1
|
Qiao S, Wang T, Sun J, Han J, Dai H, Du M, Yang L, Guo CJ, Liu C, Liu SJ, Liu H. Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases. Gut Microbes 2025; 17:2489765. [PMID: 40190016 PMCID: PMC11980479 DOI: 10.1080/19490976.2025.2489765] [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: 04/29/2024] [Revised: 12/13/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Mengxuan Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Lan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
2
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Cai Z, Zhou Z, Huang S, Ma S, Chen Y, Cao Y, Ma Y. Gut microbiome in patients with early-stage and late-stage endometriosis. BMC Womens Health 2025; 25:163. [PMID: 40189515 PMCID: PMC11974225 DOI: 10.1186/s12905-025-03689-0] [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: 04/29/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Endometriosis is a chronic inflammatory gynecological disease. Previous studies have explored relationships between endometriosis and the microbiota, but none have focused on differences in gut microbiota between early-stage and late-stage endometriosis patients or their connections to dysmenorrhea symptoms. This study compared gut microbiota compositions between early-stage and late-stage endometriosis patients using amplicon sequencing and further analyzed their dysmenorrhea symptoms. METHODS To minimize seasonal and dietary impacts, we recruited Guangdong residents hospitalized for surgery at Zhujiang Hospital. Participants underwent preoperative screening based on enrollment criteria and fecal samples were collected. Endometriosis was classified according to the American Society for Reproductive Medicine (ASRM) staging system based on surgincal and pathological findings. Stage I-II cases were designated as early-stage endometriosis, and Stage III-IV as late-stage endometriosis. RESULTS A total of 112 patient fecal samples were collected, with 75 (median age, 32 years [range, 18-49 years]) meeting the enrollment criteria, including 39 early-stage (32 Stage I and 7 Stage II) and 36 late-stage (16 Stage III and 20 Stage IV) patients. The gut microbiota structure and functions in early-stage patients significantly differed from those in late-stage cases. Dysmenorrhea was associated with specific microbial traits. Late-stage patients with dysmenorrhea displayed distinctly different gut profiles compared to other endometriosis groups. Bartonella, Snodgrassella, and other taxa were enriched in late-stage cases, while Bacteroides, and Prevotella were decreased. CONCLUSION The gut microbial community structure in early-stage endometriosis patients significantly differs from that in late-stage cases, with late-stage patients experiencing dysmenorrhea displaying particularly distinct gut profiles. Predicted functional analysis indicated suppressed steroid biosynthesis pathways in the gut of late-stage endometriosis patients. In conclusion, it is plausible that the multiple effects of steroids on the lower gastrointestinal tract may involve microbiota alterations, suggesting the need for further investigations.
Collapse
Affiliation(s)
- Zhaoxia Cai
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangzhou Liwan Maternal and Child Health Hospital, Guangzhou, 510375, China
| | - Ziwei Zhou
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Sixia Huang
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Obstetrics and Gynecology Medical Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Song Ma
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuying Chen
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuzhen Cao
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ying Ma
- Obstetrics and Gynecology Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
4
|
Yang H, Zhang X, Wu J, Xiao Y, Dai L, Wang G, Zhang X, Hu C, He S, Yuan Z. Probiotic Membrane-Modified Nanocomposite Alleviates Inflammation and Microbiota Dysbiosis in Colitis by Scavenging Oxidative Stress and Restoring Immune Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40184333 DOI: 10.1021/acsami.4c22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a complex chronic intestinal disorder in which excessive oxidative stress, dysregulated immune response, and microbiota dysbiosis contribute to recurrent episodes of inflammation in the colonic mucosa. Current clinical treatments focusing solely on inflammation resolution often exhibit limited efficacy due to the inability to fundamentally improve the pathological microenvironment. Herein, a probiotic membrane-modified drug delivery nanocomposite, namely, MPDA@Cur@EM, is developed for the comprehensive treatment of IBD. It contains two components: the curcumin-loaded mesoporous polydopamine nanoparticle (MPDA@Cur) as the core and the Escherichia coli Nissle 1917 outer membrane (EM) as the surface. For MPDA@Cur, the pathological microenvironment triggers the responsive release of curcumin. Importantly, MPDA@Cur can effectively alleviate the inflammatory response of LPS-activated macrophages through MPDA-mediated ROS scavenging and curcumin-induced M2 polarization. In the dextran sulfate sodium (DSS)-induced colitis model, the EM coating not only allows for the targeting enrichment of orally administered MPDA@Cur@EM to the inflamed colonic mucosa, but also participates in the regulation of intestinal flora. Consequently, MPDA@Cur@EM efficiently attenuates the inflammatory reaction and restores the intestinal barrier functions, demonstrated by the multipronged manner of restoring redox balance, remodeling immune homeostasis, and reshaping the gut microecology. Collectively, this work provides a safe and promising codelivery strategy of probiotic product, antioxidative nanoenzyme, and therapeutic drug for comprehensive management of IBD.
Collapse
Affiliation(s)
- Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xu Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Jianshuang Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Yao Xiao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Gaoyang Wang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xiaohong Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Chenghu Hu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| |
Collapse
|
5
|
Rahim MA, Seo H, Barman I, Hossain MS, Shuvo MSH, Song HY. Insights into Autophagy in Microbiome Therapeutic Approaches for Drug-Resistant Tuberculosis. Cells 2025; 14:540. [PMID: 40214493 PMCID: PMC11989032 DOI: 10.3390/cells14070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Tuberculosis, primarily caused by Mycobacterium tuberculosis, is an airborne lung disease and continues to pose a significant global health threat, resulting in millions of deaths annually. The current treatment for tuberculosis involves a prolonged regimen of antibiotics, which leads to complications such as recurrence, drug resistance, reinfection, and a range of side effects. This scenario underscores the urgent need for novel therapeutic strategies to combat this lethal pathogen. Over the last two decades, microbiome therapeutics have emerged as promising next-generation drug candidates, offering advantages over traditional medications. In 2022, the Food and Drug Administration approved the first microbiome therapeutic for recurrent Clostridium infections, and extensive research is underway on microbiome treatments for various challenging diseases, including metabolic disorders and cancer. Research on microbiomes concerning tuberculosis commenced roughly a decade ago, and the scope of this research has broadened considerably over the last five years, with microbiome therapeutics now viewed as viable options for managing drug-resistant tuberculosis. Nevertheless, the understanding of their mechanisms is still in its infancy. Although autophagy has been extensively studied in other diseases, research into its role in tuberculosis is just beginning, with preliminary developments in progress. Against this backdrop, this comprehensive review begins by succinctly outlining tuberculosis' characteristics and assessing existing treatments' strengths and weaknesses, followed by a detailed examination of microbiome-based therapeutic approaches for drug-resistant tuberculosis. Additionally, this review focuses on establishing a basic understanding of microbiome treatments for tuberculosis, mainly through the lens of autophagy as a mechanism of action. Ultimately, this review aims to contribute to the foundational comprehension of microbiome-based therapies for tuberculosis, thereby setting the stage for the further advancement of microbiome therapeutics for drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| |
Collapse
|
6
|
Koneru H, Bamba S, Bell A, Estrada-Graf AA, Johnson ZI. Integrating microbial communities into algal biotechnology: a pathway to enhanced commercialization. Front Microbiol 2025; 16:1555579. [PMID: 40236480 PMCID: PMC11997694 DOI: 10.3389/fmicb.2025.1555579] [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: 01/07/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Microalgae are increasingly recognized for their potential in wastewater treatment and the sustainable production of feedstock for fuel, feed, food, and other bioproducts. Like conventional agricultural systems, algal cultivation involves complex microbial communities. However, despite their pivotal role in cultivation outcomes, especially at the commodity-scale, the critical interactions between microalgae and their microbiomes are often overlooked. Here we synthesize current knowledge on the taxonomic diversity, ecological roles, and biotechnological potential of algal microbiomes, with a focus on their interactions with algal hosts through nutrient exchange, growth modulation, pathogen defense, and environmental conditioning. We also examine how environmental factors such as nutrient availability, salinity, and temperature influence these interactions. Advances in microbiome engineering, including synthetic biology and ecological approaches, offer opportunities to enhance beneficial algal-microbiome interactions, thereby improving growth, resilience, and yield. These advancements could lead to more sustainable and economically viable microalgae cultivation, with far-reaching implications for environmental management and biotechnological innovation. By addressing key economic and environmental barriers, microbiome engineering holds transformative potential to revolutionize large-scale algae cultivation and provide sustainable solutions to global challenges.
Collapse
Affiliation(s)
- Hari Koneru
- Marine Laboratory, Nicholas School of the Environment, Duke University, Beaufort, NC, United States
| | - Safiatou Bamba
- Marine Laboratory, Nicholas School of the Environment, Duke University, Beaufort, NC, United States
| | - Aksel Bell
- Marine Laboratory, Nicholas School of the Environment, Duke University, Beaufort, NC, United States
| | - Adrian A. Estrada-Graf
- Marine Laboratory, Nicholas School of the Environment, Duke University, Beaufort, NC, United States
| | - Zackary I. Johnson
- Marine Laboratory, Nicholas School of the Environment, Duke University, Beaufort, NC, United States
- Biology, Civil and Environmental Engineering, Duke Microbiome Center, Duke University, Durham, NC, United States
| |
Collapse
|
7
|
Yu T, Chae M, Wang Z, Ryu G, Kim GB, Lee SY. Microbial Technologies Enhanced by Artificial Intelligence for Healthcare Applications. Microb Biotechnol 2025; 18:e70131. [PMID: 40100535 PMCID: PMC11917392 DOI: 10.1111/1751-7915.70131] [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: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
The combination of artificial intelligence (AI) with microbial technology marks the start of a major transformation, improving applications throughout biotechnology, especially in healthcare. With the capability of AI to process vast amounts of biological big data, advanced microbial technology allows for a comprehensive understanding of complex biological systems, advancing disease diagnosis, treatment and the development of microbial therapeutics. This mini review explores the impact of AI-integrated microbial technologies in healthcare, highlighting advancements in microbial biomarker-based diagnosis, the development of microbial therapeutics and the microbial production of therapeutic compounds. This exploration promises significant improvements in the design and implementation of health-related solutions, steering a new era in biotechnological applications.
