between sleep apnea and GM diversity along with shifts in bacterial populations. Additionally, the concept of "leaky gut," a compromised intestinal barrier with potentially increased inflammation, has emerged as another key player in the potential bidirectional relationship between GM and sleep apnea. One of the potential effectors could be extracellular vesicles (EVs) underlying gut-brain communication pathways that are relevant to sleep regulation and function. Thus, therapeutic implications afforded by targeting the GM or exosomes for sleep apnea management have surfaced as promising areas of research. This review explores current understanding of the relationship between GM, exosomes and sleep apnea, highlighting key research dynamics and potential mechanisms. A comprehensive review of the literature was conducted, focusing on studies investigating GM composition, intestinal barrier function and gut-brain communication in relation to sleep apnea.
Collapse
Affiliation(s)
- Rebecca Hicks
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Sarfraz Ahmed
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
9
|
Tüsüz Önata E, Özdemir Ö. Fecal microbiota transplantation in allergic diseases. World J Methodol 2025; 15:101430. [DOI: 10.5662/wjm.v15.i2.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024] Open
Abstract
Microorganisms such as bacteria, fungi, viruses, parasites living in the human intestine constitute the human intestinal microbiota. Dysbiosis refers to compositional and quantitative changes that negatively affect healthy gut microbiota. In recent years, with the demonstration that many diseases are associated with dysbiosis, treatment strategies targeting the correction of dysbiosis in the treatment of these diseases have begun to be investigated. Faecal microbiota transplantation (FMT) is the process of transferring faeces from a healthy donor to another recipient in order to restore the gut microbiota and provide a therapeutic benefit. FMT studies have gained popularity after probiotic, prebiotic, symbiotic studies in the treatment of dysbiosis and related diseases. FMT has emerged as a potential new therapy in the treatment of allergic diseases as it is associated with the maintenance of intestinal microbiota and immunological balance (T helper 1/T helper 2 cells) and thus suppression of allergic responses. In this article, the definition, application, safety and use of FMT in allergic diseases will be discussed with current data.
Collapse
Affiliation(s)
- Ece Tüsüz Önata
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| | - Öner Özdemir
- Division of Pediatric Allergy and Immunology, Medical Faculty, Sakarya University, Adapazarı 54100, Sakarya, Türkiye
| |
Collapse
|
10
|
Rocabert A, Martín-Pérez J, Pareras L, Egea R, Alaraby M, Cabrera-Gumbau JM, Sarmiento I, Martínez-Urtaza J, Rubio L, Barguilla I, Marcos R, García-Rodríguez A, Hernández A. Nanoplastic exposure affects the intestinal microbiota of adult Drosophila flies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 980:179545. [PMID: 40311335 DOI: 10.1016/j.scitotenv.2025.179545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/27/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Micro- and nanoplastics (MNPLs) are emerging environmental pollutants that have garnered significant attention over the past few decades due to their detrimental effects on human health through various exposure pathways. This study investigates the impact of MNPLs on gut microbiota, utilizing Drosophila melanogaster as a model organism. Drosophila was selected for its microbiota's similarities to humans and its established role as an accessible and well-characterized model system. To analyze microbiota, full-length 16S rRNA gene sequencing was performed using the Nanopore sequencing platform, enabling comprehensive profiling of the microbial populations present in the samples. As models of MNPLs, two commercial polystyrene nanoplastics (PS-NPLs, 61.20 and 415.22 nm) and one lab-made polylactic acid nanoplastic (PLA-NPLs, 463.90 nm) were selected. As a positive control, zinc oxide nanoparticles (ZnO-NPs) were used. The observed findings revealed that exposure to MNPLs induced notable alterations in gut microbiota, including a reduction in bacterial abundance and shifts in species composition. These results suggest that MNPLs exposure can lead to microbial dysbiosis and potential gut health disruptions through its interaction, either with the gut epithelial barrier or directly with the resident microorganisms.
Collapse
Affiliation(s)
- Arnau Rocabert
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Joan Martín-Pérez
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Laia Pareras
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Raquel Egea
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Mohamed Alaraby
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jordi Manuel Cabrera-Gumbau
- Group of Genomics, Bioinformatics & Evolutionary Biology, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Iris Sarmiento
- Group of Genomics, Bioinformatics & Evolutionary Biology, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jaime Martínez-Urtaza
- Group of Genomics, Bioinformatics & Evolutionary Biology, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Laura Rubio
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Irene Barguilla
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ricard Marcos
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Alba García-Rodríguez
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Alba Hernández
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| |
Collapse
|
11
|
Chen J, Li H, Long X, Tong H, Xin X, Zhang H, Li Y, Liu P, He X, Chen Z. Causal association between gut microbiota and endometrial cancer in European and East Asian populations: a two-sample Mendelian randomization study. BMC Womens Health 2025; 25:283. [PMID: 40483441 PMCID: PMC12145594 DOI: 10.1186/s12905-025-03789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a significant global health concern. While observational epidemiological studies suggest a potential link between gut microbiota dysbiosis and the development of EC, the direction and causality of this association remain uncertain. METHODS We performed Mendelian randomization (MR) analysis to investigate the causal relationship between gut microbiota and EC. Exposure data were obtained from the MiBioGen study consortium (N = 18,340), and outcome data were sourced from the IEU OpenGWAS database, specifically datasets "ebi-a-GCST006464" (N = 121,885) and "bbj-a-113" (N = 90,730). The inverse variance-weighted(IVW) method was applied to evaluate the association between gut microbiota composition and EC risk. Sensitivity analyses were conducted to ensure the robustness of the findings. RESULTS Our study identified several microbial taxa linked to EC risk. In Europeans, genera such as Marvinbryantia, RuminococcaceaeUCG014, and Dorea exhibited protective effects, while family Erysipelotrichaceae (OR:1.224) and FamilyXI (OR:1.090) were significantly correlated with high EC risk. In East Asians, genera Lachnospira (OR:3.561) and family Bifidobacteriaceae (OR:1.715) were found associated with EC risk. Genera Lachnoclostridium and ErysipelotrichaceaeUCG003, family Coriobacteriaceae positively served as protective factors. Sensitivity analyses confirmed the reliability of our results, and there was no evidence of pleiotropy or heterogeneity. Our analysis identified several microbial taxa associated with EC risk. In Europeans, genera such as Marvinbryantia, Ruminococcaceae UCG014, and Dorea demonstrated protective effects, while the families Erysipelotrichaceae (OR: 1.224) and FamilyXI (OR: 1.090) were significantly associated with increased EC risk. In East Asians, the genus Lachnospira (OR: 3.561) and the family Bifidobacteriaceae (OR: 1.715) were linked to higher EC risk, whereas the genera Lachnoclostridium and Erysipelotrichaceae UCG003 and the family Coriobacteriaceae were identified as protective factors. Sensitivity analyses confirmed the reliability of these results, with no evidence of pleiotropy or heterogeneity. CONCLUSION This study highlights a relationship between gut microbiota and EC, emphasizing the potential of gut microbiota as therapeutic targets and biomarkers for assessing EC prognosis and treatment efficacy. These findings provide novel insights into the role of gut microbiota in the development and progression of EC.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
- Department of Radiology, Union Hospital, Tongji Medical College,, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan, 430022, China
| | - Haiqing Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xinrui Long
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Tong
- Department of Spine Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Xin Xin
- Department of Orthopedics of Yan'an Hospital, Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Han Zhang
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yeyao Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Ping Liu
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaolin He
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhuoyuan Chen
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China.
- Department of Orthopedics of Yan'an Hospital, Affiliated to Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
12
|
Ortiz AM, Casta FC, Bodykevich EG, Flynn JK, Fennessey CM, Brooks K, Ruiz D, Yee DS, Simpson J, Rahmberg AR, Keele BF, Brenchley JM. Repeated enema administration in rhesus macaques is not sufficient to promote bacterial dysbiosis or gastrointestinal dysfunction. Mucosal Immunol 2025:S1933-0219(25)00058-3. [PMID: 40490097 DOI: 10.1016/j.mucimm.2025.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 06/04/2025] [Accepted: 06/04/2025] [Indexed: 06/11/2025]
Abstract
Chronic gastrointestinal diseases are a significant global health burden that can require the use of gastrointestinal-cleansing regimens for diagnostics or therapeutic treatment. These regimens are beneficial for facilitating surgical preparation, drug delivery, colorectal cancer screenings, and personal use is common among proponents of natural health and among certain populations at high risk of HIV acquisition. It remains unclear, however, whether repeated clearance of the colonic microbiome induces persistent changes in the microbiome, intestinal immunity, and viral disease susceptibility. We addressed these parameters by repeatedly administering iso-osmolar enemas to rhesus macaques prior to low-dose intra-rectal challenge with simian immunodeficiency virus (SIV). Considering both longitudinal and cross-sectional analyses, we observed no consistent changes in the fecal microbiome or intestinal immune parameters of treated animals, nor were significant differences observed in susceptibility to SIV acquisition. Unexpectedly, enema-treated animals exhibited significantly lower setpoint viral loads after infection, although we were unable to clearly identify attributing causes. Our study demonstrates that repeated microbiome clearance using clinically administered iso-osmolar enemas is not sufficient to restructure the fecal microbiome, perturb intestinal immune parameters, or increase susceptibility to mucosal SIV challenge. This research framework serves as a model for the development of colonic-administered diagnostics and interventions.
