201
|
Guo D, Ning X, Bai T, Tan L, Zhou Y, Guo Z, Li X. Interaction between Vitamin D homeostasis, gut microbiota, and central precocious puberty. Front Endocrinol (Lausanne) 2024; 15:1449033. [PMID: 39717097 PMCID: PMC11663660 DOI: 10.3389/fendo.2024.1449033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Central precocious puberty (CPP) is an endocrine disease in children, characterized by rapid genital development and secondary sexual characteristics before the age of eight in girls and nine in boys. The premature activation of the hypothalamic-pituitary-gonadal axis (HPGA) limits the height of patients in adulthood and is associated with a higher risk of breast cancer. How to prevent and improve the prognosis of CPP is an important problem. Vitamin D receptor (VDR) is widely expressed in the reproductive system, participates in the synthesis and function of regulatory sex hormones, and affects the development and function of gonads. In addition, gut microbiota plays an important role in human health by mainly regulating metabolites, energy homeostasis, and hormone regulation. This review aims to clarify the effect of vitamin D deficiency on the occurrence and development of CPP and explore the role of gut microbiota in it. Although evidence on the interaction between vitamin D deficiency, gut microbiota, and sexual development remains limited, vitamin D supplementation and gut microbiota interventions offer a promising, non-invasive strategy for managing CPP.
Collapse
Affiliation(s)
- Doudou Guo
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ning
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingfang Tan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanfen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichen Guo
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
202
|
Heras-Molina A, Estellé J, Vázquez-Gómez M, López-García A, Pesantez-Pacheco JL, Astiz S, Garcia-Contreras C, Escudero R, Isabel B, Gonzalez-Bulnes A, Óvilo C. The impact of host genetics on porcine gut microbiota composition excluding maternal and postnatal environmental influences. PLoS One 2024; 19:e0315199. [PMID: 39652543 PMCID: PMC11627362 DOI: 10.1371/journal.pone.0315199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
The gut microbiota of the pig is being increasingly studied due to its implications for host homeostasis and the importance of the pig as a meat source and biomedical model of human diseases. However, most studies comparing the microbiome between different breeds do not consider the influence of maternal environment during the colonization of the microbiota. The aim of the present study was to compare the gut microbiota during postnatal growth between two pig genotypes (purebred Iberian vs. crossbreds Iberian x Large White pigs), gestated in a single maternal environment (pure Iberian mothers) inseminated with heterospermic semen. Postnatally, piglets were maintained in the same environmental conditions, and their microbiota was studied at 60 and 210 days old. Results showed that age had the greatest influence on alpha and beta diversity, and genotype also affected beta diversity at both ages. There were differences in the microbiome profile between genotypes at the ASV and genus levels when jointly analyzing the total number of samples, which may help to explain phenotypical differences. When each time-point was analyzed individually, there were more differences at 210 days-old than 60 days-old. Fecal short-chain fatty acids (SCFA) were also affected by age, but not by genotype. These results may be a basis for further research on host genotype interactions with the gut microbiota.
Collapse
Affiliation(s)
- Ana Heras-Molina
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
- CSIC-INIA, Madrid, Spain
| | - Jordi Estellé
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Marta Vázquez-Gómez
- Sorbonne université, INSERM, Nutrition et obésités: approaches systémiques, Nutriomics, Paris, France
| | | | - José-Luis Pesantez-Pacheco
- CSIC-INIA, Madrid, Spain
- School of Veterinary Medicine and Zootechnics, Faculty of Agricultural Sciences, University of Cuenca, Cuenca, Ecuador
| | | | | | - Rosa Escudero
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
| | - Beatriz Isabel
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n, Madrid, Spain
| | - Antonio Gonzalez-Bulnes
- Faculty of Veterinary Medicine, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | | |
Collapse
|
203
|
Leung HKM, Lo EKK, Chen C, Zhang F, Felicianna, Ismaiah MJ, El-Nezami H. Probiotic Mixture Attenuates Colorectal Tumorigenesis in Murine AOM/DSS Model by Suppressing STAT3, Inducing Apoptotic p53 and Modulating Gut Microbiota. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10405-1. [PMID: 39641861 DOI: 10.1007/s12602-024-10405-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. The standard CRC chemo drug, 5-Fluorouracil (5-FU), has a poor response rate and chemoresistance, prompting the need for a more effective and affordable treatment. In this study, we aimed to evaluate whether Prohep, a novel probiotic mixture, would alleviate azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colorectal tumorigenesis and enhance 5-FU efficacy and its mechanism. Our results suggested that Prohep showed stronger anti-tumorigenesis effects than 5-FU alone or when combined in the AOM/DSS model. Prohep significantly reduced the total tumor count, total tumor size, caecum weight, colonic crypt depth, colonic inflammation, and collagen fibrosis. Prohep downregulated pro-inflammatory TNF-α and proliferative p-STAT3 and upregulated apoptotic p53. Metagenomics analysis indicated that Prohep-enriched Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, and Candidatus Borkfalkia ceftriaxoniphila were inversely correlated to the total tumor count. In addition, Prohep-enriched Prevotella sp. PTAC and Desulfovibrio porci were negatively correlated to AOM/DSS enriched bacteria, while forming a co-existing community with other beneficial bacteria. From KEGG analysis, Prohep downregulated CRC-related pathways and enhanced pathways related to metabolites suppressing CRC like menaquinone, tetrapyrrole, aminolevulinic acid, and tetrahydrofolate. From Metacyc analysis, Prohep downregulated CRC-related peptidoglycan, LPS, and uric acid biosynthesis, and conversion. Prohep elevated the biosynthesis of the beneficial L-lysine, lipoic acid, pyrimidine, and palmitate. Prohep also elevated metabolic pathways related to energy utilization of lactic acid-producing bacteria (LAB) and acetate producers. Similarly, fecal acetate concentration was upregulated by Prohep. To sum up, Prohep demonstrated exceptional anti-tumorigenesis effects in the AOM/DSS model, which revealed its potential to develop into a novel CRC therapeutic in the future.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China.
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland.
| |
Collapse
|
204
|
Tang M, Wu Y, Liang J, Yang S, Huang Z, Hu J, Yang Q, Liu F, Li S. Gut microbiota has important roles in the obstructive sleep apnea-induced inflammation and consequent neurocognitive impairment. Front Microbiol 2024; 15:1457348. [PMID: 39712898 PMCID: PMC11659646 DOI: 10.3389/fmicb.2024.1457348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a state of sleep disorder, characterized by repetitive episodes of apnea and chronic intermittent hypoxia. OSA has an extremely high prevalence worldwide and represents a serious challenge to public health, yet its severity is frequently underestimated. It is now well established that neurocognitive dysfunction, manifested as deficits in attention, memory, and executive functions, is a common complication observed in patients with OSA, whereas the specific pathogenesis remains poorly understood, despite the likelihood of involvement of inflammation. Here, we provide an overview of the current state of the art, demonstrating the intimacy of OSA with inflammation and cognitive impairment. Subsequently, we present the recent findings on the investigation of gut microbiota alteration in the OSA conditions, based on both patients-based clinical studies and animal models of OSA. We present an insightful discussion on the role of changes in the abundance of specific gut microbial members, including short-chain fatty acid (SCFA)-producers and/or microbes with pathogenic potential, in the pathogenesis of inflammation and further cognitive dysfunction. The transplantation of fecal microbiota from the mouse model of OSA can elicit inflammation and neurobehavioral disorders in naïve mice, thereby validating the causal relationship to inflammation and cognitive abnormality. This work calls for greater attention on OSA and the associated inflammation, which require timely and effective therapy to protect the brain from irreversible damage. This work also suggests that modification of the gut microbiota using prebiotics, probiotics or fecal microbiota transplantation may represent a potential adjuvant therapy for OSA.
Collapse
Affiliation(s)
- Mingxing Tang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Yongliang Wu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Junyi Liang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuai Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Zuofeng Huang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Jing Hu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Qiong Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Fei Liu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuo Li
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
205
|
Zhu W, Zhang X, Wang D, Yao Q, Ma GL, Fan X. Simulator of the Human Intestinal Microbial Ecosystem (SHIME ®): Current Developments, Applications, and Future Prospects. Pharmaceuticals (Basel) 2024; 17:1639. [PMID: 39770481 PMCID: PMC11677124 DOI: 10.3390/ph17121639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The human gastrointestinal microbiota plays a vital role in maintaining host health and preventing diseases, prompting the creation of simulators to replicate this intricate system. The Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), a multicompartment dynamic simulator, has emerged as a pivotal in vitro model for studying the interactions and interferences within the human gut microbiota. The continuous and real-time monitoring hallmarks, along with the programmatically flexible setup, bestow SHIME® with the ability to mimic the entire human intestinal ecosystem with high dynamics and stability, allowing the evaluation of various treatments on the bowel microbiota in a controlled environment. This review outlines recent developments in SHIME® systems, including the M-SHIME®, Twin-SHIME®, Triple-SHIME®, and Toddle SHIME® models, highlighting their applications in the fields of food and nutritional science, drug development, gut health research, and traditional Chinese medicine. Additionally, the prospect of SHIME® integrating with other advanced technologies is also discussed. The findings underscore the versatility of SHIME® technology, demonstrating its significant contributions to current gut ecosystem research and its potential for future innovation in microbiome-related fields.
Collapse
Affiliation(s)
- Wei Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
| | - Xiaoyong Zhang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310000, China;
| | - Dong Wang
- Department of Orthopaedics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China;
| | - Qinghua Yao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310005, China;
| | - Guang-Lei Ma
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (W.Z.); (G.-L.M.)
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
- The Joint-Laboratory of Clinical Multi-Omics Research Between Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo 315010, China
| |
Collapse
|
206
|
Zou J, Xu B, Gao H, Luo P, Chen T, Duan H. Microbiome in urologic neoplasms: focusing on tumor immunity. Front Immunol 2024; 15:1507355. [PMID: 39703512 PMCID: PMC11655508 DOI: 10.3389/fimmu.2024.1507355] [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: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024] Open
Abstract
Urological tumors are an important disease affecting global human health, and their pathogenesis and treatment have been the focus of medical research. With the in - depth study of microbiomics, the role of the microbiome in urological tumors has gradually attracted attention. However, the current research on tumor - associated microorganisms mostly focuses on one type or one site, and currently, there is a lack of attention to the microbiome in the immunity and immunotherapy of urological tumors. Therefore, in this paper, we systematically review the distribution characteristics of the microbiome (including microorganisms in the gut, urine, and tumor tissues) in urologic tumors, the relationship with disease prognosis, and the potential mechanisms of microbial roles in immunotherapy. In particular, we focus on the molecular mechanisms by which the microbiome at different sites influences tumor immunity through multiple "messengers" and pathways. We aim to further deepen the understanding of microbiome mechanisms in urologic tumors, and also point out the direction for the future development of immunotherapy for urologic tumors.
Collapse
Affiliation(s)
- Jun Zou
- Department of Otorhinolaryngology, The Affiliated Fengcheng Hospital of Yichun University, Fengcheng, Jiangxi, China
| | - Baisheng Xu
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Hongbing Gao
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huanglin Duan
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| |
Collapse
|
207
|
Wang Y, Shi Q, Zhang M, Xu L, Wei Q, Zhang R, Sun A, Lu Y, Zhang Z, Shi X. Combined ecotoxicity of polystyrene microplastics and Di-(2-ethylhexyl) phthalate increase exposure risks to Mytilus coruscus based on the bioaccumulation, oxidative stress, metabolic profiles, and nutritional interferences. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136381. [PMID: 39500187 DOI: 10.1016/j.jhazmat.2024.136381] [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: 07/10/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 12/01/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and microplastics (MPs) are emerging contaminants frequently detected in the marine environment. However, the influence of MPs on DEHP bioaccumulation and their combined effects on eco-environmental risks remain underexplored. Mytilus coruscus (M. coruscus) were exposed to DEHP (200.0 µg/L), polystyrene (PS) (0.050, 0.50, and 5.0 mg/L), and their combination at environmentally relevant concentrations for 15-day, followed by a 7-day depuration period. The amount of DEHP accumulation followed the order of digestive gland > gills > muscles > gonad, with PS dose-dependently amplifying DEHP bioaccumulation in digestive gland. The changes in antioxidant enzyme activity indicated disruptions in oxidative defense. Furthermore, metabolomic analysis revealed that PS and DEHP considerably altered the lipid, energy, and citric acid cycles in digestive gland and gonad. Post-depuration analysis showed combined exposure resulted in persistent effects. Compared with single exposures, combined exposure had a greater adverse effect on the metabolism of essential amino acids, fatty acids, and volatile compounds, potentially influencing edibility and nutritional value of M. coruscus. This study underscores cumulative eco-environmental toxicity of PS and DEHP toward M. coruscus and highlights the potential increased risks of co-pollution.