Collapse
Affiliation(s)
- Taeho Yu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
| | - Minjee Chae
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
- Graduate School of Engineering Biology, KAIST, Daejeon, Republic of Korea
| | - Ziling Wang
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
| | - Gahyeon Ryu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
| | - Gi Bae Kim
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
- BioProcess Engineering Research Center, KAIST, Daejeon, Republic of Korea
| | - Sang Yup Lee
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, Republic of Korea
- Graduate School of Engineering Biology, KAIST, Daejeon, Republic of Korea
- BioProcess Engineering Research Center, KAIST, Daejeon, Republic of Korea
- Center for Synthetic Biology, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
8
|
Xu Y, Wang Z, Li C, Tian S, Du W. Droplet microfluidics: unveiling the hidden complexity of the human microbiome. LAB ON A CHIP 2025; 25:1128-1148. [PMID: 39775305 DOI: 10.1039/d4lc00877d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The human body harbors diverse microbial communities essential for maintaining health and influencing disease processes. Droplet microfluidics, a precise and high-throughput platform for manipulating microscale droplets, has become vital in advancing microbiome research. This review introduces the foundational principles of droplet microfluidics, its operational capabilities, and wide-ranging applications. We emphasize its role in enhancing single-cell sequencing technologies, particularly genome and RNA sequencing, transforming our understanding of microbial diversity, gene expression, and community dynamics. We explore its critical function in isolating and cultivating traditionally unculturable microbes and investigating microbial activity and interactions, facilitating deeper insight into community behavior and metabolic functions. Lastly, we highlight its broader applications in microbial analysis and its potential to revolutionize human health research by driving innovations in diagnostics, therapeutic development, and personalized medicine. This review provides a comprehensive overview of droplet microfluidics' impact on microbiome research, underscoring its potential to transform our understanding of microbial dynamics and their relevance to health and disease.
Collapse
Affiliation(s)
- Yibin Xu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhiyi Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caiming Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuiquan Tian
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Wenbin Du
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Kong D, Xu J, Zhang Q, Luo D, Lv Q, Li S, Chen X, Wei L, Zhu X, Liu Y, Zhang Z. Selenomethionine Attenuates Aflatoxin B 1-induced Liver Injury by Modulating the Gut Microbiota and Metabolites in Rabbits. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3080-3094. [PMID: 39854169 DOI: 10.1021/acs.jafc.4c09084] [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: 01/26/2025]
Abstract
Dietary contamination with aflatoxin B1 (AFB1), which can lead to severe liver damage, poses a great threat to livestock and poultry breeding and has detrimental impacts on food safety. Selenomethionine (SeMet), with anti-inflammatory, antioxidative, and detoxifying effects, is regarded as a beneficial food additive. However, whether SeMet can reduce AFB1-induced liver injury and intestinal microbial disorders in rabbits remains to be revealed. Forty 35-day-old rabbits were randomly divided into a control group, an AFB1 group, and 0.2 mg/kg Se and 0.4 mg/kg Se groups. The SeMet treatment group was fed different doses of the SeMet diet every day for 21 days. On Days 17-21, the AFB1 group, 0.2 mg/kg Se, and 0.4 mg/kg Se groups were intragastrically administered 0.3 mg AFB1/kg b.w. Results showed that SeMet restored alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, alleviating AFB1-induced liver function damage. This was linked to changes in intestinal metabolites and activation of the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway. In this study, the relationships between intestinal microorganisms and their metabolites and AFB1-induced liver injury are investigated, and the potential protective role of SeMet against liver damage induced by AFB1 offers novel insights into strategies for the prevention and treatment of AFB1-related toxicity.
Collapse
Affiliation(s)
- Dejing Kong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Jingyi Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Qianwen Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Dongliu Luo
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Shuangjun Li
- Henan Rabbit Industry Research and Development Center, Henan Delin Biological Products Co., Luoyang 471023, China
| | - Xiaoguang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Lan Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Xuemin Zhu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
- Henan Rabbit Industry Research and Development Center, Henan Delin Biological Products Co., Luoyang 471023, China
| |
Collapse
|
10
|
Pribyl AL, Hugenholtz P, Cooper MA. A decade of advances in human gut microbiome-derived biotherapeutics. Nat Microbiol 2025; 10:301-312. [PMID: 39779879 DOI: 10.1038/s41564-024-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
Microbiome science has evolved rapidly in the past decade, with high-profile publications suggesting that the gut microbiome is a causal determinant of human health. This has led to the emergence of microbiome-focused biotechnology companies and pharmaceutical company investment in the research and development of gut-derived therapeutics. Despite the early promise of this field, the first generation of microbiome-derived therapeutics (faecal microbiota products) have only recently been approved for clinical use. Next-generation therapies based on readily culturable and as-yet-unculturable colonic bacterial species (with the latter estimated to comprise 63% of all detected species) have not yet progressed to pivotal phase 3 trials. This reflects the many challenges involved in developing a new class of drugs in an evolving field. Here we discuss the evolution of the live biotherapeutics field over the past decade, from the development of first-generation products to the emergence of rationally designed second- and third-generation live biotherapeutics. Finally, we present our outlook for the future of this field.
Collapse
Affiliation(s)
| | - Philip Hugenholtz
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, Brisbane, Queensland, Australia.
| | | |
Collapse
|
11
|
Zhang S, Zhou R, Xie X, Xiong S, Li L, Li Y. Polysaccharides from Lycium barbarum, yam, and sunflower ameliorate colitis in a structure and intrinsic flora-dependent manner. Carbohydr Polym 2025; 349:122905. [PMID: 39643421 DOI: 10.1016/j.carbpol.2024.122905] [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: 06/05/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Polysaccharides have been suggested to ameliorate metabolic diseases. However, their differential colitis-mitigating effects in mouse models with different colony structures remain poorly understood. Therefore, this study investigated the effects of polysaccharides from Lycium barbarum (LBP), sunflower (SP), and yam (YP) on colitis in C57BL/6 J (B6) mice born via vaginal delivery (VD) and in both caesarean section (CS)- and VD-born Institute of Cancer Research (ICR) mice. LBP was mainly composed of glucose (30.2 %), galactose (27.5 %), and arabinose (26.9 %). The main components of SP and YP were galacturonic acid (75.8 %) and glucose (98.1 %), respectively. Interestingly, LBP effectively alleviated body weight loss, reduced inflammatory cytokine levels, and restored intestinal barrier function in all three mouse models. Moreover, LBP decreased the abundance of norank_f__norank_o__Clostridia_UCG-014, Coriobacteriaceae_UCG-002, and norank_f_Eubacterium_coprostanoligenes_group in B6 mice, and the abundance of these genera positively correlated with pro-inflammatory cytokine levels. LBP increased the abundance of Lactobacillus, which was positively correlated with the levels of the protective factor, IL-10, in CS-born ICR mice. Collectively, our study suggests the potential application of LBP in the treatment of ulcerative colitis. We also provide an alternative method for restoring intestinal homeostasis in CS-born offspring.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ruchen Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoran Xie
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shanshan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Koudonas A, Tsiakaras S, Tzikoulis V, Papaioannou M, de la Rosette J, Anastasiadis A, Dimitriadis G. Lifestyle Factors and the Microbiome in Urolithiasis: A Narrative Review. Nutrients 2025; 17:465. [PMID: 39940323 PMCID: PMC11820711 DOI: 10.3390/nu17030465] [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: 12/31/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
Urolithiasis represents one of the most common urologic diseases, and its incidence demonstrates, globally, an increasing trend. The application of preventive measures is an established strategy to reduce urolithiasis-related morbidity, and it is based mostly on the adaptation of lifestyle factors and pharmacotherapy. Furthermore, other research areas demonstrate promising results, such as the research on the microbiome. In the current review, we searched for the latest data on lifestyle-based prevention and microbiome alterations in urolithiasis patients. The majority of the proposed lifestyle measures are already included in the urological guidelines, while additional factors, such as vitamin D supplementation, seem to have a putative positive effect. From the microbiome studies, several microbial composition patterns and metabolic pathways demonstrated an inhibiting or promoting role in lithogenesis. Up to the present, stone prevention has not shown satisfying results, which suggests that lifestyle measures are not adequate. Moreover, microbiome studies are prone to bias, since microbes are strongly affected by numerous clinical factors, while the analysis procedures are not standardized yet. Analysis standardization and data pooling from extensive registration of clinical and microbiome data are essential steps in order to improve the existing prevention strategy with targeted microbiome manipulations.
Collapse
Affiliation(s)
- Antonios Koudonas
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
| | - Stavros Tsiakaras
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
| | - Vasileios Tzikoulis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
| | - Maria Papaioannou
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Jean de la Rosette
- Department of Urology, Istanbul Medipol Mega University Hospital, 34810 Istanbul, Turkey;
| | - Anastasios Anastasiadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
| | - Georgios Dimitriadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (S.T.); (V.T.); (A.A.); (G.D.)
| |
Collapse
|
13
|
Del Prete V, Piazzesi A, Scanu M, Toto F, Pane S, Berrilli F, Paterno G, Putignani L, di Cave D. Pneumocystis Pneumonia Severity Is Associated with Taxonomic Shifts in the Respiratory Microbiota. Pathogens 2025; 14:82. [PMID: 39861043 PMCID: PMC11768410 DOI: 10.3390/pathogens14010082] [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: 12/19/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Pneumonia caused by Pneumocystis jirovecii infection (PCP) is a potentially life-threatening illness, particularly affecting the immunocompromised. The past two decades have shown an increase in PCP incidence; however, the underlying factors that promote disease severity and fatality have yet to be fully elucidated. Recent evidence suggests that the microbiota of the respiratory tract may play a role in stimulating or repressing pulmonary inflammation, as well as the progression of both bacterial and viral pneumonia. Here, we employed 16S rRNA metataxonomic sequencing to profile the respiratory microbiota of patients with mild-moderate and severe PCP. Our results show that the upper and lower airways of PCP patients have bacterial profiles which have been associated with a pro-inflammatory response. Furthermore, we find that severe PCP is associated with lower bacterial diversity and an increase in Prevotella and a decrease in Neisseria. Functionally, severe PCP was associated with a decrease in metabolic pathways of molecules with anti-inflammatory and antimicrobial properties. To our knowledge, this is the first study showing an association of PCP severity with shifts in the respiratory microbiome and may provide some insight into which patients are more susceptible to the more severe manifestations of the disease.