Collapse
Affiliation(s)
- Alexandra M Ortiz
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fabiola Castello Casta
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth G Bodykevich
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacob K Flynn
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Delmy Ruiz
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Debra S Yee
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jennifer Simpson
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew R Rahmberg
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
14
|
Sogard AS, Emerson TS, Chandler CA, Cobb EA, Shei RJ, Paris HL, Lindley MR, Mickleborough TD. The role of nutritional factors in exercise-induced bronchoconstriction: a narrative review. Am J Physiol Regul Integr Comp Physiol 2025; 328:R651-R684. [PMID: 40257056 DOI: 10.1152/ajpregu.00249.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/20/2024] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
Exercise-induced bronchoconstriction (EIB) describes an acute narrowing of the airways that develops following vigorous physical activity. Clinical responses to current asthma therapy, such as leukotriene antagonists and corticosteroids, are heterogeneous, even with optimal treatment. Epidemiological studies indicate an increasing use of complementary and alternative medicine therapy in asthma patients due to the lack of efficacy of conventional treatment, concerns about potentially harmful side effects of pharmacological treatment, cost barriers to asthma care, and the accessibility of complementary and alternative medicine therapy. Plausible physiological mechanisms now exist for many nutrients as potential modifiers of EIB severity, primarily because of their role in inflammatory processes, airway smooth muscle function, and modulation of lung microvascular volume and pressure. Dietary supplementation as a treatment for EIB has generally shown evidence of significant yet incomplete inhibition of EIB with low-salt diets, omega-3 fatty acids, and vitamin C when supplemented for up to 3 weeks. However, larger, randomized, placebo-controlled, double-blinded trials are needed to clarify the effectiveness of nutritional intervention in individuals with EIB. Additionally, many studies have focused on nonathletes with EIB, and therefore, more studies are required to evaluate the efficacy of nutritional intervention on EIB in elite athletes. In conclusion, if dietary supplementation or restriction is prescribed, it should be seen as an option to lessen the reliance on pharmaceutical interventions and not as an alternative to established pharmacotherapies.
Collapse
Affiliation(s)
- Abigail S Sogard
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Travis S Emerson
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Christopher A Chandler
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Emily A Cobb
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Ren-Jay Shei
- Indiana University Alumni Association, Indiana University, Bloomington, Indiana, United States
| | - Hunter L Paris
- Department of Sports Medicine, Pepperdine University, Malibu, California, United States
| | - Martin R Lindley
- School of Health Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Timothy D Mickleborough
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
15
|
Fehringer M, Vogl T. Molecular mimicry in the pathogenesis of autoimmune rheumatic diseases. J Transl Autoimmun 2025; 10:100269. [PMID: 39877080 PMCID: PMC11773492 DOI: 10.1016/j.jtauto.2025.100269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Autoimmune rheumatic diseases (ARDs) are a heterogeneous group of conditions characterized by excessive and misdirected immune responses against the body's own musculoskeletal tissues. Their exact aetiology remains unclear, with genetic, demographic, behavioural and environmental factors implicated in disease onset. One prominent hypothesis for the initial breach of immune tolerance (leading to autoimmunity) is molecular mimicry, which describes structural or sequence similarities between human and microbial proteins (mimotopes). This similarity can lead to cross-reactive antibodies and T-cell receptors, resulting in an immune response against autoantigens. Both commensal microbes in the human microbiome and pathogens can trigger molecular mimicry, thereby potentially contributing to the onset of ARDs. In this review, we focus on the role of molecular mimicry in the onset of rheumatoid arthritis and systemic lupus erythematosus. Moreover, implications of molecular mimicry are also briefly discussed for ankylosing spondylitis, systemic sclerosis and myositis.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| |
Collapse
|
16
|
Ngana GS, Di Bernardo MA, Surette MG, MacNeil LT. Actinomyces viscosus promotes neuroprotection in C. elegans models of Parkinson's disease. Mech Ageing Dev 2025; 225:112061. [PMID: 40258426 DOI: 10.1016/j.mad.2025.112061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/04/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Parkinson's Disease is characterized by selective degeneration of dopaminergic neurons, primarily in the substantia nigra pars compacta, as well as accumulation of alpha-synuclein enriched protein aggregates within neurons. The pathogenesis of PD is still not completely understood, and no treatments exist that alter disease progression. Obvious genetic causes are detected in only a small number of PD patients (5-10 %), suggesting that environmental factors play a significant role the development of PD. Correlative studies suggest that the microbiota could be an important environmental modifier of neurodegeneration. We identified a microbiotal isolate, Actinomyces viscosus, that reduced neurodegeneration in C. elegans expressing a pathological mutant form (G2019S) of leucine-rich repeat kinase 2 (LRRK2) in dopaminergic neurons. A. viscosus also suppressed autophagic dysfunction in these animals and reduced alpha-synuclein aggregation in a synucleinopathy model. Global gene expression analysis revealed increased expression of aspartic cathepsins in response to A. viscosus. Consistent with the involvement of these proteins in neuroprotection, we found that reducing aspartic cathepsin function increased neurodegeneration in the LRRK2 transgenic model. Our findings contribute to the current understanding of how the gut microbiota may influence PD, elucidating one potential mechanism of microbiota-mediated neuroprotection.
Collapse
Affiliation(s)
- G Sophie Ngana
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Mercedes A Di Bernardo
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada; Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W., Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton ON, Canada.
| |
Collapse
|
17
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Ko B, Son J, In Won J, Kang BM, Choi CW, Kim R, Sung JH. Gut microbe-skin axis on a chip for reproducing the inflammatory crosstalk. LAB ON A CHIP 2025; 25:2609-2619. [PMID: 40042226 DOI: 10.1039/d4lc01010h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
The gut-skin axis has emerged as a crucial mediator of skin diseases, with mounting evidence highlighting the influence of gut microbiota on skin health. However, investigating these mechanisms has been hindered by the lack of experimental systems that enable direct study of gut microbiota-skin interactions. Here, we present the gut microbe-skin chip (GMS chip), a novel microfluidic platform designed to model microbiome-gut-skin axis interactions. The GMS chip allows the coculture of intestinal epithelial cells (Caco-2), human epidermal keratinocytes (HEKa), and gut microbes with fluidic connection mimicking the blood flow. We validated that the gut compartment, with a self-sustaining oxygen gradient, enabled coculturing gut bacteria such as Escherichia coli (E. coli) and Lactobacillus rhamnosus GG (LGG), and the skin cells properly differentiated in the chip in the presence of fluid flow. Disruption of intestinal epithelial integrity by dextran sodium sulfate (DSS) combined with lipopolysaccharides (LPS) selectively decreased skin cell viability while sparing gut cells. Notably, pretreatment with LGG showed a protective effect against the skin cell damage by enhancing the intestinal barrier function. The GMS chip effectively recapitulates the influence of gut microbiota on skin health, representing a pivotal step forward in studying gut-skin axis mechanisms and the role of the gut microbiome in skin diseases.
Collapse
Affiliation(s)
- Byungho Ko
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Jimin Son
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Jong In Won
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| | - Bo Mi Kang
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Chong Won Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, Republic of Korea.
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea.
| |
Collapse
|
19
|
Imoto N, Kano C, Morita H, Hirota T, Amanuma F, Maruyama H, Nojiri S, Watanabe S. Impact of antimicrobial exposure at delivery and siblings on early Bifidobacterium succession and allergy development up to 24 months of age. BMC Microbiol 2025; 25:332. [PMID: 40426074 PMCID: PMC12117752 DOI: 10.1186/s12866-025-04056-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Allergic diseases such as asthma, eczema, and food allergies are rising globally. The infant gut microbiota, particularly the dominance of Bifidobacterium, shapes immune development and allergy risk. In Japan-where Bifidobacterium prevalence is notably high-longitudinal investigations focusing on the pre-weaning period, when external influences are relatively limited, remain scarce. Therefore, based on consistent hypotheses and findings from previous studies, we investigated how two important early factors-antibiotic exposure at birth and the presence of older siblings-influence the gut environment in early infancy and subsequent allergy development. RESULTS In a prospective cohort of 121 Japanese infants, stool samples were collected at seven time points from birth through 24 months. We quantified the relative abundances of Bifidobacterium, Bacteroides, Clostridium, and Faecalibacterium and recorded allergic outcomes at 2 years. Both antimicrobial exposure at delivery and sibling presence significantly altered gut microbiota composition and overall diversity in early infancy. Although the full cohort showed no consistent diversity or Bifidobacterium differences by allergic status, in several subgroups where these two factors were excluded, infants who had an allergy by 24 months exhibited marked shifts in early gut microbiota community structure-particularly in beta diversity-and reduced Bifidobacterium occupancy during the pre-weaning period (1-6 months) versus non-allergic peers. Moreover, infants whose gut microbiota was initially affected by these factors showed a recovery in diversity after weaning, a rebound more pronounced in non-allergic individuals. CONCLUSIONS These findings indicate that both the initial community configuration and its capacity to rebound after perturbation are critical determinants of allergy risk. By focusing on dynamic changes through weaning and adjusting for decisive confounders, this study refines insight beyond prior cross-sectional work. Early interventions that preserve or restore microbial diversity and Bifidobacterium dominance may therefore offer a promising strategy to mitigate allergic disease development.