Collapse
Affiliation(s)
- Yinan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Qiangqiang Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Menglan Zhang
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Lingyan Xu
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Qiang Wei
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, PR China
| | - Rongrong Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Aili Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Yin Lu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, PR China
| | - Zeming Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China.
| | - Xizhi Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
208
|
Mir R, Albarqi SA, Albalawi W, Alatwi HE, Alatawy M, Bedaiwi RI, Almotairi R, Husain E, Zubair M, Alanazi G, Alsubaie SS, Alghabban RI, Alfifi KA, Bashir S. Emerging Role of Gut Microbiota in Breast Cancer Development and Its Implications in Treatment. Metabolites 2024; 14:683. [PMID: 39728464 DOI: 10.3390/metabo14120683] [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: 10/15/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background: The human digestive system contains approximately 100 trillion bacteria. The gut microbiota is an emerging field of research that is associated with specific biological processes in many diseases, including cardiovascular disease, obesity, diabetes, brain disease, rheumatoid arthritis, and cancer. Emerging evidence indicates that the gut microbiota affects the response to anticancer therapies by modulating the host immune system. Recent studies have explained a high correlation between the gut microbiota and breast cancer: dysbiosis in breast cancer may regulate the systemic inflammatory response, hormone metabolism, immune response, and the tumor microenvironment. Some of the gut bacteria are related to estrogen metabolism, which may increase or decrease the risk of breast cancer by changing the number of hormones. Further, the gut microbiota has been seen to modulate the immune system in respect of its ability to protect against and treat cancers, with a specific focus on hormone receptor-positive breast cancer. Probiotics and other therapies claiming to control the gut microbiome by bacterial means might be useful in the prevention, or even in the treatment, of breast cancer. Conclusions: The present review underlines the various aspects of gut microbiota in breast cancer risk and its clinical application, warranting research on individualized microbiome-modulated therapeutic approaches to breast cancer treatment.
Collapse
Affiliation(s)
- Rashid Mir
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, Prince Fahd Bin Sultan Research Chair for Biomedical Research, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Shrooq A Albarqi
- Molecular Medicine, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Wed Albalawi
- Molecular Medicine, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Hanan E Alatwi
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Marfat Alatawy
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Ruqaiah I Bedaiwi
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, Prince Fahd Bin Sultan Research Chair for Biomedical Research, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Reema Almotairi
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, Prince Fahd Bin Sultan Research Chair for Biomedical Research, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Eram Husain
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, Prince Fahd Bin Sultan Research Chair for Biomedical Research, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Mohammad Zubair
- Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Ghaida Alanazi
- Molecular Medicine, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Shouq S Alsubaie
- Molecular Medicine, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Razan I Alghabban
- Molecular Medicine, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Khalid A Alfifi
- Department of Laboratory and Blood Bank, King Fahd Special Hospital, Tabuk 47717, Saudi Arabia
| | - Shabnam Bashir
- Mubarak Hospital, Srinagar 190002, Jammu and Kashmir, India
| |
Collapse
|
209
|
Nazir A, Hussain FHN, Raza A. Advancing microbiota therapeutics: the role of synthetic biology in engineering microbial communities for precision medicine. Front Bioeng Biotechnol 2024; 12:1511149. [PMID: 39698189 PMCID: PMC11652149 DOI: 10.3389/fbioe.2024.1511149] [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: 10/14/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Over recent years, studies on microbiota research and synthetic biology have explored novel approaches microbial manipulation for therapeutic purposes. However, fragmented information is available on this aspect with key insights scattered across various disciplines such as molecular biology, genetics, bioengineering, and medicine. This review aims to the transformative potential of synthetic biology in advancing microbiome research and therapies, with significant implications for healthcare, agriculture, and environmental sustainability. By merging computer science, engineering, and biology, synthetic biology allows for precise design and modification of biological systems via cutting edge technologies like CRISPR/Cas9 gene editing, metabolic engineering, and synthetic oligonucleotide synthesis, thus paving the way for targeted treatments such as personalized probiotics and engineered microorganisms. The review will also highlight the vital role of gut microbiota in disorders caused by its dysbiosis and suggesting microbiota-based therapies and innovations such as biosensors for real-time gut health monitoring, non-invasive diagnostic tools, and automated bio foundries for better outcomes. Moreover, challenges including genetic stability, environmental safety, and robust regulatory frameworks will be discussed to understand the importance of ongoing research to ensure safe and effective microbiome interventions.
Collapse
Affiliation(s)
- Asiya Nazir
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | | | | |
Collapse
|
210
|
Kim N, Yang C. Butyrate as a Potential Modulator in Gynecological Disease Progression. Nutrients 2024; 16:4196. [PMID: 39683590 DOI: 10.3390/nu16234196] [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: 11/15/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
This review investigates the therapeutic potential of butyrate, a short-chain fatty acid (SCFA) produced by gut microbiota, in the prevention and treatment of various gynecological diseases, including polycystic ovary syndrome (PCOS), endometriosis, and gynecologic cancers like cervical and ovarian cancer. These conditions often pose treatment challenges, with conventional therapies offering limited and temporary relief, significant side effects, and a risk of recurrence. Emerging evidence highlights butyrate's unique biological activities, particularly its role as a histone deacetylase (HDAC) inhibitor, which allows it to modulate gene expression, immune responses, and inflammation. In PCOS, butyrate aids in restoring hormonal balance, enhancing insulin sensitivity, and reducing chronic inflammation. For endometriosis, butyrate appears to suppress immune dysregulation and minimize lesion proliferation. Additionally, in cervical and ovarian cancers, butyrate demonstrates anticancer effects through mechanisms such as cell cycle arrest, apoptosis induction, and suppression of tumor progression. Dietary interventions, particularly high-fiber and Mediterranean diets, that increase butyrate production are proposed as complementary approaches, supporting natural microbiota modulation to enhance therapeutic outcomes. However, butyrate's short half-life limits its clinical application, spurring interest in butyrate analogs and probiotics to maintain stable levels and extend its benefits. This review consolidates current findings on butyrate's multifaceted impact across gynecological health, highlighting the potential for microbiota-centered therapies in advancing treatment strategies and improving women's reproductive health.
Collapse
Affiliation(s)
- Nayeon Kim
- Department of Science Education, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Changwon Yang
- Department of Science Education, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
211
|
Lei P, Li X, Jiang L, Yu H, Zhang P, Han L, Jiang M. Alisma plantago-aquatica polysaccharides ameliorate acetaminophen-induced acute liver injury by regulating hepatic metabolic profiles and modulating gut microbiota. Int J Biol Macromol 2024; 285:138345. [PMID: 39631232 DOI: 10.1016/j.ijbiomac.2024.138345] [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: 03/14/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Acetaminophen (APAP) has emerged as a predominant contributor to acute liver failure (ALF) in United States. Alismatis rhizoma, a commonly used traditional herbal medicine, contains small molecular components with extensive hepatoprotective activity. However, the specific role of Alismatis rhizoma polysaccharide (ARP) in liver protection remains unclear. ARP50 and ARP70, derived through graded alcohol precipitation and refinement, predominantly consisted of varying proportions of glucose, galactose, and arabinose. In vitro experiments on free radical scavenging demonstrated notable antioxidant capabilities of ARP50 and ARP70. To investigate the hepatoprotective effects, an APAP-induced acute liver injury (ALI) model was established in mice. ARP50 and ARP70 exerted dose-dependent therapeutic effects on APAP-induced liver injury. Further analysis of liver metabolites revealed that ARPs facilitated the reconstruction of the liver antioxidant system by modulating the metabolism network centered on l-glutamine. In addition, the abundance of gut microbiota was altered under the influence of ARPs. ARP50 significantly reduced the levels of Pseudarthrobacter and markedly increased the levels of Faecalibacterium,At the same time, ARP50 could increase the levels of acetic acid in the liver and serum. Meanwhile, ARP70 significantly increased the abundance of Dubosiella, Muribaculum, Ileibacterium, and Prevotellaceae UCG 001, while reducing the abundance of Escherichia Shigella and Pseudarthrobacter. The results indicated that ARPs could exert a protective effect against APAP-induced acute liver injury by reshaping the liver metabolic profile and modulating the gut microbiota.
Collapse
Affiliation(s)
- Peng Lei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoge Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lei Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Heshui Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lifeng Han
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miaomiao Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
212
|
Zhang QY, Lai MQ, Chen YK, Zhong MT, Gi M, Wang Q, Xie XL. Inulin alleviates GenX-induced intestinal injury in mice by modulating the MAPK pathway, cell cycle, and cell adhesion proteins. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124974. [PMID: 39332800 DOI: 10.1016/j.envpol.2024.124974] [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: 07/03/2024] [Revised: 08/17/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024]
Abstract
GenX, a substitute for perfluorooctanoic acid, has demonstrated potential enterotoxicity. The enterotoxic effects of GenX and effective interventions need further investigation. In the present study, the mice were administered GenX (2 mg/kg/day) with or without inulin supplementation (5 g/kg/day) for 12 weeks. Histopathological assessments revealed that GenX induced colonic gland atrophy, inflammatory cell infiltration, a reduction in goblet cell numbers, and decreased mucus secretion. Furthermore, a significant decrease in the protein levels of ZO-1, occludin, and claudin-5 indicated compromised barrier integrity. Transcriptomic analysis identified 2645 DEGs, which were mapped to 39 significant pathways. The TGF-β, BMP6, and β-catenin proteins were upregulated in the intestinal mucosa following GenX exposure, indicating activation of the TGF-β pathway. Conversely, the protein expression of PAK3, CyclinD2, contactin1, and Jam2 decreased, indicating disruptions in cell cycle progression and cell adhesion. Inulin cotreatment ameliorated these GenX-induced alterations, partially through modulating the MAPK pathway, as evidenced by the upregulation of the cell cycle and cell adhesion proteins. Collectively, these findings suggested that GenX exposure triggered intestinal injury in mice by activating the TGF-β pathway and disrupting proteins crucial for the cell cycle and cell adhesion, whereas inulin supplementation mitigated this injury by modulating the MAPK pathway.
Collapse
Affiliation(s)
- Qin-Yao Zhang
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China
| | - Ming-Quan Lai
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China
| | - Yu-Kui Chen
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China
| | - Mei-Ting Zhong
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China
| | - Min Gi
- Department of Environmental Risk Assessment, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Qi Wang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, No. 1838 North Guangzhou Road, 510515, Guangzhou, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China.
| |
Collapse
|
213
|
Manokasemsan W, Jariyasopit N, Poungsombat P, Kaewnarin K, Wanichthanarak K, Kurilung A, Duangkumpha K, Limjiasahapong S, Pomyen Y, Chaiteerakij R, Tansawat R, Srisawat C, Sirivatanauksorn Y, Sirivatanauksorn V, Khoomrung S. Quantifying fecal and plasma short-chain fatty acids in healthy Thai individuals. Comput Struct Biotechnol J 2024; 23:2163-2172. [PMID: 38827233 PMCID: PMC11141283 DOI: 10.1016/j.csbj.2024.05.007] [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: 02/24/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are involved in important physiological processes such as gut health and immune response, and changes in SCFA levels can be indicative of disease. Despite the importance of SCFAs in human health and disease, reference values for fecal and plasma SCFA concentrations in healthy individuals are scarce. To address this gap in current knowledge, we developed a simple and reliable derivatization-free GC-TOFMS method for quantifying fecal and plasma SCFAs in healthy individuals. We targeted six linear- and seven branched-SCFAs, obtaining method recoveries of 73-88% and 83-134% in fecal and plasma matrices, respectively. The developed methods are simpler, faster, and more sensitive than previously published methods and are well suited for large-scale studies. Analysis of samples from 157 medically confirmed healthy individuals showed that the total SCFAs in the feces and plasma were 34.1 ± 15.3 µmol/g and 60.0 ± 45.9 µM, respectively. In fecal samples, acetic acid (Ace), propionic acid (Pro), and butanoic acid (But) were all significant, collectively accounting for 89% of the total SCFAs, whereas the only major SCFA in plasma samples was Ace, constituting of 93% of the total plasma SCFAs. There were no statistically significant differences in the total fecal and plasma SCFA concentrations between sexes or among age groups. The data revealed, however, a positive correlation for several nutrients, such as carbohydrate, fat, iron from vegetables, and water, to most of the targeted SCFAs. This is the first large-scale study to report SCFA reference intervals in the plasma and feces of healthy individuals, and thereby delivers valuable data for microbiome, metabolomics, and biomarker research.