Collapse
Affiliation(s)
- Valentina Del Prete
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (V.D.P.); (F.B.)
| | - Antonia Piazzesi
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00144 Rome, Italy; (A.P.); (M.S.); (F.T.)
| | - Matteo Scanu
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00144 Rome, Italy; (A.P.); (M.S.); (F.T.)
| | - Francesca Toto
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00144 Rome, Italy; (A.P.); (M.S.); (F.T.)
| | - Stefania Pane
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00144 Rome, Italy;
| | - Federica Berrilli
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (V.D.P.); (F.B.)
| | - Giovangiacinto Paterno
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00144 Rome, Italy
| | - David di Cave
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (V.D.P.); (F.B.)
| |
Collapse
|
14
|
Guo J, Zhao Z, Broadwater C, Tobin I, Liu J, Whitmore M, Zhang G. Is Intestinal Microbiota Fully Restored After Chickens Have Recovered from Coccidiosis? Pathogens 2025; 14:81. [PMID: 39861042 PMCID: PMC11768824 DOI: 10.3390/pathogens14010081] [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: 12/31/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The intestinal microbiota is known to be altered by Eimeria-induced coccidiosis, but it remains unclear whether the microbiota is fully restored after recovery. To address this, 110 newly hatched Cobb male broiler chickens were challenged with 2 × 104 sporulated oocysts of Eimeria maxima (EM) strain M6 or mock-infected with saline on day 10. Body weight and feed intake were recorded. Additionally, 10 mock- and 12 EM-infected birds were randomly selected to assess the small intestinal lesion, fecal oocyst shedding, and ileal and cecal microbiota compositions using 16S rRNA gene sequencing at 3, 5, 7, 14, and 21 days post-infection (dpi). EM infection significantly decreased (p < 0.001) body weight by 5 dpi, persisting through 21 dpi. The infection also reduced (p < 0.05) weight gain, feed intake, and feed efficiency in the first week; however, these parameters became comparable in the second and third weeks. At 7 dpi, during the peak of infection, major lactic acid bacteria were enriched, while short-chain fatty acid-producing bacteria were mostly suppressed in both the ileum and cecum. Opportunistic pathogens such as Escherichia and Clostridium perfringens transiently bloomed at 7 dpi. By 14 dpi, differential bacterial enrichment subsided, and nearly all commensal bacteria returned to healthy levels by 21 dpi. Coupled with comparable growth performance between healthy and EM-recovered chickens, we conclude that the intestinal microbiota is largely restored to its healthy state after recovery. Understanding the microbiota's responses to coccidiosis may inform probiotic-based mitigation strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.G.); (Z.Z.); (C.B.); (I.T.); (J.L.); (M.W.)
| |
Collapse
|
15
|
Qu Z, Liu H, Yang J, Zheng L, Huang J, Wang Z, Xie C, Zuo W, Xia X, Sun L, Zhou Y, Xie Y, Lu J, Zhu Y, Yu L, Liu L, Zhou H, Dai L, Leung ELH. Selective utilization of medicinal polysaccharides by human gut Bacteroides and Parabacteroides species. Nat Commun 2025; 16:638. [PMID: 39809740 PMCID: PMC11733155 DOI: 10.1038/s41467-025-55845-7] [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: 07/16/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Human gut Bacteroides and Parabacteroides species play crucial roles in human health and are known for their capacity to utilize diverse polysaccharides. Understanding how these bacteria utilize medicinal polysaccharides is foundational for developing polysaccharides-based prebiotics and drugs. Here, we systematically mapped the utilization profiles of 20 different medicinal polysaccharides by 28 human gut Bacteroides and Parabacteroides species. The growth profiles exhibited substantial variation across different bacterial species and medicinal polysaccharides. Ginseng polysaccharides promoted the growth of multiple Bacteroides and Parabacteroides species; in contrast, Dendrobium polysaccharides selectively promoted the growth of Bacteroides uniformis. This distinct utilization profile was associated with genomic variation in carbohydrate-active enzymes, rather than monosaccharides composition variation among medicinal polysaccharides. Through comparative transcriptomics and genetical manipulation, we validated that the polysaccharide utilization locus PUL34_Bu enabled Bacteroides uniformis to utilize Dendrobium polysaccharides (i.e. glucomannan). In addition, we found that the GH26 enzyme in PUL34_Bu allowed Bacteroides uniformis to utilize multiple plant-derived mannan. Overall, our results revealed the selective utilization of medicinal polysaccharide by Bacteroides and Parabacteroides species and provided insights into the use of polysaccharides in engineering the human gut microbiome.
Collapse
Affiliation(s)
- Zepeng Qu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hongbin Liu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ji Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
| | - Linggang Zheng
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jumin Huang
- Cancer Center, Faculty of Health Sciences, Ministry of Education (MOE) Frontiers Science Center for Precision Oncology, University of Macau, Macau, Macau, SAR, China
| | - Ziming Wang
- Cancer Center, Faculty of Health Sciences, Ministry of Education (MOE) Frontiers Science Center for Precision Oncology, University of Macau, Macau, Macau, SAR, China
| | - Chun Xie
- Cancer Center, Faculty of Health Sciences, Ministry of Education (MOE) Frontiers Science Center for Precision Oncology, University of Macau, Macau, Macau, SAR, China
| | - Wenlong Zuo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiong Xia
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lin Sun
- Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, Northeast Normal University, Changchun, China
| | - Yifa Zhou
- Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, Northeast Normal University, Changchun, China
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingguang Lu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau, China
| | - Lili Yu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau, China
| | - Lihua Liu
- School of Economics and Management, Yanbian University, Yanji, China
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Lei Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, Ministry of Education (MOE) Frontiers Science Center for Precision Oncology, University of Macau, Macau, Macau, SAR, China.
- State Key Laboratory of Quality Research in Chinese Medicine, University of, Macau, Macau.
| |
Collapse
|
16
|
Su D, Li M, Xie Y, Xu Z, Lv G, Jiu Y, Lin J, Chang CJ, Chen H, Cheng F. Gut commensal bacteria Parabacteroides goldsteinii-derived outer membrane vesicles suppress skin inflammation in psoriasis. J Control Release 2025; 377:127-145. [PMID: 39532207 DOI: 10.1016/j.jconrel.2024.11.014] [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: 07/01/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Despite gut microbiota-derived extracellular vesicles (EVs) serving as pivotal mediators in bacteria-host cell interactions, their potential role in modulating skin inflammation remains poorly understood. Here, we developed strategies for mass production of Parabacteroides goldsteinii-derived outer membrane vesicles (Pg OMVs), commonly known as EVs. We found that orally administered Pg OMVs can reach the colon, traverse the intestinal barrier, and circulate to the inflamed skin of psoriasis-like mice, resulting in reduced epidermal hyperplasia, suppressed infiltration of inflammatory cells in the skin lesions, and effective amelioration of both skin and systemic inflammation. Additionally, subcutaneous injection of thermosensitive PF-127 hydrogel loaded with Pg OMVs exerts similar immunomodulatory effects, allowing sustained release of Pg OMVs into skin cells, effectively suppressing skin inflammation and ameliorating symptoms of psoriasis. This study unveils the importance of gut microbiota-derived OMVs, which can target inflamed skin via both the gut-skin axis and local skin administration, providing a promising alternative to live bacteria therapy for the treatment of skin inflammatory diseases.
Collapse
Affiliation(s)
- Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Manchun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yuedong Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zhanxue Xu
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Guowen Lv
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Jingxiong Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chih-Jung Chang
- Medical Research Center and Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen 361028, China; School of Medicine, Huaqiao University, Quanzhou 362021, China.
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
17
|
Chiaro TR, Greenewood M, Bauer KM, Ost KS, Stephen-Victor E, Murphy M, Weis AM, Nelson MC, Hill JH, Bell R, Voth W, Jackson T, Klag KA, O'Connell RM, Zac Stephens W, Round JL. Clec12a controls colitis by tempering inflammation and restricting expansion of specific commensals. Cell Host Microbe 2025; 33:89-103.e7. [PMID: 39788099 PMCID: PMC11824846 DOI: 10.1016/j.chom.2024.12.009] [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: 02/24/2023] [Revised: 11/02/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Microbiota composition regulates colitis severity, yet the innate immune mechanisms that control commensal communities and prevent disease remain unclear. We show that the innate immune receptor, Clec12a, impacts colitis severity by regulating microbiota composition. Transplantation of microbiota from a Clec12a-/- animal is sufficient to worsen colitis in wild-type mice. Clec12a-/- mice have expanded Faecalibaculum rodentium, and treatment with F. rodentium similarly exacerbates disease. However, Clec12a-/- animals are resistant to colitis development when rederived into an 11-member community, underscoring the role of specific species. Colitis in Clec12a-/- mice is dependent on monocytes, and cytokine and sequencing analysis in Clec12a-/- macrophages and serum shows enhanced inflammation with a reduction in phagocytic genes. F. rodentium specifically binds to Clec12a, and Clec12a-/--deficient macrophages are impaired in their ability to phagocytose F. rodentium. Thus, Clec12a contributes to an innate-immune-surveillance mechanism that controls the expansion of potentially harmful commensals while limiting inflammation.