Collapse
Affiliation(s)
- Naruaki Imoto
- Advanced Research Institute for Health Science, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan.
| | - Chie Kano
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Hiroto Morita
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Tatsuhiko Hirota
- Core Technology Laboratories, Asahi Quality & Innovations Ltd, Midori, Moriya, 302- 0106, Ibaraki, Japan
| | - Fumitaka Amanuma
- Department of Paediatrics, Department of Neonatology, Iwate Prefectural Iwai Hospital, Ichinoseki, 029-0192, Iwate, Japan
| | - Hidekazu Maruyama
- Department of Paediatrics, Department of Neonatology, Iwate Prefectural Iwai Hospital, Ichinoseki, 029-0192, Iwate, Japan
| | - Shuko Nojiri
- Clinical Research Support Centre, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan
| | - Shin Watanabe
- Advanced Research Institute for Health Science, Juntendo University, Bunkyo Ward, Tokyo, 113-8421, Japan
| |
Collapse
|
20
|
Liu L, Guo L, Dai J, Cai X, Wu B. Fecal 16S rRNA sequencing and metabolomics reveal abnormal metabolism activity in preterm infants with different gestational ages. Front Cell Infect Microbiol 2025; 15:1530653. [PMID: 40491434 PMCID: PMC12146292 DOI: 10.3389/fcimb.2025.1530653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/28/2025] [Indexed: 06/11/2025] Open
Abstract
Objective This study aims to conduct a comprehensive analysis of the differences in gut microbiota and metabolomics in preterm infants stratified by gestational age. Methods Fresh fecal samples were collected from neonates within the first 3 days after birth. The gut microbiota composition and the changes in specific taxa abundance were analyzed using 16S rRNA sequencing. Metabolomic profiling was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Participants were categorized into four groups based on gestational age at birth: PreA group (34-36 weeks), PreB group (32-33 weeks), PreC group (28-31 weeks), and control group (37-42 weeks). Metabolic pathways were identified through metabolomics analysis, referencing the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Results Notably, Principal Coordinates Analysis (PCoA) showed clear separation among samples from all groups, with significant differences noted in the PreC group when compared with the other three. We found a strong association between Escherichia-Shigella and Ureaplasma genera with infants born before 32 weeks of gestation, suggesting a higher risk of opportunistic infections for preterm infants under this gestational threshold. As gestational age increases, Megamonas and Prevotella gradually emerged, while Escherichia-Shigella and Ureaplasma progressively diminished. KEGG enrichment analysis indicated that Pyrimidine metabolism was a differentially regulated pathway between the PreA group and the control group. Interestingly, the only major differential metabolic pathway between the PreB group and the control group was Arachidonic acid metabolism. The bubble diagram revealed significant enrichment of differential metabolites in Pyrimidine and beta-Alanine metabolism pathways when comparing the PreC group with the control group. Conclusion Significant differences were observed in the fecal microbiome and metabolome between preterm and full-term infants, particularly in those born before 32 weeks of gestation. These findings suggested that the gut microbial system in preterm infants undergone progressive maturation, approaching a "healthy" state characteristic of full-term infants as gestational age increases.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Liang Guo
- Department of Neonatology, Guangming District People’s Hospital, Shenzhen, China
| | - Jincheng Dai
- Department of Pediatrics, Guangming District People’s Hospital, Shenzhen, China
| | - Xiangsheng Cai
- Clinical Laboratory, Guangzhou Eleventh People’s Hospital, Guangzhou Carde and Talent Health Management Center, Guangzhou, China
| | - Benqing Wu
- Department of Neonatology, Guangming District People’s Hospital, Shenzhen, China
- Department of Pediatrics, Guangming District People’s Hospital, Shenzhen, China
| |
Collapse
|
21
|
Jan A, Bayle P, Mohellibi N, Lemoine C, Pepke F, Béguet-Crespel F, Jouanin I, Tremblay-Franco M, Laroche B, Serror P, Rigottier-Gois L. A consortium of seven commensal bacteria promotes gut microbiota recovery and strengthens ecological barrier against vancomycin-resistant enterococci. MICROBIOME 2025; 13:129. [PMID: 40414934 DOI: 10.1186/s40168-025-02127-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 04/29/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Vancomycin-resistant enterococci (VRE) often originate from the gastrointestinal tract, where their proliferation precedes dissemination into the bloodstream, and can lead to systemic infection. Uncovering the actors and mechanisms reducing the intestinal colonisation by VRE is essential to control infection. We aimed to identify commensal bacteria that interfere with VRE gut colonisation or act as an ecological barrier. RESULTS We performed a 3-week longitudinal analysis of the gut microbiota composition and VRE carriage levels during microbiota recovery in mice colonised with VRE after antibiotic-induced dysbiosis. By combining biological data and mathematical modelling, we identified 15 molecular species (OTUs) that negatively correlated with VRE overgrowth. Six strains representative of these OTUs were collected, cultivated and used in mixture with a seventh strain (Mix7) in two different mouse lines challenged with VRE. Of the seven strains, three belonged to Lachnospiraceae, one to Muribaculaceae, one to Ruminococcaceae and two to Lactobacillaceae. We found that Mix7 led to a better recovery of the gut microbiota composition and reduced VRE carriage. Differences in the effect of Mix7 were observed between responder and non-responder mice. These differences were associated with variations in the composition of the initial microbiota and during recovery and represent potential biomarkers for predicting response to Mix7. In a mouse model of alternative stable state of dysbiosis, response to Mix7 was associated with higher concentrations of short-chain fatty acids (acetate, propionate, butyrate) and a range of metabolites including bile acids, reflecting the recovery of the microbiota back to initial state. Furthermore, Muribaculum intestinale strain was required to obtain the Mix7 effect on VRE reduction in vivo, but the presence of at least one of the other six strains was needed. None of the supernatant of the seven strains, alone or in combination, inhibited VRE growth in vitro. Interestingly, five strains belong to species shared among humans and mice, and the other two have human functional equivalents. CONCLUSIONS An innovative approach based on mathematical modelling of the microbiota composition permitted to identify a mixture of commensal bacterial strains, which improves the ecological barrier effect against VRE. The mechanisms are dependent on the recovery and initial composition of the microbiota. Ultimately, this work will enable a move towards a personalised medicine by targeting predisposed patients presenting a risk of infection, such as neutropenic or bone-marrow transplant patients, and likely to respond to supplementation with commensal strains, providing new live biotherapeutic products and biomarkers to predict response to supplementation. Video Abstract.
Collapse
Affiliation(s)
- Alan Jan
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Perrine Bayle
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Nacer Mohellibi
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Clara Lemoine
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Frédéric Pepke
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Fabienne Béguet-Crespel
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Isabelle Jouanin
- Toxalim - Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, Toulouse University, UT3, Toulouse, F-31300, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | - Marie Tremblay-Franco
- Toxalim - Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, Toulouse University, UT3, Toulouse, F-31300, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | - Béatrice Laroche
- MaIAGE, INRAE, Université Paris-Saclay, Jouy-en-Josas, 78350, France
- MUSCA, INRIA, Université Paris-Saclay, Palaiseau, 91120, France
| | - Pascale Serror
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Lionel Rigottier-Gois
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France.
| |
Collapse
|
22
|
Dash HR, Patel A. Genealogically bewildered individuals and forensic identification: a review of current and emerging solutions. Int J Legal Med 2025:10.1007/s00414-025-03513-2. [PMID: 40411594 DOI: 10.1007/s00414-025-03513-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 05/10/2025] [Indexed: 05/26/2025]
Abstract
The increasing use of assisted reproductive technologies (ART) with donor gametes is driven by rising infertility rates, delayed parenthood, and the need to prevent hereditary diseases. Greater social acceptance of diverse family structures, advancements in reproductive medicine, and improving success rates also contribute. Accessibility, affordability, and cross-border reproductive care further expand ART's reach, making donor gametes a preferred option for many individuals and couples worldwide. The widespread application of ART has led to an increasing number of donor-conceived individuals, many of whom are now reaching reproductive maturity. This demographic shift introduces significant challenges for traditional forensic genetic identification methods, which rely on biological reference samples from genetically related individuals. The absence of such samples complicates the identification process, particularly for individuals conceived via gamete donation or adoption, where biological and legal parentage are incongruent. Conventional forensic genetic analyses, including short tandem repeat (STR) and single nucleotide polymorphism (SNP) profiling of autosomal, Y-chromosome, X-chromosome, and mitochondrial DNA, exhibit limited efficacy in these scenarios. While these methods can sometimes identify individuals conceived using a single donor gamete, they are insufficient for cases involving dual donor gametes or mitochondrial replacement therapy. Emerging methodologies such as forensic genetic genealogy, DNA methylation profiling, and human microbiome analysis offer innovative approaches but necessitate further clinical validation and standardization.
Collapse
Affiliation(s)
- Hirak Ranjan Dash
- Department of Forensic Science, National Forensic Sciences University, Delhi Campus, New Delhi, 110085, India.