Collapse
Affiliation(s)
- Weerawan Manokasemsan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Narumol Jariyasopit
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Patcha Poungsombat
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Khwanta Kaewnarin
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- SingHealth Duke-NUS Institute of Biodiversity Medicine, National Cancer Centre Singapore, Singapore
| | - Kwanjeera Wanichthanarak
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Alongkorn Kurilung
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kassaporn Duangkumpha
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Suphitcha Limjiasahapong
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Yotsawat Pomyen
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Roongruedee Chaiteerakij
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Division of Gastroenterology, Department of Medicine, Faculty of Medicine Chulalongkorn University, Chulalongkorn University, Bangkok, Thailand
| | - Rossarin Tansawat
- Thailand Metabolomics Society, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Chulalongkorn University, Bangkok, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Yongyut Sirivatanauksorn
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vorapan Sirivatanauksorn
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
| | - Sakda Khoomrung
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellent in Metabolomics and Systems Biology (SiCORE-MSB), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Thailand Metabolomics Society, Bangkok, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
214
|
Wang Q, Li G, Qin W, Cai J, Wang N. Evaluation of in vitro simulated digestion and fermentation characteristics of the crude exopolysaccharide from Levilactobacillus brevis M-14. J Food Sci 2024; 89:9860-9878. [PMID: 39475350 DOI: 10.1111/1750-3841.17467] [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: 07/04/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 12/28/2024]
Abstract
The present study employed an in vitro static digestion model to simulate the saliva-gastrointestinal digestion and fecal fermentation of exopolysaccharides (EPSs) extracted from Levilactobacillus brevis M-14, aiming to understand the dynamic changes in physicochemical properties, biological potential of EPS, as well as their impact on the human enteric microorganism. EPS was mainly composed of polysaccharides. The molecular weight (Mw) showed that EPS comprised two parts with a high Mw and a low Mw fraction of 42.81 × 104 and 1.23 × 104 Da, respectively. EPSs mainly consisted of mannose, rhamnose, galacturonic acid, glucose, and galactose in a molar ratio of 0.42∶0.13∶0.21∶0.13∶0.11. In the simulated digestion process, EPS was relatively stable. Furthermore, simulated digestion increased the antioxidant and hypoglycemic capacities of EPS. During the fermentation stage, the total carbohydrate contents of EPS decreased by 20.19%. The Mw of the two components of EPS decreased by 16.37% and 61.67%, respectively, and accompanied by the production of free monosaccharides. EPS had the potential to modulate the composition of gut microbiota, increasing the relative abundance of Enterococcus and Parabacteroides, while decreasing the relative abundance of Bacteroides. The pH decreased and total short-chain fatty acids contents increased, especially acetic acid and propionic acid. This research provided valuable insights into the potential application of EPS as a prebiotic agent.
Collapse
Affiliation(s)
- Qi Wang
- School of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Gen Li
- School of Life Science, Shanxi University, Taiyuan, Shanxi, China
| | - Wenjun Qin
- Nutritional Department, Shanxi Traditional Chinese Medical Hospital, Taiyuan, Shanxi, China
| | - Jin Cai
- Institute of Applied Chemistry, Shanxi University, Taiyuan, Shanxi, China
| | - Nifei Wang
- Institute of Biotechnology, Shanxi University, Taiyuan, Shanxi, China
| |
Collapse
|
215
|
Zhao C, Chen F, Li Q, Zhang W, Peng L, Yue C. Causal relationship between oral microbiota and epilepsy risk: Evidence from Mendelian randomization analysis in East Asians. Epilepsia Open 2024; 9:2419-2428. [PMID: 39382490 PMCID: PMC11633697 DOI: 10.1002/epi4.13074] [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: 08/19/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVE Gut microbiota can traverse into the brain, activate the vagus nerve, and modulate immune responses and inflammatory processes, thereby influencing the onset of epileptic seizures. However, research on oral microbiota and epilepsy remains limited, and observational studies have been inconsistent. We aim to estimate the potential links between oral microbiota and epilepsy and elucidate which specific oral microbes may directly influence the pathogenesis of epilepsy. METHODS A two-sample MR analysis was conducted using genome-wide association study (GWAS) data specific to OM and epilepsy in East Asian individuals. Single nucleotide polymorphisms (SNPs) independent of confounders served as instrumental variables (IVs) to deduce causality. MR methodologies, including inverse variance weighted (IVW), MR-Egger, weighted median, and weighed mode methods, were utilized. Sensitivity analysis, including Cochrane's Q test, MR-Egger intercept test, and leave-one-out analysis, was applied to confirm the robustness of results. RESULTS Among the 3117 bacterial taxa examined, we observed that 14 OM, like s_Streptococcus_mitis, s_Streptococcus_pneumoniae, and s_Haemophilus, were positively associated with epilepsy, while 7 OM, like g_Fusobacterium and g_Aggregatibacter, were negatively related to epilepsy. The MR-Egger intercept suggested that no evidence of horizontal pleiotropy was observed (p > 0.05). The leave-one-out analysis validated the robustness of the results. SIGNIFICANCE This study underscores the effect of OM on epilepsy, suggesting potential mechanisms between the OM and epilepsy. Further investigation into the potential role of the OM is needed to enhance our in-depth understanding of the pathogenesis of epilepsy. PLAIN LANGUAGE SUMMARY Previous research has demonstrated that the microbiota may influence the onset of epileptic seizures. We applied 3117 oral microbiota from the newest publicly available database of East Asian populations. Mendelian randomization analysis was utilized to estimate the causal relationship between oral microbiota and epilepsy. Our results showed that a causal effect exists between 21 oral microbiota and epilepsy. We provided genetic evidence for risk assessment and early intervention in epilepsy.
Collapse
Affiliation(s)
- Chenyang Zhao
- The First People's Hospital of ChenzhouChenzhouChina
- The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Fei Chen
- Huadong HospitalFudan UniversityShanghaiChina
| | - Qiong Li
- The First People's Hospital of ChenzhouChenzhouChina
- The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Wei Zhang
- The First People's Hospital of ChenzhouChenzhouChina
| | - Lixiu Peng
- The First People's Hospital of ChenzhouChenzhouChina
- The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Chaoyan Yue
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
216
|
Ban SY, Yun DY, Yum SJ, Jeong HG, Park JT. Development of Saccharomyces cerevisiae accumulating excessive amount of glycogen and its effects on gut microbiota in a mouse model. Int J Biol Macromol 2024; 283:137589. [PMID: 39557260 DOI: 10.1016/j.ijbiomac.2024.137589] [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: 05/20/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Saccharomyces cerevisiae accumulates glycogen, a hyperbranched glucose polymer with multiple bio-functionalities. In this study, mutants of S. cerevisiae that accumulate excessive amounts of glycogen were developed through UV mutagenesis. From over 30,000 mutants, the mutant strain CEY1, which exhibited the highest glycogen production, was selected using iodine vapor screening. The glycogen structures of wild type (WT) and CEY1 were analyzed and found to be relatively similar in molecular weight, hydrodynamic diameter, and side-chain distribution. The glycogen from CEY1 contained long branches (DP >12) 23.6 % greater than those in Escherichia coli TBP38. In addition, WT and CEY1 glycogen showed 32 %-34 % digestibility, which is significantly lower than E. coli glycogen. The glycogen content in dried CEY1 cells was increased to 21.7 % during laboratory-scale fed-batch fermentation. Glycogen with a homogeneous structure was accumulated to 17.5 % (w/w dried cell), and the total glucan content was increased by 33.2 % during large-scale fed-batch fermentation. In a mouse model, a diet containing 30 % CEY1 increased the production of butyrate and populations of beneficial bacteria, including Bacteroides and Parabacteroides. Therefore, glycogen from CEY1 exhibits a distinct structure from other polysaccharides, with notably slow and low digestibility, thereby indicating its potential application as a dietary supplement.
Collapse
Affiliation(s)
- So Young Ban
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea
| | - Da-Young Yun
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Su-Jin Yum
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hee-Gon Jeong
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Jong-Tae Park
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea.
| |
Collapse
|
217
|
Jang AR, Jung DH, Lee TS, Kim JK, Lee YB, Lee JY, Kim SY, Yoo YC, Ahn JH, Hong EH, Kim CW, Kim SM, Yoo HH, Huh JY, Ko HJ, Park JH. Lactobacillus plantarum NCHBL-004 modulates high-fat diet-induced weight gain and enhances GLP-1 production for blood glucose regulation. Nutrition 2024; 128:112565. [PMID: 39326237 DOI: 10.1016/j.nut.2024.112565] [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: 05/01/2024] [Revised: 06/27/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVES This study investigated the therapeutic potential of Lactobacillus plantarum NCHBL-004 (NCHBL-004) in the treatment of obesity and associated metabolic disorders. METHODS Mice were fed either a normal diet (ND) or a high-fat diet (HFD) with oral administration of NCHBL-004. After euthanasia, blood, liver and adipose tissue were collected. Furthermore, the microbiome and short-chain fatty acids (SCFAs) were analyzed from feces. RESULTS Oral administration of live NCHBL-004 to mice fed a HFD resulted in notable reductions in weight gain, improvements in glucose metabolism, and maintenance of balanced lipid levels. A comparative analysis with other Lactobacillus strains highlighted the superior efficacy of NCHBL-004. Moreover, heat-killed NCHBL-004 demonstrated beneficial effects similar to those of live NCHBL-004. Additionally, administration of live NCHBL-004 induced glucagon-like peptide 1 (GLP-1) production and increased the levels of short-chain fatty acids (SCFAs), including acetate and propionate, in feces, positively influencing liver lipid metabolism and mitigating inflammation. Consistent with this, analysis of the gut microbiome following NCHBL-004 administration showed increases in SCFA-producing microbes with increased proportions of Lactobacillus spp. and a significant increase in the proportion of microbes capable of promoting GLP-1 secretion. CONCLUSIONS These findings underscore the potential of both live and inactivated NCHBL-004 as potential therapeutic approaches to managing obesity and metabolic disorders, suggesting avenues for further investigation and clinical applications.
Collapse
Affiliation(s)
- Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; Nodcure, INC., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Do-Hyeon Jung
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jeon-Kyung Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Yu-Bin Lee
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae-Young Lee
- Nodcure, INC., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - So-Yeon Kim
- Department of Microbiology, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Yung-Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; KNU Researcher training program for Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon 24341, Republic of Korea; Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Eun-Hye Hong
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; KNU Researcher training program for Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon 24341, Republic of Korea; Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Chae-Won Kim
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; KNU Researcher training program for Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon 24341, Republic of Korea; Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Su Min Kim
- Pharmacomicrobiomics Research Center, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hye Hyun Yoo
- Pharmacomicrobiomics Research Center, College of Pharmacy, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea; KNU Researcher training program for Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon 24341, Republic of Korea; Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine and Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; Nodcure, INC., 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea.
| |
Collapse
|
218
|
Ke JP, Li JY, Yang Z, Wu HY, Yu JY, Yang Y, Chen CH, Zhou P, Hua F, Wang W, Hu F, Chu GX, Wan XC, Bao GH. Unraveling anti-aging mystery of green tea in C. elegans: Chemical truth and multiple mechanisms. Food Chem 2024; 460:140510. [PMID: 39033639 DOI: 10.1016/j.foodchem.2024.140510] [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: 03/17/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Tea drinking impacts aging and aging-related diseases. However, knowledge of anti-aging molecules other than the major catechins in complex tea extracts remains limited. Here we used Caenorhabditis elegans to analyze the longevity effects of tea extracts and constituents comprehensively. We found that the hot water extract of green tea prolonged lifespan and heathspan. Further, the MeOH fraction prolonged lifespan significantly longer than other fractions. Correlation analysis between mass spectroscopic data and anti-aging activity suggests that ester-type catechins (ETCs) are the major anti-aging components, including 4 common ETCs, 6 phenylpropanoid-substituted ester-type catechins (PSECs), 5 cinnamoylated catechins (CCs), 7 ester-type flavoalkaloids (ETFs), and 4 cinnamoylated flavoalkaloids (CFs). CFs (200 μM) are the strongest anti-aging ETCs (with the longest 73% lifespan extension). Green tea hot water extracts and ETCs improved healthspan by enhancing stress resistance and reducing ROS accumulation. The mechanistic study suggests that they work by multiple pathways. Moreover, ETCs modulated gut microbial homeostasis, increased the content of short-chain fatty acids, and reduced fat content. Altogether, our study provides new evidence for the anti-aging benefits of green tea and insights into a deep understanding of the chemical truth and multi-target mechanism.