Collapse
Affiliation(s)
- Tyson R Chiaro
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Morgan Greenewood
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Kaylyn M Bauer
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Kyla S Ost
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Emmanuel Stephen-Victor
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Michaela Murphy
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Allison M Weis
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Morgan C Nelson
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Jennifer H Hill
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Rickesha Bell
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Warren Voth
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Taylor Jackson
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Kendra A Klag
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - Ryan M O'Connell
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - W Zac Stephens
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA
| | - June L Round
- University of Utah School of Medicine, Department of Pathology, Division of Microbiology and Immunology, Salt Lake City, UT 84211, USA.
| |
Collapse
|
18
|
Novelle MG, Naranjo-Martínez B, López-Cánovas JL, Díaz-Ruiz A. Fecal microbiota transplantation, a tool to transfer healthy longevity. Ageing Res Rev 2025; 103:102585. [PMID: 39586550 DOI: 10.1016/j.arr.2024.102585] [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: 06/14/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
The complex gut microbiome influences host aging and plays an important role in the manifestation of age-related diseases. Restoring a healthy gut microbiome via Fecal Microbiota Transplantation (FMT) is receiving extensive consideration to therapeutically transfer healthy longevity. Herein, we comprehensively review the benefits of gut microbial rejuvenation - via FMT - to promote healthy aging, with few studies documenting life length properties. This review explores how preconditioning donors via standard - lifestyle and pharmacological - antiaging interventions reshape gut microbiome, with the resulting benefits being also FMT-transferable. Finally, we expose the current clinical uses of FMT in the context of aging therapy and address FMT challenges - regulatory landscape, protocol standardization, and health risks - that require refinement to effectively utilize microbiome interventions in the elderly.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Genetics, Physiology and Microbiology (Unity of Animal Physiology), Faculty of Biology, Complutense University of Madrid (UCM), Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain.
| |
Collapse
|
19
|
Chugh S, Létisse F, Neyrolles O. The exometabolome as a hidden driver of bacterial virulence and pathogenesis. Trends Microbiol 2024:S0966-842X(24)00312-3. [PMID: 39701858 DOI: 10.1016/j.tim.2024.11.009] [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: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
The traditional view of metabolism as merely supplying energy and biosynthetic precursors is undergoing a paradigm shift. Metabolic dynamics not only regulates gene expression but also orchestrates cellular processes with remarkable precision. Most bacterial pathogens exhibit exceptional metabolic plasticity, enabling them to adapt to diverse environments, including hostile conditions within a host. While the role of intracellular bacterial metabolism in pathogen-host interactions has been extensively studied, the contributions of the extracellularly released or secreted bacterial metabolites (referred to here as the bacterial 'exometabolome') to metabolic adaptations and disease pathogenesis remain largely unexplored. In this review, we highlight the significant and intriguing roles of bacterial exometabolomes in drug tolerance, immune suppression, and disease pathogenesis, opening a new frontier in our understanding of bacterial-host interactions.
Collapse
Affiliation(s)
- Saurabh Chugh
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Fabien Létisse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
20
|
Wang H, Li S, Zhang L, Zhang N. The role of fecal microbiota transplantation in type 2 diabetes mellitus treatment. Front Endocrinol (Lausanne) 2024; 15:1469165. [PMID: 39735647 PMCID: PMC11671274 DOI: 10.3389/fendo.2024.1469165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/22/2024] [Indexed: 12/31/2024] Open
Abstract
In contemporary microbial research, the exploration of interactions between microorganisms and multicellular hosts constitutes a burgeoning field. The gut microbiota is increasingly acknowledged as a pivotal contributor to various disorders within the endocrine system, encompassing conditions such as diabetes and thyroid diseases. A surge in research activities has been witnessed in recent years, elucidating the intricate interplay between the gut microbiota and disorders of the endocrine system. Simultaneously, fecal microbiota transplantation (FMT) has emerged as a focal point, garnering substantial attention in both biomedical and clinical spheres. Research endeavors have uncovered the remarkable therapeutic efficacy of FMT across diverse diseases, with particular emphasis on its application in addressing type 2 diabetes mellitus (T2DM) and associated com-plications. Consequently, this manuscript accentuates the intimate connection between the gut microbiota and disorders within the endocrine system, with a specific focus on exploring the potential of FMT as an intervention in the therapeutic landscape of T2DM and its complications. Furthermore, the article scrutinizes concerns inherent in treatment modalities centered around the gut microbiota, proposing viable solutions to address these issues.
Collapse
Affiliation(s)
| | | | | | - Nan Zhang
- *Correspondence: Nan Zhang, ; Luping Zhang,
| |
Collapse
|
21
|
Tong CQ, Li MJ, Liu Y, Zhou Q, Sun WQ, Chen JY, Wang D, Li F, Chen ZJ, Song YH. Regulation of hippocampal miRNA expression by intestinal flora in anxiety-like mice. Eur J Pharmacol 2024; 984:177016. [PMID: 39369876 DOI: 10.1016/j.ejphar.2024.177016] [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: 04/25/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
This study investigated the possible interaction between gut flora and miRNAs and the effect of both on anxiety disorders. The model group was induced with chronic restraint stress (CRS) and each group was tested for anxiety-like behaviour by open field test and elevated plus maze test. Meanwhile, the gut flora was analysed by 16S rRNA high-throughput sequencing. The miRNAs in hippocampus were analysed by high-throughput sequencing, and the key miRNAs were obtained by using the method of bioinformatics analysis. PCR was used to verify the significantly related key miRNAs. Spearman correlation analysis was used to explore the correlation between behaviour, key miRNAs and differential gut microbiota. The 16S rRNA high-throughput sequencing result showed that the gut flora was dysregulated in the model group. In particular, Verrucomicrobia, Akkermansia, Anaerostipes, Ralstonia, Burkholderia and Anaeroplasma were correlated with behaviour. The results of miRNA high-throughput sequencing analysis and bioinformatics analysis showed that 7 key miRNAs influenced the pathogenesis of anxiety, and qRT-PCR results were consistent with the high-throughput sequencing results. Mmu-miR-543-3p and mmu-miR-26a-5p were positively correlated with Verrucomicrobia, Akkermansia and Anaerostipes. Therefore, we infer that chronic stress caused the decrease of Akkermansia abundance, which may aggravate the decrease of mmu-miR-543-3p and mmu-miR-26a-5p expression, leading to the increase of SLC1A2 expression. In conclusion, gut flora has played an important influence on anxiety with changes in miRNAs.
Collapse
Affiliation(s)
- Chang-Qing Tong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Meng-Jia Li
- College of Life Science, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Yan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Qin Zhou
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Wen-Qi Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Jia-Yi Chen
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Di Wang
- Department of traditional Chinese Medicine, Beijing ANDing hospital affiliated to capital University of medical sciences, Beijing, BJ, China
| | - Feng Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China
| | - Zi-Jie Chen
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China.
| | - Yue-Han Song
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, BJ, China.
| |
Collapse
|
22
|
Seo H, Kim S, Beck S, Song HY. Perspectives on Microbiome Therapeutics in Infectious Diseases: A Comprehensive Approach Beyond Immunology and Microbiology. Cells 2024; 13:2003. [PMID: 39682751 PMCID: PMC11640688 DOI: 10.3390/cells13232003] [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: 11/02/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Although global life expectancy has increased over the past 20 years due to advancements in managing infectious diseases, one-fifth of people still die from infections. In response to this ongoing threat, significant efforts are underway to develop vaccines and antimicrobial agents. However, pathogens evolve resistance mechanisms, complicating their control. The COVID-19 pandemic has underscored the limitations of focusing solely on the pathogen-killing strategies of immunology and microbiology to address complex, multisystemic infectious diseases. This highlights the urgent need for practical advancements, such as microbiome therapeutics, that address these limitations while complementing traditional approaches. Our review emphasizes key outcomes in the field, including evidence of probiotics reducing disease severity and insights into host-microbiome crosstalk that have informed novel therapeutic strategies. These findings underscore the potential of microbiome-based interventions to promote physiological function alongside existing strategies aimed at enhancing host immune responses and pathogen destruction. This narrative review explores microbiome therapeutics as next-generation treatments for infectious diseases, focusing on the application of probiotics and their role in host-microbiome interactions. While offering a novel perspective grounded in a cooperative defense system, this review also addresses the practical challenges and limitations in translating these advancements into clinical settings.
Collapse
Affiliation(s)
- Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Sukyung Kim
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Samuel Beck
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, J-607, 609 Albany, Boston, MA 02118, USA
| | - Ho-Yeon Song
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Cheonan-si 31151, Chungnam-do, Republic of Korea
| |
Collapse
|
23
|
Menezes AA, Shah ZA. A Review of the Consequences of Gut Microbiota in Neurodegenerative Disorders and Aging. Brain Sci 2024; 14:1224. [PMID: 39766423 PMCID: PMC11726757 DOI: 10.3390/brainsci14121224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/15/2025] Open
Abstract
Age-associated alterations in the brain lead to cognitive deterioration and neurodegenerative disorders (NDDs). This review with a particular focus on Alzheimer's disease (AD), emphasizes the burgeoning significance of the gut microbiota (GMB) in neuroinflammation and its impact on the gut-brain axis (GBA), a communication conduit between the gut and the central nervous system (CNS). Changes in the gut microbiome, including diminished microbial diversity and the prevalence of pro-inflammatory bacteria, are associated with AD pathogenesis. Promising therapies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, may restore gut health and enhance cognitive performance. Clinical data remain insufficient, necessitating further research to elucidate causes, enhance therapy, and consider individual variances. This integrative approach may yield innovative therapies aimed at the GMB to improve cognitive function and brain health in older people.
Collapse
Affiliation(s)
| | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
24
|
Ren Q, Chen G, Wan Q, Xiao L, Zhang Z, Feng Y. Unravelling the role of natural and synthetic products as DNA topoisomerase inhibitors in hepatocellular carcinoma. Bioorg Chem 2024; 153:107860. [PMID: 39442463 DOI: 10.1016/j.bioorg.2024.107860] [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: 05/17/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Topoisomerase is a ubiquitous enzyme in the control of DNA chain topology. There have been extensive research on topoisomerase inhibitors derived from natural sources, which act as partial inducers of tumor cell apoptosis. However, their specific efficacy in treating hepatocellular carcinoma is relatively unexplored. Hence, this comprehensive review focuses on the structural characteristics and anti-cancer properties of topoisomerase inhibitors in hepatocellular carcinoma. Furthermore, this review is also elucidating the mechanism of action, structure-activity relationships, therapeutic limitations, stage of clinical trials of described classes of natural bioactive compounds as well as their potential application in cancer chemotherapies. This broad understanding of topoisomerase medical biology will provide indispensable framework for enhancing the efficiency of rational anti-hepatocellular carcinoma drug discovery.
Collapse
Affiliation(s)
- Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Qi Wan
- Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Liangman Xiao
- Acupuncture Rehabilitation Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhitong Zhang
- Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
25
|
Yuan X, Chai J, Xu W, Zhao Y. Exploring the Potential of Probiotics and Prebiotics in Major Depression: From Molecular Function to Clinical Therapy. Probiotics Antimicrob Proteins 2024; 16:2181-2217. [PMID: 39078446 DOI: 10.1007/s12602-024-10326-z] [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] [Accepted: 07/10/2024] [Indexed: 07/31/2024]
Abstract
Major depressive disorder (MDD) represents a complex and challenging mental health condition with multifaceted etiology. Recent research exploring the gut-brain axis has shed light on the potential influence of gut microbiota on mental health, offering novel avenues for therapeutic intervention. This paper reviews current evidence on the role of prebiotics and probiotics in the context of MDD treatment. Clinical studies assessing the effects of prebiotic and probiotic interventions have demonstrated promising results, showcasing improvements in depression symptoms and metabolic parameters in certain populations. Notably, prebiotics and probiotics have shown the capacity to modulate inflammatory markers, cortisol levels, and neurotransmitter pathways linked to MDD. However, existing research presents varied outcomes, underscoring the need for further investigation into specific microbial strains, dosage optimization, and long-term effects. Future research should aim at refining personalized interventions, elucidating mechanisms of action, and establishing standardized protocols to integrate these interventions into clinical practice. While prebiotics and probiotics offer potential adjunctive therapies for MDD, continued interdisciplinary efforts are vital to harnessing their full therapeutic potential and reshaping the landscape of depression treatment paradigms.