- School of Forensic Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, 752050, India.
| | - Anubhuti Patel
- Department of Reproductive Medicine and the Center for Human Reproduction, IMS and SUM Hospital, Bhubaneswar, Odisha, 751003, India
| |
Collapse
|
23
|
Quijano L, Fischer D, Ferrero-Regis T, Navone L. Exploring bacterial cellulose as an engineered living and programmable biomaterial across disciplines through qualitative thematic analysis. Sci Rep 2025; 15:17980. [PMID: 40410284 PMCID: PMC12102144 DOI: 10.1038/s41598-025-01931-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
Bacterial cellulose is an engineered living material which holds significant potential due to its material properties and broad applicability across scientific and design disciplines. However, challenges in interdisciplinary collaboration, scalability and commercialization have slowed its widespread adoption and integration into industry applications such as fashion and textiles. This study addresses the gap in understanding how bacterial cellulose is perceived, developed, and utilized across scientific and design disciplines. Through 20 semi-structured interviews with scientists and designers around the world, this paper explores the following themes: (1) the human-living material relationship spectrum, which highlights the distinct ways science and design stakeholders interact with bacterial cellulose as a living material; (2) perceptions of a living material made from bacteria; and (3) bacterial cellulose's potential as a programmable biomaterial. Additionally, we employ Bruno Latour's Actor-Network Theory to map the complex network of human and non-human actors shaping bacterial cellulose's trajectory, identifying critical factors such as consumer acceptance, interdisciplinary collaboration, and material culture. By bridging perspectives from science and design, this study offers actionable insights into bacterial cellulose's future as a sustainable and programmable engineered living material, guiding its responsible development and broader adoption across industries.
Collapse
Affiliation(s)
- Luis Quijano
- Faculty of Creative Industries, Education and Social Justice, Queensland University of Technology (QUT), Brisbane, QLD, 4059, Australia.
- Faculty of Science, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.
- ARC Centre of Excellence in Synthetic Biology, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.
| | - Dagmar Fischer
- Division of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstraße 4, 91058, Erlangen, Germany
- FAU NeW-Research Center for New Bioactive Compounds, Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Tiziana Ferrero-Regis
- Faculty of Creative Industries, Education and Social Justice, Queensland University of Technology (QUT), Brisbane, QLD, 4059, Australia
| | - Laura Navone
- Faculty of Science, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
- ARC Centre of Excellence in Synthetic Biology, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| |
Collapse
|
24
|
Ley D, Saha S. Menopause and gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01075-7. [PMID: 40410564 DOI: 10.1038/s41575-025-01075-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2025] [Indexed: 05/25/2025]
Abstract
Menopause has far-reaching effects on human physiology, including the gastrointestinal tract, and can negatively influence the quality of life of women who are affected. Menopause can have multiple effects on gastrointestinal function, including altering gut motility and changing the composition of the gut microbiota. As a result, some gastrointestinal and hepatic conditions are more common among individuals in peri- and postmenopause, and people with these conditions before menopause might also experience greater symptom severity and worse health-related quality of life during this time. The use of hormone replacement therapy to treat menopausal symptoms might also affect gastrointestinal health and well-being. Individuals in postmenopause are at risk for bone remodelling and osteoporosis due to ageing and loss of sex hormones. However, secondary osteoporosis can also occur due to medications used to treat gastrointestinal conditions (for example, glucocorticoids and other immunosuppressive medications) and the conditions themselves (for example, autoimmune disease or coeliac disease). Although all people who menstruate will eventually transition to menopause, there are relatively few studies evaluating the effect of menopause on gastrointestinal symptoms and quality of life. This Review aims to summarize available evidence and highlight areas where research is needed.
Collapse
Affiliation(s)
- Dana Ley
- Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sumona Saha
- Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
25
|
Turnier JL, Canna SW. Insights from the 2024 pediatric rheumatology basic/translational years in review. Pediatr Rheumatol Online J 2025; 23:57. [PMID: 40410775 PMCID: PMC12100868 DOI: 10.1186/s12969-025-01102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Advances in Pediatric Rheumatology are driven by mechanistic insights from basic and translational science. We have selected and reviewed the most impactful basic/translational science from our "Year in Review (YIR)" presentations from the 2024 Pediatric Rheumatology European Society and American College of Rheumatology Convergence meetings (September and November 2024, respectively). MAIN BODY We drew from fundamental immunology, human genetics, animal models, and computational & "omic" manuscripts published in the year preceding these meetings. Avoiding overlap with other topics presented in this "Perspectives" series, summarized herein are the major themes we gleaned from that process. These include (1) innovative concepts and tools to study immune health, (2) new mechanistic insights into pediatric rheumatic diseases and (3) novel therapeutic targets and treatment approaches in rheumatic disease. CONCLUSIONS As part of a living relationship with the basic/translational literature that shapes our field and practice, we hope readers will be inspired to delve more deeply into the topics and manuscripts highlighted in this YIR summary.
Collapse
Affiliation(s)
- Jessica L Turnier
- Division of Pediatric Rheumatology, Department of Pediatrics, C.S. Mott Children's Hospital, University of Michigan, 2800 Plymouth Road NCRC Building 20, Rm 1842, Ann Arbor, Michigan, 48109, USA.
| | - Scott W Canna
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
- Rheumatology and Immune Dysregulation Program, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| |
Collapse
|
26
|
Wang R, Ma F, Yin D, Wang H, Wei X. Intestinal Microbes, Metabolites, and Hormones in Alcohol-Associated Liver Disease. Semin Liver Dis 2025. [PMID: 40334703 DOI: 10.1055/a-2601-9480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Alcohol-associated liver disease (ALD)-encompassing conditions including steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma-refers to hepatic damage arising from excessive or hazardous alcohol consumption, and is now recognized as a significant global health burden. Although the mechanisms underlying ALD remain incompletely understood, several pathways have been substantiated over the last five decades, notably the involvement of intestinal microorganisms and the involvement of the gut-liver axis in alcohol metabolism and ALD pathogenesis. Ethanol intake disrupts the intestinal microbial balance and compromises the gut barrier, resulting in increased permeability to microbial products. The subsequent translocation of microbial metabolites and other antigenic substances to the liver activates hepatic immune responses, thereby contributing to liver injury. In addition, gastrointestinal hormones are also implicated in ALD progression through various mechanisms. Although no therapies for ALD have been approved by the Food and Drug Administration, various therapeutic strategies targeting the intestinal microbiota and gut barrier have been identified. In conclusion, this review discusses the role of the gut-liver axis in alcohol metabolism and ALD pathogenesis and explores the emerging therapeutic strategies.
Collapse
Affiliation(s)
- Ruimeng Wang
- Second Clinical Medical College, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Dou Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaohui Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Hillman EBM, Baumgartner M, Carson D, Amos GCA, Wazir I, Khan HA, Khan MA, Rijpkema S, Walters JRF, Wellington EMH, Arasaradnam R, Lewis SJ. Changing Gastrointestinal Transit Time Alters Microbiome Composition and Bile Acid Metabolism: A Cross-Over Study in Healthy Volunteers. Neurogastroenterol Motil 2025:e70075. [PMID: 40394972 DOI: 10.1111/nmo.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/21/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND The specific influence of whole gut transit time (WGTT) on microbiome dynamics and bile acid metabolism remains unclear, despite links between changes in WGTT and certain gastrointestinal disorders. Our investigation aimed to determine the impact of WGTT changes on the composition of the fecal microbiome and bile acid profile. METHODS Healthy volunteers (n = 18) received loperamide, to decrease bowel movement frequency, and senna, a laxative, each over a 6-day period, in a randomized sequence, with a minimum 16-day interval between each treatment. Stool samples were analyzed for microbiome by shotgun sequencing and bile acid composition determined with high-performance liquid chromatography coupled to tandem mass spectrometry. Sera were examined for markers of bile acid synthesis. KEY RESULTS Senna or loperamide decreased or increased WGTT, respectively. Treatment altered stool characteristics, bowel movement frequency, and stool weight. The senna-treated group had increased primary and secondary fecal bile acids; serum levels of fibroblast growth factor 19 were significantly reduced. Increasing WGTT with loperamide led to an increase in bile salt hydrolase genes, along with elevated bacterial species richness (p = 0.04). Thirty-six species exhibiting significant differences were identified, several of which have notable implications for gut health. WGTT displayed negative correlations with total primary (particularly chenodeoxycholic acid) and secondary bile acids (ursodeoxycholic acid and glycochenodeoxycholic acid). Treatment-induced changes in microbiome composition and bile acid metabolism reverted back to baseline within 16 days. CONCLUSION Whole gut transit time changes significantly affect fecal microbiome composition and function, as well as bile acid composition and synthesis in healthy subjects. This consideration is likely to have long-term implications.