Collapse
Affiliation(s)
- Jia-Ping Ke
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Jia-Yi Li
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Zi Yang
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Hao-Yue Wu
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Jing-Ya Yu
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Yi Yang
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Chen-Hui Chen
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China
| | - Peng Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Fang Hua
- School of Pharmacy, Anhui Xinhua University, Hefei, Anhui, People's Republic of China
| | - Wei Wang
- Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, West Anhui University, Lu'an 237012, China
| | - Fenglin Hu
- Engineering Research Center of Fungal Biotechnology, Ministry of Education, Anhui Agricultural University, Hefei, 230036, China.
| | - Gang-Xiu Chu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, 230036, China.
| | - Xiao-Chun Wan
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China.
| | - Guan-Hu Bao
- Natural Products Laboratory, International Joint Laboratory of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, People's Republic of China; Joint Research Center for Food Nutrition and Health of IHM, Hefei, China.
| |
Collapse
|
219
|
Catussi BLC, Lo Turco EG, Pereira DM, Teixeira RMN, Castro BP, Massaia IFD. Metabolomics: Unveiling biological matrices in precision nutrition and health. Clin Nutr ESPEN 2024; 64:314-323. [PMID: 39427750 DOI: 10.1016/j.clnesp.2024.10.148] [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: 03/11/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Precision nutrition, an expanding field at the intersection of nutrition science and personalized medicine, is rapidly evolving with metabolomics integration. Metabolomics, facilitated by advanced technologies like mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy, facilitates comprehensive profiling of metabolites across diverse biological samples. From the perspective of health care systems, precision nutrition gains relevance due to the substantial impact of prevalent non-communicable diseases (NCDs) on societal well-being, which is directly linked with dietary habits and eating behavior. Furthermore, biomarker products derived from metabolomics have been utilized in Europe, the USA, and Brazil to understand metabolic dysregulations and tailor diets accordingly. Despite its burgeoning status, metabolomics holds great potential in revolutionizing nutritional science, particularly with the integration of artificial intelligence and machine learning, offering novel insights into personalized dietary interventions and disease prediction. This narrative review emphasizes the transformative impact of metabolomics in precision and delineates avenues for future research and application, paving the way for a more tailored and practical approach to nutrition management.
Collapse
|
220
|
Gopal RK, Ganesh PS, Pathoor NN. Synergistic Interplay of Diet, Gut Microbiota, and Insulin Resistance: Unraveling the Molecular Nexus. Mol Nutr Food Res 2024; 68:e2400677. [PMID: 39548908 DOI: 10.1002/mnfr.202400677] [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: 08/21/2024] [Revised: 10/04/2024] [Indexed: 11/18/2024]
Abstract
This comprehensive review explores the intricate relationship between gut microbiota, diet, and insulin resistance, emphasizing the novel roles of diet-induced microbial changes in influencing metabolic health. It highlights how diet significantly influences gut microbiota composition, with different dietary patterns fostering diverse microbial communities. These diet-induced changes in the microbiome impact human metabolism by affecting inflammation, energy balance, and insulin sensitivity, particularly through microbial metabolites like short-chain fatty acids (SCFAs). Focusing the key mediators like endotoxemia and systemic inflammation, and introduces personalized microbiome-based therapeutic strategies, it also investigates the effects of dietary components-fiber, polyphenols, and lipids-on microbiota and insulin sensitivity, along with the roles of protein intake and amino acid metabolism. The study compares the effects of Western and Mediterranean diets on the microbiota-insulin resistance axis. Therapeutic implications, including probiotics, fecal microbiota transplantation (FMT), and personalized diets, are discussed. Key findings reveal that high-fat diets, especially those rich in saturated fats, contribute to dysbiosis and increased intestinal permeability, while high-fiber diets promote beneficial bacteria and SCFAs. The review underscores the future potential of food and microbiota interventions for preventing or managing insulin resistance.
Collapse
Affiliation(s)
- Rajesh Kanna Gopal
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Naji Naseef Pathoor
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| |
Collapse
|
221
|
Cheng X, Meng X, Chen R, Song Z, Li S, Wei S, Lv H, Zhang S, Tang H, Jiang Y, Zhang R. The molecular subtypes of autoimmune diseases. Comput Struct Biotechnol J 2024; 23:1348-1363. [PMID: 38596313 PMCID: PMC11001648 DOI: 10.1016/j.csbj.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by their complexity and a wide range of clinical differences. Despite patients presenting with similar symptoms and disease patterns, their reactions to treatments may vary. The current approach of personalized medicine, which relies on molecular data, is seen as an effective method to address the variability in these diseases. This review examined the pathologic classification of ADs, such as multiple sclerosis and lupus nephritis, over time. Acknowledging the limitations inherent in pathologic classification, the focus shifted to molecular classification to achieve a deeper insight into disease heterogeneity. The study outlined the established methods and findings from the molecular classification of ADs, categorizing systemic lupus erythematosus (SLE) into four subtypes, inflammatory bowel disease (IBD) into two, rheumatoid arthritis (RA) into three, and multiple sclerosis (MS) into a single subtype. It was observed that the high inflammation subtype of IBD, the RA inflammation subtype, and the MS "inflammation & EGF" subtype share similarities. These subtypes all display a consistent pattern of inflammation that is primarily driven by the activation of the JAK-STAT pathway, with the effective drugs being those that target this signaling pathway. Additionally, by identifying markers that are uniquely associated with the various subtypes within the same disease, the study was able to describe the differences between subtypes in detail. The findings are expected to contribute to the development of personalized treatment plans for patients and establish a strong basis for tailored approaches to treating autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Zerun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuai Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuhao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
222
|
Raghani N, Postwala H, Shah Y, Chorawala M, Parekh P. From Gut to Brain: Unraveling the Intricate Link Between Microbiome and Stroke. Probiotics Antimicrob Proteins 2024; 16:2039-2053. [PMID: 38831225 DOI: 10.1007/s12602-024-10295-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
Stroke, a neurological disorder, is intricately linked to the gut microbiota, influencing microbial composition and elevating the risk of ischemic stroke. The neuroprotective impact of short-chain fatty acids (SCFAs) derived from dietary fiber fermentation contrasts with the neuroinflammatory effects of lipopolysaccharide (LPS) from gut bacteria. The pivotal role of the gut-brain axis, facilitating bidirectional communication between the gut and the brain, is crucial in maintaining gastrointestinal equilibrium and influencing cognitive functions. An in-depth understanding of the interplay among the gut microbiota, immune system, and neurological outcomes in stroke is imperative for devising innovative preventive and therapeutic approaches. Strategies such as dietary adjustments, probiotics, prebiotics, antibiotics, or fecal transplantation offer promise in modulating stroke outcomes. Nevertheless, comprehensive research is essential to unravel the precise mechanisms governing the gut microbiota's involvement in stroke and to establish effective therapeutic interventions. The initiation of large-scale clinical trials is warranted to assess the safety and efficacy of interventions targeting the gut microbiota in stroke management. Tailored strategies that reinstate eubiosis and foster a healthy gut microbiota hold potential for both stroke prevention and treatment. This review underscores the gut microbiota as a promising therapeutic target in stroke and underscores the need for continued research to delineate its precise role and develop microbiome-based interventions effectively.
Collapse
Affiliation(s)
- Neha Raghani
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Humzah Postwala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Yesha Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Mehul Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India.
| | - Priyajeet Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| |
Collapse
|
223
|
Chin KW, Khoo SC, Paul RPM, Luang-In V, Lam SD, Ma NL. Potential of Synbiotics and Probiotics as Chemopreventive Agent. Probiotics Antimicrob Proteins 2024; 16:2085-2101. [PMID: 38896220 DOI: 10.1007/s12602-024-10299-z] [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] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
Cancer remains a global problem, with millions of new cases diagnosed yearly and countless lives lost. The financial burden of cancer therapy, along with worries about the long-term safety of existing medicines, necessitates the investigation of alternative approaches to cancer prevention. Probiotics generate chemopreventive compounds such as bacteriocins, short-chain fatty acids (SCFA), and extracellular polymeric substances (EPS), which have demonstrated the ability to impede cancer cell proliferation, induce apoptosis, and bolster the expression of pro-apoptotic genes. On the other hand, prebiotics, classified as non-digestible food ingredients, promote the proliferation of probiotics within the colon, thereby ensuring sustained functionality of the gut microbiota. Consequently, the synergistic effect of combining prebiotics with probiotics, known as the synbiotic effect, in dietary interventions holds promise for potentially mitigating cancer risk and augmenting preventive measures. The utilization of gut microbiota in cancer treatment has shown promise in alleviating adverse health effects. This review explored the potential and the role of probiotics and synbiotics in enhancing health and contributing to cancer prevention efforts. In this review, the applications of functional probiotics and synbiotics, the mechanisms of action of probiotics in cancer, and the relationship of probiotics with various drugs were discussed, shedding light on the potential of probiotics and synbiotics to alleviate the burdens of cancer treatment.
Collapse
Affiliation(s)
- Kah Wei Chin
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Shing Ching Khoo
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Richard Paul Merisha Paul
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia
| | - Vijitra Luang-In
- Natural Antioxidant Innovation Research Unit, Department of Biotechnology, Faculty of Technology, Mahasarakham University, Khamriang, 44150, Kantarawichai, Maha Sarakham, Thailand
| | - Su Datt Lam
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Selangor, Malaysia
| | - Nyuk Ling Ma
- Bioses Research Interest Group (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus, 21030, Terengganu, Malaysia.
- Department of Sustainable Engineering, Institute of Biotechnology, Saveetha School of Engineering, SIMATS, Chennai, 602105, India.
| |
Collapse
|
224
|
Catassi G, Mateo SG, Occhionero AS, Esposito C, Giorgio V, Aloi M, Gasbarrini A, Cammarota G, Ianiro G. The importance of gut microbiome in the perinatal period. Eur J Pediatr 2024; 183:5085-5101. [PMID: 39358615 PMCID: PMC11527957 DOI: 10.1007/s00431-024-05795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
This narrative review describes the settlement of the neonatal microbiome during the perinatal period and its importance on human health in the long term. Delivery methods, maternal diet, antibiotic exposure, feeding practices, and early infant contact significantly shape microbial colonization, influencing the infant's immune system, metabolism, and neurodevelopment. By summarizing two decades of research, this review highlights the microbiome's role in disease predisposition and explores interventions like maternal vaginal seeding and probiotic and prebiotic supplementation that may influence microbiome development. CONCLUSION The perinatal period is a pivotal phase for the formation and growth of the neonatal microbiome, profoundly impacting long-term health outcomes. WHAT IS KNOWN • The perinatal period is a critical phase for the development of the neonatal microbiome, with factors such as mode of delivery, maternal diet, antibiotic exposure, and feeding practices influencing its composition and diversity, which has significant implications for long-term health. • The neonatal microbiome plays a vital role in shaping the immune system, metabolism, and neurodevelopment of infants. WHAT IS NEW • Recent studies have highlighted the potential of targeted interventions, such as probiotic and prebiotic supplementation, and innovative practices like maternal vaginal seeding, to optimize microbiome development during the perinatal period. • Emerging evidence suggests that specific bacterial genera and species within the neonatal microbiome are associated with reduced risks of developing chronic conditions, indicating new avenues for promoting long-term health starting from early life.
Collapse
Affiliation(s)
- Giulia Catassi
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Pediatric Gastroenterology and Liver Unit, Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Sandra Garcia Mateo
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Gastroenterology, Lozano Blesa University Hospital, 50009, Zaragossa, Spain
| | - Annamaria Sara Occhionero
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Chiara Esposito
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Giorgio
- Department of Woman and Child Health and Public Health, UOC Pediatria, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato DigerenteMedicina Interna E Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
225
|
Thakur P, Baraskar K, Shrivastava VK, Medhi B. Cross-talk between adipose tissue and microbiota-gut-brain-axis in brain development and neurological disorder. Brain Res 2024; 1844:149176. [PMID: 39182900 DOI: 10.1016/j.brainres.2024.149176] [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: 04/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
Collapse
Affiliation(s)
- Pratibha Thakur
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India.
| | - Kirti Baraskar
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Vinoy K Shrivastava
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab 160012, India.
| |
Collapse
|
226
|
Zheng M, Chao X, Zheng Y, Hong T, Wu W, Zhu Y, Ni H, Jiang Z. A polysaccharide from edible red seaweed Bangia fusco-purpurea prevents obesity in high-fat diet-induced C57BL/6 mice. Int J Biol Macromol 2024; 283:137545. [PMID: 39542298 DOI: 10.1016/j.ijbiomac.2024.137545] [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: 08/05/2024] [Revised: 10/26/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
The study aimed to investigate the impacts of a polysaccharide (BFP) from Bangia fusco-purpurea on high-fat diet (HFD)-induced obesity in C57BL/6 mice, as well as its underlying mechanisms. Our results showed that orally administrated BFP was more effective than inulin (INU) in reducing body weight and fat accumulation in obese mice, indicating its anti-obesity effect. BFP effectively improved the compositions and metabolites of intestinal microbiota in obese mice, leading to enhanced energy metabolism and lipid metabolism, thus contributing to its anti-obesity effect. Notably, the better anti-obesity effect of BFP compared to INU was attributed to their varying degrees of modulation of specific intestinal microbial taxa, such as Clostridium and Aerococcus, as well as the regulation of differential metabolites (including biotin, piperine, G6P, etc.) also varied. Also, both in vitro (3T3-L1 preadipocytes) and in vivo (HFD-induced obese mice) models confirmed that BFP achieved anti-obesity effect attributed to enhance energy metabolism, promote lipolysis, increase fatty acid oxidation, and inhibit adipogenesis via activating the AMP-activated protein kinase and Acetyl-CoA carboxylase signaling pathways and suppressing the peroxisome proliferator-activated receptor γ expression. Our findings suggest that BFP has the potential to be used as prebiotics, dietary agents, and nutritional supplements against obesity.
