1
|
Lv LX, Yin JN, Sun YL, Wei MY, Jiang WQ, Gu YC, Yang XP, Shao CL. Marine natural products as potential anti-Pseudomonas aeruginosa agents: challenges and advances. Eur J Med Chem 2025; 292:117670. [PMID: 40305937 DOI: 10.1016/j.ejmech.2025.117670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
Antimicrobial resistance (AMR) has become a pressing need to address in the major global public health challenges, posing a serious threat to human health. Pseudomonas aeruginosa (PA) is one of the most concerning Gram-negative pathogens and is typically treated with broad-spectrum antibiotics. PA exhibits resistance to multiple antibiotics, multifactorial virulence, and dynamic hyperadaptation, which results in a particularly formidable challenge in eliminating PA from patients. The problem of drug resistance is becoming increasingly serious, and the development of new antibiotics is extremely lagging behind, resulting in no drug with a new structure and mechanism being approved for the treatment of infections caused by drug-resistant Gram-negative bacteria over the past half-century. Consequently, the development of new antibiotics is of utmost urgency and importance. Marine natural products (MNPs) have become an important source for developing new antibiotics due to their unique properties. So far, 44 potential molecules with significant anti-PA activity have been isolated from marine organisms, of which 19 have been reported as quorum-sensing system inhibitors (QSIs) with potential for further development. In this review, we provide a comprehensive summary of the current status of drug resistance, pathogenic mechanisms, and resistance mechanisms associated with PA infections. We also highlight the challenges and opportunities presented by MNPs in the development of anti-PA drugs, and offer recommendations to accelerate the antibiotic development process, thereby providing valuable insights for the study and exploitation of novel antibiotics.
Collapse
Affiliation(s)
- Liu-Xia Lv
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Jun-Na Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Yi-Lin Sun
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Wen-Qing Jiang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Xiao-Ping Yang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China.
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China; Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, People's Republic of China.
| |
Collapse
|
2
|
Chen X, Li H, Wang G, Wang Z, Lv Y, Xie H, Zhu S. Exploring the role of intestinal pathogenic bacteria in metronidazole-induced bone loss: focus on Klebsiella variicola. Gut Pathog 2025; 17:42. [PMID: 40483510 PMCID: PMC12144832 DOI: 10.1186/s13099-025-00713-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/15/2025] [Indexed: 06/11/2025] Open
Abstract
Antibiotic use is known to contribute to the development of osteoporosis, although the exact mechanisms remain poorly understood. Metronidazole (MET), a commonly prescribed antibiotic for treating anaerobic infections, has been linked to alterations in the gut microbiota (GM), which in turn are associated with various adverse side effects in the host. Recent studies have shown that the GM plays a key role in regulating bone homeostasis, though the underlying mechanisms remain under investigation. In this study, we demonstrate for the first time that MET promotes inflammatory osteoporosis through gut dysbiosis, with Klebsiella variicola (K. variicola) identified as a major pathogen influencing bone metabolism. The pro-inflammatory extracellular vesicles (EVs) secreted by K. variicola induce enhanced inflammatory responses and osteoclastic differentiation in both bone macrophages and bone tissue. Notably, the use of antibiotics that target K. variicola effectively mitigates MET-induced bone loss in vivo. This study expands our understanding of the mechanisms underlying antibiotic-induced bone loss and underscores the significant role of the pathogenic bacterium K. variicola in the development of osteoporosis, providing new avenues for future research on the microbiota-gut-bone axis in bone-related diseases.
Collapse
Affiliation(s)
- Xia Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongming Li
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guang Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhenxing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yan Lv
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Sheng Zhu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Angmedicine, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
3
|
Garfinkle R, Bennett RD, Dantu S, Gasior A, Hawkins AT, Holland J, Ore AS, Shaffer VO, Taylor JP, Sylla P, McLemore EC, Boutros M. SAGES white paper on antibiotic omission in the management of acute uncomplicated diverticulitis: why, when, who, and most importantly, how. Surg Endosc 2025; 39:3456-3465. [PMID: 40263135 DOI: 10.1007/s00464-025-11738-w] [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: 03/16/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Historically, the management of acute uncomplicated diverticulitis was centered on antibiotics. However, modern theories regarding the pathogenesis of diverticulitis have challenged the notion that antibiotics are necessary in all cases. Despite major reform in many societal guidelines, the adoption of non-antibiotic therapy for uncomplicated diverticulitis has been limited, especially in North America. The purpose of this SAGES White Paper was to review the available evidence on antibiotic omission in uncomplicated diverticulitis and to explore methods of safe implementation. METHODS A task force within the SAGES Colorectal Surgery Committee was formed to work on this White Paper. The committee and its leadership approved an outline that would focus on the following topics: (1) Defining the problem with unnecessary antibiotic exposure; (2) Evaluating the evidence on antibiotic omission in uncomplicated diverticulitis; (3) Identifying the appropriate patient for antibiotic omission; (4) Outlining how to counsel patients who are treated without antibiotics; (5) Reviewing methods to safely implement this practice in both the hospital and community setting. These topics were divided up among members of the task force who performed a structured literature search in preparation for their assignments. RESULTS Antibiotics are associated with several patient and societal adverse effects, including the rising problem of antimicrobial resistance. Randomized controlled trials have demonstrated no superiority to the routine administration of antibiotics in acute uncomplicated diverticulitis. Appropriate patients for antibiotic omission include those who are immunocompetent, non-septic, and have mild symptoms/disease severity on imaging. Existing frameworks for the safe implementation of new practices can be referenced to help increase adoption of non-antibiotic therapy. CONCLUSION The existing body of evidence supports antibiotic omission in appropriate cases of acute uncomplicated diverticulitis. In order to increase the widespread adoption of this practice, buy-in from key stakeholders (both healthcare professionals and patients) is necessary.
Collapse
Affiliation(s)
- Richard Garfinkle
- Division of Colon and Rectal Surgery, Department of Surgery, Jewish General Hospital, McGill University, 3755 Cote Saint-Catherine Road, Montreal, QC, H3T1E2, Canada.
| | - Robert D Bennett
- Division of Colon and Rectal Surgery, University of South Florida Morsani College of Medicine, Tampa Bay, FL, USA
| | - Siva Dantu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Alessandra Gasior
- Nationwide Children's Hospital, Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Alexander T Hawkins
- Section of Colon and Rectal Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jessica Holland
- Department of Surgery, Thunder Bay Regional Health Sciences Center, Thunder Bay, ON, USA
| | - Ana Sofia Ore
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | | | - James P Taylor
- Department of Surgery, Montefiore Medical Center, Bronx, NY, USA
| | - Patricia Sylla
- Department of Surgery, Mount Sinai Hospital, New York, NY, USA
| | - Elisabeth C McLemore
- Department of Surgery, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA, USA
| | - Marylise Boutros
- Department of Colorectal Surgery, Cleveland Clinic Florida, Weston, FL, USA
| |
Collapse
|
4
|
Chen J, Feng J, Cui X, Huang L, Du B, Xia Y, Xue G, Feng Y, Ke Y, Zhao H, Cui J, Yan C, Gan L, Fan Z, Fu T, Xu Z, Yang Y, Yu Z, Zhao S, Wang Z, Kong Y, Jiang B, Wang M, Ling M, Yuan J. Genomic analysis and therapeutic efficacy evaluation of bacteriophage PK2420 for pneumonia caused by hypervirulent Klebsiella pneumoniae (K20 serotype). mSystems 2025; 10:e0163224. [PMID: 40237452 PMCID: PMC12090786 DOI: 10.1128/msystems.01632-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Hypervirulent Klebsiella pneumoniae (hvKp) strains are increasingly recognized for their aggressive nature, which leads to severe clinical outcomes. The emergence of multidrug-resistant strains constitutes a substantial challenge for clinical management. Phage therapy offers a potential solution to the antibiotic resistance crisis. A multidrug-resistant hvKp strain, K2420 (K20 serotype), was used to isolate bacteriophages from hospital sewage. Phage morphology, biological properties, and genome characteristics were analyzed using transmission electron microscopy, plaque assays, and whole-genome sequencing. Therapeutic safety and efficacy were assessed in an acute pneumonia murine model induced by intratracheal injection of K2420. Assessment parameters included bacterial load, phage titer, body temperature, cytokine levels, histopathological findings, and other relevant indicators. Phage PK2420, a member of the Autographiviridae family and Przondovirus genus, was identified. It rapidly lyses K. pneumoniae (K20 serotype), inhibits biofilm formation, and exhibits a burst size of 37.4 plaque-forming units/cell. The phage is stable at temperatures ranging from 0°C to 40°C and pH values between 6 and 9. Its genome, 41,155 bp in length, contains 46 coding sequences. The phage has no genes associated with antibiotic resistance, virulence, or lysogeny. In vivo, PK2420 substantially reduced K. pneumoniae bacterial loads, improved survival rates, and alleviated pneumonia severity without observable side effects. Phage PK2420 exhibits lytic activity against K. pneumoniae both in vitro and in murine models, providing a promising and safe option for the treatment of hvKp infections.IMPORTANCEOur investigation provides insights into the interaction mechanism among hypervirulent Klebsiella pneumoniae (hvKp) (K20 serotype), phage, and the host in a mouse pneumonia model, offering a valuable reference for future research on phage pharmacokinetics. This study demonstrated that bacteriophage PK2420 exhibits promising biosafety and therapeutic efficacy against hvKp-induced pulmonary infections and dissemination in a murine model. These findings suggest that phage PK2420 may be a potential option for the clinical treatment of hvKp infections.
Collapse
Affiliation(s)
- Jinfeng Chen
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Xiaohu Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lijuan Huang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yuyan Xia
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yang Yang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Shuo Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Zhen Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yiming Kong
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Boyi Jiang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Mingxuan Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Mengyao Ling
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jing Yuan
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
- Children’s Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Zou S, Yang X, Zhou L. Gut microbiota in epilepsy: How antibiotics induce dysbiosis and influence seizure susceptibility. Microbiol Res 2025; 298:128225. [PMID: 40398011 DOI: 10.1016/j.micres.2025.128225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/27/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Epilepsy, a widespread chronic neurological disorder, has recently come under scrutiny for its potential association with the intricate dynamics of gut microbiota. Numerous investigations into the microbiota-gut-brain axis have revealed a close relationship between gut microbiota and epilepsy, suggesting gut microbiota as a potential treatment strategy. In clinical practice, a longstanding correlation has been observed between some kinds of antibiotics and the potential to induce seizures. Consequently, we have conceived a hypothesis that antibiotics might impact seizure activity by modulating the gut microbiota and influencing the physiological processes within the microbiota-gut-brain axis. In this review, our primary objective is to present the existing evidence and theoretical foundations supporting the hypothesis that dysbiosis within the gut microbiota may play a significant role in the pathophysiology of epilepsy. Furthermore, we aim to summarize the possible mechanisms between microbiota-gut-brain axis and epilepsy, offering insights into the selection of appropriate antibiotics for long-term epilepsy management and enhancing therapeutic efficacy through modulation of the gut microbiota. Further research is necessary to fully elucidate the intricate relationship between gut microbiota ecosystem and epilepsy. Exploring these connections holds promise for advancing our understanding of epilepsy pathogenesis and improving patient treatment and care.
Collapse
Affiliation(s)
- Shangnan Zou
- Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Clinical Neuroscience Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Xiaofeng Yang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Basic Medicine, Guangzhou National Laboratory, Guangzhou, Guangdong, China.
| | - Liemin Zhou
- Clinical Neuroscience Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Neurology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
He Y, Shaoyong W, Chen Y, Li M, Gan Y, Sun L, Liu Y, Wang Y, Jin M. The functions of gut microbiota-mediated bile acid metabolism in intestinal immunity. J Adv Res 2025:S2090-1232(25)00307-8. [PMID: 40354934 DOI: 10.1016/j.jare.2025.05.015] [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: 02/14/2025] [Revised: 04/19/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Bile acids, derived from cholesterol in the liver, consist a steroidal core. Primary bile acids and secondary bile acids metabolized by the gut microbiota make up the bile acid pool, which modulate nuclear hormone receptors to regulate immunity. Disruptions in the crosstalk between bile acids and the gut flora are intimately associated with the development and course of gastrointestinal inflammation. AIM OF REVIEW This review provides an extensive summary of bile acid production, transport and metabolism. It also delves into the impact of bile acid metabolism on the body and explores the involvement of bile acid-microbiota interactions in various disease states. Furthermore, the potential of targeting bile acid signaling as a means to prevent and treat inflammatory bowel disease is proposed. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we primarily address the functions of bile acid-microbiota crosstalk in diseases. Firstly, we summarize bile acid signalling and the factors influencing bile acid metabolism, with highlighting the immune function of microbially conjugated bile acids and the unique roles of different receptors. Subsequently, we emphasize the vital role of bile acids in maintaining a healthy gut microbiota and regulating the intestinal barrier function, energy metabolism and immunity. Finally, we explore differences of bile acid metabolism in different disease states, offering new perspectives on restoring the host's health and the gastrointestinal ecosystem by targeting the gut microbiota-bile acid-bile acid receptor axis.