Collapse
Affiliation(s)
- Evette B M Hillman
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - Maximilian Baumgartner
- Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Danielle Carson
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Gregory C A Amos
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Imad Wazir
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Haider A Khan
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Malik A Khan
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Sjoerd Rijpkema
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Julian R F Walters
- Division of Digestive Diseases, Imperial College London, London, UK
- Imperial College Healthcare Trust, London, UK
| | | | - Ramesh Arasaradnam
- Department of Gastroenterology, University Hospitals Coventry & Warwickshire, Coventry, UK
- Warwick Medical School, The University of Warwick, Coventry, UK
| | - Stephen J Lewis
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| |
Collapse
|
28
|
Lopez JA, McKeithen-Mead S, Shi H, Nguyen TH, Huang KC, Good BH. Abundance measurements reveal the balance between lysis and lysogeny in the human gut microbiome. Curr Biol 2025; 35:2282-2294.e11. [PMID: 40300605 DOI: 10.1016/j.cub.2025.03.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/27/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
The human gut contains diverse communities of bacteriophage, whose interactions with the broader microbiome and potential roles in human health are only beginning to be uncovered. Here, we combine multiple types of data to quantitatively estimate gut phage population dynamics and lifestyle characteristics in human subjects. Unifying results from previous studies, we show that an average human gut contains a low ratio of phage particles to bacterial cells (∼1:100) but a much larger ratio of phage genomes to bacterial genomes (∼4:1), implying that most gut phage are effectively temperate (e.g., integrated prophage and phage-plasmids). By integrating imaging and sequencing data with a generalized model of temperate phage dynamics, we estimate that phage induction and lysis occur at a low average rate (∼0.001-0.01 per bacterium per day), imposing only a modest fitness burden on their bacterial hosts. Consistent with these estimates, we find that the phage composition of a diverse synthetic community in gnotobiotic mice can be quantitatively predicted from bacterial abundances alone while still exhibiting phage diversity comparable to native human microbiomes. These results provide a foundation for interpreting existing and future studies on links between the gut virome and human health.
Collapse
Affiliation(s)
- Jamie Alcira Lopez
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Saria McKeithen-Mead
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Handuo Shi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taylor H Nguyen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Benjamin H Good
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
29
|
Liu D, Ton PM, Zong D, Zarrinpar A, Ding Y. Expression of Fluorescence Reporters and Natural Products in Native Gut Escherichia coli. ACS Synth Biol 2025; 14:1557-1566. [PMID: 40138712 DOI: 10.1021/acssynbio.4c00835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Escherichia coli is a widely studied model organism and an integral component of the human gut microbiome, offering significant potential for bacteria-based therapeutic applications. Despite this promise, engineering native E. coli strains remains challenging. In this study, we employed the chassis-independent recombinase-assisted genome engineering (CRAGE) technique to genetically engineer the native gut strain E. coli EcAZ-1 and the probiotic strain E. coli Nissle 1917 (EcN). We successfully expressed a suite of heterologous genes, including the bioluminescent lux operon, green fluorescent protein (GFP), and oxygen-independent fluorescent protein IFP2.0, in both strains. Optimization of IFP2.0 fluorescence was achieved under both aerobic and anaerobic conditions by coexpressing a heme oxygenase gene and/or supplementing the chromophore biliverdin or hemin. Additionally, we engineered these strains to biosynthesize the bioactive compounds naringenin and mycosporine-like amino acids. This work highlights the potential of native E. coli strains as versatile platforms for synthetic biology, paving the way for innovative applications in biomedical research and therapeutic development.
Collapse
Affiliation(s)
- Dake Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - Phuong M Ton
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| | - David Zong
- Division of Gastroenterology, University of California, San Diego, La Jolla, California 92093, United States
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California 92093, United States
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California 92093, United States
- Division of Gastroenterology, Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, California 92093, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
30
|
Creighton RL, Hughes SM, Hladik F, Gornalusse GG. The intestinal interferon system and specialized enterocytes as putative drivers of HIV latency. Front Immunol 2025; 16:1589752. [PMID: 40438119 PMCID: PMC12116432 DOI: 10.3389/fimmu.2025.1589752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
The barrier to HIV cure is the HIV reservoir, which is composed of latently infected CD4+ T cells and myeloid cells that carry stably integrated and replication-competent provirus. The gastrointestinal tract (GIT) contains a substantial part of the HIV reservoir and its immunophysiology could be especially conducive for HIV persistence and reactivation. However, the exact cellular microenvironment and molecular mechanisms that govern the renewal of provirus-harboring cells and proviral reactivation in the GIT remain unclear. In this review, we outline the evidence supporting an overarching hypothesis that interferon activity driven by specialized enterocytes creates a microenvironment that fosters proliferation of latently infected CD4+ T cells and sporadic HIV reactivation from these cells. First, we describe unique immunologic features of the gastrointestinal associated lymphoid tissue (GALT), specifically highlighting IFN activity in specialized enterocytes and potential interactions between these cells and neighboring HIV susceptible cells. Then, we will describe dysregulation of IFN signaling in HIV infection and how IFN dysregulation in the GALT may contribute to the persistence and reactivation of the latent HIV reservoir. Finally, we will speculate on the clinical implications of this hypothesis for HIV cure strategies and outline the next steps.
Collapse
Affiliation(s)
- Rachel L. Creighton
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sean M. Hughes
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Florian Hladik
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Germán G. Gornalusse
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Global Health, Schools of Medicine and Public Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
31
|
Adelfio M, Callen GE, He X, Paster BJ, Hasturk H, Ghezzi CE. Engineered Tissue Models to Decode Host-Microbiota Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417687. [PMID: 40364768 DOI: 10.1002/advs.202417687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/13/2025] [Indexed: 05/15/2025]
Abstract
A mutualistic co-evolution exists between the host and its associated microbiota in the human body. Bacteria establish ecological niches in various tissues of the body, locally influencing their physiology and functions, but also contributing to the well-being of the whole organism through systemic communication with other distant niches (axis). Emerging evidence indicates that when the composition of the microbiota inhabiting the niche changes toward a pathogenic state (dysbiosis) and interactions with the host become unbalanced, diseases may present. In addition, imbalances within a single niche can cause dysbiosis in distant organs. Current research efforts are focused on elucidating the mechanisms leading to dysbiosis, with the goal of restoring tissue homeostasis. In vitro models can provide critical experimental platforms to address this need, by reproducing the niche cyto-architecture and physiology with high fidelity. This review surveys current in in vitro host-microbiota research strategies and provides a roadmap that can guide the field in further developing physiologically relevant in vitro models of ecological niches, thus enabling investigation of the role of the microbiota in human health and diseases. Lastly, given the Food and Drug Administration Modernization Act 2.0, this review highlights emerging in vitro strategies to support the development and validation of new therapies on the market.
Collapse
Affiliation(s)
- Miryam Adelfio
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Grace E Callen
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Xuesong He
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Bruce J Paster
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Hatice Hasturk
- ADA Forsyth Institute, 245 First St, Cambridge, MA, 02142, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| |
Collapse
|
32
|
Lan J, Zou J, Xin H, Sun J, Han T, Sun M, Niu M. Nanomedicines as disruptors or inhibitors of biofilms: Opportunities in addressing antimicrobial resistance. J Control Release 2025; 381:113589. [PMID: 40032007 DOI: 10.1016/j.jconrel.2025.113589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
The problem of antimicrobial resistance (AMR) has caused global concern due to its great threat to human health. Evidences are emerging for a critical role of biofilms, one of the natural protective mechanisms developed by bacteria during growth, in resisting commonly used clinical antibiotics. Advances in nanomedicines with tunable physicochemical properties and unique anti-biofilm mechanisms provide opportunities for solving AMR risks more effectively. In this review, we summarize the five "A" stages (adhesion, amplification, alienation, aging and allocation) of biofilm formation and mechanisms through which they protect the internal bacteria. Aimed at the characteristics of biofilms, we emphasize the design "THAT" principles (targeting, hacking, adhering and transport) of nanomedicines in their interactions with biofilms and internal bacteria. Furthermore, recent progresses in multimodal antibacterial nanomedicines, including biofilms disruption and bactericidal activity, and the types of currently available antibiofilm nanomedicines contained organic and inorganic nanomedicines are outlined and highlighted their potential applications in the development of preclinical research. Last but not least, we offer a perspective for the effectiveness of nanomedicines designed to address AMR and challenges associated with their clinical translation.
Collapse
Affiliation(s)
- Jiaming Lan
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jingyu Zou
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - He Xin
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Tao Han
- Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| | - Meng Niu
- Department of Interventional Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
33
|
Nohesara S, Mostafavi Abdolmaleky H, Dickerson F, Pinto-Tomas AA, Jeste DV, Thiagalingam S. Associations of microbiome pathophysiology with social activity and behavior are mediated by epigenetic modulations: Avenues for designing innovative therapeutic strategies. Neurosci Biobehav Rev 2025; 174:106208. [PMID: 40350003 DOI: 10.1016/j.neubiorev.2025.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
A number of investigations have shown that gut microbiome influences humans' ability to communicate with others, and impairments in social interactions are linked to alterations in gut microbiome composition and diversity, via epigenetic mechanisms. This article reviews the links among gut microbiome, social behavior, and epigenetic shifts relevant to gut microbiome-derived metabolites. First, we discuss how different social determinants of health, such as socioeconomic status, diet, environmental chemicals, migration, ecological conditions, and seasonal changes may influence gut microbiome composition, diversity, and functionality, along with epigenetic alterations and thereby affect social behavior. Next, we consider how gut microbiome-derived metabolites, diet, probiotics, and fecal microbiome transplantation may reduce impairments in social interactions through the adjustment of epigenetic aberrations (e.g., DNA methylation, histone modifications, and microRNAs expression) which may suppress or increase gene expression patterns. Finally, we present the potential benefits and unresolved challenges with the use of gut microbiome-targeted therapeutics in reducing social deficits.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt, Baltimore, MD, USA
| | - Adrian A Pinto-Tomas
- University of Costa Rica, Center for Research in Microscopic Structures and Biochemistry Department, School of Medicine, San Jose, Costa Rica
| | - Dilip V Jeste
- Global Research Network on Social Determinants of Mental Health and Exposomics, La Jolla, CA 92037, USA.