Collapse
Affiliation(s)
- Mingjing Zheng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Xiaoling Chao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yajun Zheng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Tao Hong
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Weijing Wu
- Laboratory of nutrition and food safety, Xiamen Medical College, Xiamen, Fujian 361023, China.
| | - Yanbing Zhu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China
| | - Hui Ni
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China; Xiamen Ocean Vocational College, Xiamen, Fujian 361102, China
| | - Zedong Jiang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China; Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, Xiamen, Fujian 361021, China.
| |
Collapse
|
227
|
Yilmaz Y. Postbiotics as Antiinflammatory and Immune-Modulating Bioactive Compounds in Metabolic Dysfunction-Associated Steatotic Liver Disease. Mol Nutr Food Res 2024; 68:e2400754. [PMID: 39499063 DOI: 10.1002/mnfr.202400754] [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: 09/16/2024] [Revised: 10/12/2024] [Indexed: 11/07/2024]
Abstract
Postbiotics, defined as products or metabolic byproducts secreted by live bacteria or released after bacterial lysis, are emerging as promising therapeutic agents for metabolic dysfunction-associated steatotic liver disease (MASLD). This review explores the antiinflammatory and immunomodulatory properties of various postbiotics, including exopolysaccharides, lipoteichoic acid, short-chain fatty acids, hydrogen sulfide, polyamines, tryptophan derivatives, and polyphenol metabolites. These compounds have demonstrated potential in mitigating steatotic liver infiltration, reducing inflammation, and slowing fibrosis progression in preclinical studies. Notably, postbiotics exert their beneficial effects by modulating gut microbiota composition, enhancing intestinal barrier function, optimizing lipid metabolism, reducing hepatic inflammation and steatosis, and exhibiting hepatoprotective properties. However, translating these findings into clinical practice requires well-designed trials to validate efficacy and safety, standardize production and characterization, and explore personalized approaches and synergistic effects with other therapeutic modalities. Despite challenges, the unique biological properties of postbiotics, such as enhanced safety compared to probiotics, make them attractive candidates for developing novel nutritional interventions targeting the multifactorial pathogenesis of MASLD. Further research is needed to establish their clinical utility and potential to improve liver and systemic outcomes in this increasingly prevalent condition.
Collapse
Affiliation(s)
- Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
- The Global NASH Council, Washington, DC, 53020, USA
| |
Collapse
|
228
|
Joshi DD, Deb L, Kaul K, Somkuwar BG, Rana VS, Singh R. Relevance of Indian Traditional Herbal Brews for Gut Microbiota Balance. Indian J Microbiol 2024; 64:1425-1444. [PMID: 39678955 PMCID: PMC11645388 DOI: 10.1007/s12088-024-01251-4] [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: 12/05/2023] [Accepted: 03/02/2024] [Indexed: 12/17/2024] Open
Abstract
The considerable changes in lifestyle patterns primarily affect the human gut microbiota and result in obesity, diabetes, dyslipidemia, renal complications, etc. though there are few traditional safeguards such as herbal brews to maintain the ecological stability under intestinal dysbiosis. The present article is designed to collect all the scientific facts in a place to decipher the role of the Indian traditional herbal brews used to balance gut health for centuries. Computerized databases, commercial search engines, research papers, articles, and books were used to search by using different keywords to select the most appropriate published articles from 2000 onward to September 2023. A total of 1907 articles were scrutinized, 46 articles were finally selected from the 254 screened, and targeted information was compiled. Interaction of herbal brews to the gut microflora and resulting metabolites act as prebiotics due to antimicrobial, anti-inflammatory, and antioxidant properties, and modulate the pH of the gut. The effect of brews on gut microbiota has a drastic impact on various gut-related diseases and has gained popularity as an alternative to antibiotics against bacteria, fungi, viruses, parasites, and boosting the immune system and strengthening the intestinal barrier. Berberine, kaempferol, piperine, and quercetin have been found in more than one brew discussed in the present article. Practically, these brews balance the gut microbiota, prevent chronic and degenerative diseases, and reduce organ inflammation, though, there is a knowledge gap on the molecular mechanism to explain their efficacy. Indian traditional herbal brews used to reboot and heal the gut microbiota since centuries-old practice with successful history without toxicity. The systematic consumption of these brews under specific dietary prescriptions has a hope of arrays for a healthy human gut microbiome in the present hasty lifestyle with overall health and well-being. Graphical Abstract
Collapse
Affiliation(s)
- Devi Datt Joshi
- Amity Food and Agriculture Foundation, Amity University Uttar Pradesh, Noida, Sector-125, J-1 Block, Noida, UP 201313 India
| | - Lokesh Deb
- Institute of Bioresources and Sustainable Development (IBSD)-Regional Centre, Sikkim, 5th Mile, Tadong, Gangtok, Sikkim 737102 India
| | - Kanak Kaul
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Sector-125, E-2 Block, Noida, UP 201313 India
| | - Bharat G. Somkuwar
- Institute of Bioresources and Sustainable Development (IBSD), Node Mizoram, A-1, C/O P. Lalthangzauva Building, Chawnga Road, Nursery Veng, Aizawl, Mizoram 796005 India
| | - Virendra Singh Rana
- Division of Agricultural Chemicals, ICAR-Indian Agricultural Research Institute (IARI), Pusa Campus, New Delhi, 110 012 India
| | - Rajni Singh
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Sector-125, E-2 Block, Noida, UP 201313 India
| |
Collapse
|
229
|
Caprara GL, von Ameln Lovison O, Martins AF, Bernardi JR, Goldani MZ. Characterization of newborn gut microbiota according to the pre-gestational maternal nutritional status and delivery mode. Arch Gynecol Obstet 2024; 310:2889-2898. [PMID: 39387928 DOI: 10.1007/s00404-024-07772-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE The aim of this study is to characterize the composition of the newborn gut microbiota based on the maternal pre-pregnancy nutritional status and the delivery mode. METHODS A biological sample was collected from the anal mucosa of the newborns between 24 and 48 h after delivery, as it was not possible to collect a meconium sample at that time. A general data collection questionnaire was administered. The microbiome of the samples was analyzed by next-generation sequencing of the hypervariable regions v3-v4 of the 16S gene. Alpha diversity analyses were performed using the Observed Richness and Shannon diversity index metrics and Beta diversity analyses were conducted using Nonmetric multidimensional scaling with Weighted Unifrac, Differential abundance analysis was performed using a Negative Binomial Wald Test with maximum likelihood estimation for coefficients of Generalized Linear Models. RESULTS Newborns of obese mothers exhibited lower alpha diversity compared to newborns of mothers with adequate BMI (body mass index). We observed variation in the composition of the microbial community in newborn stool samples, both from mothers with overweight/obesity and those with adequate pre-pregnancy BMI. We observed a visible correlation between the mode of delivery and the newborn's microbiota. We found variation in the overall composition of the microbial community in the stools of newborns, regardless of the delivery mode. CONCLUSIONS The results of our study demonstrate differences in the microbiota of neonates born via cesarean section compared to those born vaginally as well as differences in newborns of mothers with overweight/obesity compared to those with an adequate pre-pregnancy BMI.
Collapse
Affiliation(s)
- Gabriele Luiza Caprara
- Programa de Pós-Graduação Em Saúde da Criança E Do Adolescente, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| | - Otávio von Ameln Lovison
- Laboratório de Pesquisa Em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Andreza Francisco Martins
- Laboratório de Pesquisa Em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Juliana Rombaldi Bernardi
- Programa de Pós-Graduação Em Saúde da Criança E Do Adolescente, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Alimentação, Nutrição E Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Marcelo Zubaran Goldani
- Programa de Pós-Graduação Em Saúde da Criança E Do Adolescente, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
230
|
Wei J, Liu C, Qin D, Ren F, Duan J, Chen T, Wu A. Targeting inflammation and gut microbiota with antibacterial therapy: Implications for central nervous system health. Ageing Res Rev 2024; 102:102544. [PMID: 39419400 DOI: 10.1016/j.arr.2024.102544] [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: 09/02/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
The complex symbiotic relationship between inflammation, the gut microbiota, and the central nervous system (CNS) has become a pivotal focus of contemporary biomedical research. Inflammation, as a physiological defense mechanism, plays a dual role as both a protective and pathological factor, and is intricately associated with gut microbiota homeostasis, often termed the "second brain." The gutbrain axis (GBA) exemplifies this multifaceted interaction, where gut health exerts significantly regulatory effects on CNS functions. Antibacterial therapies represent both promising and challenging strategies for modulating inflammation and gut microbiota composition to confer CNS benefits. However, while such therapies may exert positive modulatory effects on the gut microbiota, they also carry the potential to disrupt microbial equilibrium, potentially exacerbating neurological dysfunction. Recent advances have provided critical insights into the therapeutic implications of antibacterial interventions; nevertheless, the application of these therapies in the context of CNS health warrants a judicious and evidence-based approach. As research progresses, deeper investigation into the microbial-neural interface is essential to fully realize the potential of therapies targeting inflammation and the gut microbiota for CNS health. Future efforts should focus on refining antibacterial interventions to modulate the gut microbiota while minimizing disruption to microbial balance, thereby reducing risks and enhancing efficacy in CNS-related conditions. In conclusion, despite challenges, a more comprehensive understanding of the GBA, along with precise modulation through targeted antibacterial therapies, offers significant promise for advancing CNS disorder treatment. Continued research in this area will lead to innovative interventions and improved patient outcomes.
Collapse
Affiliation(s)
- Jing Wei
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China; School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Chunmeng Liu
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China.
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, China.
| | - Ting Chen
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua 418000, China.
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Department of Cardiology, the Affiliated Hospital of Southwest Medical University and Key Laboratory of Medical Electrophysiology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
231
|
Oluseyifunmi IW, Lourenco J, Olukosi OA. The interactivity of sources and dietary levels of resistant starches - impact on growth performance, starch, and nutrient digestibility, digesta oligosaccharides profile, cecal microbial metabolites, and indicators of gut health in broiler chickens. Poult Sci 2024; 103:104337. [PMID: 39388980 PMCID: PMC11752116 DOI: 10.1016/j.psj.2024.104337] [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: 07/16/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
In a 21-d study, 480 Cobb 500 (off-sex) male broiler chicks were used to investigate the effects of feeding different sources and levels of resistant starches (RS) on growth performance, nutrient and energy utilization, and intestinal health in broiler chickens. The birds were allocated to 10 dietary treatments in a 3 × 3 + 1 factorial arrangement. The factors were 3 RS-sources (RSS): banana starch (BS), raw potato starch (RPS), and high-amylose corn starch (HCS); each at 3 levels (RSL) 25, 50, or 100 g/kg plus a corn-soybean meal control diet. Birds and feed were weighed on d 0, 8, and 21. On d 21, samples of jejunal tissue and digesta were collected for chemical analysis. Data were analyzed using the mixed model procedure of JMP with factor levels nested with the control. In the 0 to 21 phase, the birds fed the RPS diets had higher (P = 0.011) FI than those fed HCS or control diets, and FCR was greater (P = 0.030) in birds that received BS diets than in other diets. RSS × RSL was significant (P < 0.05) for total tract nutrient retention, AME, and AMEn on d 21. The starch digestibility was higher (P < 0.001) in birds that received the control diet than in RS diets, and decreased as RS levels increased, except for HCS. The apparent metabolizable energy (AME) and nitrogen-corrected AME (AMEn) were higher (P < 0.001) in birds fed 100 g/kg HCS diet, with both decreasing with increasing levels of BS and RPS, except for HCS. Relative ileal oligosaccharides profile showed significant (P < 0.05) RSS × RSL with a higher relative abundance of Hex(3) (P = 0.01) and Pent(3) (P = 0.001) in HCS diets. In conclusion, RS may influence gut health and growth performance in broiler chickens through modulation of cecal SCFA and nutrient digestion, but these depend largely on the botanical origin and concentrations of individual RS.
Collapse
Affiliation(s)
| | - Jeferson Lourenco
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Oluyinka A Olukosi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
232
|
Cheng J, Ye K, Fu C, Zhou Y, Chen Y, Ma G, Chen S, Tu J, Xiao H. Comprehensive assessment of rice bran dietary fiber on gut microbiota composition and metabolism during in vitro fermentation. Food Res Int 2024; 197:115231. [PMID: 39577956 DOI: 10.1016/j.foodres.2024.115231] [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: 07/14/2024] [Revised: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024]
Abstract
Rice bran, a by-product of rice processing, is rich in various nutrients. As one of the main components of rice bran, dietary fiber has a variety of potential health benefits, especially its probiotic effects on gut health. This study involved the preparation and characterization of soluble rice bran dietary fibers (RB-SDF) and insoluble rice bran dietary fibers (RB-IDF), followed by an investigation into their gastrointestinal probiotic impact and principal metabolites. These results showed that rice bran dietary fiber could promote the production of short-chain fatty acids and the growth of probiotics during the fermentation in vitro. Specifically, RB-SDF significantly stimulated the growth of Bacteroides, Parabacteroides, and Acinetobacter, while RB-IDF encouraged the expansion of Tyzzerella, Pseudoflavonifractor, and Lachnospiraceae_UCG_004. Both dietary fibers could reduce the relative abundance of Escherichia_Shigella and Fusobacterium. The differential metabolites identified by untargeted metabolomics were l-pyroglutamic acid, d-(+)-tryptophan, indole-3-lactic acid, sulfolithocholic acid, 4-hydroxybenzaldehyde, indicating that different carbohydrates could significantly affect the metabolic profile of gut microbiota. Our finding indicated that rice bran dietary fiber can produce beneficial metabolites and modulate microbial ecosystems, which deserve further development for health applications.