Collapse
Affiliation(s)
- Xin Yuan
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jianbo Chai
- Heilongjiang Mental Hospital, Harbin, 150036, China
| | - Wenqiang Xu
- Harbin Jiarun Hospital, Harbin, 150040, China
| | - Yonghou Zhao
- Heilongjiang Mental Hospital, Harbin, 150036, China.
| |
Collapse
|
26
|
Feng M, Zou Z, Shou P, Peng W, Liu M, Li X. Gut microbiota and Parkinson's disease: potential links and the role of fecal microbiota transplantation. Front Aging Neurosci 2024; 16:1479343. [PMID: 39679259 PMCID: PMC11638248 DOI: 10.3389/fnagi.2024.1479343] [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: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide and seriously affects the quality of life of elderly patients. PD is characterized by the loss of dopaminergic neurons in the substantia nigra as well as abnormal accumulation of α-synuclein in neurons. Recent research has deepened our understanding of the gut microbiota, revealing that it participates in the pathological process of PD through the gut-brain axis, suggesting that the gut may be the source of PD. Therefore, studying the relationship between gut microbiota and PD is crucial for improving our understanding of the disease's prevention, diagnosis, and treatment. In this review, we first describe the bidirectional regulation of the gut-brain axis by the gut microbiota and the mechanisms underlying the involvement of gut microbiota and their metabolites in PD. We then summarize the different species of gut microbiota found in patients with PD and their correlations with clinical symptoms. Finally, we review the most comprehensive animal and human studies on treating PD through fecal microbiota transplantation (FMT), discussing the challenges and considerations associated with this treatment approach.
Collapse
Affiliation(s)
- Maosen Feng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhiyan Zou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Pingping Shou
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Wei Peng
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingxue Liu
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
27
|
Huai M, Pei M, Chen J, Duan X, Zhu Y, Yang F, Ge W. Oral creatine-modified selenium-based hyaluronic acid nanogel mediated mitochondrial energy recovery to drive the treatment of inflammatory bowel disease. J Nanobiotechnology 2024; 22:740. [PMID: 39609811 PMCID: PMC11603945 DOI: 10.1186/s12951-024-03007-0] [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: 06/18/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
The damnification of mitochondrion is often considered to be an important culprit of inflammatory bowel disease (IBD), however, there are fewer reports of mechanisms of mitochondria-mediated IBD treatment. Therefore, we first proposed to reboot mitochondrial energy metabolism to treat IBD by capturing the double-sided factor of ROS and creatine (Cr)-assisted energy adjustment. Herein, an oral Cr-modified selenium-based hyaluronic acid (HA) nanogel (HASe-Cr nanogel) was fabricated for treatment of IBD, through ROS elimination and energy metabolism upgradation. More concretely, due to IBD lesion-specific positive charge and the high expression of CD44, HASe-Cr nanogel exhibited dual targeted inflammatory bio-functions, and ROS-driven degradation properties in high-yield ROS levels in inflammation areas. As expected, multifunctional HASe-Cr nanogel could effectively ameliorate IBD-related symptoms, such as mitochondrial biological function restoration, inhibition of M1-like macrophage polarization, gut mucosal reconstruction, microbial ecological repair, etc., thus excellently treating IBD. Overall, the proposed strategy underlined that the great potentiality of HASe-Cr nanogel by restarting mitochondrial metabolic energy in colitis lesions, providing new a pavement of mitochondrion-mediated colitis treatment in clinical applications.
Collapse
Affiliation(s)
- Manxiu Huai
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China
| | - Mingliang Pei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jie Chen
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 West Huaihai Road, Shanghai, 200030, P. R. China
| | - Xiaoyan Duan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China
| | - Yun Zhu
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wensong Ge
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China.
| |
Collapse
|
28
|
Du C, Zhao Y, Shen F, Qian H. Effect of Brassica rapa L. Polysaccharide on Lewis Lung Cancer Mice by Inflammatory Regulation and Gut Microbiota Modulation. Foods 2024; 13:3704. [PMID: 39594117 PMCID: PMC11593872 DOI: 10.3390/foods13223704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related fatalities globally, related to inflammatory and gut microbiota imbalance. Brassica rapa L. polysaccharide (BP) is a functional compound, which is utilized by the gut microbiota to regulate immunity and metabolism. However, the effect of BP on lung cancer and whether it affects the "gut-lung" axis remains unclear. This study explored the intervention of BP in Lewis lung cancer (LLC) mice and its effect on the gut microbiota. The results revealed that BP reduced tumor weight and downregulated the expression of Ki67 protein. Additionally, BP reduced the content of inflammatory factors and growth factors, promoting tumor cell apoptosis and inhibiting the growth of LLC. The intervention of BP suppressed intestinal inflammation, preserved intestinal barrier integrity, and augmented the level of beneficial microbiota, such as Blautia and Bifidobacterium. Furthermore, BP significantly increased the production of short-chain fatty acids (SCFAs), particularly butyrate and propionate. A correlation analysis showed significant correlations among the gut microbiota, SCFAs, inflammatory factors, and tight junction proteins. A functional analysis indicated that BP promoted amino acid metabolism and fatty acid metabolism. These findings suggested that BP had the potential to act as prebiotics to prevent disease and improve lung cancer progression by regulating the gut microbiota.
Collapse
Affiliation(s)
- Changhui Du
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Yong Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China;
| | - Fanglin Shen
- School of Environmental Engineering, Wuxi University, Wuxi 214105, China;
| | - He Qian
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| |
Collapse
|
29
|
Hurtado-Lorenzo A, Swantek JL. The landscape of new therapeutic opportunities for IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:1-83. [PMID: 39521596 DOI: 10.1016/bs.apha.2024.10.011] [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: 11/16/2024]
Abstract
This chapter presents an overview of the emerging strategies to address the unmet needs in the management of inflammatory bowel diseases (IBD). IBD poses significant challenges, as over half of patients experience disease progression despite interventions, leading to irreversible complications, and a substantial proportion do not respond to existing therapies, such as biologics. To overcome these limitations, we describe a diverse array of novel therapeutic approaches. In the area of immune homeostasis restoration, the focus is on targeting cytokine networks, leukocyte trafficking, novel immune pathways, and cell therapies involving regulatory T cells and mesenchymal stem cells (MSC). Recognizing the critical role of impaired intestinal barrier integrity in IBD, we highlight therapies aimed at restoring barrier function and promoting mucosal healing, such as those targeting cell proliferation, tight junctions, and lipid mediators. Addressing the challenges posed by fibrosis and fistulas, we describe emerging targets for reversing fibrosis like kinase and cytokine inhibitors and nuclear receptor agonists, as well as the potential of MSC for fistulas. The restoration of a healthy gut microbiome, through strategies like fecal microbiota transplantation, rationally defined bacterial consortia, and targeted antimicrobials, is also highlighted. We also describe innovative approaches to gut-targeted drug delivery to enhance efficacy and minimize side effects. Reinforcing these advancements is the critical role of precision medicine, which emphasizes the use of multiomics analysis for the discovery of biomarkers to enable personalized IBD care. Overall, the emerging landscape of therapeutic opportunities for IBD holds great potential to surpass the therapeutic ceiling of current treatments.
Collapse
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States.
| | - Jennifer L Swantek
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States
| |
Collapse
|
30
|
Ye ZN, Eslick GD, Huang SG, He XX. Faecal microbiota transplantation for eradicating Helicobacter pylori infection: clinical practice and theoretical postulation. EGASTROENTEROLOGY 2024; 2:e100099. [PMID: 39944265 PMCID: PMC11770466 DOI: 10.1136/egastro-2024-100099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
The sustained increase in antibiotic resistance leads to a declining trend in the eradication rate of Helicobacter pylori (H. pylori) infection with antibiotic-based eradication regimens. Administration of a single probiotic shows limited efficacy in eradicating H. pylori infection. This review indicates that faecal microbiota transplantation (FMT), a novel therapeutic approach, either as a monotherapy or adjunctive therapy, exhibits beneficial effects in terms of the eradication of H. pylori infection and the prevention of adverse events. The role of FMT in H. pylori eradication may be associated directly or indirectly with some therapeutic constituents within the faecal suspension, including bacteria, viruses, antimicrobial peptides and metabolites. In addition, variations in donor selection, faecal suspension preparation and delivery methods are believed to be the main factors determining the effectiveness of FMT for the treatment of H. pylori infection. Future research should refine the operational procedures of FMT to achieve optimal efficacy for H. pylori infection and explore the mechanisms by which FMT acts against H. pylori.
Collapse
Affiliation(s)
- Zhi-Ning Ye
- The Affiliated Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Guy D Eslick
- The Australian Paediatric Surveillance Unit, The University of Sydney, The Children's Hospital, Sydney, New South Wale, Australia
| | - Shao-Gang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xing-Xiang He
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
31
|
Saha M, Dittami SM, Chan CX, Raina JB, Stock W, Ghaderiardakani F, Valathuparambil Baby John AM, Corr S, Schleyer G, Todd J, Cardini U, Bengtsson MM, Prado S, Skillings D, Sonnenschein EC, Engelen AH, Wang G, Wichard T, Brodie J, Leblanc C, Egan S. Progress and future directions for seaweed holobiont research. THE NEW PHYTOLOGIST 2024; 244:364-376. [PMID: 39137959 DOI: 10.1111/nph.20018] [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: 03/19/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Abstract
In the marine environment, seaweeds (i.e. marine macroalgae) provide a wide range of ecological services and economic benefits. Like land plants, seaweeds do not provide these services in isolation, rather they rely on their associated microbial communities, which together with the host form the seaweed holobiont. However, there is a poor understanding of the mechanisms shaping these complex seaweed-microbe interactions, and of the evolutionary processes underlying these interactions. Here, we identify the current research challenges and opportunities in the field of seaweed holobiont biology. We argue that identifying the key microbial partners, knowing how they are recruited, and understanding their specific function and their relevance across all seaweed life history stages are among the knowledge gaps that are particularly important to address, especially in the context of the environmental challenges threatening seaweeds. We further discuss future approaches to study seaweed holobionts, and how we can apply the holobiont concept to natural or engineered seaweed ecosystems.