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
34
|
Pietrangeli T, Foffi R, Stocker R, Ybert C, Cottin-Bizonne C, Detcheverry F. Universal Law for the Dispersal of Motile Microorganisms in Porous Media. PHYSICAL REVIEW LETTERS 2025; 134:188303. [PMID: 40408679 DOI: 10.1103/physrevlett.134.188303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/09/2025] [Indexed: 05/25/2025]
Abstract
Dispersal is essential to the plethora of motile microorganisms living in porous environments, yet how it relates to movement patterns and pore space structure remains largely unknown. Here we investigate numerically the long-time dispersal of a run-and-tumble microorganism that remains trapped at solid surfaces and escapes from them by tumbling. We find that dispersal and mean run time are connected by a universal relation, that applies for a variety of porous microstructures and swimming strategies. We explain how this generic dependence originates in the invariance of the mean free path with respect to the movement pattern, and we discuss the optimal strategy that maximizes dispersal. Finally, we extend our approach to microorganisms moving along the surface. Our results provide a general framework to quantify dispersal that works across the vast diversity of movement patterns and porous media.
Collapse
Affiliation(s)
- T Pietrangeli
- Institut Lumière Matière, University of Lyon, Université Claude Bernard Lyon 1, CNRS, F-69622 Villeurbanne, France
| | - R Foffi
- ETH Zurich, Department of Civil, Environmental and Geomatic Engineering, Institute of Environmental Engineering, Zurich, Switzerland
| | - R Stocker
- ETH Zurich, Department of Civil, Environmental and Geomatic Engineering, Institute of Environmental Engineering, Zurich, Switzerland
| | - C Ybert
- Institut Lumière Matière, University of Lyon, Université Claude Bernard Lyon 1, CNRS, F-69622 Villeurbanne, France
| | - C Cottin-Bizonne
- Institut Lumière Matière, University of Lyon, Université Claude Bernard Lyon 1, CNRS, F-69622 Villeurbanne, France
| | - F Detcheverry
- Institut Lumière Matière, University of Lyon, Université Claude Bernard Lyon 1, CNRS, F-69622 Villeurbanne, France
| |
Collapse
|
35
|
Banerjee R. Tiny but Mighty: Small RNAs-The Micromanagers of Bacterial Survival, Virulence, and Host-Pathogen Interactions. Noncoding RNA 2025; 11:36. [PMID: 40407594 PMCID: PMC12101431 DOI: 10.3390/ncrna11030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Bacterial pathogens have evolved diverse strategies to infect hosts, evade immune responses, and establish successful infections. While the role of transcription factors in bacterial virulence is well documented, emerging evidence highlights the significant contribution of small regulatory RNAs (sRNAs) in bacterial pathogenesis. These sRNAs function as posttranscriptional regulators that fine-tune gene expression, enabling bacteria to adapt rapidly to challenging environments. This review explores the multifaceted roles of bacterial sRNAs in host-pathogen interactions. Firstly, it examines how sRNAs regulate pathogenicity by modulating the expression of key virulence factors, including fimbriae, toxins, and secretion systems, followed by discussing the role of sRNAs in bacterial stress response mechanisms that counteract host immune defenses, such as oxidative and envelope stress. Additionally, this review investigates the involvement of sRNAs in antibiotic resistance by regulating efflux pumps, biofilm formation, and membrane modifications, which contribute to multi-drug resistance phenotypes. Lastly, this review highlights how sRNAs contribute to intra- and interspecies communication through quorum sensing, thereby coordinating bacterial behavior in response to environmental cues. Understanding these regulatory networks governed by sRNAs is essential for the development of innovative antimicrobial strategies. This review highlights the growing significance of sRNAs in bacterial pathogenicity and explores their potential as therapeutic targets for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Rajdeep Banerjee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
36
|
Reynolds J, Yoon JY. Fluorescence-based spectrometric and imaging methods and machine learning analyses for microbiota analysis. Mikrochim Acta 2025; 192:334. [PMID: 40323435 DOI: 10.1007/s00604-025-07159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/06/2025] [Indexed: 06/11/2025]
Abstract
Most microbiota determination (skin, gut, soil, etc.) are currently conducted in a laboratory using expensive equipment and lengthy procedures, including culture-dependent methods, nucleic acid amplifications (including quantitative PCR), DNA microarray, immunoassays, 16S rRNA sequencing, shotgun metagenomics, and sophisticated mass spectrometric methods. In situ and rapid analysis methods are desirable for fast turnaround time and low assay cost. Fluorescence identification of bacteria and their mixtures is emerging to meet this demand, thanks to the recent development in various machine learning methods. High-dimensional spectroscopic or microscopic imaging data can be obtained to identify the bacterial makeup and its implications for human health and the environment. For example, we can classify healthy versus non-healthy skin microbiome, inflammatory versus non-inflammatory gut microbiome, degraded versus non-degraded soil microbiome, etc. This tutorial summarizes the various machine-learning algorithms used in bacteria identification and microbiota determinations. It also summarizes the various fluorescence spectroscopic methods used to identify bacteria and their mixtures, including fluorescence lifetime spectroscopy, fluorescence resonance energy transfer (FRET), and synchronous fluorescence (SF) spectroscopy. Finally, various fluorescence microscopic imaging methods were summarized that have been used to identify bacteria and their mixtures, including epi-fluorescence microscopy, confocal microscopy, two-photon/multi-photon microscopy, and super-resolution imaging methods (STED, SIM, PALM, and STORM). Finally, it discusses how these methods can be applied to microbiota determinations, what can be demonstrated in the future, opportunities and challenges, and future directions.
Collapse
Affiliation(s)
- Jocelyn Reynolds
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jeong-Yeol Yoon
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
37
|
Gao M, Zhang S, Zhao J, Zhao W, Ommati MM, Miao C, Zhou B, Wang HW. The NRF2/HO- 1 Pathway: a Potential Regulatory Factor in Fluoride-Induced Colonic Injury under Estrogen Deficiency. Biol Trace Elem Res 2025:10.1007/s12011-025-04633-x. [PMID: 40317343 DOI: 10.1007/s12011-025-04633-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Our previous studies have demonstrated that fluoride (F) overexposure is a risk factor for colonic microenvironment, yet its underlying mechanisms and the influencing factors remain poorly understood. Here, a rat model of F exposure (0, 25, 50, 100 mg/L in drinking water) combined with ovariectomy (OVX)-induced estrogen deficiency was established to investigate the roles of nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in F-induced colonic damage under the state of estrogen deficiency. Result showed that F exposure significantly reduced occludin and claudin-1 expression, further resulting in the colon's morphology impairment. Concurrently, F suppressed epithelial proliferation, decreased goblet cell numbers, and diminished short-chain fatty acid (SCFA) production. OVX-induced estrogen deficiency exacerbated F-induced colonic barrier damage and SCFA decreased. Mechanistically, estrogen deficiency aggravated F intestinal toxicity by further inhibiting the protein expression of Nrf2 and HO-1 and upregulating Keap1 protein expression, following downregulated Bcl-2 mRNA levels and upregulated Bax and caspase-3 mRNA levels, and promoting colonic epithelial cell apoptosis. These findings identify that Nrf2/HO-1 key protein disorders are involved in F-induced colonic barrier injury, and estrogen deficiency further aggravated F intestinal toxicity.
Collapse
Affiliation(s)
- Meng Gao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Sai Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Jing Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Wenpeng Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Chengyi Miao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Bianhua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, People's Republic of China.
| |
Collapse
|
38
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
39
|
Gurevich VV. Assembling the jigsaw puzzle of life. Trends Biochem Sci 2025; 50:374-375. [PMID: 40102125 PMCID: PMC12048255 DOI: 10.1016/j.tibs.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
The cell is a dynamic system where millions of molecules of thousands different kinds act within a complex network with numerous feedback loops. Because we cannot pursue many targets simultaneously, 'big data' rarely yield useful leads. Comprehensive models can place the snippets obtained in simplified experimental conditions into a coherent picture.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 27232, USA.
| |
Collapse
|
40
|
D’Alessandro A. Red blood cell metabolism: a window on systems health towards clinical metabolomics. Curr Opin Hematol 2025; 32:111-119. [PMID: 40085132 PMCID: PMC11949704 DOI: 10.1097/moh.0000000000000863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
PURPOSE OF REVIEW This review focuses on recent advances in the understanding of red blood cell (RBC) metabolism as a function of hypoxia and oxidant stress. In particular, we will focus on RBC metabolic alterations during storage in the blood bank, a medically relevant model of erythrocyte responses to energy and redox stress. RECENT FINDINGS Recent studies on over 13 000 healthy blood donors, as part of the Recipient Epidemiology and Donor Evaluation Study (REDS) III and IV-P RBC omics, and 525 diversity outbred mice have highlighted the impact on RBC metabolism of biological factors (age, BMI), genetics (sex, polymorphisms) and exposure (dietary, professional or recreational habits, drugs that are not grounds for blood donor deferral). SUMMARY We review RBC metabolism from basic biochemistry to storage biology, briefly discussing the impact of inborn errors of metabolism and genetic factors on RBC metabolism, as a window on systems metabolic health. Expanding on the concept of clinical chemistry towards clinical metabolomics, monitoring metabolism at scale in large populations (e.g., millions of blood donors) may thus provide insights into population health as a complementary tool to genetic screening and standard clinical measurements.