Collapse
Affiliation(s)
- Jingni Cheng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Kai Ye
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Chujing Fu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yu Zhou
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yang Chen
- School of Biotechnology, Jiangsu University of Science and Technology, Jiangsu Province, Zhenjiang 212008, China
| | - Gaoxing Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jie Tu
- School of Biotechnology, Jiangsu University of Science and Technology, Jiangsu Province, Zhenjiang 212008, China
| | - Hang Xiao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Department of Food Science, University of Massachusetts, Amherst 01003, USA.
| |
Collapse
|
233
|
Ranjan A, Arora J, Chauhan A, Basniwal RK, Kumari A, Rajput VD, Prazdnova EV, Ghosh A, Mukerjee N, Mandzhieva SS, Sushkova S, Minkina T, Jindal T. Advances in characterization of probiotics and challenges in industrial application. Biotechnol Genet Eng Rev 2024; 40:3226-3269. [PMID: 36200338 DOI: 10.1080/02648725.2022.2122287] [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: 05/09/2022] [Accepted: 08/05/2022] [Indexed: 11/02/2022]
Abstract
An unbalanced diet and poor lifestyle are common reasons for numerous health complications in humans. Probiotics are known to provide substantial benefits to human health by producing several bioactive compounds, vitamins, short-chain fatty acids and short peptides. Diets that contain probiotics are limited to curd, yoghurt, kefir, kimchi, etc. However, exploring the identification of more potential probiotics and enhancing their commercial application to improve the nutritional quality would be a significant step to utilizing the maximum benefits. The complex evolution patterns among the probiotics are the hurdles in their characterization and adequate application in the industries and dairy products. This article has mainly discussed the molecular methods of characterization that are based on the analysis of ribosomal RNA, whole genome, and protein markers and profiles. It also has critically emphasized the emerging challenges in industrial applications of probiotics.
Collapse
Affiliation(s)
- Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Jayati Arora
- Amity Institute of Environmental Sciences, Amity University, Noida, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| | - Rupesh Kumar Basniwal
- Amity Institute of Advanced Research and Studies (M&D), Amity University, Noida, India
| | - Arpna Kumari
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Vishnu D Rajput
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Evgeniya V Prazdnova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati, India
| | - Nobendu Mukerjee
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
- Department of Health Sciences, Novel Global Community Educational Foundation, New South Wales, Australia
| | - Saglara S Mandzhieva
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Svetlana Sushkova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Tatiana Minkina
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Tanu Jindal
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| |
Collapse
|
234
|
Horn PA, Zeni ALB, Herkenhoff ME, Curbani L, Pereira Gonçalves GH, Rutkoski CF, Israel NG, de Almeida EA. Brewer's spent yeast improves human gut microbiota and ameliorates clinical blood parameters: A randomized, double-blind, placebo-controlled trial. BIOACTIVE CARBOHYDRATES AND DIETARY FIBRE 2024; 32:100442. [DOI: 10.1016/j.bcdf.2024.100442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
235
|
Bai Y, Zhao Y, Jin J, Ye Z, Fan H, Zhao D, Gao S. Jiang Tang San Hao Formula exerts its anti-diabetic effect by affecting the gut-microbiota-brain axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156100. [PMID: 39388919 DOI: 10.1016/j.phymed.2024.156100] [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: 05/04/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Type 2 diabetes is a complex metabolic disorder characterized by insulin resistance and impaired insulin secretion, with growing evidence highlighting the critical role of the gut-microbiota-brain axis in modulating glucose and lipid metabolism. OBJECTIVE To evaluate the effects of Jiang Tang San Hao Formula (JTSHF) on blood glucose control in type 2 diabetic mouse model and to explore its mechanism through the gut- microbiota-brain axis. METHODS A type 2 diabetes model was established using six-week-old male C57BL6/J mice, induced by a high-fat diet combined with streptozotocin injection. The diabetic mice then randomly assigned to the model group, metformin (Glucophage) group and JTSHF group, receiving 11 weeks of treatment by gavage. Body weight and fasting blood glucose were monitored biweekly. The oral glucose tolerance test was performed during the fifth and 10th weeks of the intervention. The measurements of body composition were conducted pre- and post-treatment. After the intervention, serum insulin, lipid levels, glucagon like peptide-1 (GLP-1), peptide YY, ghrelin, and leptin were detected. The fresh feces of mice were collected before sacrifice for gut microbiota analysis and short chain fatty acids quantification. The colon tissues of mice in each group were collected to observe the morphological structure and to measure the expression levels of GPR41 and GPR43. The hypothalamus was collected to assess the expression of POMC, AgRP and NPY. RESULTS JTSHF significantly boosted sugar and lipid metabolism and contributed to weight reduction in diabetic mice (p < 0.05). At the genus level, JTSHF increased the relative abundance of Bacteroides, Prevotella, and Parabacteroides, and decreased Clostridium, Lactobacillus, and Oscillibacter in the gut microbiota. JTSHF enhanced the content of short chain fatty acids, improved the expression level of GPR43/41 in colonic tissue (p < 0.05), and increased POMC expression while decreasing AgRP and NPY expression in the hypothalamus (p < 0.05). Serum GLP-1 was increased, and ghrelin was decreased significantly after JTSHF intervention (p < 0.05). CONCLUSION By affecting the composition, relative abundance, and metabolites of gut microbiota, JTSHF regulates various gut brain peptides, affects the hypothalamic feeding center, improves glucose and lipid metabolism, and thus plays the anti-diabetic role. The study provides novel insights into how traditional Chinese medicine modulates the gut-brain connection to exert anti-diabetic effects, highlighting the innovative potential of JTSHF in metabolic disease management.
Collapse
Affiliation(s)
- Ying Bai
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yi Zhao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jialin Jin
- China Science and Technology Development Center for Chinese Medicine, Beijing, China
| | - Zimengwei Ye
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Fan
- Guangdong Pharmaceutical University, Guangdong, China
| | - Dandan Zhao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Sihua Gao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
236
|
Wei J, Tian Y, Guan M, Wei J, Ji Y, Tao G, Sylvester KG. Sodium formate induces development-dependent intestinal epithelial injury via necroptosis and apoptosis. Redox Rep 2024; 29:2433393. [PMID: 39620924 PMCID: PMC11613409 DOI: 10.1080/13510002.2024.2433393] [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] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES Necrotizing enterocolitis (NEC) is a common and sometimes fatal disease affecting premature infants. Elevated formate has been found in the stool of patients with NEC. Sodium formate (NaF) is used to explore the role of formate in the intestinal epithelial injury. METHODS In this study, 150 mM NaF solution was intraluminally injected in 14-day-old and 28-day-old mice. Mice were sacrificed after 24 h of feces collection, and the blood and small intestinal tissues were collected to detect the pathological damage of intestinal tissue, intestinal permeability, oxidative stress indicators including SOD, HO-1, MDA, and 4-HNE, inflammatory cytokines including IL-1β, TNF-α and IL-6, mitochondrial function such as ATP and PGC-1α in mice intestinal tissue, indicators of the cell death modes including necroptosis-related protein RIPK1 and p-MLKL, and apoptosis- related protein cleaved-caspase-3 and p-AKT (S473). RESULTS NaF treatment significantly damaged intestinal epithelial tissue and barrier function, caused mitochondrial dysfunction, manifesting as decreased ATP and PGC-1α levels, increased lipid peroxidation products MDA and 4-HNE, depleted antioxidant enzyme SOD, and upregulated the expression of HO-1. Furthermore, NaF treatment induced inflammatory responses by promoting the release of IL-1β, IL-6 and TNF-α in a development-dependent manner, eventually inducing necroptosis and apoptosis. CONCLUSIONS Formate may be a source of metabolic intestinal injury contributing to the pathogenesis of NEC in human newborns.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yuan Tian
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Meiqi Guan
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jinshu Wei
- Department of Pediatrics, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yong Ji
- Department of Neonatal Intensive Care Unit, Shanxi Children’s Hospital, Taiyuan, People’s Republic of China
| | - Guozhong Tao
- Pediatric Surgery-Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karl G. Sylvester
- Pediatric Surgery-Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
237
|
Basnet J, Eissa MA, Cardozo LLY, Romero DG, Rezq S. Impact of Probiotics and Prebiotics on Gut Microbiome and Hormonal Regulation. GASTROINTESTINAL DISORDERS 2024; 6:801-815. [PMID: 39649015 PMCID: PMC11623347 DOI: 10.3390/gidisord6040056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
The gut microbiome plays a crucial role in human health by influencing various physiological functions through complex interactions with the endocrine system. These interactions involve the production of metabolites, signaling molecules, and direct communication with endocrine cells, which modulate hormone secretion and activity. As a result, the microbiome can exert neuroendocrine effects and contribute to metabolic regulation, adiposity, and appetite control. Additionally, the gut microbiome influences reproductive health by altering levels of sex hormones such as estrogen and testosterone, potentially contributing to conditions like polycystic ovary syndrome (PCOS) and hypogonadism. Given these roles, targeting the gut microbiome offers researchers and clinicians novel opportunities to improve overall health and well-being. Probiotics, such as Lactobacillus and Bifidobacterium, are live beneficial microbes that help maintain gut health by balancing the microbiota. Prebiotics, non-digestible fibers, nourish these beneficial bacteria, promoting their growth and activity. When combined, probiotics and prebiotics form synbiotics, which work synergistically to enhance the gut microbiota balance and improve metabolic, immune, and hormonal health. This integrated approach shows promising potential for managing conditions related to hormonal imbalances, though further research is needed to fully understand their specific mechanisms and therapeutic potential.
Collapse
Affiliation(s)
- Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Manar A. Eissa
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Damian G. Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
238
|
Lim JJ, Reginald K, Say YH, Liu MH, Chew FT. Frequent intake of high fiber and probiotic diets lowers risks associated with atopic dermatitis and house dust mite allergy: a cross-sequential study of young Chinese adults from Singapore and Malaysia. Eur J Nutr 2024; 64:38. [PMID: 39614888 PMCID: PMC11608386 DOI: 10.1007/s00394-024-03524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/18/2024] [Indexed: 12/06/2024]
Abstract
PURPOSE Dietary fiber intake may influence the risk and severity of atopic dermatitis (AD), a common chronic allergic skin condition. This cross-sequential study investigated the association between dietary fiber intake and various characteristics of AD, including house dust mites (HDM) allergy and dry skin, in 13,561 young Chinese adults (mean years = 22.51, SD ± 5.90) from Singapore and Malaysia. METHODS Dietary habits were assessed using a validated semi-quantitative, investigator-administered food frequency questionnaire from the International Study of Asthma and Allergies in Childhood. We derived an amount-based dietary index to estimate fiber intake while studying its correlation with probiotic drinks intake. AD status was determined by skin prick tests for HDM and symptomatic histories of eczema. Multivariable logistic regression analysis, adjusting for demographic, genetic predisposition, body mass index and lifestyle factors, and synergy factor analysis were used to explore the association and interaction of dietary factors on disease outcomes. RESULTS High fiber intake (approximately 98.25 g/serving/week) significantly lowered the associated risks for HDM allergy (Adjusted Odds Ratio [AOR]: 0.895; 95% Confidence Intervals [CI]: 0.810-0.989; adjusted p-value < 0.05) and AD (AOR: 0.831; 95% CI: 0.717-0.963; adjusted p-value < 0.05), but not dry skin. While probiotic intake was not associated with AD, it was significantly correlated with fiber intake (R2 = 0.324, p-value < 0.0001). Among those frequently consuming probiotics, moderate fiber intake sufficiently lowered the AD risk (AOR: 0.717; 95% CI: 0.584-0.881; adjusted p-value < 0.01). Moreover, a fibre-rich diet independently mitigated risks associated with high intake of fats, saturated fats, and protein. CONCLUSION A high-fiber diet is associated with AD and HDM allergy. Moderate-to-high fiber intake, particularly in conjunction with probiotics, may further mitigate AD risks.