Collapse
Affiliation(s)
- Yanmin He
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Weike Shaoyong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yanli Chen
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Menglin Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yujie Gan
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Lu Sun
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yalin Liu
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Yizhen Wang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China
| | - Mingliang Jin
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou 310058, China; Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou 310058, China; National Engineering Research Center for Green Feed and Healthy Breeding, Hangzhou 310058, China.
| |
Collapse
|
7
|
Nohesara S, Mostafavi Abdolmaleky H, Dickerson F, Pinto-Tomas AA, Jeste DV, Thiagalingam S. Associations of microbiome pathophysiology with social activity and behavior are mediated by epigenetic modulations: Avenues for designing innovative therapeutic strategies. Neurosci Biobehav Rev 2025; 174:106208. [PMID: 40350003 DOI: 10.1016/j.neubiorev.2025.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
A number of investigations have shown that gut microbiome influences humans' ability to communicate with others, and impairments in social interactions are linked to alterations in gut microbiome composition and diversity, via epigenetic mechanisms. This article reviews the links among gut microbiome, social behavior, and epigenetic shifts relevant to gut microbiome-derived metabolites. First, we discuss how different social determinants of health, such as socioeconomic status, diet, environmental chemicals, migration, ecological conditions, and seasonal changes may influence gut microbiome composition, diversity, and functionality, along with epigenetic alterations and thereby affect social behavior. Next, we consider how gut microbiome-derived metabolites, diet, probiotics, and fecal microbiome transplantation may reduce impairments in social interactions through the adjustment of epigenetic aberrations (e.g., DNA methylation, histone modifications, and microRNAs expression) which may suppress or increase gene expression patterns. Finally, we present the potential benefits and unresolved challenges with the use of gut microbiome-targeted therapeutics in reducing social deficits.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt, Baltimore, MD, USA
| | - Adrian A Pinto-Tomas
- University of Costa Rica, Center for Research in Microscopic Structures and Biochemistry Department, School of Medicine, San Jose, Costa Rica
| | - Dilip V Jeste
- Global Research Network on Social Determinants of Mental Health and Exposomics, La Jolla, CA 92037, USA.
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
8
|
Farooq S, Talat A, Dhariwal A, Petersen FC, Khan AU. Transgenerational gut dysbiosis: Unveiling the dynamics of antibiotic resistance through mobile genetic elements from mothers to infants. Int J Antimicrob Agents 2025; 65:107458. [PMID: 39921114 DOI: 10.1016/j.ijantimicag.2025.107458] [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: 08/20/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES The initial microbial colonization of the gut is seeded by microbes transmitted from the mother's gut, skin, and vaginal tract. As the gut microbiome evolves, a few transmitted microbes persist throughout life. Understanding the impact of mother-to-neonate gut microbiome and antibiotic resistance genes (ARGs) transmission is crucial for establishing its role in infants' immunity against pathogens. METHODS This study primarily explores mother-neonate ARG transmission through 125 publicly available fecal metagenomes, isolated from eighteen mother-neonate pairs. RESULTS The core ARGs, detected in both mothers and their respective infants at all stages (birth, 1st, 2nd, 3rd, 4th, 8th and 12th months) included aminoglycosidases APH(3')-IIIa, Bifidobacterium adolescentis rpoB mutants conferring resistance to rifampicin, β-lactamases CblA-1, CfxA2, multidrug resistance gene CRP, diaminopyrimidine resistance gene dfrF, fluoroquinolone-resistance gene emrR, macrolide; lincosamide; streptogramin resistance gene ErmB, ErmG, macrolide resistance gene Mef(En2), nucleosidase SAT-4, and tetracycline-resistance genes tet(O), tet(Q), and tet(W). Most of these infants and mothers were not administered any antibiotics. In infants, ARGs were predominantly carried by Bacillota, Pseudomonadota, and Actinomycetota, similar to the mothers. The dominant ARG-carrying opportunistic pathogens were Escherichia coli, Klebsiella, and Streptococcus, found across all infant cohorts. All the core ARGs were associated with mobile genetic elements, signifying the role of horizontal gene transfer(HGT). We detected 132 virulence determinants, mostly E. coli-specific, including pilus chaperones, general secretion pathway proteins, type III secretion system effectors, and heme-binding proteins. CONCLUSIONS Maternal-neonate transmission of ARGs along with possible nosocomial infections, mode of delivery, breastfeeding versus formula feeding, and gestation period, must be considered for mother-neonate health.
Collapse
Affiliation(s)
- Samiya Farooq
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Absar Talat
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Achal Dhariwal
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | | | - Asad U Khan
- Antimicrobial Resistance Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India; Bioinformatics and Computational Biology Centre of DBT Government of India, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India.
| |
Collapse
|
9
|
Zhang T, Zhong H, Yang M, Shi X, Yang L, Yang J, Liu H, Luo Y, Xie Y, Zhong Z, Peng G, Zhang K, Zheng C, Zhang M, Zhou Z. Lactobacillus salivary LSbg3 is a Potential Food Probiotic Having Excellent Anti-pathogen Effect That Might Improve Antibiotic-Resistant Diarrhea in Dogs. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10527-0. [PMID: 40259196 DOI: 10.1007/s12602-025-10527-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/23/2025]
Abstract
Antibiotics may disrupt the intestinal microbiota balance and induce antimicrobial resistance. Although probiotics should be a priority treatment for animal diarrhea, it still has chance to be used as same/or behind as antibiotics in the clinic. Among the probiotics, Lactobacillus (Lact.) was the most frequently utilized in clinical setting since its excellent ability of safety, anti-pathogen, stress resistance, and easy colonization in intestine. In this study, we screened 24 strains of Lact. in the presence of antibiotics from clinical common antibiotic-treated feces, identified L. salivarius LSbg3 exhibiting good stress resistance, potent antibacterial activity, and exceptional intestinal adhesion capability. Its genome showed a good function of regulating intestinal nutrition while lack of transmission antibiotic-resistance genes. Additionally, in a simulated canine diarrhea with failed antibiotic treatment, LSbg3 had a good efficacy in the releasing diarrhea, balancing the microbiome and suppressing typical pathogens, positioning a potential food probiotic have excellent effect on anti-pathogen that can effectively improve antibiotic-resistant diarrhea in dogs.
Collapse
Affiliation(s)
- Ting Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hongyu Zhong
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Min Yang
- Pet Nutrition and Health Research Center, Chengdu Agricultural College, Chengdu, 611130, China
| | - Xin Shi
- Sichuan Institute of Musk Deer Breeding, Sichuan Institute for Drug Control, Chengdu, 611731, Sichuan, China
| | - Liuqing Yang
- Sichuan Institute of Musk Deer Breeding, Sichuan Institute for Drug Control, Chengdu, 611731, Sichuan, China
| | - Jie Yang
- Sichuan Institute of Musk Deer Breeding, Sichuan Institute for Drug Control, Chengdu, 611731, Sichuan, China
| | - Haifeng Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yan Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yue Xie
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhijun Zhong
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Guangneng Peng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Kun Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Chengli Zheng
- Sichuan Institute of Musk Deer Breeding, Sichuan Institute for Drug Control, Chengdu, 611731, Sichuan, China.
| | - Ming Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| | - Ziyao Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
10
|
Yin L, Lv B, Ge J, Qi Y, Xia J, Ma S, Wang Y, Liu Y, Zhou D, Cao J, Yan Z, Qi K, Sang W, Li D, Cheng H, Chen W, Xu K, Gu W, Li Z, Zhu F. The impact of antibiotic use on outcomes of relapsed/refractory multiple myeloma patients treated with CAR-T therapy. Front Immunol 2025; 16:1566016. [PMID: 40313953 PMCID: PMC12043596 DOI: 10.3389/fimmu.2025.1566016] [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: 01/24/2025] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background In recent years, chimeric antigen receptor (CAR)-T cell therapy has achieved tremendous efficacy in relapsed/refractory multiple myeloma (R/R MM). However, the impact of antibiotic (ATB) use on R/R MM patients treated with CAR-T is still not known. The aim of our study was to analyse the influence of ATB on the clinical outcomes of R/R MM patients treated with CAR-T cells. Methods In this retrospective study, 199 patients with R/R MM who received CAR-T cells between January 2018 and December 2023 were evaluated from two hospitals in China. They were stratified into ATB-group and No ATB-group according to whether ATB was administered in the 4 weeks before therapy. We mainly analyzed the efficacy, survival outcomes and cytotoxicity of CAR-T cell therapy in two groups of patients. Result In the ATB group (90 patients), the overall response rate (ORR) was 70% comparable to the No ATB group (109 patients: ORR, 81.7%; P = 0.054). The complete response rate (CRR) was 40%, which was significantly lower compared with No ATB group (CRR, 57.8%; P = 0.012). The median progression-free survival (PFS) was 6.7 months while the median overall survival (OS) was 21.9 months for the ATB group. The median PFS and OS for the No ATB group were 13.9 months and 36.1 months. There were significant differences in PFS (P = 0.007) and OS (P = 0.004) between the evaluated groups. Nonetheless, multivariate analysis found ATB use did not reduce the CRR (odds ratio [OR], 0.947; 95% confidence interval [CI], 0.251 to 3.565, P = 0.936). Besides, administration of ATB did not affect the PFS (hazard ratio [HR], 0.634; 95% CI, 0.28 to 1.436, P = 0.275) and OS (HR, 2.259; 95% CI, 0.755 to 6.762, P = 0.145) in R/R MM patients treated with CAR-T cells. Additionally, both groups of patients had similar incidences of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Conclusion Our results point to a detrimental effect of ATB on treatment outcomes to CAR-T cell therapy. However, the use of ATB is not associated with the incidence of CRS or ICANS.
Collapse
Affiliation(s)
- Lingling Yin
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Bin Lv
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Jiao Ge
- Department of Hematology, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Yuekun Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Jieyun Xia
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Sha Ma
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Ying Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Yang Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Dian Zhou
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Jiang Cao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Zhiling Yan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Kunming Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Wei Sang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Depeng Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Hai Cheng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Wei Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Weiying Gu
- Department of Hematology, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Feng Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| |
Collapse
|
11
|
Kyriazopoulou E, Stylianakis E, Damoraki G, Koufargyris P, Kollias I, Katrini K, Drakou E, Marousis K, Spyrou A, Symbardi S, Alexiou N, Alexiou Z, Lada M, Poulakou G, Chrysos G, Adamis G, Giamarellos-Bourboulis EJ. Procalcitonin-guided early cessation of antibiotics prevents gut inflammation and preserves gut microbiome: Data from the PROGRESS controlled trial. Int J Antimicrob Agents 2025; 66:107507. [PMID: 40216091 DOI: 10.1016/j.ijantimicag.2025.107507] [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: 11/03/2024] [Revised: 03/09/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025]
Abstract
The PROGRESS randomised trial (ClinicalTrials.gov: NCT03333304) showed that early stopof antibiotics guided by procalcitonin (PCT) decreased the incidence of infections by multidrug-resistant organisms and/or Clostridioides difficile and was associated with survival benefit. This study was conducted to investigate whether this survival benefit is associated with microbiome dysbiosis. Patients with sepsis due to lung infection, acute pyelonephritis or primary bacteraemia were randomised to standard-of-care (SoC) duration of antibiotics or early stop using PCT. Faecal samples were collected before, and 7 and 28 days after randomisation and analysed using 16S rRNA Nanopore sequencing. Calprotectin was measured using an enzyme immunoassay. Median (Q1-Q3) antimicrobial duration was 5 (5-7.5) days in the PCT arm and 11 (8-15) days in the SoC arm (P < 0.001). Faecal calprotectin levels were similar in the two treatment arms at baseline. By day 7, the levels of faecal calprotectin were significantly increased in the SoC arm (P = 0.002) but were unchanged in the PCT arm. Microbiome α- and β-diversity was similar at baseline in the PCT (n=81) and SoC (n=76) treatment arms. Shannon's index was significantly lower in the SoC arm on day 7 compared with baseline (median [Q1-Q3], 2.88 [2.37-3.39] at day 1 vs. 2.24 [1.52-3.08] at day 7; Pt-test = 0.0013). This was not the case for the PCT arm (median [Q1-Q3], 2.73 [2.26-3.4] at day 1 vs. 2.43 [1.81-3.21] at day 7; Pt-test = 0.037, Bonferroni corrected α = 0.0125). The relative abundance of Actinomycetota and Pseudomonadota was decreased in the PCT arm by day 7 and that of Bacillota was increased. Early PCT-guided stop of antibiotics contributes to decreased microbiome dysbiosis by day 7.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Emmanouil Stylianakis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Georgia Damoraki
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Panagiotis Koufargyris
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioannis Kollias
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Konstantina Katrini
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Elina Drakou
- 1st Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Konstantinos Marousis
- 1st Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Andronikos Spyrou
- 1st Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Styliani Symbardi
- 1st Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Nikolaos Alexiou
- 1st Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Zoi Alexiou
- 2nd Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Malvina Lada
- 2nd Department of Internal Medicine, Sismanogleio General Hospital of Athens, Athens, Greece
| | - Garyfallia Poulakou
- 3rd Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Georgios Chrysos
- 2nd Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - George Adamis
- 1st Department of Internal Medicine, G. Gennimatas General Hospital of Athens, Athens, Greece
| | - Evangelos J Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
12
|
Vitetta L, Bambling M, Strodl E. Persister Intestinal Bacteria, Epigenetics and Major Depression. FRONT BIOSCI-LANDMRK 2025; 30:26837. [PMID: 40302324 DOI: 10.31083/fbl26837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 05/02/2025]
Abstract
The microbiota-gut-brain axis has been proposed as a potential modulator of mood disorders such as major depression. Complex bidirectional biochemical activities in this axis have been posited to participate in adverse mood disorders. Environmental and genetic factors have dominated recent discussions on depression. The prescription of antibiotics, antidepressants, adverse negative DNA methylation reactions and a dysbiotic gut microbiome have been cited as causal for the development and progression of depression. While research continues to investigate the microbiome-gut-brain axis, this review will explore the state of persistence of gut bacteria that underpins bacterial dormancy, possibly due to adverse environmental conditions and/or pharmaceutical prescriptions. Bacterial dormancy persistence in the intestinal microbial cohort could affect the role of bacterial epigenomes and DNA methylations. DNA methylations are highly motif driven exerting significant control on bacterial phenotypes that can disrupt bacterial metabolism and neurotransmitter formation in the gut, outcomes that can support adverse mood dispositions.