Collapse
Affiliation(s)
- Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
41
|
Karabekmez ME. Harnessing Human Holobiome and Meta-Multi-Omics Analyses for Medical Applications. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:179-182. [PMID: 40197113 DOI: 10.1089/omi.2025.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Next-generation sequencing technology has revolutionized all fields of living systems, and its applications almost reinvented some research areas including metagenomics. The microbiotas in our body, including those of the oral, nasal, ocular, alveolar, skin regions, and particularly gut microbiota, have close linkages with our health status. Maturation of experimental techniques for metagenomics has been followed by other related omics platforms, for example, metatranscriptomics, metaproteomics, and all possible metacounterparts of multiomics studies. Now, we are on the eve of a meta-multi-omics era for the analysis of human holobiome in medical research. This era will help buttress the current efforts for systems medicine by illuminating the relationships between human holobiome and health or all human diseases including not only cancers but also infectious diseases, autoimmune diseases, obesity, aging, genetic disorders, and psychiatric conditions. Equally important, meta-multi-omics era is also poised to inform the determinants of human health and, by extension, help build individually tailored precision medicine interventions.
Collapse
Affiliation(s)
- Muhammed Erkan Karabekmez
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Türkiye
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts
| |
Collapse
|
42
|
Wang T, Ahmad S, Cruz-Lebrón A, Ernst SE, Olivos Caicedo KY, Jeong Y, Binion B, Mbuvi P, Dutta D, Fernandez-Materan FV, Breister AM, Tang E, Lee JW, Kang JD, Harris SC, Ikegawa S, Gaskins HR, Erdman JW, Yang G, Cann I, Daniel SL, Hylemon PB, Anantharaman K, Bernardi RC, Alves JMP, Sfanos KS, Irudayaraj J, Ridlon JM. An expanded metabolic pathway for androgen production by commensal bacteria. Nat Microbiol 2025; 10:1084-1098. [PMID: 40259019 DOI: 10.1038/s41564-025-01979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/06/2025] [Indexed: 04/23/2025]
Abstract
Commensal bacteria have been implicated in the modulation of steroid hormones, including circulating androgen levels in the host. However, the microbial genetic pathways involved in androgen production have not been fully characterized. Here we identify a microbial gene encoding an enzyme that catalyses the conversion of androstenedione to epitestosterone in the gut microbiome member Clostridium scindens and named this gene desF. We demonstrate that epitestosterone impacts androgen receptor-dependent prostate cancer cell proliferation in vitro. We also demonstrate that stool desF levels are elevated in patients with prostate cancer who are unresponsive to abiraterone/prednisone therapy. Bacterial isolates from urine or prostatectomy tissue produced androgens, and 17β-hydroxysteroid dehydrogenase activity encoded by the desG gene was detected in strains of the urinary tract bacterium Propionimicrobium lymphophilum. Furthermore, we demonstrate that urinary androgen-producing bacterial strains can promote prostate cancer cell growth through metabolism of cortisol and prednisone. Abiraterone, which targets host desmolase (CYP17A1), a rate-limiting enzyme in adrenal steroidogenesis, does not inhibit bacterial desmolase (DesAB), whereas the conversion of prednisone to androgens by DesAB, DesF and DesG drives androgen-receptor-dependent prostate cancer cell line proliferation in vitro. Our results are a significant advance in steroid microbiology and highlight a potentially important role for gut and urinary tract bacteria in host endocrine function and drug metabolism.
Collapse
Affiliation(s)
- Taojun Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Saeed Ahmad
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Angélica Cruz-Lebrón
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah E Ernst
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Yoon Jeong
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Briawna Binion
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Pauline Mbuvi
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Debapriya Dutta
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Francelys V Fernandez-Materan
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Adam M Breister
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Tang
- Department of Physics, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jae Won Lee
- Department of Biotechnology, Sungshin Women's University, Seoul, South Korea
| | - Jason D Kang
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Spencer C Harris
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | | | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John W Erdman
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Glen Yang
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Isaac Cann
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Steven L Daniel
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Phillip B Hylemon
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | | | - João M P Alves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karen S Sfanos
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Joseph Irudayaraj
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle-Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
- Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
43
|
Ford D. Interactions between the intestinal microbiota and drug metabolism - Clinical implications and future opportunities. Biochem Pharmacol 2025; 235:116809. [PMID: 39983848 DOI: 10.1016/j.bcp.2025.116809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/10/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
The importance of the intestinal microbita in a multitude of physiological processes is well-evidenced. These include metabolism of nutrients and xenobiotics, biosynthesis of vitamin K and vitamin B12, immunomodulation, maintenance of the gut mucosal barrier integrity and protection against some pathogens. Interindividual differences in the intestinal microbiota composition have impacts on health. The bioavailability and activity of some pharmaceuticals are heavily influenced by interindividual variability in metabolism, which has a genetic basis. This variability, primarily occurring in the liver but also in the intestine, has been studied extensively. Despite the advancement of this field - pharmacogenetics - its integration into clinical practice remains limited for reasons discussed herein. This highlights the even greater challenge of applying emerging knowledge on variability in the gut microbiota to drug therapy. However, ignoring these opportunities would be a mistake. While clinical applications of microbiota-guided drug therapy are currently absent and the ideas in this article are largely theoretical, research is uncovering that in cases where a substantial portion of a drug or its metabolites reaches the colon, or where drugs are formulated for colonic delivery, the gut microbiota can significantly affect drug metabolism and activity. Greater focus should be placed on research into how interindividual variability in the intestinal microbiome can modify pharmaceutical bioavailability and activity. This article is deliberately speculative and exploratory but proposes that, though there are still no clinical examples of microbiome-guided drug therapy, these interactions could afford opportunities for improvements in personalised medicine and also for drug design.
Collapse
Affiliation(s)
- Dianne Ford
- Faculty of Health and Life Sciences, Northumberland Building, Northumbria University,Newcastle Upon Tyne NE1 8ST, UK.
| |
Collapse
|
44
|
Kuo IC. Disinfection of outpatient ophthalmic devices: a critique of "semicritical" designation. J Cataract Refract Surg 2025; 51:355-358. [PMID: 39982123 PMCID: PMC12018133 DOI: 10.1097/j.jcrs.0000000000001635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Ophthalmic devices used in outpatient clinics are undergoing scrutiny by regulatory agencies and hospital infection control groups. Applying a nearly century-old classification for disinfection and sterilization of reusable devices and instruments, they are faulting ophthalmologists for not using high-level disinfection (HLD) listed in newer manufacturer instructions for use. By this classification, A-scan and B-scan ultrasound probes, fundus and laser contact lenses, and gonioscopy lenses are grouped with other "semicritical" devices, such as gastrointestinal endoscopes, laryngoscope blades, and anorectal manometers, and therefore require sterilization or HLD. HLD, however, varies by geographic region, compatibility with material, and disinfectant availability. Because HLD necessitates turnover time, clinics must buy more devices to maintain patient volume. Therefore, manufacturers have no incentive to develop sustainable or reusable devices. Most important, however, none of the ophthalmic devices in question has been associated with spread of infection. In short, the mandate for HLD seems to be arbitrary, reactive to perception of possible infection risk from classification of the eye as a "mucous membrane," and devoid of evidence that HLD decreases risk of infection transmission relative to current cleaning and disinfection methods. In the manner that the Ophthalmic Instrument Cleaning and Sterilization Task Force Regulatory raised concerns that regulatory agencies arbitrarily were imposing costly infection control measures for intraocular ophthalmic surgery without evidence that they improved patient safety, a similar task force is needed to address regulatory HLD mandates for clinic devices used in tens of millions of patient encounters for decades without infection transmission.
Collapse
Affiliation(s)
- Irene C Kuo
- From the Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
Longden T, Isaacs D. Pericyte Electrical Signalling and Brain Haemodynamics. Basic Clin Pharmacol Toxicol 2025; 136:e70030. [PMID: 40159653 PMCID: PMC11955720 DOI: 10.1111/bcpt.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Dynamic control of membrane potential lies at the nexus of a wide spectrum of biological processes, ranging from the control of individual cell secretions to the orchestration of complex thought and behaviour. Electrical signals in all vascular cell types (smooth muscle cells, endothelial cells and pericytes) contribute to the control of haemodynamics and energy delivery across spatiotemporal scales and throughout all tissues. Here, our goal is to review and synthesize key studies of electrical signalling within the brain vasculature and integrate these with recent data illustrating an important electrical signalling role for pericytes, in doing so attempting to work towards a holistic description of blood flow control in the brain by vascular electrical signalling. We use this as a framework for generating further questions that we believe are important to pursue. Drawing parallels with electrical signal integration in the nervous system may facilitate deeper insights into how signalling is organized within the vasculature and how it controls blood flow at the network level.