Collapse
Affiliation(s)
- Jun Jie Lim
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Allergy and Molecular Immunology Laboratory, Lee Hiok Kwee Functional Genomics Laboratories, Block S2, Level 5, 14 Science Drive 4, off Lower Kent Ridge Road, Singapore, 117543, Singapore
| | - Kavita Reginald
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Allergy and Molecular Immunology Laboratory, Lee Hiok Kwee Functional Genomics Laboratories, Block S2, Level 5, 14 Science Drive 4, off Lower Kent Ridge Road, Singapore, 117543, Singapore
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, 47500, Petaling Jaya, Selangor, Malaysia
| | - Yee-How Say
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Allergy and Molecular Immunology Laboratory, Lee Hiok Kwee Functional Genomics Laboratories, Block S2, Level 5, 14 Science Drive 4, off Lower Kent Ridge Road, Singapore, 117543, Singapore
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, 47500, Petaling Jaya, Selangor, Malaysia
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR), 31900, Kampar, Perak, Malaysia
| | - Mei Hui Liu
- Department of Food Science & Technology, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Fook Tim Chew
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Allergy and Molecular Immunology Laboratory, Lee Hiok Kwee Functional Genomics Laboratories, Block S2, Level 5, 14 Science Drive 4, off Lower Kent Ridge Road, Singapore, 117543, Singapore.
| |
Collapse
|
239
|
Gul S, Shi Y, Hu J, Song S. The Influence of Microbiota on Wild Birds' Parental Coprophagy Behavior: Current Advances and Future Research Directions. Microorganisms 2024; 12:2468. [PMID: 39770671 PMCID: PMC11677090 DOI: 10.3390/microorganisms12122468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive review provides an in-depth exploration of the intriguing phenomenon of parental coprophagy in wild birds and its profound implications on the influence of adult avian parents' health. This review investigates the composition and dynamics of avian feces' microbiota, casting light on the various dietary, environmental, and genetic factors that influence its diversity. Furthermore, it emphasizes parental coprophagy, a behavior observed in numerous bird species, particularly among herbivorous and passerine birds. The review investigates multiple hypotheses proposed to explain the occurrence of coprophagy. It delves into its function as a potential mechanism for transmitting microorganisms, particularly feces bacteria, from nestlings to their parents. This microbial transfer may affect the health and well-being of adult avian parents. In addition, the review highlights the current research deficits and debates surrounding coprophagy. These gaps include crucial aspects such as the onset of coprophagy, its long-term effects on both parents and offspring, the nutritional implications of consuming nestling feces, the potential risks of pathogen transmission, and the ecological and evolutionary factors that drive this behavior. As the review synthesizes existing knowledge and identifies areas requiring additional research, it emphasizes the significance of future studies that comprehensively address these gaps. By doing so, we can understand coprophagy's ecological and evolutionary significance in wild birds, advancing our knowledge on avian biology. This information can improve conservation efforts to protect migratory bird populations and their complex ecosystems.
Collapse
Affiliation(s)
- Saba Gul
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| | - Yurou Shi
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| | - Jie Hu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
- Institute of Environmental Sciences, Leiden University, 2333CC Leiden, The Netherlands
| | - Sen Song
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (S.G.); (Y.S.); (J.H.)
| |
Collapse
|
240
|
Beldie LA, Dica CC, Moța M, Pirvu BF, Burticală MA, Mitrea A, Clenciu D, Efrem IC, Vladu BE, Timofticiuc DCP, Roșu MM, Gheonea TC, Amzolini AM, Moța E, Vladu IM. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024; 16:4131. [PMID: 39683525 DOI: 10.3390/nu16234131] [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: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Recent studies have revealed that dysbiosis, defined as alterations in gut microbiota, plays an important role in the development and the progression of many non-communicable diseases, including metabolic disorders, such as type 2 diabetes mellitus and gestational diabetes mellitus (GDM). The high frequency of GDM makes this disorder an important public health issue, which needs to be addressed in order to reduce both the maternal and fetal complications that are frequently associated with this disease. The studies regarding the connections between gut dysbiosis and GDM are still in their early days, with new research continuously emerging. This narrative review seeks to outline the mechanisms through which a healthy diet that protects the gut microbiota is able to prevent the occurrence of GDM, thus providing medical nutritional therapeutic perspectives for the management of GDM.
Collapse
Affiliation(s)
- Luiza-Andreea Beldie
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Cristina-Camelia Dica
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Maria Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bianca-Florentina Pirvu
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Marilena-Alexandra Burticală
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
| | - Adina Mitrea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Clenciu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Cristian Efrem
- Department of Medical Semiology, Faculty of Dentistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Beatrice Elena Vladu
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Diana Cristina Protasiewicz Timofticiuc
- Department of Diabetes, Nutrition and Metabolic Diseases, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Magdalena Roșu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Midwives and Nursing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Theodora Claudia Gheonea
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Maria Amzolini
- Department of Medical Semiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Moța
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ionela Mihaela Vladu
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
241
|
Stolarczyk E, Vong CT, Garrido-Mesa N, Marks E, Abdel-Aziz D, Ju Q, Jackson I, Powell N, Lord GM, Howard JK. Global deletion of the immune cell transcription factor, T-bet, alters gut microbiota and insulin sensitivity in mice. Front Genet 2024; 15:1502832. [PMID: 39664730 PMCID: PMC11631911 DOI: 10.3389/fgene.2024.1502832] [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: 09/27/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
The gut microbiota plays a role in energy homeostasis: its composition differs in lean and obese mice and may impact insulin sensitivity. The immune system has co-evolved with the gut microbiota, but direct regulation of microbial communities by the immune system and its metabolic impact is unclear. Mice lacking the immune cell specific transcription factor T-bet (Tbx21) are insulin sensitive. Compared with wild-type mice, T-bet deficient mice were found to have a higher proportion of colonic regulatory T cells despite significantly fewer colonic T cells, B cells and NK cells. Microbiota deletion by administration of antibiotics, increased colonic immune cell numbers. Furthermore, we report that T-bet -/- mice have an altered gut microbial composition and fecal short-chain fatty acid content, with an increase in butyrate production, compared with wild-type mice. Finally, in a proof-of concept study, we show that the enhanced insulin sensitivity observed in T-bet -/- mice is temporarily transmissible to antibiotic-treated wild-type mice through fecal transfer. Immune regulation of the gut microbiota by T-bet may be a novel pathway modulating insulin sensitivity.
Collapse
Affiliation(s)
- E. Stolarczyk
- Diabetes and Obesity Theme, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, United Kingdom
| | - C. T. Vong
- Diabetes and Obesity Theme, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, United Kingdom
| | - N. Garrido-Mesa
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - E. Marks
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - D. Abdel-Aziz
- Diabetes and Obesity Theme, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, United Kingdom
| | - Q. Ju
- Diabetes and Obesity Theme, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, United Kingdom
| | - I. Jackson
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - N. Powell
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - G. M. Lord
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - J. K. Howard
- Diabetes and Obesity Theme, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
242
|
Saleem J, Zakar R, Iqbal S, Arshad M, Shahzad R, Batool M, Nawaz M, Butt MS, Fischer F. Effects of prebiotics on microbial diversity and abundance in young children with acute malnutrition: study protocol for a multi-centered, double-blinded randomized controlled trial. Trials 2024; 25:798. [PMID: 39593072 PMCID: PMC11590257 DOI: 10.1186/s13063-024-08647-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The anti-inflammatory and antimicrobial benefits of prebiotics may present an affordable and cost-effective strategy for not only the prevention but also treatment of malnutrition. Therefore, the present trial has been designed with the aim to evaluate the role of prebiotics on the gut microbiome of severe acute malnourished (SAM) children. METHODS The study is designed as a prospective, double-blinded, triple-armed, multi-centered randomized controlled trial, with 6-59 months old uncomplicated SAM children recruited to the experimental group receiving ready-to-use therapeutic food (RUTF) plus prebiotics and the active comparator group receiving RUTF plus starch for 2 months duration (8 weeks). Healthy children with matching age and gender will be recruited to placebo comparator group and will receive starch as a placebo during the study period. A total of 58 participants will be recruited to each arm with 1:1:1 allocation ratio following a pre-defined inclusion and exclusion criteria. The results of the gut microbiome diversity will serve as the primary outcome, while weight-for-height/length z-score, mid-upper-arm circumference, neurodevelopment assessment, and body mass accumulation will serve as the secondary outcome. Data collection and evaluations will be conducted at baseline and at the end of the trial (week 8), while the safety monitoring will be conducted at every second week. For analysis, the principles of intention-to-treat will be followed. CONCLUSIONS Conclusively, the results of the present trial would provide useful insights and high-quality data for the treatment and management of SAM children by evaluating the effect of RUTF plus prebiotic on the gut microbiome diversity of children, leading to medical evidence for designing the large-scale studies. TRIAL REGISTRATION The present trial is registered at ClinicalTrials.gov with identifier No: NCT06155474 and registration date 4 December 2023.
Collapse
Affiliation(s)
- Javeria Saleem
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Rubeena Zakar
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Sanaullah Iqbal
- Department of Food Science and Human Nutrition, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Arshad
- Centre for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ruhma Shahzad
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Munazza Batool
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Muhammad Nawaz
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | | | - Florian Fischer
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
243
|
Saedi S, Derakhshan S, Hasani A, Khoshbaten M, Poortahmasebi V, Milani PG, Sadeghi J. Recent Advances in Gut Microbiome Modulation: Effect of Probiotics, Prebiotics, Synbiotics, and Postbiotics in Inflammatory Bowel Disease Prevention and Treatment. Curr Microbiol 2024; 82:12. [PMID: 39589525 DOI: 10.1007/s00284-024-03997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
The human gastrointestinal tract contains trillions of microbes that affect the body. Dysbiosis in the composition of gut microbiota is one of the leading causes of chronic inflammatory diseases such as inflammatory bowel disease (IBD). IBD is a global public health challenge and millions of people in the world are suffering from this disease. It is a recurring inflammatory disease that affects different parts of the human digestive system. Ulcerative colitis and Crohn's disease are the two main types of IBD with similar clinical symptoms. The increasing incidence and severity of IBD require new treatment methods. The composition of the gut microbiota can be modified using dietary supplements such as prebiotics and bacterial supplements called probiotics. Furthermore, the effects of the microbiome can be improved by using paraprobiotics (non-viable, inactivated bacteria or their components) and/or postbiotics (products of bacterial metabolism).
Collapse
Affiliation(s)
- Samira Saedi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Reasearch Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safoura Derakhshan
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alka Hasani
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Khoshbaten
- Department of Internal Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Payam Gonbari Milani
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadeghi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
244
|
Szczuko M, Duliban G, Drozd A, Sochaczewska D, Pokorska-Niewiada K, Ziętek M. The Association of Short-Chain Fatty Acids with the Occurrence of Gastrointestinal Symptoms in Infants. Int J Mol Sci 2024; 25:12487. [PMID: 39684199 DOI: 10.3390/ijms252312487] [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: 09/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are produced by the fermentation of undigested polysaccharides; they are a group of metabolites resulting from the activity of intestinal bacteria. The main SCFAs are acetic, butyric, propionic, valeric, and caproic acid, and their levels and proportions depend on various factors. The aim of this study was to investigate the relationship between the concentration of SCFAs and the occurrence of specific gastrointestinal symptoms in infants. This study was conducted using faecal samples obtained at 1, 3, 6, and 12 months of age. The SCFA content was measured using gas chromatography. At 1 month, an association was found between butyric acid and flatulence. At 3 months, an association was found between butyric acid and flatulence/gas and between 3,4-methylovaleric acid and mucus in the stool. At 6 months, an association was found between butyric and valeric acids and flatulence. By 12 months, the gastrointestinal symptoms had decreased significantly. This study confirms that there is an association between SCFA levels and the presence of bloating, gas, mucus in the stool, and constipation in the gastrointestinal tract. Higher levels of butyric and valeric acids may lead to an increase in troublesome symptoms, such as flatulence and gas, in the first few months of life but are not associated with the occurrence of intestinal colic. The level of 3,4-methylovaleric acid is associated with the presence of allergies, whereas a decrease in acetic acid and an increase in isovaleric acid may exacerbate defecation problems in infants.