Collapse
Affiliation(s)
- Luis Vitetta
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2005, Australia
| | - Matthew Bambling
- Faculty of Medicine and Health, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Esben Strodl
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4058, Australia
| |
Collapse
|
13
|
Peng T, Song B, Wang Y, Yuan J, Yang Z, Tang L. Trophic transfer of sulfonamide antibiotics in aquatic food chains: A comprehensive review with a focus on environmental health risks. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 369:125823. [PMID: 39923974 DOI: 10.1016/j.envpol.2025.125823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Antibiotics, which have been identified as emerged pollutants, are creating an increase in environmental concerns, with sulfonamide antibiotics (SAs) being among the most commonly discovered antibiotics. Due to their widespread usage and inadequate sewage treatment, SAs are frequently released into the aquatic environment. The introduction of SAs into aquatic environments can kill or inhibit the growth or metabolic activity of microorganisms, thereby affecting biological communities and ecological functions and disrupting the equilibrium of aquatic ecosystems. The transmission of SAs to human beings can occur through trophic transfer of food chains, particularly when humans consume aquatic food. This study examines the trophic transfer of SAs along the aquatic food chain, provides a summarize of the spatial distribution of SAs in aquatic environments, and evaluates the environmental risks associated with it. The prevalence of SAs was predominantly noted in the aqueous phase, with relatively lower concentrations detected in sediments, solidifying their status as one of the most widespread antibiotics among aquatic organisms. SAs, characterized by their high biomagnification capacity and strong bioaccumulative properties in invertebrates, emerge as the antibiotic type with the greatest ecological risks. The ecological risk posed by sulfonamide antibiotics to aquatic organisms is more pronounced than the health risk to humans, suggesting that the adverse effects on aquatic life warrant greater attention. Additionally, this study offers practical recommendations to address the limitations of previous research, emphasizing the importance of regulating exposure and establishing a robust health risk prediction system as effective measures for antibiotic control.
Collapse
Affiliation(s)
- Tianwei Peng
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Biao Song
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China.
| | - Yuchen Wang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Jie Yuan
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Zhengqing Yang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Lin Tang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China.
| |
Collapse
|
14
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [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: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
15
|
Nie F, Guo J, Pan J, Guo Z, Wang C, Yan J, Ma W. Effects of antibiotics on the anti-tumor efficacy of immune checkpoint inhibitor therapy. Clin Transl Oncol 2025; 27:790-799. [PMID: 39046682 DOI: 10.1007/s12094-024-03615-0] [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: 05/05/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE Immunotherapy using immune checkpoint inhibitors (ICIs) has shown several benefits over traditional therapies. However, the eligible population remains small. Antibiotic (ATB) use might reduce immunotherapy efficacy by disrupting the gut microbiota. However, in China, ATB effect on ICI therapy efficacy remains unelucidated. We aimed to assess the effects of ATBs on the anti-tumor efficacy of ICIs to provide a reference for clinical use. METHODS We included 134 patients with advanced tumors undergoing ICI therapy at Shanghai Jiading District Central Hospital from January 1, 2021, to October 1, 2023. They were divided into Non-ATB and ATB groups based on ATB use within 30 days before and after ICI administration. Moreover, we compared progression-free (PFS) and overall (OS) survival between the groups. RESULTS Median PFS and OS were lower in the ATB than in the Non-ATB group (PFS: 4.0 vs. 5.5 months; OS: 5.4 vs. 6.5 months). Univariate analysis revealed that ATB use significantly affected PFS (hazard ratio [HR] = 2.318, 95% confidence interval [CI] = 1.281-4.194, P = 0.005) and OS (HR = 2.115, 95% CI = 1.161-3.850, P = 0.014). Moreover, multivariate analysis revealed poor PFS (HR = 2.573, 95% CI = 1.373-4.826, P = 0.003) and OS (HR = 2.452, 95% CI = 1.298-4.632, P = 0.006) in patients who received ATBs during ICI therapy. CONCLUSIONS ATB use is negatively correlated with ICI therapy efficacy, leading to reduced PFS and OS in patients undergoing such treatment. Owing to the significant impact of ATBs on the human gut microbiome, regulation of the gut microbiome may emerge as a novel therapeutic target that can enhance the clinical activity of ICIs.
Collapse
Affiliation(s)
- Fangfang Nie
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JunGang Guo
- Department of Thoracic Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - JiaYi Pan
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China
| | - ZhaoJiao Guo
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Chun Wang
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Jun Yan
- Department of Oncology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China.
| | - WeiNa Ma
- Department of Pharmacy, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, No.1 Cheng Bei Road, Jia ding District, Shanghai, 201800, China.
| |
Collapse
|
16
|
Bano Y, Shrivastava A, Shukla P, Chaudhary AA, Khan SUD, Khan S. The implication of microbiome in lungs cancer: mechanisms and strategies of cancer growth, diagnosis and therapy. Crit Rev Microbiol 2025; 51:128-152. [PMID: 38556797 DOI: 10.1080/1040841x.2024.2324864] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 04/02/2024]
Abstract
Available evidence illustrates that microbiome is a promising target for the study of growth, diagnosis and therapy of various types of cancer. Lung cancer is a leading cause of cancer death worldwide. The relationship of microbiota and their products with diverse pathologic conditions has been getting large attention. The novel research suggests that the microbiome plays an important role in the growth and progression of lung cancer. The lung microbiome plays a crucial role in maintaining mucosal immunity and synchronizing the stability between tolerance and inflammation. Alteration in microbiome is identified as a critical player in the progression of lung cancer and negatively impacts the patient. Studies suggest that healthy microbiome is essential for effective therapy. Various clinical trials and research are focusing on enhancing the treatment efficacy by altering the microbiome. The regulation of microbiota will provide innovative and promising treatment strategies for the maintenance of host homeostasis and the prevention of lung cancer in lung cancer patients. In the current review article, we presented the latest progress about the involvement of microbiome in the growth and diagnosis of lung cancer. Furthermore, we also assessed the therapeutic status of the microbiome for the management and treatment of lung cancer.
Collapse
Affiliation(s)
- Yasmin Bano
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
| | - Abhinav Shrivastava
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
| | - Piyush Shukla
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
- Laboratory of Natural Products, Department of Rural Technology and Social Development, Guru Ghasidas University, Bilaspur, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health Technology (IIHT), Deoband, Saharanpur, UP, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, Australia
| |
Collapse
|
17
|
Bermúdez-Sánchez S, Bahl MI, Hansen EB, Licht TR, Laursen MF. Oral amoxicillin treatment disrupts the gut microbiome and metabolome without interfering with luminal redox potential in the intestine of Wistar Han rats. FEMS Microbiol Ecol 2025; 101:fiaf003. [PMID: 39779288 PMCID: PMC11775830 DOI: 10.1093/femsec/fiaf003] [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: 01/03/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025] Open
Abstract
Oral antibiotic treatment is well known to be one of the main factors affecting gut microbiota composition by altering bacterial diversity. It decreases the abundance of butyrate-producing bacteria such as Lachnospiraceae and Ruminococcaceae, while increasing abundance of Enterobacteriaceae. The recovery time of commensal bacteria post-antibiotic treatment varies among individuals, and often, complete recovery is not achieved. Recently, gut microbiota disruption has been associated with increased gut oxygen levels and higher redox potential in faecal samples. Given that redox balance is crucial for microbial metabolism and gut health, influencing fermentation processes and maintaining anaerobic conditions, we investigated the impact of oral amoxicillin treatment on the redox potential in the caecum. We used 24 Wistar Han male rats and measured caecal redox potential in situ with a probe, before and after 7 days of amoxicillin treatment, as well as after 7 days of recovery. Additionally, we analysed caecal weight, pH, antioxidant capacity, caecal microbiota, metabolome, and colonic tissue expression of relevant genes involved in the redox potential state. Our findings show that oral amoxicillin treatment significantly reduced archaeal load, and decreased the bacterial alpha diversity and affected bacterial composition of the caecal microbiome. The caecal metabolome was also significantly affected, exemplified by reduced amounts of short chain fatty acids during amoxicillin treatment. While the caecal metabolome fully recovered 7 days post amoxicillin treatment, the microbiome did not fully recover within this time frame. However, amoxicillin did not lead to an increase in luminal redox potential in the cecum during or post amoxicillin treatment. Limited differences were observed for colonic expression of genes involved in intestinal barrier function and generation of reactive oxygen species, except for the catalase gene, which was significantly upregulated post-amoxicillin treatment. Our results suggest that while oral amoxicillin disrupts the gut microbiome and metabolome, it does not directly interfere with gut luminal redox state.
Collapse
Affiliation(s)
- Sandra Bermúdez-Sánchez
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Egon Bech Hansen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Frederik Laursen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
18
|
Guo F, Qiao J, Hu Z, Huang J, Bi R, Abbas W, Zhen W, Guo Y, Wang Z. Yeast cell wall polysaccharides accelerate yet in-feed antibiotic delays intestinal development and maturation via modulating gut microbiome in chickens. J Anim Sci Biotechnol 2025; 16:14. [PMID: 39856758 PMCID: PMC11763161 DOI: 10.1186/s40104-024-01145-x] [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: 09/10/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND It is important to promote intestinal development and maturation of chicks for feed digestion and utilization, intestinal health, and disease resistance. This study aimed to investigate the effects of dietary yeast cell wall polysaccharides (YCWP) addition on intestinal development and maturation of chickens and its potential action mechanism. METHODS 180 one-day-old male Arbor Acres broilers were randomly assigned to three groups containing control (basal diets without any antibiotics or anticoccidial drug), bacitracin methylene disalicylate (BMD)-treated group (50 mg/kg) and YCWP-supplemented group (100 mg/kg). RESULTS Compared with control group, in-feed antibiotic BMD continuous administration significantly decreased crypt depth (d 21) and villus height (d 42) along with mucosal maltase activity (d 42) in the ileum (P < 0.05). Also, BMD markedly downregulated gene expression levels of β-catenin, lysozyme, occludin and FABP-2 (d 21) and innate immune related genes CD83 and MHC-I mRNA levels (d 42, P < 0.05), and decreased goblet cell counts in the ileum of chickens (d 21 and d 42, P < 0.05). While, TLR-2, TLR-6 and iNOS mRNA abundances were notably upregulated by BMD treatment (d 42, P < 0.05). Nevertheless, dietary YCWP addition significantly increased the ratio of villus height to crypt depth (d 21), villus surface area (d 21 and d 42), ileal alkaline phosphatase and maltase activities as well as goblet cell (d 21 and d 42) and IgA-producing plasma cell numbers as compared to BMD treatment (d 21, P < 0.05). YCWP addition also upregulated gene expression levels of Lgr5, Wnt/β-catenin signaling pathway related gene (Wnt3, β-catenin, d 21; β-catenin, d 42), intestinal cells proliferation marker Ki-67 and barrier function related genes (occludin, d 21 and d 42, P < 0.05). Moreover, YCWP significantly increased antigen presenting cell marker related genes (MHC-II, d 21; CD83 and MHC-I, d 42), TLR-1, TLR-2 and TLR-6 mRNA levels (d 21, P < 0.05). Cecal microbiome analysis showed that YCWP addition obviously improved cecal microbial composition, as indicated by increasing relative abundance of Fournierella, Psychrobacter and Ruminiclostridium on d 21, and Alistipes and Lactobacillus on d 42, which were positively related with gut development and maturation related indexes (P < 0.05). CONCLUSION Collectively, YCWP promoted yet antibiotic BMD delayed intestinal morphological and immunological development linked with modulating gut microbiome in chickens.