Collapse
Affiliation(s)
- Thomas A. Longden
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Dominic Isaacs
- Department of Pharmacology and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
- Program in NeuroscienceUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
46
|
Cao Z, Li P, Zhang M, Cai S, Li N, Luo M, Li Y, Wu H, Mao X, Ren R, Xie H, Shi S. Erythrocyte Extracellular Vesicles Amalgamate into the Hair and Skin to Maintain Homeostasis. J Extracell Vesicles 2025; 14:e70080. [PMID: 40314064 PMCID: PMC12046290 DOI: 10.1002/jev2.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Erythrocytes are a major cell type in the circulation, numbering between 20 and 30 trillion. The function of erythrocytes is to bring oxygen to the tissues and release carbon dioxide to the lungs. Anaemic patients, who have low levels of erythrocytes, show significant symptoms affecting the hair and skin; however, the detailed relationship between erythrocytes and the integumentary system is not fully understood. Here, we show that erythrocyte extracellular vesicle (EV) can transfer haemoglobin, ABO antigens and keratin into the hair and promote hair regeneration through miR-20a-5p- and miR-22-3p-mediated upregulation of Wnt/β-catenin signalling in dermal papilla cells. Moreover, we show that local injection of autologous erythrocyte EVs ameliorates hair growth in androgenic alopecia (AGA) patients. Interestingly, we found that erythrocyte EVs exit the body from the hair/skin and their membranes contribute to the formation of the outer barrier of the skin. In summary, we identify a previously unknown role of erythrocytes in amalgamating into hair structures and reveal a new therapeutic approach using erythrocyte EVs to promote hair regeneration in AGA patients.
Collapse
Affiliation(s)
- Zeyuan Cao
- Hospital of Stomatology, Guanghua School of StomatologySun Yat‐sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of StomatologyGuangzhouChina
| | - Peiyi Li
- Hospital of Stomatology, Guanghua School of StomatologySun Yat‐sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of StomatologyGuangzhouChina
| | - Manjin Zhang
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| | - Simin Cai
- Hospital of Stomatology, Guanghua School of StomatologySun Yat‐sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of StomatologyGuangzhouChina
| | - Na Li
- Department of Plastic Surgerythe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Mingtao Luo
- Department of Plastic Surgerythe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Yinghui Li
- Department of Orthodontics, Hospital of StomatologyHebei Medical UniversityShijiazhuangChina
| | - Haolin Wu
- International Center for Aging and CancerHainan Medical UniversityHaikouChina
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of StomatologySun Yat‐sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of StomatologyGuangzhouChina
| | - Ruibao Ren
- International Center for Aging and CancerHainan Medical UniversityHaikouChina
| | - Hongju Xie
- Department of Plastic Surgerythe Second Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of StomatologySun Yat‐sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of StomatologyGuangzhouChina
- International Center for Aging and CancerHainan Medical UniversityHaikouChina
| |
Collapse
|
47
|
Furst B, González‐Alonso J. The heart, a secondary organ in the control of blood circulation. Exp Physiol 2025; 110:649-665. [PMID: 38126953 PMCID: PMC12053891 DOI: 10.1113/ep091387] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Circulation of the blood is a fundamental physiological function traditionally ascribed to the pressure-generating function of the heart. However, over the past century the 'cardiocentric' view has been challenged by August Krogh, Ernst Starling, Arthur Guyton and others, based on haemodynamic data obtained from isolated heart preparations and organ perfusion. Their research brought forth experimental evidence and phenomenological observations supporting the concept that cardiac output occurs primarily in response to the metabolic demands of the tissues. The basic tenets of Guyton's venous return model are presented and juxtaposed with their critiques. Developmental biology of the cardiovascular system shows that the blood circulates before the heart has achieved functional integrity and that its movement is intricately connected with the metabolic demands of the tissues. Long discovered, but as yet overlooked, negative interstitial pressure may play a role in assisting the flow returning to the heart. Based on these phenomena, an alternative circulation model has been proposed in which the heart functions like a hydraulic ram and maintains a dynamic equilibrium between the arterial (centrifugal) and venous (centripetal) forces which define the blood's circular movement. In this focused review we introduce some of the salient arguments in support of the proposed circulation model. Finally, we present evidence that exercising muscle blood flow is subject to local metabolic control which upholds optimal perfusion in the face of a substantive rise in muscle vascular conductance, thus lending further support to the permissive role of the heart in the overall control of blood circulation.
Collapse
Affiliation(s)
- Branko Furst
- Department of AnesthesiologyAlbany Medical CenterAlbanyNew YorkUSA
| | - José González‐Alonso
- Sport, Health and Exercise Sciences, Department of Life Sciences, College of Health, Medicine and Life SciencesBrunel University LondonUxbridgeUK
| |
Collapse
|
48
|
Torshizi Esfahani A, Zafarjafarzadeh N, Vakili F, Bizhanpour A, Mashaollahi A, Karimi Kordestani B, Baratinamin M, Mohammadpour S. Gut microbiome in colorectal cancer: metagenomics from bench to bedside. JNCI Cancer Spectr 2025; 9:pkaf026. [PMID: 40045177 DOI: 10.1093/jncics/pkaf026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/27/2024] [Accepted: 02/27/2025] [Indexed: 05/27/2025] Open
Abstract
Colorectal cancer (CRC) is a major global health challenge. Emerging research highlights the pivotal role of the gut microbiota in influencing CRC risk, progression, and treatment response. Metagenomic approaches, especially high-throughput shotgun sequencing, have provided unprecedented insights into the intricate connections between the gut microbiome and CRC. By enabling comprehensive taxonomic and functional profiling, metagenomics has revealed microbial signatures, activities, and biomarkers associated with colorectal tumorigenesis. Furthermore, metagenomics has shown a potential to guide patient stratification, predict treatment outcomes, and inform microbiome-targeted interventions. Despite remaining challenges in multi-omics data integration, taxonomic gaps, and validation across diverse cohorts, metagenomics has propelled our comprehension of the intricate gut microbiome-CRC interplay. This review underscores the clinical relevance of microbial signatures as potential diagnostic and prognostic tools in CRC. Furthermore, it discusses personalized treatment strategies guided by this omics' approach.
Collapse
Affiliation(s)
- Amir Torshizi Esfahani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Zafarjafarzadeh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Fatemeh Vakili
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Anahita Bizhanpour
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Amirhesam Mashaollahi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Bita Karimi Kordestani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Mahdieh Baratinamin
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Somayeh Mohammadpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Adlakha YK, Chhabra R. The human microbiome: redefining cancer pathogenesis and therapy. Cancer Cell Int 2025; 25:165. [PMID: 40296128 PMCID: PMC12039184 DOI: 10.1186/s12935-025-03787-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The human microbiome has always been an important determinant of health and recently, its role has also been described in cancer. The altered microbiome could aid cancer progression, modulate chemoresistance and significantly alter drug efficacy. The broad implications of microbes in cancer have prompted researchers to investigate the microbe-cancer axis and identify whether modifying the microbiome could sensitize cancer cells for therapy and improve the survival outcome of cancer patients. The preclinical data has shown that enhancing the number of specific microbial species could restore the patients' response to cancer drugs and the microbial biomarkers may play a vital role in cancer diagnostics. The elucidation of detailed interactions of the human microbiota with cancer would not only help identify the novel drug targets but would also enhance the efficacy of existing drugs. The field exploring the emerging roles of microbiome in cancer is at a nascent stage and an in-depth scientific perspective on this topic would make it more accessible to a wider audience. In this review, we discuss the scientific evidence connecting the human microbiome to the origin and progression of cancer. We also discuss the potential mechanisms by which microbiota affects initiation of cancer, metastasis and chemoresistance. We highlight the significance of the microbiome in therapeutic outcome and evaluate the potential of microbe-based cancer therapy.
Collapse
Affiliation(s)
- Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector-125, Noida, Uttar Pradesh, 201303, India.
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
50
|
Krishnan D, Ghosh P, Lakshman N, Justin A, Ramasamy S. Oro-pharyngeal mucosal microbiome alternations causing immune system dysregulation in schizophrenia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:125-156. [PMID: 40414631 DOI: 10.1016/bs.irn.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Schizophrenia is a chronic and thoughtful psychological disorder that affects a person's thinking, feelings, and behaviours. Multi-factorial genetic, environmental, and neurological variables cause it. Recently, more research focused on the human microbiome, which alters the immune system and develops adverse health effects on the human body. The study discusses a possible relationship between the oropharyngeal microbiome and schizophrenia. According to recent studies, the oropharyngeal microbiome may alter the immune system in the human body and cause various psychiatric disorders, including schizophrenia. The oropharyngeal microbiome can cause schizophrenia either by affecting the genes, chromosomes, and immune system in the human body. Additionally, it examines the combined mechanism of how the oropharyngeal microbiome's alterations lead to genetic abnormalities and immune dysregulation in schizophrenia. By combining the various approaches, this chapter offers a comprehensive view of the oropharyngeal microbiome's role in schizophrenia and suggests that microbial alterations could serve as biomarkers or therapeutic targets for the disorder.
Collapse
Affiliation(s)
- Deena Krishnan
- Molecular Genetics and Cancer Biology Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Puja Ghosh
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Nathish Lakshman
- Molecular Genetics and Cancer Biology Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Antony Justin
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Sivasamy Ramasamy
- Molecular Genetics and Cancer Biology Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|