Collapse
Affiliation(s)
- Mahasweta Saha
- Plymouth Marine Laboratory, Marine Ecology and Biodiversity, Prospect Place, Plymouth, PL1 3DH, UK
| | - Simon M Dittami
- CNRS, Integrative Biology of Marine Models Laboratory (LBI2M, UMR 8227), Station Biologique de Roscoff, Place Georges Teissier, Sorbonne Université, Roscoff, 29680, France
| | - Cheong Xin Chan
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Jean-Baptiste Raina
- Climate Change Cluster, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Willem Stock
- Phycology Research Group, Ghent University, Krijgslaan 281 Sterre S8, Ghent, 9000, Belgium
| | - Fatemeh Ghaderiardakani
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Lessingstr. 8, Jena, 07743, Germany
| | | | - Shauna Corr
- Plymouth Marine Laboratory, Marine Ecology and Biodiversity, Prospect Place, Plymouth, PL1 3DH, UK
| | - Guy Schleyer
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology (HKI), Beutenbergstr. 11a, Jena, 07745, Germany
| | - Jonathan Todd
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Frontiers Science Center for Deep Ocean Multispheres and Earth System, and College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Ulisse Cardini
- Department of Integrative Marine Ecology (EMI), Genoa Marine Centre, Stazione Zoologica Anton Dohrn - National Institute of Marine Biology, Ecology and Biotechnology, Genoa, 16126, Italy
| | - Mia M Bengtsson
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Str. 8, Greifswald, 17489, Germany
| | - Soizic Prado
- National Museum of Natural History, Unit Molecules of Communication and Adaptation of Microorganisms (UMR 7245), Paris, France
| | - Derek Skillings
- Department of Philosophy, University of North Carolina Greensboro, Greensboro, NC, 27402, USA
| | - Eva C Sonnenschein
- Department of Biosciences, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | | | - Gaoge Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- MoE Key Laboratory of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Thomas Wichard
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Lessingstr. 8, Jena, 07743, Germany
| | - Juliet Brodie
- Natural History Museum, Research, Cromwell Road, London, SW7 5BD, UK
| | - Catherine Leblanc
- CNRS, Integrative Biology of Marine Models Laboratory (LBI2M, UMR 8227), Station Biologique de Roscoff, Place Georges Teissier, Sorbonne Université, Roscoff, 29680, France
| | - Suhelen Egan
- Centre for Marine Science and Innovation (CMSI), School of Biological, Earth and Environmental Sciences (BEES), UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
32
|
Ferguson Toll J, Solà E, Perez MA, Piano S, Cheng A, Subramanian AK, Kim WR. Infections in decompensated cirrhosis: Pathophysiology, management, and research agenda. Hepatol Commun 2024; 8:e0539. [PMID: 39365139 PMCID: PMC11458171 DOI: 10.1097/hc9.0000000000000539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/01/2024] [Indexed: 10/05/2024] Open
Abstract
Bacterial infections in patients with cirrhosis lead to a 4-fold increase in mortality. Immune dysfunction in cirrhosis further increases the risk of bacterial infections, in addition to alterations in the gut microbiome, which increase the risk of pathogenic bacteria. High rates of empiric antibiotic use contribute to increased incidence of multidrug-resistant organisms and further increases in mortality. Despite continous advances in the field, major unknowns regarding interactions between the immune system and the gut microbiome and strategies to reduce infection risk and improve mortality deserve further investigation. Here, we highlight the unknowns in these major research areas and make a proposal for a research agenda to move toward improving disease progression and outcomes in patients with cirrhosis and infections.
Collapse
Affiliation(s)
- Jessica Ferguson Toll
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Elsa Solà
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | | | - Salvatore Piano
- Department of Medicine, University Hospital of Padova, Padova, Italy
| | - Alice Cheng
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Aruna K. Subramanian
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - W. Ray Kim
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| |
Collapse
|
33
|
Lu K, Li C, Men J, Xu B, Chen Y, Yan P, Gai Z, Zhang Q, Zhang L. Traditional Chinese medicine to improve immune imbalance of asthma: focus on the adjustment of gut microbiota. Front Microbiol 2024; 15:1409128. [PMID: 39411430 PMCID: PMC11473343 DOI: 10.3389/fmicb.2024.1409128] [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: 03/29/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Asthma, being the prevailing respiratory ailment globally, remains enigmatic in terms of its pathogenesis. In recent times, the advancement of traditional Chinese medicine pertaining to the intestinal microbiota has yielded a plethora of investigations, which have substantiated the potential of traditional Chinese medicine in disease prevention and treatment through modulation of the intestinal microbiota. Both animal models and clinical trials have unequivocally demonstrated the indispensable role of the intestinal microbiota in the pathogenesis of asthma. This article presents a summary of the therapeutic effects of traditional Chinese medicine in the context of regulating gut microbiota and its metabolites, thereby achieving immune regulation and inhibiting airway inflammation associated with asthma. It elucidates the mechanism by which traditional Chinese medicine modulates the gut microbiota to enhance asthma management, offering a scientific foundation for the utilization of traditional Chinese medicine in the treatment of asthma.
Collapse
Affiliation(s)
- Ke Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingwen Men
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bin Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizheng Yan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhibo Gai
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingxiang Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lu Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
34
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
35
|
Gao Y, Wang K, Lin Z, Cai S, Peng A, He L, Qi H, Jin Z, Qian X. The emerging roles of microbiome and short-chain fatty acids in the pathogenesis of bronchopulmonary dysplasia. Front Cell Infect Microbiol 2024; 14:1434687. [PMID: 39372498 PMCID: PMC11449852 DOI: 10.3389/fcimb.2024.1434687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/28/2024] [Indexed: 10/08/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects premature infants and leads to long-term pulmonary complications. The pathogenesis of BPD has not been fully elucidated yet. In recent years, the microbiome and its metabolites, especially short-chain fatty acids (SCFAs), in the gut and lungs have been demonstrated to be involved in the development and progression of the disease. This review aims to summarize the current knowledge on the potential involvement of the microbiome and SCFAs, especially the latter, in the development and progression of BPD. First, we introduce the gut-lung axis, the production and functions of SCFAs, and the role of SCFAs in lung health and diseases. We then discuss the evidence supporting the involvement of the microbiome and SCFAs in BPD. Finally, we elaborate on the potential mechanisms of the microbiome and SCFAs in BPD, including immune modulation, epigenetic regulation, enhancement of barrier function, and modulation of surfactant production and the gut microbiome. This review could advance our understanding of the microbiome and SCFAs in the pathogenesis of BPD, which also helps identify new therapeutic targets and facilitate new drug development.
Collapse
Affiliation(s)
- Yuan Gao
- Neonatal Intensive Care Unit (NICU), Jinhua Maternal and Child Health Care Hospital, Jinhua, China
| | - Kaixuan Wang
- Department of Pediatrics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zupan Lin
- Neonatal Intensive Care Unit (NICU), Jinhua Maternal and Child Health Care Hospital, Jinhua, China
| | - Shujing Cai
- Neonatal Intensive Care Unit (NICU), Jinhua Maternal and Child Health Care Hospital, Jinhua, China
| | - Aohui Peng
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Le He
- Department of Pediatrics, Jinhua Hospital of TCM Affiliated to Zhejiang University of Traditional Chinese Medicine, Jinhua, China
| | - Hui Qi
- China National Clinical Research Center of Respiratory Diseases, Respiratory Department, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Zhigang Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xubo Qian
- Department of Pediatrics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- Department of Pediatrics, Jinhua Hospital of TCM Affiliated to Zhejiang University of Traditional Chinese Medicine, Jinhua, China
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
36
|
Van den Berghe L, Masschelein J, Pinheiro VB. From competition to cure: the development of live biotherapeutic products for anticancer therapy in the iGEM competition. Front Bioeng Biotechnol 2024; 12:1447176. [PMID: 39351063 PMCID: PMC11439766 DOI: 10.3389/fbioe.2024.1447176] [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: 06/11/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer is a leading cause of mortality globally, often diagnosed at advanced stages with metastases already present, complicating treatment efficacy. Traditional treatments like chemotherapy and radiotherapy face challenges such as lack of specificity and drug resistance. The hallmarks of cancer, as defined by Hanahan and Weinberg, describe tumors as complex entities capable of evolving traits that promote malignancy, including sustained proliferation, resistance to cell death, and metastasis. Emerging research highlights the significant role of the microbiome in cancer development and treatment, influencing tumor progression and immune responses. This review explores the potential of live biotherapeutic products (LBPs) for cancer diagnosis and therapy, focusing on projects from the International Genetically Engineered Machines (iGEM) competition that aim to innovate LBPs for cancer treatment. Analyzing 77 projects from 2022, we highlight the progress and ongoing challenges within this research field.