Collapse
Affiliation(s)
- Fangshen Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jianing Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zeqiong Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jia Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Ruichen Bi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Waseem Abbas
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Wenrui Zhen
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, People's Republic of China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
19
|
Văcărean-Trandafir IC, Amărandi RM, Ivanov IC, Dragoș LM, Mențel M, Iacob Ş, Muşină AM, Bărgăoanu ER, Roată CE, Morărașu Ș, Țuțuianu V, Ciobanu M, Dimofte MG. Impact of antibiotic prophylaxis on gut microbiota in colorectal surgery: insights from an Eastern European stewardship study. Front Cell Infect Microbiol 2025; 14:1468645. [PMID: 39872941 PMCID: PMC11770057 DOI: 10.3389/fcimb.2024.1468645] [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: 07/22/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Antibiotic overuse is driving a global rise in antibiotic resistance, highlighting the need for robust antimicrobial stewardship (AMS) initiatives to improve prescription practices. While antimicrobials are essential for treating sepsis and preventing surgical site infections (SSIs), they can inadvertently disrupt the gut microbiota, leading to postoperative complications. Treatment methods vary widely across nations due to differences in drug choice, dosage, and therapy duration, affecting antibiotic resistance rates, which can reach up to 51% in some countries. In Romania and the Republic of Moldova, healthcare practices for surgical antibiotic prophylaxis differ significantly despite similarities in genetics, culture, and diet. Romania's stricter healthcare regulations result in more standardized antibiotic protocols, whereas Moldova's limited healthcare funding leads to less consistent practices and greater variability in treatment outcomes. Methods This study presents the results of a prospective cross-border investigation involving 86 colorectal cancer patients from major oncological hospitals in Romania and Moldova. We analyzed fecal samples collected from patients before and 7 days post-antibiotic treatment, focusing on the V3-V4 region of the 16S rRNA gene. Results Our findings indicate that inconsistent antibiotic prophylaxis policies-varying in type, dosage, or therapy duration-significantly impacted the gut microbiota and led to more frequent dysbiosis compared to stricter prophylactic antibiotic practices (single dose, single product, limited time). Discussion We emphasize the need for standardized antibiotic prophylaxis protocols to minimize dysbiosis and its associated risks, promoting more effective antimicrobial use, particularly in low- and middle-income countries (LMICs).
Collapse
Affiliation(s)
| | | | | | | | - Mihaela Mențel
- TRANSCEND Research Centre, Regional Institute of Oncology, Iasi, Romania
| | - Ştefan Iacob
- Second Surgical Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ana-Maria Muşină
- Second Surgical Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | | | - Cristian Ene Roată
- Second Surgical Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Ștefan Morărașu
- Second Surgical Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Valeri Țuțuianu
- Scientific Laboratory of Cancer Biology, Institute of Oncology, Chișinău, Moldova
| | - Marcel Ciobanu
- Surgical Oncology Department, Proctology, Institute of Oncology, Chișinău, Moldova
| | - Mihail-Gabriel Dimofte
- Second Surgical Oncology Department, Regional Institute of Oncology, Iasi, Romania
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
20
|
Davarci İ, Davarci PZ. Microbiota awareness levels of medical students: the case of Trakya university. BMC MEDICAL EDUCATION 2025; 25:44. [PMID: 39794753 PMCID: PMC11720333 DOI: 10.1186/s12909-025-06662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Human microbiota is noteworthy for its ability to encode a much greater variety and quantity of proteins than human cells. Despite its dynamic and complex nature, it has been associated with healthy living for over a century. The knowledge and attitudes of healthcare professionals, especially physicians, influence the use of pro-prebiotic interventions to modulate the microbiota. This study aimed to examine the awareness of medical students about the microbiota and the factors influencing their awareness. METHODS The study was conducted between May 15 and August 15, 2023, with the participation of 235 medical students. The students were administered a 13-item questionnaire and the Microbiota Awareness Scale (MAS). The questionnaire and scale were administered face-to-face, and the results were statistically analyzed. RESULTS The students' MAS total mean score was 70.34 ± 8.15. The MAS scores of medical students in the 4th, 5th, and 6th grades who underwent clinical training, those who conducted research on microbiota independently, those who had previously used prebiotics/probiotics, and those who rated themselves highly on microbiota knowledge were statistically higher. CONCLUSIONS The study revealed that students' awareness of the microbiota increased with advancing years of medical education, reaching a high level by the time of graduation. To enable future physicians to effectively communicate this knowledge to the public and incorporate it into clinical practice, it is crucial to provide comprehensive education on microbiota, probiotics, and prebiotics during medical training. Additionally, promoting scientific research and organizing activities in these areas are essential for fostering deeper understanding and practical application.
Collapse
Affiliation(s)
- İsmail Davarci
- Faculty of Medicine, Department of Medical Microbiology, Trakya University, Edirne, Türkiye.
| | - Pınar Zehra Davarci
- Faculty of Medicine, Department of Public Health, Trakya University, Edirne, Türkiye
| |
Collapse
|
21
|
Duong QA, Curtis N, Zimmermann P. The association between prenatal antibiotic exposure and adverse long-term health outcomes in children: A systematic review and meta-analysis. J Infect 2025; 90:106377. [PMID: 39675435 DOI: 10.1016/j.jinf.2024.106377] [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: 07/06/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Antibiotics are the most commonly prescribed drugs during pregnancy. The long-term health risks to children associated with prenatal antibiotic exposure are uncertain. OBJECTIVE To identify the association between prenatal antibiotics and adverse long-term health outcomes in children. METHODS A systematic search was done to identify original studies investigating the association between prenatal antibiotic exposure and adverse long-term health outcomes in children. Studies were excluded if: (i) antibiotics were only given during delivery or (ii) the outcome was present before antibiotic exposure. RESULTS We included 158 studies, reporting 23 outcomes in 21,943,763 children, in our analysis. For the following adverse health outcomes, there was a significant association with antibiotic exposure found in two or more studies: atopic dermatitis (OR 1.27, 95% CI 1.06-1.52, p=0.01), food allergies (OR 1.25, 95% CI 1.09-1.44, p<0.01), allergic rhinoconjunctivitis (OR 1.16, 95% CI 1.15-1.17, p<0.01), wheezing (OR 1.39, 95% CI 1.14-1.69, p<0.01), asthma (OR 1.36, 95% CI 1.24-1.50, p<0.01), obesity (OR 1.36, 95% CI 1.12-1.64, p<0.01), cerebral palsy (OR 1.25, 95% CI 1.10-1.43, p<0.01), epilepsy or febrile seizure (OR 1.16, 95% CI 1.08-1.24, p<0.01), and cancer (OR 1.13, 95% CI 1.01-1.26, p=0.04). CONCLUSION Although causality cannot be implied, these findings support antibiotic stewardship efforts to ensure judicious use of antibiotics during pregnancy to avoid potential long-term health risks.
Collapse
Affiliation(s)
- Quynh A Duong
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| | - Petra Zimmermann
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland.
| |
Collapse
|
22
|
Wu Y, Zhang L, Zheng H, Huang W, Zhang X, Ji W, Ma R, Mao X, Huang Y, Liu X, Zeng J, Guo S, Cheng Z, Wang T, Wang M, Wang W, Huang J, Lu X, Li L, Li Y, Bai X, Wei Y, Li X, Wang X. Oral bacteriophage therapy effectively prevent and control of Clostridium perfringens type c infections in newborn piglets. Vet Microbiol 2025; 300:110330. [PMID: 39674030 DOI: 10.1016/j.vetmic.2024.110330] [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: 07/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Clostridium perfringens (C. perfringens) type C exhibits strong pathogenicity, often leading to swine dysentery, severely affecting the economic efficiency of the pig farming industry. Bacteriophages as bacterial viruses have many natural advantages and are potent candidates for controlling bacterial infections. In this study, a lytic C. perfringens phage designed as vB_CpeP_15N3 was isolated with the host C. perfringens type C CVCC1155, and its potential for therapy was determined in vitro and in vivo. Despite the narrow host range, phage vB_CpeP_15N3 exhibited a large burst size of 102 PFU/cell following a short latent period of 10 min. In addition, phage vB_CpeP_15N3 remained stable at temperatures ranging from 4 to 50°C and pH levels from 5 to 9 and had a strong antibacterial effect in vitro. Through whole-genome analysis, phage vB_CpeP_15N3 belongs to the family Guelinviridae, genus Brucesealvirus with no genes related to lysogeny and bacterial virulence or resistance. We further demonstrated that phage vB_CpeP_15N3 by oral administration for preventive purposes could significantly alleviate clinical symptoms and jejunal lesions of newborn piglets through the reduced colonization of C. perfringens type C in the jejunum and the level of CPB toxin in the content of jejunum in the newborn piglet model of CVCC1155 infection. In addition, phage vB_CpeP_15N3 by oral administration for preventive purposes could improve the diversity and abundance of the jejunum microbiota in newborn piglets. Moreover, the prevention by phage vB_CpeP_15N3 obtained more effective therapeutic results than phage and gentamicin treatments. Taken together, these findings suggested that phage vB_CpeP_15N3 is a promising alternative of antibiotics for preventing and controlling C. perfringens type C infection of newborn piglets.
Collapse
Affiliation(s)
- Yuxing Wu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Lingyuan Zhang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Haodong Zheng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Weiqian Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China; Guangdong Rongda Biology Co., LTD, Qingyuan, 511517, PR China
| | - Xiaochen Zhang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Wantong Ji
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Runwen Ma
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xinyu Mao
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yonghua Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xiaoxin Liu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Jialing Zeng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Shaowen Guo
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Zixin Cheng
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Tong Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Mei Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Wenqi Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Jingjing Huang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xuanyi Lu
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Lixuan Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yinan Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xue Bai
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Yixian Wei
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xun Li
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China
| | - Xiaoye Wang
- College of Animal Science and Technology, Guangxi University, PR China; Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, PR China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, PR China.
| |
Collapse
|
23
|
Sakalli Kani A, Süleyman A. Because of γ-Aminobutyric Acid-Glutamate Imbalance, Gut Microbiota, or Both? Delirious Mania Induced by Ciprofloxacin Use: A Case Report and Review of the Literature. J Psychiatr Pract 2025; 31:46-48. [PMID: 39836682 DOI: 10.1097/pra.0000000000000836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Ciprofloxacin is an antibiotic from the fluoroquinolone group that is frequently used in many clinical practices. In addition to its peripheral neuropathic side effects, it is an antibiotic that can pass through the blood-brain barrier due to its lipophilic features and cause rare central nervous system symptoms. Although cases of neuropsychiatric symptoms developing after treatment with ciprofloxacin have been reported in the literature, the number of reports of manic episodes after ciprofloxacin use is limited, and there have been no reports of delirious mania developing after ciprofloxacin use until the case presented in this report. Here we report the case of a 52-year-old woman who developed manic symptoms after receiving ciprofloxacin, which evolved into delirious mania. Clinical factors that may predispose to neurotoxicity are discussed and compared with features of the mania cases in the literature. The underlying neurobiological mechanisms are also reviewed.
Collapse
Affiliation(s)
- Ayşe Sakalli Kani
- Department of Psychiatry, School of Medicine, Marmara University, Istanbul, Turkey
| | | |
Collapse
|
24
|
Letchumanan G, Marlini M, Baharom N, Lawley B, Syed Mohideen FB, Jogulu SR, Addnan FH, Nur Fariha MM, Omar MR, Pathmanathan SG. Ethnicity-matched case-control study reveals significant gut microbiota differences in Malaysian adults with type 2 diabetes. J Med Microbiol 2025; 74. [PMID: 39886920 DOI: 10.1099/jmm.0.001963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Introduction. Type 2 diabetes mellitus (T2DM) is a major global health issue projected to exceed 700 million cases by 2045. In Malaysia, T2DM prevalence has risen, with notable ethnic disparities.Gap statement. The gut microbiota's role in T2DM pathogenesis is well recognized, yet its composition in Malaysia's ethnically diverse population remains underexplored.Aim. This study aimed to characterize gut microbiota composition among T2DM and ethnicity-matched adults without diabetes (nonDM) in Malaysia.Methodology. A case-control study was conducted with 45 T2DM and 45 nonDM participants matched by ethnicity from a primary care clinic in Klang Valley, Malaysia. Faecal DNA was subjected to 16S rRNA sequencing to identify microbiota diversity and composition differences and compare predicted functional capabilities. Correlations between bacterial taxa, clinical characteristics and dietary intake were analysed.Results. T2DM participants showed decreased alpha diversity (observed, P-value=0.002, r=0.69; Shannon, P-value<0.001, r=0.73) and significant differences in beta diversity (permutational multivariate ANOVA, R²=0.036, P-value=0.001). Linear discriminant analysis effect size and multiple regression analysis, adjusted for covariates age, gender, BMI and intakes of protein, fat, carbohydrate and fibre, identified the phylum Proteobacteria and genera Escherichia-Shigella to be increased, while the genera Anaerostipes and Romboutsia decreased in T2DM. These bacteria were associated with various clinical characteristics and dietary intake. However, these 'potential biomarkers' were not uniformly present across all participants, suggesting that individual bacterial taxa may not serve as universal biomarkers.Conclusion. Significant gut microbiota differences exist between T2DM and nonDM individuals in Malaysia, indicating a dysbiosis characterized by increased pro-inflammatory bacteria and reduced short-chain fatty acid-producing bacteria in T2DM. While these findings highlight the potential functional relevance of gut microbiota in T2DM pathogenesis, addressing limitations such as participant matching for confounding factors in future studies could uncover additional significant differences in microbiota composition. Furthermore, the variability in taxa prevalence across individuals suggests that targeting microbial metabolic products may offer more promising strategies to inform microbiota-targeted interventions than relying solely on specific bacterial taxa as biomarkers.