Collapse
Affiliation(s)
- Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
- Department of Human Nutrition and Bromatology, Pomeranian Medical University in Szczecin, 71-210 Szczecin, Poland
| | - Gabriela Duliban
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Diana Sochaczewska
- Department of Neonatology, Pomeranian Medical University in Szczecin, 72-009 Police, Poland
| | - Kamila Pokorska-Niewiada
- Department of Toxicology, Dairy Technology and Food Storage, West Pomeranian University of Technology in Szczecin, 71-459 Szczecin, Poland
| | - Maciej Ziętek
- Department of Perinatology, Obstetrics and Gynecology Pomeranian Medical University in Szczecin, 72-009 Police, Poland
| |
Collapse
|
245
|
Gao M, Li J, Han X, Zhang B, Chen J, Lang J, Zhang Q. Effect of melatonin on gut microbiome and metabolomics in diabetic cognitive impairment. Front Pharmacol 2024; 15:1489834. [PMID: 39640487 PMCID: PMC11619431 DOI: 10.3389/fphar.2024.1489834] [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: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Diabetic cognitive impairment(DCI) presents as a central nervous complication of diabetes especially among aging population. Melatonin (MEL) is known for its antioxidant and anti-inflammation effects in neuroprotective aspects. Recent evidence has demonstrated that the gut microbiome plays a key role in DCI by modulating cognitive function through the gut-brain crosstalk. MEL has been shown to modulate gut microbiota composition in diabetic model. However, the underlying mechanism through which the gut microbiome contributes to DCI remains unclear. This study aims to investigate the effect and mechanism of MEL in attenuating DCI in relation to regulating the gut microbiome and metabolomics. Methods Cognitive and memory function were assessed by the Morris water maze test, histopathological assessment of brain tissues, and immunoblotting of neuroinflammation and apoptosis. The levels of serum tumor necrosis factor-α (TNF-α) and Interleukin-18 (IL-18) were measured by enzyme-linked immunoassays to reflect the circulatory inflammation level.16S rRNA microbiome sequencing analysis was performed on control mice(db-m group), diabetic mice(db-db group) and MEL-treated diabetic mice(db-dbMEL group). Gut metabolites changes were characterized using liquid chromatography tandem mass spectrometry (LC-MS/MS). Results Our study confirmed that MEL alleviated diabetes-induced cognition and memory dysfunction. MEL protected against neuroinflammation and apoptosis in hippocampus of db-db mice. MEL corrected the increased abundance of Bacteroides and Dorea and the reduced abundance of Prevotella in db-db mice. The vast majority of differential metabolites among the three groups were lipids and lipid-like molecules. MEL significantly restored the reduced levels of pyruvate and lactic acid. Discussion Our results supported the use of MEL as a promising therapeutic agent for DCI, in which the underlying mechanism may be associated with gut microbiome and metabolomics regulation.
Collapse
Affiliation(s)
- Ming Gao
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Rare Disease, Hebei Provincial Department of Science and Technology, Shijiazhuang, Hebei, China
| | - Jie Li
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Rare Disease, Hebei Provincial Department of Science and Technology, Shijiazhuang, Hebei, China
| | - Xu Han
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Beiyao Zhang
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinting Chen
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiadong Lang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiangqiang Zhang
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
246
|
Coll E, Cigarran S, Portolés J, Cases A. Gut Dysbiosis and Its Role in the Anemia of Chronic Kidney Disease. Toxins (Basel) 2024; 16:495. [PMID: 39591250 PMCID: PMC11598790 DOI: 10.3390/toxins16110495] [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: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The gut dysbiosis present in chronic kidney disease (CKD) has been associated with anemia. Factors such as the accumulation of gut-derived uremic toxins, increased gut barrier permeability-induced inflammation, and a reduced intestinal production of short-chain fatty acids (SCFAs), all associated with changes in the intestinal microbiota composition in CKD, may lead to the development or worsening of anemia in renal patients. Understanding and addressing these mechanisms related to gut dysbiosis in CKD patients can help to delay the development of anemia and improve its control in this population. One approach is to avoid or reduce the use of drugs linked to gut dysbiosis in CKD, such as phosphate binders, oral iron supplementation, antibiotics, and others, unless they are indispensable. Another approach involves introducing dietary changes that promote a healthier microbiota and/or using prebiotics, probiotics, or symbiotics to improve gut dysbiosis in this setting. These measures can increase the presence of SCFA-producing saccharolytic bacteria and reduce proteolytic bacteria, thereby lowering the production of gut-derived uremic toxins and inflammation. By ameliorating CKD-related gut dysbiosis, these strategies can also improve the control of renal anemia and enhance the response to erythropoiesis-stimulating agents (ESAs) in ESA-resistant patients. In this review, we have explored the relationship between gut dysbiosis in CKD and renal anemia and propose feasible solutions, both those already known and potential future treatments.
Collapse
Affiliation(s)
- Elisabet Coll
- Servei de Nefrologia, Fundacio Puigvert, 08025 Barcelona, Spain
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
| | | | - Jose Portolés
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Ressearch Net RICORS 2030 Instituto de Salud Carlos III ISCIII, 28029 Madrid, Spain
- Nephrology Department, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain
- Medicine Department, Facultad de Medicina, Research Institute Puerta de Hierro Segovia de Arana (IDIPHISA), Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Aleix Cases
- Anemia Working Group of the Spanish Society of Nephrology, 39008 Santander, Spain; (J.P.); (A.C.)
- Nephrology Unit, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
247
|
Ostrowska M, Nowosad K, Mikoluc B, Szczerba H, Komon-Janczara E. Changes in the Gut and Oral Microbiome in Children with Phenylketonuria in the Context of Dietary Restrictions-A Preliminary Study. Nutrients 2024; 16:3915. [PMID: 39599702 PMCID: PMC11597790 DOI: 10.3390/nu16223915] [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: 10/22/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Phenylketonuria (PKU) is a metabolic disorder that necessitates dietary restrictions, potentially impacting the composition of gut and oral microbiota. This study aimed to compare the microbiota composition between children with PKU and healthy controls. METHODS Using 16S rRNA gene sequencing, we analysed microbial communities at six phylogenetic levels. RESULTS Our findings revealed significant differences in the gut microbiota: Euryarchaeota was more abundant in controls (p = 0.01), while Bacilli and Lactobacillales were higher in PKU children (p = 0.019). Methanobacteriales were significantly elevated in controls (p = 0.01). At the genus and species levels, PKU children had higher Streptococcus and Eubacterium dolichum (p = 0.019, p = 0.015), whereas controls had more Barnesiella, Coprococcus, and Faecalibacterium prausnitzii (p = 0.014, p = 0.019, p = 0.014). In the oral microbiota, control children exhibited significantly higher Bacteroidetes (p = 0.032), while PKU children had increased Bacilli and Betaproteobacteria (p = 0.0079, p = 0.016). Streptococcus and Neisseria were more prevalent in PKU (p = 0.0079, p = 0.016). CONCLUSIONS These results suggest that PKU and its dietary management significantly alter the gut and oral microbiota composition. Understanding these microbial shifts could have implications for managing PKU and improving patient outcomes.
Collapse
Affiliation(s)
- Malgorzata Ostrowska
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Karolina Nowosad
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Bozena Mikoluc
- Department of Pediatrics, Rheumatology, Immunology and Metabolic Bone Diseases, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Hubert Szczerba
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Elwira Komon-Janczara
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| |
Collapse
|
248
|
Enache RM, Profir M, Roşu OA, Creţoiu SM, Gaspar BS. The Role of Gut Microbiota in the Onset and Progression of Obesity and Associated Comorbidities. Int J Mol Sci 2024; 25:12321. [PMID: 39596385 PMCID: PMC11595101 DOI: 10.3390/ijms252212321] [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: 10/05/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity, a global public health problem, is constantly increasing, so the concerns in preventing and combating it are increasingly focused on the intestinal microbiota. It was found that the microbiota is different in lean people compared to obese individuals, but the exact mechanisms by which energy homeostasis is influenced are still incompletely known. Numerous studies show the involvement of certain bacterial species in promoting obesity and associated diseases such as diabetes, hypertension, cancer, etc. Our aim is to summarize the main findings regarding the influence of several factors such as lifestyle changes, including diet and bariatric surgery, on the diversity of the gut microbiota in obese individuals. The second purpose of this paper is to investigate the potential effect of various microbiota modulation techniques on ameliorating obesity and its comorbidities. A literature search was conducted using the PubMed database, identifying articles published between 2019 and 2024. Most studies identified suggest that obesity is generally associated with alterations of the gut microbiome such as decreased microbial diversity, an increased Firmicutes-to-Bacteroidetes ratio, and increased SCFAs levels. Our findings also indicate that gut microbiota modulation techniques could represent a novel strategy in treating obesity and related metabolic diseases. Although some mechanisms (e.g., inflammation or hormonal regulation) are already considered a powerful connection between gut microbiota and obesity development, further research is needed to enhance the knowledge on this particular topic.
Collapse
Affiliation(s)
- Robert-Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
249
|
Hu Y, Wang H, Zhong Y, Sun Y. Retrospective analysis of diet and gut microbiota diversity and clinical pharmacology outcomes in patients with Parkinsonism syndrome. Heliyon 2024; 10:e38645. [PMID: 39512468 PMCID: PMC11539248 DOI: 10.1016/j.heliyon.2024.e38645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/15/2024] Open
Abstract
Background Parkinson's disease (PD) is a global neurodegenerative ailment impacting millions, causes significant degeneration of dopaminergic neurons in the substantia nigra. Emerging research underscores the gut microbiota's role in PD onset. Yet, investigations in Chinese demographics are lacking. This study thus targets a retrospective examination of gut microbiota variety and clinical aspects in Chinese PD patients. Methods We retrospectively analyzed 50 PD patients' clinical data (admitted May 2021-April 2022) and compared their gut microbiota composition and abundance via 16S rDNA V3-V4 region sequencing against 50 healthy controls. The study also explored links between disease severity and PD patients' gut microbiota. Results We found that the gut microbiota diversity was increased in PD patients. Specifically, phyla Firmicutes, Actinobacteria, and Proteobacteria were more abundant, whereas Bacteroidetes was less abundant compared to controls. Gut microbiota diversity did not vary between early-vs. late-onset PD, tremor-dominant vs. non-tremor-dominant, or constipation-associated vs. non-constipation-associated subtypes. The abundance of Firmicutes was positively correlated with UPDRS (Unified Parkinson's Disease Rating Scale) III score, NMSS (Non-Motor Symptoms Scale) score, Wexner score, and PDQ-39 (Parkinson's Disease Questionnaire-39) score; the abundance of Actinobacteria was positively correlated with UPDRS III, NMSS, Wexner, and PDQ-39 scores; and the abundance of Bacteroidetes was negatively correlated with UPDRS III, NMSS, Wexner, and PDQ-39 scores. At the family level, the abundance of Bifidobacteriaceae, Enterobacteriaceae, and Porphyromonadaceae was positively correlated with UPDRS III, NMSS, and PDQ-39 scores. Conclusion Compared with healthy individuals, PD patients have increased gut microbiota diversity, and the abundance of Bifidobacteriaceae, Enterobacteriaceae, and Porphyromonadaceae is associated with the severity of both motor and non-motor symptoms in PD patients.
Collapse
Affiliation(s)
- Yunxin Hu
- The Sixth Department of Neurology, Guangdong 999 Brain Hospital, Baiyun District, Guangzhou City, Guangdong Province, China
| | - Haoyue Wang
- The Sixth Department of Neurology, Guangdong 999 Brain Hospital, Baiyun District, Guangzhou City, Guangdong Province, China
| | - Yongkang Zhong
- The Sixth Department of Neurology, Guangdong 999 Brain Hospital, Baiyun District, Guangzhou City, Guangdong Province, China
| | - Yongqi Sun
- The Sixth Department of Neurology, Guangdong 999 Brain Hospital, Baiyun District, Guangzhou City, Guangdong Province, China
| |
Collapse
|
250
|
Zhou J, Chen L, Foo HL, Cao Z, Lin Q. Changes in microbial diversity and volatile metabolites during the fermentation of Bulang pickled tea. Food Chem 2024; 458:140293. [PMID: 38970959 DOI: 10.1016/j.foodchem.2024.140293] [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: 02/27/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
The present study aimed to determine microbial community, short-chain fatty acids (SCFAs), and volatilome of Bulang pickled tea during fermentation. Sequencing of 16S rRNA and ITS revealed that Bualng pickled tea was dominated by Lactobacillus plantarum, unclassified Enterobacteriaceae, unclassified Debaryomyces, Candida metapsilosis, Cladosporium sphaerospermum, and unclassified Aspergillus. The overall contents of SCFAs increased, with acetic acid showing the highest content. A total of 398 differential volatile metabolites were detected using differential metabolomics analysis. Out of these different volatile compounds, ten key volatile compounds including (Z)-4-heptenal, 1-(2-thienyl)-ethanone, 5-methyl-(E)-2-hepten-4-one, 2-ethoxy-3-methylpyrazine, p-cresol, 2-methoxy-phenol, ethy-4-methylvalerate, 3-ethyl-phenol, p-menthene-8-thiol, and 2-s-butyl-3-methoxypyrazinewere were screened based on odor activity value (OAV). The Spearman correlation analysis showed a high correlation of SCFAs and volatile compounds with microorganisms, especially L. plantarum and C. sphaerospermum. This study provided a theoretical basis for elucidating the flavor quality formation mechanism of Bulang pickled tea.
Collapse
Affiliation(s)
- Jinping Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Laifeng Chen
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China
| | - Hooi Ling Foo
- Department of Bioprocess Technology, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Research Laboratory of Probiotics and Cancer Therapeutics, UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Zhenhui Cao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China.
| | - Qiuye Lin
- College of Food Science and Technology, Yunnan Agricultural University, Heilongtan, Kunming 650201, People's Republic of China.
| |
Collapse
|