Collapse
Affiliation(s)
- Luka Van den Berghe
- Laboratory for Biomolecular Discovery and Engineering, Department of Biology, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Joleen Masschelein
- Laboratory for Biomolecular Discovery and Engineering, Department of Biology, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Vitor B Pinheiro
- Department of Pharmaceutical and Pharmacological Sciences, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Zhu Y, Huang X, Deng Z, Bai T, Gao B, Xu C, Fu J, Zhao Y, Zhang Y, Zhang M, Zhang M, Yang M, Chen L. Orally biomimetic metal-phenolic nanozyme with quadruple safeguards for intestinal homeostasis to ameliorate ulcerative colitis. J Nanobiotechnology 2024; 22:545. [PMID: 39238009 PMCID: PMC11378530 DOI: 10.1186/s12951-024-02802-z] [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: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is defined by persistent inflammatory processes within the gastrointestinal tract of uncertain etiology. Current therapeutic approaches are limited in their ability to address oxidative stress, inflammation, barrier function restoration, and modulation of gut microbiota in a coordinated manner to maintain intestinal homeostasis. RESULTS This study involves the construction of a metal-phenolic nanozyme (Cur-Fe) through a ferric ion-mediated oxidative coupling of curcumin. Cur-Fe nanozyme exhibits superoxide dismutase (SOD)-like and •OH scavenging activities, demonstrating significant anti-inflammatory and anti-oxidant properties for maintaining intracellular redox balance in vitro. Drawing inspiration from Escherichia coli Nissle 1917 (EcN), a biomimetic Cur-Fe nanozyme (CF@EM) is subsequently developed by integrating Cur-Fe into the EcN membrane (EM) to improve the in vivo targeting ability and therapeutic effectiveness of the Cur-Fe nanozyme. When orally administered, CF@EM demonstrates a strong ability to colonize the inflamed colon and restore intestinal redox balance and barrier function in DSS-induced colitis models. Importantly, CF@EM influences the gut microbiome towards a beneficial state by enhancing bacterial diversity and shifting the compositional structure toward an anti-inflammatory phenotype. Furthermore, analysis of intestinal microbial metabolites supports the notion that the therapeutic efficacy of CF@EM is closely associated with bile acid metabolism. CONCLUSION Inspired by gut microbes, we have successfully synthesized a biomimetic Cur-Fe nanozyme with the ability to inhibit inflammation and restore intestinal homeostasis. Collectively, without appreciable systemic toxicity, this work provides an unprecedented opportunity for targeted oral nanomedicine in the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Xiaoling Huang
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Uygur Autonomous Region, Urumgi, 830001, China
| | - Zhichao Deng
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Ting Bai
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710077, China
| | - Bowen Gao
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Chenxi Xu
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Junlong Fu
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yuanru Zhao
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Mingxin Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Mei Yang
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
38
|
Ettel P, Weichhart T. Not just sugar: metabolic control of neutrophil development and effector functions. J Leukoc Biol 2024; 116:487-510. [PMID: 38450755 PMCID: PMC7617515 DOI: 10.1093/jleuko/qiae057] [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: 11/30/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
The mammalian immune system is constantly surveying our tissues to clear pathogens and maintain tissue homeostasis. In order to fulfill these tasks, immune cells take up nutrients to supply energy for survival and for directly regulating effector functions via their cellular metabolism, a process now known as immunometabolism. Neutrophilic granulocytes, the most abundant leukocytes in the human body, have a short half-life and are permanently needed in the defense against pathogens. According to a long-standing view, neutrophils were thought to primarily fuel their metabolic demands via glycolysis. Yet, this view has been challenged, as other metabolic pathways recently emerged to contribute to neutrophil homeostasis and effector functions. In particular during neutrophilic development, the pentose phosphate pathway, glycogen synthesis, oxidative phosphorylation, and fatty acid oxidation crucially promote neutrophil maturation. At steady state, both glucose and lipid metabolism sustain neutrophil survival and maintain the intracellular redox balance. This review aims to comprehensively discuss how neutrophilic metabolism adapts during development, which metabolic pathways fuel their functionality, and how these processes are reconfigured in case of various diseases. We provide several examples of hereditary diseases, in which mutations in metabolic enzymes validate their critical role for neutrophil function.
Collapse
Affiliation(s)
- Paul Ettel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| | - Thomas Weichhart
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090Vienna, Austria
| |
Collapse
|
39
|
Yazici C, Priyadarshini M, Boulay B, Dai Y, Layden BT. Alterations in microbiome associated with acute pancreatitis. Curr Opin Gastroenterol 2024; 40:413-421. [PMID: 38900442 PMCID: PMC11305980 DOI: 10.1097/mog.0000000000001046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
PURPOSE OF REVIEW This review evaluates the current knowledge of gut microbiome alterations in acute pancreatitis, including those that can increase acute pancreatitis risk or worsen disease severity, and the mechanisms of gut microbiome driven injury in acute pancreatitis. RECENT FINDINGS Recent observational studies in humans showed the association of gut microbiome changes (decreased gut microbiome diversity, alterations in relative abundances of certain species, and association of unique species with functional pathways) with acute pancreatitis risk and severity. Furthermore, in-vivo studies highlighted the role of gut microbiome in the development and severity of acute pancreatitis using FMT models. The gut barrier integrity, immune cell homeostasis, and microbial metabolites appear to play key roles in acute pancreatitis risk and severity. SUMMARY Large human cohort studies that assess gut microbiome profile, its metabolites and impact on acute pancreatitis risk and severity will be crucial for development of innovative prediction, prevention and treatment strategies.
Collapse
Affiliation(s)
- Cemal Yazici
- Division of Gastroenterology and Hepatology, University of Illinois Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL USA
| | - Medha Priyadarshini
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois Chicago, Chicago, IL, USA
| | - Brian Boulay
- Division of Gastroenterology and Hepatology, University of Illinois Chicago, Chicago, IL, USA
| | - Yang Dai
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Brian T. Layden
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL USA
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
40
|
Xu Y, Du H, Chen Y, Ma C, Zhang Q, Li H, Xie Z, Hong Y. Targeting the gut microbiota to alleviate chemotherapy-induced toxicity in cancer. Crit Rev Microbiol 2024; 50:564-580. [PMID: 37439132 DOI: 10.1080/1040841x.2023.2233605] [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: 02/12/2023] [Revised: 05/22/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
Despite ongoing breakthroughs in novel anticancer therapies, chemotherapy remains a mainstream therapeutic modality in different types of cancer. Unfortunately, chemotherapy-related toxicity (CRT) often leads to dose limitation, and even results in treatment termination. Over the past few years, accumulating evidence has indicated that the gut microbiota is extensively engaged in various toxicities initiated by chemotherapeutic drugs, either directly or indirectly. The gut microbiota can now be targeted to reduce the toxicity of chemotherapy. In the current review, we summarized the clinical relationship between the gut microbiota and CRT, as well as the critical role of the gut microbiota in the occurrence and development of CRT. We then summarized the key mechanisms by which the gut microbiota modulates CRT. Furthermore, currently available strategies to mitigate CRT by targeting the gut microbiota were summarized and discussed. This review offers a novel perspective for the mitigation of diverse chemotherapy-associated toxic reactions in cancer patients and the future development of innovative drugs or functional supplements to alleviate CRT via targeting the gut microbiota.
Collapse
Affiliation(s)
- Yaning Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuchun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Chong Ma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Qian Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Hao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yanjun Hong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
41
|
Borrego-Ruiz A, Borrego JJ. Influence of human gut microbiome on the healthy and the neurodegenerative aging. Exp Gerontol 2024; 194:112497. [PMID: 38909763 DOI: 10.1016/j.exger.2024.112497] [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: 03/04/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The gut microbiome plays a crucial role in host health throughout the lifespan by influencing brain function during aging. The microbial diversity of the human gut microbiome decreases during the aging process and, as a consequence, several mechanisms increase, such as oxidative stress, mitochondrial dysfunction, inflammatory response, and microbial gut dysbiosis. Moreover, evidence indicates that aging and neurodegeneration are closely related; consequently, the gut microbiome may serve as a novel marker of lifespan in the elderly. In this narrative study, we investigated how the changes in the composition of the gut microbiome that occur in aging influence to various neuropathological disorders, such as mild cognitive impairment (MCI), dementia, Alzheimer's disease (AD), and Parkinson's disease (PD); and which are the possible mechanisms that govern the relationship between the gut microbiome and cognitive impairment. In addition, several studies suggest that the gut microbiome may be a potential novel target to improve hallmarks of brain aging and to promote healthy cognition; therefore, current and future therapeutic interventions have been also reviewed.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain.
| |
Collapse
|
42
|
Lin X, He K, Gu Z, Zhao X. Emerging chemophysiological diversity of gut microbiota metabolites. Trends Pharmacol Sci 2024; 45:824-838. [PMID: 39129061 DOI: 10.1016/j.tips.2024.07.006] [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: 05/24/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/13/2024]
Abstract
Human physiology is profoundly influenced by the gut microbiota, which generates a wide array of metabolites. These microbiota-derived compounds serve as signaling molecules, interacting with various cellular targets in the gastrointestinal tract and distant organs, thereby impacting our immune, metabolic, and neurobehavioral systems. Recent advancements have unveiled unique physiological functions of diverse metabolites derived from tryptophan (Trp) and bile acids (BAs). This review highlights the emerging chemophysiological diversity of these metabolites and discusses the role of chemical and biological tools in analyzing and therapeutically manipulating microbial metabolism and host targets, with the aim of bridging the chemical diversity with physiological complexity in host-microbe molecular interactions.
Collapse
Affiliation(s)
- Xiaorong Lin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Kaixin He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, Zhejiang, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, Zhejiang, China; Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Xiaohui Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
43
|
Zhang Y, Zhou C, Tian B, Xu J, Wang X, Dai H, Wang H, Xu F, Wang C. 3D printed spiral tube-like cellulose scaffold for oral delivery of probiotics. SCIENCE ADVANCES 2024; 10:eadp3654. [PMID: 39178258 PMCID: PMC11343031 DOI: 10.1126/sciadv.adp3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/19/2024] [Indexed: 08/25/2024]
Abstract
Introducing specific strains of probiotics into the gut microbiome is a promising way to modulate the intestinal microbiome to treat various health conditions clinically. However, oral probiotics typically have a temporary or limited impact on the gut microbiome and overall health benefits. Here, we reported a 3D printed cellulose-derived spiral tube-like scaffold that enabled high efficacy of the oral delivery of probiotics. Benefiting from the unique surface pattern, this system can effectively extend the retention time of loaded probiotics in the gut without invading nearby tissues, provide a favorable environment for the survival and long-term colonization of loaded probiotics, and influence the intestinal ecosystem as a dietary fiber after degradation. We demonstrate Roseburia intestinalis-loaded scaffold exerts noticeable impacts on the regulation of the gut microbiome to treat various gut-related diseases, including obesity and inflammatory bowel disease; thus, we provide a universal platform for oral delivery of probiotics.