Collapse
Affiliation(s)
- Geetha Letchumanan
- Department of Medical Sciences, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Muhamad Marlini
- Department of Medical Sciences, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Nizam Baharom
- Public Health Unit, Department of Primary Health Care, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Blair Lawley
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Fathima Begum Syed Mohideen
- Family Medicine Unit, Department of Primary Health Care, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | | | - Faizul Helmi Addnan
- Department of Medical Sciences, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Mohd Manzor Nur Fariha
- Department of Medical Sciences, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Mohd Rahman Omar
- Medical-Based Department, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| | - Siva Gowri Pathmanathan
- Department of Medical Sciences, Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia (USIM), Nilai, Negeri Sembilan, Malaysia
| |
Collapse
|
25
|
Zhang S, Luo C, Li K, Wang J, Wang H, Zhong R, Chen L, Ma Q, Zhang H. Baicalin alleviates intestinal inflammation and microbial disturbances by regulating Th17/Treg balance and enhancing Lactobacillus colonization in piglets. J Anim Sci Biotechnol 2024; 15:172. [PMID: 39707535 DOI: 10.1186/s40104-024-01126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Intestinal inflammation is a common and serious health problem in piglet production, especially enteritis caused by pathogenic Escherichia coli (E. coli). This condition often leads to high mortality, slow weight gain, and significant economic losses. RESULTS In this study, we isolated an E. coli strain, SKLAN202302, from the colon of diarrheal piglets to create an intestinal inflammation model for evaluating the protective effects of baicalin. Piglets infected with E. coli exhibited significant reductions in body weight, feed intake, small intestine length, and ileal goblet cell count (P < 0.05), along with deteriorated ileal morphology. However, baicalin supplementation resulted in body weights, feed intake, and intestinal morphology similar to those of the control group. Notably, there was a significant increase in the colonization of Lactobacillus species, particularly Lactobacillus_reuteri, Lactobacillus_amylovorus, and Lactobacillus_johnii, compared to the E. coli group (P < 0.05). At the metabolic and transcriptional levels, E. coli infection increased inflammatory mediators, including eicosanoids (leukotriene F4, prostaglandin F1a, leukotriene E4, thromboxane B2, prostaglandin G2, and PGH2), monosaccharides, and TCA cycle intermediates (oxoglutaric acid, glutaric acid, adipic acid, citric acid, and isocitric acid) in the ileum. It also promoted the expression of genes related to autoimmune diseases and the Th17 differentiation signaling pathway (CTLA4, IFN-ALPHA-8, IL12RB2, TRAV3, TRAV16, FOS, and VEGFA), as well as inflammatory factors. Conversely, baicalin supplementation not only counteracted these effects but also enhanced the presence of metabolites such as phospholipids [including lysoPC (P-18:1(9Z)/0:0), PC (17:0/0:0), lysoPC (16:1(9Z)/0:0), PC (18:0/0:0), lysoPC (18:0/0:0), PA (10:0/i-16:0), and PA (10:0/8:0)] and amino acids. It also regulated genes within the IL-17 signaling pathway (IL4, CCL17, CXCL10, IFNG, and CXCL2), suggesting a mechanism by which baicalin mitigates E. coli-induced intestinal and microbial disturbances. Subsequent flow cytometry analysis showed that E. coli infection increased the numbers of CD3+ and Foxp3+ cells, decreased IL-17A+ cells, and reduced Th17/Treg ratios. Baicalin supplementation restored these parameters to control levels. CONCLUSIONS Baicalin supplementation effectively alleviates E. coli-induced intestinal inflammation and microbial disturbances in piglets by enhancing beneficial Lactobacillus colonization, counteracting inflammatory mediators, and regulating immune-related gene expression and the Th17/Treg balance. These findings highlight baicalin's potential in alleviating intestinal inflammation.
Collapse
Affiliation(s)
- Shunfen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chengzeng Luo
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Kai Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Junhong Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Huixin Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qiugang Ma
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
26
|
Ardura-Garcia C, Curtis N, Zimmermann P. Systematic review of the impact of intestinal microbiota on vaccine responses. NPJ Vaccines 2024; 9:254. [PMID: 39706841 DOI: 10.1038/s41541-024-01000-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/17/2024] [Indexed: 12/23/2024] Open
Abstract
The intestinal microbiota plays a critical role in host immunity and might contribute to the significant variation between individuals' vaccine responses. A systematic search was done using MEDLINE and Embase to identify original human studies investigating the association between intestinal microbiota composition and humoral and cellular vaccine responses. In total, 30 publications (26 studies, 14 in infants, 12 in adults), were included. Of these, 26 publications found an association between intestinal microbiota composition and vaccine responses. A beneficial effect of Actynomycetota (particularly Bifidobacterium) and a detrimental effect of Pseudomonadota (particularly Gammaproteobacteria) were observed across studies. Study designs were highly heterogenous, with variation in vaccine type, outcome measure, timing of stool analysis and analysis methods. Overall, studies support the concept that the composition of the intestinal microbiota influences vaccine responses. Further adequately powered studies are needed to confirm this association and inform potential microbiota-targeted interventions to optimise vaccine responses.
Collapse
Affiliation(s)
- Cristina Ardura-Garcia
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, Cambodia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland.
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia.
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
27
|
Shahid A, Chambers S, Scott-Thomas A, Bhatia M. Gut Microbiota and Liver Dysfunction in Sepsis: The Role of Inflammatory Mediators and Therapeutic Approaches. Int J Mol Sci 2024; 25:13415. [PMID: 39769181 PMCID: PMC11678143 DOI: 10.3390/ijms252413415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sepsis is a life-threatening complication caused by an uncontrolled immune response to infection that can lead to multi-organ dysfunction, including liver injury. Recent research has shown the critical role of gut microbiota in sepsis pathogenesis, with the gut-liver axis playing a crucial role in disease progression. Mechanisms such as the disruption of the gut barrier and liver injury pathways mediated by cytokines, chemokines, adhesion molecules, hydrogen sulfide (H2S). and substance P (SP) have been the focus of recent studies. Some potential biomarkers and gut microbiota-targeted therapies have shown promise as emerging tools for predicting and managing sepsis. This review describes the role of the gut-liver axis in sepsis and the potential of microbiota-targeted therapies and biomarker-driven interventions to improve sepsis outcomes.
Collapse
Affiliation(s)
| | | | | | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (A.S.); (S.C.); (A.S.-T.)
| |
Collapse
|
28
|
Airola C, Severino A, Spinelli I, Gasbarrini A, Cammarota G, Ianiro G, Ponziani FR. "Pleiotropic" Effects of Antibiotics: New Modulators in Human Diseases. Antibiotics (Basel) 2024; 13:1176. [PMID: 39766566 PMCID: PMC11727521 DOI: 10.3390/antibiotics13121176] [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: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 01/15/2025] Open
Abstract
Antibiotics, widely used medications that have significantly increased life expectancy, possess a broad range of effects beyond their primary antibacterial activity. While some are recognized as adverse events, others have demonstrated unexpected benefits. These adjunctive effects, which have been defined as "pleiotropic" in the case of other pharmacological classes, include immunomodulatory properties and the modulation of the microbiota. Specifically, macrolides, tetracyclines, and fluoroquinolones have been shown to modulate the immune system in both acute and chronic conditions, including autoimmune disorders (e.g., rheumatoid arthritis, spondyloarthritis) and chronic inflammatory pulmonary diseases (e.g., asthma, chronic obstructive pulmonary disease). Azithromycin, in particular, is recommended for the long-term treatment of chronic inflammatory pulmonary diseases due to its well-established immunomodulatory effects. Furthermore, antibiotics influence the human microbiota. Rifaximin, for example, exerts a eubiotic effect that enhances the balance between the gut microbiota and the host immune cells and epithelial cells. These pleiotropic effects offer new therapeutic opportunities by interacting with human cells, signaling molecules, and bacteria involved in non-infectious diseases like spondyloarthritis and inflammatory bowel diseases. The aim of this review is to explore the pleiotropic potential of antibiotics, from molecular and cellular evidence to their clinical application, in order to optimize their use. Understanding these effects is essential to ensure careful use, particularly in consideration of the threat of antimicrobial resistance.
Collapse
Affiliation(s)
- Carlo Airola
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Severino
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Irene Spinelli
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (A.S.); (I.S.); (A.G.); (G.C.); (G.I.)
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| |
Collapse
|
29
|
Luo Y, Sheikh TMM, Li X, Yuan Y, Yao F, Wang M, Guo X, Wu J, Shafiq M, Xie Q, Jiao X. Exploring the dynamics of gut microbiota, antibiotic resistance, and chemotherapy impact in acute leukemia patients: A comprehensive metagenomic analysis. Virulence 2024; 15:2428843. [PMID: 39620486 PMCID: PMC11622590 DOI: 10.1080/21505594.2024.2428843] [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: 05/07/2024] [Revised: 08/24/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024] Open
Abstract
Leukemia poses significant challenges to its treatment, and understanding its complex pathogenesis is crucial. This study used metagenomic sequencing to investigate the interplay between chemotherapy, gut microbiota, and antibiotic resistance in patients with acute leukemia (AL). Pre- and post-chemotherapy stool samples from patients revealed alterations in microbial richness, taxa, and antibiotic resistance genes (ARGs). The analysis revealed a decreased alpha diversity, increased dispersion in post-chemotherapy samples, and changes in the abundance of specific bacteria. Key bacteria such as Enterococcus, Klebsiella, and Escherichia coli have been identified as prevalent ARG carriers. Correlation analysis between gut microbiota and blood indicators revealed potential links between microbial species and inflammatory biomarkers, including C-reactive protein (CRP) and adenosine deaminase (ADA). This study investigated the impact of antibiotic dosage on microbiota and ARGs, revealing networks connecting co-occurring ARGs with microbial species (179 nodes, 206 edges), and networks associated with ARGs and antibiotic dosages (50 nodes, 50 edges). Antibiotics such as cephamycin and sulfonamide led to multidrug-resistant Klebsiella colonization. Our analyses revealed distinct microbial profiles with Salmonella enterica elevated post-chemotherapy in NF patients and Akkermansia muciniphila elevated pre-chemotherapy. These microbial signatures could inform strategies to modulate the gut microbiome, potentially mitigating the risk of neutropenic fever in patients undergoing chemotherapy. Finally, a comprehensive analysis of KEGG modules shed light on disrupted metabolic pathways after chemotherapy, providing insights into potential targets for managing side effects. Overall, this study revealed intricate relationships between gut microbiota, chemotherapy, and antibiotic resistance, providing new insights into improving therapy and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ying Luo
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - YuMeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Meimei Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoling Guo
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Jilong Wu
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Shantou University Medical College, Shantou, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, China
| |
Collapse
|
30
|
Hayakawa K, Saito S, Miyoshi-Akiyama T, Fukui Y, Takemoto N, Hashimoto T, Inagaki T, Hirose K, Kobayashi K, Koizumi R, Endo M, Komatsubara M, Nomoto H, Inada M, Ide S, Kamegai K, Ashida S, Nagata N, Kato H, Ohmagari N. Comparison of the effects of cefmetazole and meropenem on microbiome: A pilot study. J Infect Chemother 2024; 30:1274-1279. [PMID: 38879077 DOI: 10.1016/j.jiac.2024.06.007] [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: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Cefmetazole (CMZ) is a carbapenem-sparing option in the treatment of extended-spectrum beta-lactamase (ESBL)-producing bacterial infection. In this pilot study, we aimed to compare the effects of antimicrobial treatment (meropenem [MP] and CMZ) with those of no antimicrobial treatment (control group) on the microbiome. METHODS The study was a multicenter, prospective, observational pilot study conducted from October 2020 to October 2022. Feces and saliva samples were collected for microbiome analyses at two time points (early-period: days 1-3; and late-period: days 4-30) for the antimicrobial treatment group, and at one time point for the control group. RESULTS Five feces (MP-F and CMZ-F) and five saliva (MP-S and CMZ-S) samples were included in the MP and the CMZ groups. Ten feces (C-F) and saliva (C-S) samples were included in the control group. Group α diversity was notably lower in the late-period MP-F group than the control group as determined with the Shannon richness index. β diversity analysis of the feces samples based on weighted and unweighted UniFrac distances revealed distinctions in both the late-period CMZ-F and MP-F groups compared with the control group. Weighted UniFrac analysis showed that only the early-period MP-F group differed from the control group. In the saliva samples, weighted and unweighted UniFrac analyses showed significant differences between the control group and the early CMZ, late CMZ, and late MP groups. CONCLUSIONS MP treatment may cause larger impact on the feces microbiome than CMZ in Japanese patients.
Collapse
Affiliation(s)
- Kayoko Hayakawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuto Fukui
- Department of Infectious Diseases, Toho University Omori Medical Center, Tokyo, Japan
| | - Norihiko Takemoto
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Takeshi Inagaki
- Department of General Internal Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Keika Hirose
- Department of Emergency Medicine and Critical Care, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kentaro Kobayashi
- Department of Emergency Medicine and Critical Care, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ryuji Koizumi
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mio Endo
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mika Komatsubara
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hidetoshi Nomoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Makoto Inada
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Ide
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kohei Kamegai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinobu Ashida
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Kanagawa, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Deng F, Du X, Zhang P, Xu J, Li Y, Yang Z. Impact of Antibiotic on Efficacy and Adverse Reactions of Chemoimmunotherapy in Non-small Cell Lung Cancer Patients: A Retrospective Cohort Study. Thorac Cancer 2024; 15:2560-2569. [PMID: 39551485 DOI: 10.1111/1759-7714.15490] [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: 09/10/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND This study aimed to evaluate the impact of antibiotic exposure on efficacy and adverse reactions in non-small cell lung cancer (NSCLC) patients receiving chemoimmunotherapy, and to explore any specific associations on the basis of antibiotic class. METHODS A retrospective study was conducted on NSCLC patients who received chemoimmunotherapy in two Shandong hospitals between January 2018 and October 2023. The association between antibiotic exposure and progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and incidence of immune related adverse reactions (irAE) of patients were evaluated. RESULTS Of the 316 patients, 134 (42.41%) received antibiotics (ATB group), and 182 (57.59%) did not (N-ATB group). There was no significant difference in PFS (aHR = 1.009, 95% CI: 0.770-1.323; p = 0.946) or OS (aHR = 1.420, 95% CI: 0.986-2.047; p = 0.060) between ATB and N-ATB groups. The impact on efficacy was related to the type of antibiotic. β-Lactams (aHR = 1.737, 95% CI: 1.148-2.629; p = 0.009), in particular β-lactam/β-lactamase inhibitor combinations (BLBLIs) (aHR = 1.885, 95% CI: 1.207-2.944, p = 0.005) were associated with poorer OS. However, quinolones (aHR = 1.192, 95% CI: 0.861-1.650; p = 0.291) were not associated with OS. The incidence of irAEs was not significantly different between ATB and N-ATB groups (p = 0.073), but was higher with BLBLIs (p = 0.013). CONCLUSIONS In NSCLC patients receiving chemoimmunotherapy, no significant difference was observed in efficacy and incidence of irAEs between the ATB and the n-ATB groups. In antibiotic class analysis, β-lactams and specifically BLBLIs were observed to be associated with worse OS.
Collapse
Affiliation(s)
- Fang Deng
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Xiuwei Du
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Ping Zhang
- Department of Oncology, Binzhou People's Hospital, Binzhou, Shandong, China
| | - Jing Xu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhongfei Yang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| |
Collapse
|
32
|
Simoni A, Schwartz L, Junquera GY, Ching CB, Spencer JD. Current and emerging strategies to curb antibiotic-resistant urinary tract infections. Nat Rev Urol 2024; 21:707-722. [PMID: 38714857 PMCID: PMC11540872 DOI: 10.1038/s41585-024-00877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/23/2024]
Abstract
Rising rates of antibiotic resistance in uropathogenic bacteria compromise patient outcomes and prolong hospital stays. Consequently, new strategies are needed to prevent and control the spread of antibiotic resistance in uropathogenic bacteria. Over the past two decades, sizeable clinical efforts and research advances have changed urinary tract infection (UTI) treatment and prevention strategies to conserve antibiotic use. The emergence of antimicrobial stewardship, policies from national societies, and the development of new antimicrobials have shaped modern UTI practices. Future UTI management practices could be driven by the evolution of antimicrobial stewardship, improved and readily available diagnostics, and an improved understanding of how the microbiome affects UTI. Forthcoming UTI treatment and prevention strategies could employ novel bactericidal compounds, combinations of new and classic antimicrobials that enhance bacterial killing, medications that prevent bacterial attachment to uroepithelial cells, repurposing drugs, and vaccines to curtail the rising rates of antibiotic resistance in uropathogenic bacteria and improve outcomes in people with UTI.
Collapse
Affiliation(s)
- Aaron Simoni
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
| | - Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Guillermo Yepes Junquera
- Department of Pediatrics, Division of Infectious Diseases, Nationwide Children's, Columbus, OH, USA
| | - Christina B Ching
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Urology, Nationwide Children's, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
33
|
Bencivenni S, Brigidi P, Zannoni A, Ventrella D, Elmi A, Bacci ML, Forni M, D'Amico F, Turroni S. Göttingen Minipigs as a Model for Assessing the Impact of Drugs on the Gut and Milk Microbiota-A Preliminary Study. Nutrients 2024; 16:4060. [PMID: 39683454 DOI: 10.3390/nu16234060] [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: 10/25/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Early gut microbiota (GM) dysbiosis can affect a child's health and has been linked to the onset of pathologies later in life. Breast milk is recognized as a major driver of the structure and dynamics of an infant's GM. In addition to nutritious and prebiotic compounds, milk contains a microbiota that is shaped by several maternal factors, including gut microorganisms and medications. However, the impact of the latter on the milk microbiota is still largely unknown. Here, we investigated the effects of amoxicillin on the milk microbiota and GM of lactating Göttingen Minipigs sows, a promising model for studying medication transfer during lactation. METHODS Three sows were given amoxicillin (7 mg/kg/day) for three weeks starting from the second week after farrowing. Fecal and milk samples were collected before and after treatment and profiled by 16S rRNA amplicon sequencing. RESULTS Göttingen Minipigs' milk microbiota showed similarities to that of humans and conventional sows, with minor compositional shifts after treatment. At the genus level, we observed a decrease in Staphylococcus and o_Bacteroidales;Other;Other, and an increasing trend in the abundance of Streptococcus, Stenotrophomonas, f_Rhodobacteraceae;Other, Proteiniclasticum, f_Propionibacteriaceae;Other and Gemella. In contrast, as expected, the GM was strongly affected by amoxicillin, even at the phylum level. CONCLUSIONS In addition to demonstrating the relevance of Göttingen Minipigs as a valid model for studying the impact of medications on maternal milk and GM, our findings suggest that the milk microbiota may be more stable during antibiotic treatment than the GM.
Collapse
Affiliation(s)
- Silvia Bencivenni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, 40126 Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, 40126 Bologna, Italy
| | - Domenico Ventrella
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, 40126 Bologna, Italy
| | - Alberto Elmi
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
- Department of Veterinary Sciences, University of Pisa, 56124 Pisa, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, 40126 Bologna, Italy
| | - Monica Forni
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, 40126 Bologna, Italy
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
34
|
Guarner F, Bustos Fernandez L, Cruchet S, Damião A, Maruy Saito A, Riveros Lopez JP, Rodrigues Silva L, Valdovinos Diaz MA. Gut dysbiosis mediates the association between antibiotic exposure and chronic disease. Front Med (Lausanne) 2024; 11:1477882. [PMID: 39568738 PMCID: PMC11576192 DOI: 10.3389/fmed.2024.1477882] [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: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Antibiotics are safe, effective drugs and continue to save millions of lives and prevent long-term illness worldwide. A large body of epidemiological, interventional and experimental evidence shows that exposure to antibiotics has long-term negative effects on human health. We reviewed the literature data on the links between antibiotic exposure, gut dysbiosis, and chronic disease (notably with regard to the "developmental origins of health and disease" ("DOHaD") approach). Molecular biology studies show that the systemic administration of antibiotic to infants has a rapid onset but also often a long-lasting impact on the microbial composition of the gut. Along with other environmental factors (e.g., an unhealthy "Western" diet and sedentary behavior), antibiotics induce gut dysbiosis, which can be defined as the disruption of a previously stable, functionally complete microbiota. Gut dysbiosis many harmful long-term effects on health. Associations between early-life exposure to antibiotics have been reported for chronic diseases, including inflammatory bowel disease, celiac disease, some cancers, metabolic diseases (obesity and type 2 diabetes), allergic diseases, autoimmune disorders, atherosclerosis, arthritis, and neurodevelopmental, neurodegenerative and other neurological diseases. In mechanistic terms, gut dysbiosis influences chronic disease through direct effects on mucosal immune and inflammatory pathways, plus a wide array of direct or indirect effects of short-chain fatty acids, the enteric nervous system, peristaltic motility, the production of hormones and neurotransmitters, and the loss of intestinal barrier integrity (notably with leakage of the pro-inflammatory endotoxin lipopolysaccharide into the circulation). To mitigate dysbiosis, the administration of probiotics in patients with chronic disease is often (but not always) associated with positive effects on clinical markers (e.g., disease scores) and biomarkers of inflammation and immune activation. Meta-analyses are complicated by differences in probiotic composition, dose level, and treatment duration, and large, randomized, controlled clinical trials are lacking in many disease areas. In view of the critical importance of deciding whether or not to prescribe antibiotics (especially to children), we suggest that the DOHaD concept can be logically extended to "gastrointestinal origins of health and disease" ("GOHaD") or even "microbiotic origins of health and disease" ("MOHaD").
Collapse
Affiliation(s)
| | - Luis Bustos Fernandez
- Centro Medico Bustos Fernandez, Instituto de Gastroenterologia, Buenos Aires, Argentina
| | - Sylvia Cruchet
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
| | - Adérson Damião
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aldo Maruy Saito
- Catedra de Pediatria, Hospital Cayetano Heredia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | |
Collapse
|
35
|
Xu WB, Wang YF, Meng SY, Zhang XT, Wang YR, Liu ZY. Effects of antibiotic and disinfectant exposure on the mouse gut microbiome and immune function. Microbiol Spectr 2024; 12:e0061124. [PMID: 39292002 PMCID: PMC11536992 DOI: 10.1128/spectrum.00611-24] [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: 03/10/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
This study explores the effects of disinfectant and antibiotic exposure on gut health, focusing on gut microbiota balance and gut immune function. Our analysis indicates that disinfectants increase the proportion of Gram-positive bacteria, particularly increasing Staphylococcus levels, while antibiotics increase the proportion of Gram-negative bacteria, especially Bacteroides levels. These changes disrupt microbial harmony and affect the gut microbiome's functional capacity. Additionally, our research reveals that both disinfectants and antibiotics reduce colon length and cause mucosal damage. A significant finding is the downregulation of NLRC4, a key immune system regulator in the gut, accompanied by changes in immune factor expression. This interaction between chemical exposure and immune system dysfunction increases susceptibility to inflammatory bowel disease and other gut conditions. Given the importance of disinfectants in disease prevention, this study advocates for a balanced approach to their use, aiming to protect public health while minimizing adverse effects on the gut microbiome and immune function. IMPORTANCE Disinfectants are extensively employed across various sectors, such as the food sector. Disinfectants are widely used in various sectors, including the food processing industry, animal husbandry, households, and pharmaceuticals. Their extensive application risks environmental contamination, impacting water and soil quality. However, the effect of disinfectant exposure on the gut microbiome and the immune function of animals remains a significant, unresolved issue with profound public health implications. This highlights the need for increased scrutiny and more regulated use of disinfectants to mitigate unintended consequences on gut health and maintain immune system integrity.
Collapse
Affiliation(s)
- Wen-Bo Xu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| | - Yun-Fan Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| | - Si-Yu Meng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| | - Xiao-Tong Zhang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| | - Yi-Rong Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| | - Zhao-Ying Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha, China
| |
Collapse
|
36
|
Zhang J, Zhang X, Xiao B, Ouyang J, Wang P, Peng X. Analyzing the causal role of blood cells in aging: a Mendelian randomization study. Biogerontology 2024; 26:7. [PMID: 39495328 DOI: 10.1007/s10522-024-10148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Blood cells are crucial components of the human body, closely linked to the aging process. This study aims to explore the causal relationship between 91 blood cell phenotypes and aging through Mendelian randomization (MR) analysis. Exposure data from genome-wide association studies (GWAS) was extracted from the GWAS of blood cell perturbation phenotypes in 2,600 European individuals. Initial analysis utilized GWAS data related to aging from the GWAS Catalog database GCST90014288, with inverse-variance weighting as the primary method for causal analysis. Sensitivity analyses included Cochran's Q test, MR-Egger intercept test, MR-PRESSO, and leave-one-out analysis. For significant associations, replication and meta-analysis were conducted using independent aging GWAS data from GCST90014300. Initial analysis revealed that environmental peroxide-impacted red blood cells and ciprofloxacin-impacted reticulocytes accelerated aging. Additionally, elevated neutrophil levels were found to accelerate aging, while LiCl-impacted neutrophils reduced aging risk. Replication and meta-analysis showed consistent results: ciprofloxacin-impacted reticulocytes and elevated neutrophil levels increased the risk of aging, while LiCl-impacted neutrophils reduced the risk. RBCs showed no significant impact on aging progression. Sensitivity analyses confirmed the robustness and reliability of these positive findings. Our study provides evidence of a causal relationship between three blood cell disturbance phenotypes and human aging.