Collapse
Affiliation(s)
- Yue Zhang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenmeng Zhou
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Bo Tian
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiaoyu Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Heng Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Laboratory for Biomaterial and Immunoengineering, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
44
|
Bostanghadiri N, Kouhzad M, Taki E, Elahi Z, Khoshbayan A, Navidifar T, Darban-Sarokhalil D. Oral microbiota and metabolites: key players in oral health and disorder, and microbiota-based therapies. Front Microbiol 2024; 15:1431785. [PMID: 39228377 PMCID: PMC11368800 DOI: 10.3389/fmicb.2024.1431785] [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: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
The review aimed to investigate the diversity of oral microbiota and its influencing factors, as well as the association of oral microbiota with oral health and the possible effects of dysbiosis and oral disorder. The oral cavity harbors a substantial microbial burden, which is particularly notable compared to other organs within the human body. In usual situations, the microbiota exists in a state of equilibrium; however, when this balance is disturbed, a multitude of complications arise. Dental caries, a prevalent issue in the oral cavity, is primarily caused by the colonization and activity of bacteria, particularly streptococci. Furthermore, this environment also houses other pathogenic bacteria that are associated with the onset of gingival, periapical, and periodontal diseases, as well as oral cancer. Various strategies have been employed to prevent, control, and treat these disorders. Recently, techniques utilizing microbiota, like probiotics, microbiota transplantation, and the replacement of oral pathogens, have caught the eye. This extensive examination seeks to offer a general view of the oral microbiota and their metabolites concerning oral health and disease, and also the resilience of the microbiota, and the techniques used for the prevention, control, and treatment of disorders in this specific area.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mobina Kouhzad
- Department of Genetics, Faculty of Science, Islamic Azad University North Tehran Branch, Tehran, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Chen Y, Xiao L, Zhou M, Zhang H. The microbiota: a crucial mediator in gut homeostasis and colonization resistance. Front Microbiol 2024; 15:1417864. [PMID: 39165572 PMCID: PMC11333231 DOI: 10.3389/fmicb.2024.1417864] [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: 04/15/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
The gut microbiota is a complex and diverse community of microorganisms that colonizes the human gastrointestinal tract and influences various aspects of human health. These microbes are closely related to enteric infections. As a foreign entity for the host, commensal microbiota is restricted and regulated by the barrier and immune system in the gut and contributes to gut homeostasis. Commensals also effectively resist the colonization of pathogens and the overgrowth of indigenous pathobionts by utilizing a variety of mechanisms, while pathogens have developed strategies to subvert colonization resistance. Dysbiosis of the microbial community can lead to enteric infections. The microbiota acts as a pivotal mediator in establishing a harmonious mutualistic symbiosis with the host and shielding the host against pathogens. This review aims to provide a comprehensive overview of the mechanisms underlying host-microbiome and microbiome-pathogen interactions, highlighting the multi-faceted roles of the gut microbiota in preventing enteric infections. We also discuss the applications of manipulating the microbiota to treat infectious diseases in the gut.
Collapse
Affiliation(s)
- Yiding Chen
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Ling Xiao
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Min Zhou
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, West China Tianfu Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Center for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Yan JH, Jin SX, Chen QW, Zhang Y, Li QR, Chen Z, Sun Y, Zhong Z, Zhang XZ. Intracellular Gelation-Mediated Living Bacteria for Advanced Biotherapeutics in Mouse Models. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:16605-16614. [PMID: 39039962 DOI: 10.1021/acs.langmuir.4c02215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Despite its significant potential in various disease treatments and diagnostics, microbiotherapy is consistently plagued by multiple limitations ranging from manufacturing challenges to in vivo functionality. Inspired by the strategy involving nonproliferating yet metabolically active microorganisms, we report an intracellular gelation approach that can generate a synthetic polymer network within bacterial cells to solve these challenges. Specifically, poly(ethylene glycol dimethacrylate) (PEGDA, 700 Da) monomers are introduced into the bacterial cytosol through a single cycle of freeze-thawing followed by the initiation of intracellular free radical polymerization by UV light to create a macromolecular PEGDA gel within the bacterial cytosol. The molecular crowding resulting from intracytoplasmic gelation prohibits bacterial division and confers robust resistance to simulated gastrointestinal fluids and bile acids while retaining the ability to secrete functional proteins. Biocompatibility assessments demonstrate that the nondividing gelatinized bacteria are effective in alleviating systemic inflammation triggered by intravenous Escherichia coli injection. Furthermore, the therapeutic efficacy of gelatinized Lactobacillus rhamnosus in colitis mice provides additional support for this approach. Collectively, intracellular gelation indicates a universal strategy to manufacture next-generation live biotherapeutics for advanced microbiotherapy.
Collapse
Affiliation(s)
- Jian-Hua Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Sheng-Xin Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yun Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhu Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yunxia Sun
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
47
|
Maier L, Stein-Thoeringer C, Ley RE, Brötz-Oesterhelt H, Link H, Ziemert N, Wagner S, Peschel A. Integrating research on bacterial pathogens and commensals to fight infections-an ecological perspective. THE LANCET. MICROBE 2024; 5:100843. [PMID: 38608681 DOI: 10.1016/s2666-5247(24)00049-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
The incidence of antibiotic-resistant bacterial infections is increasing, and development of new antibiotics has been deprioritised by the pharmaceutical industry. Interdisciplinary research approaches, based on the ecological principles of bacterial fitness, competition, and transmission, could open new avenues to combat antibiotic-resistant infections. Many facultative bacterial pathogens use human mucosal surfaces as their major reservoirs and induce infectious diseases to aid their lateral transmission to new host organisms under some pathological states of the microbiome and host. Beneficial bacterial commensals can outcompete specific pathogens, thereby lowering the capacity of the pathogens to spread and cause serious infections. Despite the clinical relevance, however, the understanding of commensal-pathogen interactions in their natural habitats remains poor. In this Personal View, we highlight directions to intensify research on the interactions between bacterial pathogens and commensals in the context of human microbiomes and host biology that can lead to the development of innovative and sustainable ways of preventing and treating infectious diseases.
Collapse
Affiliation(s)
- Lisa Maier
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Christoph Stein-Thoeringer
- Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany; Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Ruth E Ley
- Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; Max Planck Institute for Biology, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Hannes Link
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany
| | - Nadine Ziemert
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Samuel Wagner
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence "Controlling Microbes to Fight Infections" (CMFI), Tübingen, Germany; German Center for Infection Research, partner site, Tübingen, Germany.
| |
Collapse
|
48
|
Wortelboer K, Herrema H. Opportunities and challenges in phage therapy for cardiometabolic diseases. Trends Endocrinol Metab 2024; 35:687-696. [PMID: 38637223 DOI: 10.1016/j.tem.2024.03.007] [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: 02/13/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
The worldwide prevalence of cardiometabolic diseases (CMD) is increasing, and emerging evidence implicates the gut microbiota in this multifactorial disease development. Bacteriophages (phages) are viruses that selectively target a bacterial host; thus, phage therapy offers a precise means of modulating the gut microbiota, limiting collateral damage on the ecosystem. Several studies demonstrate the potential of phages in human disease, including alcoholic and steatotic liver disease. In this opinion article we discuss the potential of phage therapy as a predefined medicinal product for CMD and discuss its current challenges, including the generation of effective phage combinations, product formulation, and strict manufacturing requirements.
Collapse
Affiliation(s)
- Koen Wortelboer
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology, and Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam UMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Diabetes, and Metabolism, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology, and Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam UMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Diabetes, and Metabolism, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Veseli I, Chen YT, Schechter MS, Vanni C, Fogarty EC, Watson AR, Jabri BA, Blekhman R, Willis AD, Yu MK, Fernandez-Guerra A, Fussel J, Eren AM. Microbes with higher metabolic independence are enriched in human gut microbiomes under stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.10.540289. [PMID: 37293035 PMCID: PMC10245760 DOI: 10.1101/2023.05.10.540289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A wide variety of human diseases are associated with loss of microbial diversity in the human gut, inspiring a great interest in the diagnostic or therapeutic potential of the microbiota. However, the ecological forces that drive diversity reduction in disease states remain unclear, rendering it difficult to ascertain the role of the microbiota in disease emergence or severity. One hypothesis to explain this phenomenon is that microbial diversity is diminished as disease states select for microbial populations that are more fit to survive environmental stress caused by inflammation or other host factors. Here, we tested this hypothesis on a large scale, by developing a software framework to quantify the enrichment of microbial metabolisms in complex metagenomes as a function of microbial diversity. We applied this framework to over 400 gut metagenomes from individuals who are healthy or diagnosed with inflammatory bowel disease (IBD). We found that high metabolic independence (HMI) is a distinguishing characteristic of microbial communities associated with individuals diagnosed with IBD. A classifier we trained using the normalized copy numbers of 33 HMI-associated metabolic modules not only distinguished states of health versus IBD, but also tracked the recovery of the gut microbiome following antibiotic treatment, suggesting that HMI is a hallmark of microbial communities in stressed gut environments.
Collapse
|
50
|
de Lorenzo V, Pérez-Pantoja D, Nikel PI. Pseudomonas putida KT2440: the long journey of a soil-dweller to become a synthetic biology chassis. J Bacteriol 2024; 206:e0013624. [PMID: 38975763 PMCID: PMC11270871 DOI: 10.1128/jb.00136-24] [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: 07/09/2024] Open
Abstract
Although members of the genus Pseudomonas share specific morphological, metabolic, and genomic traits, the diversity of niches and lifestyles adopted by the family members is vast. One species of the group, Pseudomonas putida, thrives as a colonizer of plant roots and frequently inhabits soils polluted with various types of chemical waste. Owing to a combination of historical contingencies and inherent qualities, a particular strain, P. putida KT2440, emerged time ago as an archetype of an environmental microorganism amenable to recombinant DNA technologies, which was also capable of catabolizing chemical pollutants. Later, the same bacterium progressed as a reliable platform for programming traits and activities in various biotechnological applications. This article summarizes the stepwise upgrading of P. putida KT2440 from being a system for fundamental studies on the biodegradation of aromatic compounds (especially when harboring the TOL plasmid pWW0) to its adoption as a chassis of choice in metabolic engineering and synthetic biology. Although there are remaining uncertainties about the taxonomic classification of KT2440, advanced genome editing capabilities allow us to tailor its genetic makeup to meet specific needs. This makes its traditional categorization somewhat less important, while also increasing the strain's overall value for contemporary industrial and environmental uses.
Collapse
Affiliation(s)
- Victor de Lorenzo
- Systems Biology Department, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Danilo Pérez-Pantoja
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana (UTEM), Santiago, Chile
- Center of Applied Ecology and Sustainability (CAPES), Santiago, Chile
| | - Pablo I. Nikel
- The Novo Nordisk Foundation Center for Biosustainability, Systems Environmental Microbiology Group, Kgs. Lyngby, Denmark
| |
Collapse
|