Collapse
Affiliation(s)
- Jingjing Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
| | - Xin Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
| | - Boan Xiao
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
| | - Jiecai Ouyang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China
| | - Peng Wang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China.
| | - Xiaobin Peng
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, 510900, China.
| |
Collapse
|
37
|
Taghaddos D, Saqib Z, Bai X, Bercik P, Collins SM. Post-infectious ibs following Clostridioides difficile infection; role of microbiota and implications for treatment. Dig Liver Dis 2024; 56:1805-1809. [PMID: 38653643 DOI: 10.1016/j.dld.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/25/2024]
Abstract
Up to 25% of patients recovering from antibiotic-treated Clostridioides difficile infection (CDI) develop functional symptoms reminiscent of Post-Infectious Irritable Bowel Syndrome (PI-IBS). For patients with persistent symptoms following infection, a clinical dilemma arises as to whether to provide additional antibiotic treatment or to adopt a conservative symptom-based approach. Here, we review the literature on CDI-related PI-IBS and compare the findings with PI-IBS. We review proposed mechanisms, including the role of C. difficile toxins and the microbiota, and discuss implications for therapy. We suggest that gut dysfunction post-CDI may be initiated by toxin-induced damage to enteroglial cells and that a dysbiotic gut microbitota maintains the clinical phenotype over time, prompting consideration of microbiota-directed therapies. While Fecal Microbial Transplant (FMT) is currently reserved for recurrent CDI (rCDI), we propose that microbiota-directed therapies may have a role in primary CDI in order to avoid or mitigate futher antibiotic treatment that further disrupts the microbiota and thus prevent PI-IBS. We discuss novel microbial transfer therapies and as they emerge, we recommend clinical trials to determine whether microbial transfer therapy of the primary infection prevents both rCDI and CDI-related PI- IBS.
Collapse
Affiliation(s)
- Dana Taghaddos
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Zarwa Saqib
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Xiaopeng Bai
- Division of Gastroenterology, Kyushu University, Japan
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Stephen M Collins
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
38
|
Piccioni A, Spagnuolo F, Candelli M, Voza A, Covino M, Gasbarrini A, Franceschi F. The Gut Microbiome in Sepsis: From Dysbiosis to Personalized Therapy. J Clin Med 2024; 13:6082. [PMID: 39458032 PMCID: PMC11508704 DOI: 10.3390/jcm13206082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Sepsis is a complex clinical syndrome characterized by an uncontrolled inflammatory response to an infection that may result in septic shock and death. Recent research has revealed a crucial link between sepsis and alterations in the gut microbiota, showing that the microbiome could serve an essential function in its pathogenesis and prognosis. In sepsis, the gut microbiota undergoes significant dysbiosis, transitioning from a beneficial commensal flora to a predominance of pathobionts. This transformation can lead to a dysfunction of the intestinal barrier, compromising the host's immune response, which contributes to the severity of the disease. The gut microbiota is an intricate system of protozoa, fungi, bacteria, and viruses that are essential for maintaining immunity and metabolic balance. In sepsis, there is a reduction in microbial heterogeneity and a predominance of pathogenic bacteria, such as proteobacteria, which can exacerbate inflammation and negatively influence clinical outcomes. Microbial compounds, such as short-chain fatty acids (SCFAs), perform a crucial task in modulating the inflammatory response and maintaining intestinal barrier function. However, the role of other microbiota components, such as viruses and fungi, in sepsis remains unclear. Innovative therapeutic strategies aim to modulate the gut microbiota to improve the management of sepsis. These include selective digestive decontamination (SDD), probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT), all of which have shown potential, although variable, results. The future of sepsis management could benefit greatly from personalized treatment based on the microbiota. Rapid and easy-to-implement tests to assess microbiome profiles and metabolites associated with sepsis could revolutionize the disease's diagnosis and management. These approaches could not only improve patient prognosis but also reduce dependence on antibiotic therapies and promote more targeted and sustainable treatment strategies. Nevertheless, there is still limited clarity regarding the ideal composition of the microbiota, which should be further characterized in the near future. Similarly, the benefits of therapeutic approaches should be validated through additional studies.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Fabio Spagnuolo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Antonio Voza
- Department of Emergency Medicine, IRCCS-Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Medical and Surgical Science Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
39
|
Wurm J, Curtis N, Zimmermann P. The effect of antibiotics on the intestinal microbiota in children - a systematic review. FRONTIERS IN ALLERGY 2024; 5:1458688. [PMID: 39435363 PMCID: PMC11491438 DOI: 10.3389/falgy.2024.1458688] [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: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Background Children are the age group with the highest exposure to antibiotics (ABX). ABX treatment changes the composition of the intestinal microbiota. The first few years of life are crucial for the establishment of a healthy microbiota and consequently, disturbance of the microbiota during this critical period may have far-reaching consequences. In this review, we summarise studies that have investigated the effect of ABX on the composition of the intestinal microbiota in children. Methods According to the PRISMA guidelines, a systematic search was done using MEDLINE and Embase to identify original studies that have investigated the effect of systemic ABX on the composition of the intestinal microbiota in children. Results We identified 89 studies investigating a total of 9,712 children (including 4,574 controls) and 14,845 samples. All ABX investigated resulted in a reduction in alpha diversity, either when comparing samples before and after ABX or children with ABX and controls. Following treatment with penicillins, the decrease in alpha diversity persisted for up to 6-12 months and with macrolides, up to the latest follow-up at 12-24 months. After ABX in the neonatal period, a decrease in alpha diversity was still found at 36 months. Treatment with penicillins, penicillins plus gentamicin, cephalosporins, carbapenems, macrolides, and aminoglycosides, but not trimethoprim/sulfamethoxazole, was associated with decreased abundances of beneficial bacteria including Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, and/or Bifidobacterium, and Lactobacillus. The direction of change in the abundance of Enterobacteriaceae varied with ABX classes, but an increase in Enterobacteriaceae other than Escherichia coli was frequently observed. Conclusion ABX have profound effects on the intestinal microbiota of children, with notable differences between ABX classes. Macrolides have the most substantial impact while trimethoprim/sulfamethoxazole has the least pronounced effect.
Collapse
Affiliation(s)
- Juliane Wurm
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Health Science and Medicine, University Lucerne, Lucerne, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
40
|
Wang XY, Meng FH, Zhang MY, Li FX, Lei YX, Ma ZG, Li JQ, Lou YN, Chu YF, Ma K, Yu SX. Gut Lactococcus garvieae promotes protective immunity to foodborne Clostridium perfringens infection. Microbiol Spectr 2024; 12:e0402523. [PMID: 39190634 PMCID: PMC11448249 DOI: 10.1128/spectrum.04025-23] [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: 11/23/2023] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
The gut microbiota, a pivotal component of the intestinal mucosal barrier, is critical for host resistance to enteric pathogen infection. Here, we report a novel function of the potentially probiotic Lactococcus garvieae strain LG1 (L. garvieae strain LG1) in maintaining intestinal mucosal barrier integrity and protecting against foodborne Clostridium perfringens (C. perfringens) infection. L. garvieae was isolated from the intestinal contents of Chinese Mongolian sheep (MS) and exhibited potential probiotic properties. In a C. perfringens enterocolitis model, L. garvieae-pretreated mice were less susceptible to C. perfringens infection compared with Phosphate buffered solution (PBS)-pretreated mice, which manifested as higher survival rates, lower pathogen loads, less weight loss, mild clinical symptoms and intestinal damage, and minor inflammation. Further mechanistic analysis showed that L. garvieae could ameliorate the disruption of intestinal permeability and maintain the integrity of the intestinal mucosal barrier by promoting the expression of tight junction proteins and mucoproteins. Moreover, L. garvieae was also able to facilitate antimicrobial peptide expression and ameliorate dysbiosis of the gut microbiota caused by C. perfringens. Together, these findings highlight the prospect of immunomodulatory potentially probiotic L. garvieae and might offer valuable strategies for prophylaxis and/or treatment of pathogenic C. perfringens mucosal infection. IMPORTANCE C. perfringens necrotic enteritis leads to losses of about US $2 billion to the poultry industry worldwide every year. Worse, US Centers for Disease Control and Prevention (CDC) has estimated that C. perfringens causes nearly 1 million foodborne illnesses in the United States annually. Nowadays, the treatment recommendation is a combination of a broad-spectrum synergistic penicillin with clindamycin or a carbapenem, despite growing scientific concern over antibiotic resistance. The global understanding of the gut microbiome for C. perfringens infection may provide important insights into the intervention. L. garvieae originated from Mongolian sheep intestine, exhibited potentially probiotic properties, and was able to limit C. perfringens enterocolitis and pathogenic colonization. Importantly, we found that L. garvieae limits C. perfringens invasion via improving intestinal mucosal barrier function. Also, L. garvieae alleviates C. perfringens-induced gut microbiota dysbiosis. It allowed us to convince that utilization of probiotics to promote protective immunity against pathogens infection is of pivotal importance.
Collapse
Affiliation(s)
- Xue-Yin Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fan-Hua Meng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ming-Yue Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fen-Xin Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yu-Xin Lei
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zhao-Guo Ma
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jia-Qi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ya-Nan Lou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yue-Feng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy agricultural Sciences, Lanzhou, China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy agricultural Sciences, Lanzhou, China
| | - Shui-Xing Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
- Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, College of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
41
|
Zhao R, Yu T, Li J, Niu R, Liu D, Wang W. Single-cell encapsulation systems for probiotic delivery: Armor probiotics. Adv Colloid Interface Sci 2024; 332:103270. [PMID: 39142064 DOI: 10.1016/j.cis.2024.103270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/28/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Functional foods or drugs based on probiotics have gained unprecedented attention and development due to the increasingly clear relationship between probiotics and human health. Probiotics can regulate intestinal microbiota, dynamically participating in various physiological activities to directly affect human health. Some probiotic-based functional preparations have shown great potential in treating multiple refractory diseases. Currently, the survival and activity of probiotic cells in complex environments in vitro and in vivo have taken priority, and various encapsulation systems based on food-derived materials have been designed and constructed to protect and deliver probiotics. However, traditional encapsulation technology cannot achieve precise protection for a single probiotic, which makes it unable to have a significant effect after release. In this case, single-cell encapsulation systems can be assembled based on biological interfaces to protect and functionalize individual probiotic cells, maximizing their physiological activity. This review discussed the arduous challenges of probiotics in food processing, storage, human digestion, and the commonly used probiotic encapsulation system. Besides, a novel technology of probiotic encapsulation was introduced based on single-cell coating, namely, "armor probiotics". We focused on the classification, structural design, and functional characteristics of armor coatings, and emphasized the essential functional characteristics of armor probiotics in human health regulation, including regulating intestinal health and targeted bioimaging and treatment of diseased tissues. Subsequently, the benefits, limitations, potential challenges, as well as future direction of armor probiotics were put forward. We hope this review may provide new insights and ideas for developing a single-cell probiotics encapsulating system.
Collapse
Affiliation(s)
- Runan Zhao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Ting Yu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiaheng Li
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Ruihao Niu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Wenjun Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China.
| |
Collapse
|
42
|
Tu V, Ren Y, Tanes C, Mukhopadhyay S, Daniel SG, Li H, Bittinger K. A quantitative approach to measure and predict microbiome response to antibiotics. mSphere 2024; 9:e0048824. [PMID: 39230261 PMCID: PMC11423569 DOI: 10.1128/msphere.00488-24] [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] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Although antibiotics induce sizable perturbations in the human microbiome, we lack a systematic and quantitative method to measure and predict the microbiome's response to specific antibiotics. Here, we introduce such a method, which takes the form of a microbiome response index (MiRIx) for each antibiotic. Antibiotic-specific MiRIx values quantify the overall susceptibility of the microbiota to an antibiotic, based on databases of bacterial phenotypes and published data on intrinsic antibiotic susceptibility. We applied our approach to five published microbiome studies that carried out antibiotic interventions with vancomycin, metronidazole, ciprofloxacin, amoxicillin, and doxycycline. We show how MiRIx can be used in conjunction with existing microbiome analytical approaches to gain a deeper understanding of the microbiome response to antibiotics. Finally, we generate antibiotic response predictions for the oral, skin, and gut microbiome in healthy humans. Our approach is implemented as open-source software and is readily applied to microbiome data sets generated by 16S rRNA marker gene sequencing or shotgun metagenomics. IMPORTANCE Antibiotics are potent influencers of the human microbiome and can be a source for enduring dysbiosis and antibiotic resistance in healthcare. Existing microbiome data analysis methods can quantify perturbations of bacterial communities but cannot evaluate whether the differences are aligned with the expected activity of a specific antibiotic. Here, we present a novel method to quantify and predict antibiotic-specific microbiome changes, implemented in a ready-to-use software package. This has the potential to be a critical tool to broaden our understanding of the relationship between the microbiome and antibiotics.
Collapse
Affiliation(s)
- Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Yue Ren
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Scott G Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hongzhe Li
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
43
|
Kędzierska-Kapuza K, Łopuszyńska I, Niewiński G, Franek E, Szczuko M. The Influence of Non-Pharmacological and Pharmacological Interventions on the Course of Autosomal Dominant Polycystic Kidney Disease. Nutrients 2024; 16:3216. [PMID: 39339816 PMCID: PMC11434835 DOI: 10.3390/nu16183216] [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: 08/18/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Polycystic kidney disease (PKD) includes autosomal dominant (ADPKD) and autosomal recessive (ARPKD) forms, both of which are primary genetic causes of kidney disease in adults and children. ADPKD is the most common hereditary kidney disease, with a prevalence of 329 cases per million in Europe. This condition accounts for 5-15% of end-stage chronic kidney disease (ESKD) cases, and in developed countries such as Poland, 8-10% of all dialysis patients have ESKD due to ADPKD. The disease is caused by mutations in the PKD1 and PKD2 genes, with PKD1 mutations responsible for 85% of cases, leading to a more aggressive disease course. Recent research suggests that ADPKD involves a metabolic defect contributing to cystic epithelial proliferation and cyst growth. Aim: This review explores the interplay between metabolism, obesity, and ADPKD, discussing dietary and pharmacological strategies that target these metabolic abnormalities to slow disease progression. Conclusion: Metabolic reprogramming therapies, including GLP-1 analogs and dual agonists of GIP/GLP-1 or glucagon/GLP-1 receptors, show promise, though further research is needed to understand their potential in ADPKD treatment fully.
Collapse
Affiliation(s)
- Karolina Kędzierska-Kapuza
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Inga Łopuszyńska
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Grzegorz Niewiński
- Department of Gastroenterological Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Edward Franek
- Department of Internal Diseases, Endocrinology and Diabetology, National Medical Institute of the Ministry of Interior and Administration, 137 Wołoska St., 02-507 Warsaw, Poland
| | - Małgorzata Szczuko
- Department of Human Nutrition and Metabolomic, Pomeranian Medical University, 24 W. Broniewskiego St., 71-460 Szczecin, Poland
| |
Collapse
|
44
|
Dong H, Li R, Zhao N, Dadhania DM, Suthanthiran M, Lee JR, Ling W. Antibiotic subclasses differentially perturb the gut microbiota in kidney transplant recipients. FRONTIERS IN TRANSPLANTATION 2024; 3:1400067. [PMID: 39371270 PMCID: PMC11451434 DOI: 10.3389/frtra.2024.1400067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/27/2024] [Indexed: 10/08/2024]
Abstract
Introduction The impact of antibiotics on the gut microbiota in kidney transplant recipients is not well characterized. In this study, we determine the impact of different subclasses of antibiotics on the gut microbiota in a cohort of 168 kidney transplant recipients. Methods Gut microbiome profiling was performed on 510 fecal specimens using 16S rRNA gene sequencing of the V4-V5 hypervariable region. We classified fecal specimens by antibiotic exposure into 5 categories: Beta-lactam, Fluoroquinolone (FQ), Beta-lactam & FQ Group, Other Antibiotics, and No Antibiotic (No Abx). Mixed-effects regression models were utilized to identify changes in microbial diversity and in the centered log-ratio (CLR) transformed abundance of genera while adjusting for important covariates. Results Antibiotic administration was associated with a significant decrease in the Shannon alpha diversity index, a decreased abundance of 11 taxa including Eubacterium and Ruminococcus, and an increased abundance of 16 taxa including Enterococcus and Staphylococcus. Exposure to Beta-lactam antibiotics was associated with an increased abundance of 10 taxa including Enterococcus and a decreased abundance of 5 taxa including Eubacterium while exposure to FQ antibiotics was associated with an increased abundance of 3 taxa and a decreased abundance of 4 taxa including Ruminococcus. Conclusions Beta-lactam antibiotics and FQ antibiotics have a profound impact on the gut microbiota in kidney transplant recipients. Given the link of the gut microbiota to infectious complications, antibiotic associated changes in the microbiota may lead to an increased risk for further infections.
Collapse
Affiliation(s)
- Hanbo Dong
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Runzhe Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Darshana M. Dadhania
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - John R. Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - Wodan Ling
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
45
|
Kouraki A, Zheng AS, Miller S, Kelly A, Ashraf W, Bazzani D, Bonadiman A, Tonidandel G, Bolzan M, Vijay A, Nightingale J, Menni C, Ollivere BJ, Valdes AM. Metagenomic changes in response to antibiotic treatment in severe orthopedic trauma patients. iScience 2024; 27:110783. [PMID: 39286492 PMCID: PMC11403444 DOI: 10.1016/j.isci.2024.110783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
We investigated changes in microbiome composition and abundance of antimicrobial resistance (AMR) genes post-antibiotic treatment in severe trauma patients. Shotgun sequencing revealed beta diversity (Bray-Curtis) differences between 16 hospitalized multiple rib fractures patients and 10 age- and sex-matched controls (p = 0.043), and between antibiotic-treated and untreated patients (p = 0.015). Antibiotic-treated patients had lower alpha diversity (Shannon) at discharge (p = 0.003) and 12-week post-discharge (p = 0.007). At 12 weeks, they also exhibited a 5.50-fold (95% confidence interval [CI]: 2.86-8.15) increase in Escherichia coli (p = 0.0004) compared to controls. Differential analysis identified nine AMRs that increased in antibiotic-treated compared to untreated patients between hospital discharge and 6 and 12 weeks follow-up (false discovery rate [FDR] < 0.20). Two aminoglycoside genes and a beta-lactamase gene were directly related to antibiotics administered, while five were unrelated. In trauma patients, lower alpha diversity, higher abundance of pathobionts, and increases in AMRs persisted for 12 weeks post-discharge, suggesting prolonged microbiome disruption. Probiotic or symbiotic therapies may offer future treatment avenues.
Collapse
Affiliation(s)
- Afroditi Kouraki
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Amy S. Zheng
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Suzanne Miller
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Anthony Kelly
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Waheed Ashraf
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | | | | - Amrita Vijay
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Jessica Nightingale
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Cristina Menni
- Department of Twin Research, King’s College London, London SE1 7EH, UK
| | - Benjamin J. Ollivere
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Ana M. Valdes
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
46
|
Li D, Liu Z, Fan X, Zhao T, Wen D, Huang X, Li B. Lactic Acid Bacteria-Gut-Microbiota-Mediated Intervention towards Inflammatory Bowel Disease. Microorganisms 2024; 12:1864. [PMID: 39338538 PMCID: PMC11433943 DOI: 10.3390/microorganisms12091864] [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: 08/02/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), arises from intricate interactions involving genetics, environment, and pharmaceuticals with an ambiguous pathogenic mechanism. Recently, there has been an increasing utilization of lactic acid bacteria (LAB) in managing IBD, attributed to their ability to enhance intestinal barrier function, mitigate inflammatory responses, and modulate gut microbiota. This review initiates by elucidating the pathogenesis of IBD and its determinants, followed by an exploration of the mechanisms underlying LAB therapy in UC and CD. Special attention is directed towards their influence on intestinal barrier function and homeostasis regulated by gut microbiota. Furthermore, the review investigates the complex interplay among pivotal gut microbiota, metabolites, and pathways associated with inflammation. Moreover, it underscores the limitations of LAB in treating IBD, particularly in light of their varying roles in UC and CD. This comprehensive analysis endeavors to offer insights for the optimized application of LAB in IBD therapy.
Collapse
Affiliation(s)
- Diantong Li
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Zhenjiang Liu
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xueni Fan
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Tingting Zhao
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Dongxu Wen
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
| | - Xiaodan Huang
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Bin Li
- Institute of Animal Husbandry and Veterinary, Xizang Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa 850000, China; (D.L.); (Z.L.); (X.F.); (T.Z.); (D.W.)
| |
Collapse
|
47
|
Gottwald J, Balke J, Stellmacher J, van Vorst K, Ghazisaeedi F, Fulde M, Alexiev U. Cy3-Based Nanoviscosity Determination of Mucus: Effect of Mucus Collection Methods and Antibiotics Treatment. Macromol Biosci 2024; 24:e2300437. [PMID: 38625085 DOI: 10.1002/mabi.202300437] [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: 09/25/2023] [Revised: 12/21/2023] [Indexed: 04/17/2024]
Abstract
The integrity of the protective mucus layer as a primary defense against pathogen invasion and microbial leakage into the intestinal epithelium can be compromised by the effects of antibiotics on the commensal microbiome. Changes in mucus integrity directly affect the solvent viscosity in the immediate vicinity of the mucin network, that is, the nanoviscosity, which in turn affects both biochemical reactions and selective transport. To assess mucus nanoviscosity, a reliable readout via the viscosity-dependent fluorescence lifetime of the molecular rotor dye cyanine 3 is established and nanoviscosities from porcine and murine ex vivo mucus are determined. To account for different mucin concentrations due to the removal of digestive residues during mucus collection, the power law dependence of mucin concentration on viscosity is used. The impact of antibiotics combinations (meropenem/vancomycin, gentamycin/ampicillin) on ex vivo intestinal mucus nanoviscosity is presented. The significant increase in viscosity of murine intestinal mucus after treatment suggests an effect of antibiotics on the microbiota that affects mucus integrity. This method will be a useful tool to assess how drugs, directly or indirectly, affect mucus integrity. Additionally, the method can be utilized to analyze the role of mucus nanoviscosity in health and disease, as well as in drug development.
Collapse
Affiliation(s)
- Jacqueline Gottwald
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Jens Balke
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Johannes Stellmacher
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Kira van Vorst
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Ulrike Alexiev
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| |
Collapse
|
48
|
Shuai Y, Li N, Zhang Y, Bao Q, Wei T, Yang T, Cheng Q, Wang W, Hu B, Mao C, Yang M. Aptamer-free upconversion nanoparticle/silk biosensor system for low-cost and highly sensitive detection of antibiotic residues. Biosens Bioelectron 2024; 258:116335. [PMID: 38710144 DOI: 10.1016/j.bios.2024.116335] [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: 11/19/2023] [Revised: 04/11/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
The detection of antibiotics is crucial for safeguarding the environment, ensuring food safety, and promoting human health. However, developing a rapid, convenient, low-cost, and sensitive method for antibiotic detection presents significant challenges. Herein, an aptamer-free biosensor was successfully constructed using upconversion nanoparticles (UCNPs) coated with silk fibroin (SF), based on Förster resonance energy transfer (FRET) and the charge-transfer effect, for detecting roxithromycin (RXM). A synergistic FRET efficiency was achieved by utilizing alizarin red and RXM complexes as energy acceptors, with UCNP as the energy donor, and immobilizing an ultrathin SF protein corona within 10 nm. The biosensor detects RXM in deionized water with high sensitivity primarily through monolayer adsorption, with a detection range of 1.0 nM-141.6 nM and a detection limit as low as 0.68 nM. The performance of this biosensor was compared with the ultra-performance liquid chromatography-mass spectrometry (UPLC-MS/MS) method for detecting antibiotics in river water separately and a strong correlation between the two methods was observed. The biosensor exhibited long-term stability in aqueous solutions (up to 60 d) with no attenuation of fluorescence intensity. Furthermore, the biosensor's applicability extended to the highly sensitive detection of other antibiotics, such as azithromycin. This study introduces a low-cost, eco-friendly, and highly sensitive method for antibiotic detection, with broad potential for future applications in environmental, healthcare, and food-related fields.
Collapse
Affiliation(s)
- Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Na Li
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Ying Zhang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Tiancheng Wei
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Qichao Cheng
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wei Wang
- Department of Environmental Science, Zhejiang University, Hangzhou, 310058, PR China
| | - Baolan Hu
- Department of Environmental Science, Zhejiang University, Hangzhou, 310058, PR China
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, PR China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
49
|
Spigaglia P. Clostridioides difficile and Gut Microbiota: From Colonization to Infection and Treatment. Pathogens 2024; 13:646. [PMID: 39204246 PMCID: PMC11357127 DOI: 10.3390/pathogens13080646] [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: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.
Collapse
Affiliation(s)
- Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Roma, Italy
| |
Collapse
|
50
|
Li Y, Fu S, Klein MS, Wang H. High Prevalence of Antibiotic Resistance in Traditionally Fermented Foods as a Critical Risk Factor for Host Gut Antibiotic Resistome. Microorganisms 2024; 12:1433. [PMID: 39065201 PMCID: PMC11279133 DOI: 10.3390/microorganisms12071433] [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: 06/13/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
This study aimed to assess the suitability of fermented food interventions to replenish damaged gut microbiota. Metagenomic assessment of published sequencing data found that fermented food interventions led to a significant increase in the gut antibiotic resistome in healthy human subjects. Antibiotic resistome and viable antibiotic-resistant (AR) bacteria were further highly prevalent in retail kimchi and artisan cheeses by metagenomic and culture analyses. Representative AR pathogens of importance in nosocomial infections, such as Klebsiella pneumoniae, Serratia marcescens, and vancomycin-resistant Enterococcus (VRE), as well as commensals and lactic acid bacteria, were characterized; some exhibited an extremely high minimum inhibitory concentration (MIC) against antibiotics of clinical significance. Exposing fermented food microbiota to representative antibiotics further led to a boost of the corresponding antibiotic and multidrug-resistance gene pools, as well as disturbed microbiota, including the rise of previously undetectable pathogens. These results revealed an underestimated public health risk associated with fermented food intervention at the current stage, particularly for susceptible populations with compromised gut integrity and immune functions seeking gut microbiota rescue. The findings call for productive intervention of foodborne AR via technology innovation and strategic movements to mitigate unnecessary, massive damages to the host gut microbiota due to orally administered or biliary excreted antibiotics.
Collapse
Affiliation(s)
| | | | | | - Hua Wang
- Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Court, Columbus, OH 43210, USA (M.S.K.)
| |
Collapse
|