151
|
Hemachandra S, Rathnayake SN, Jayamaha AA, Francis BS, Welmillage D, Kaur DN, Zaw HK, Zaw LT, Chandra HA, Abeysekera ME. Fecal Microbiota Transplantation as an Alternative Method in the Treatment of Obesity. Cureus 2025; 17:e76858. [PMID: 39901991 PMCID: PMC11788455 DOI: 10.7759/cureus.76858] [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] [Accepted: 01/03/2025] [Indexed: 02/05/2025] Open
Abstract
Fecal microbiota transplantation (FMT) has emerged as a promising therapeutic approach for various health conditions, particularly obesity and metabolic disorders. This review examines the mechanisms underlying FMT, including its role in restoring gut microbiota diversity and enhancing immunomodulatory functions, which are essential for maintaining overall health. Recent studies indicate that FMT can significantly improve body weight and metabolic parameters, suggesting its potential as an alternative or complementary treatment to current obesity therapies. However, the effectiveness of FMT depends on several factors, including the composition of the donor microbiota, recipient characteristics, and concomitant medications or dietary interventions. Despite its great promise, challenges such as standardized protocols, donor screening, and the need for a deeper understanding of gut microbiota dynamics remain key hurdles. Future research should focus on elucidating the specific microbial compositions necessary for optimal therapeutic outcomes and exploring personalized FMT approaches tailored to individual patient profiles. This evolving field presents exciting opportunities for innovative strategies in obesity treatment, warranting further investigation and clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hein K Zaw
- Gastroenterology, Nanjing Medical University, Nanjing, CHN
| | - Lin T Zaw
- Gastroenterology, Nanjing Medical University, Nanjing, CHN
| | | | | |
Collapse
|
152
|
Suresh MG, Mohamed S, Yukselen Z, Hatwal J, Venkatakrishnan A, Metri A, Bhardwaj A, Singh A, Bush N, Batta A. Therapeutic Modulation of Gut Microbiome in Cardiovascular Disease: A Literature Review. HEART AND MIND 2025; 9:68-79. [DOI: 10.4103/hm.hm-d-24-00044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/30/2024] [Indexed: 03/20/2025] Open
Abstract
Abstract
The cardiovascular diseases (CVDs) are a primary global health concern with significant mortality and morbidity. Recent findings suggest that gut microbiota, through its complex interactions with host metabolism, immune response, and inflammation, plays a crucial role in the pathogenesis and progression of CVD. The mechanisms linking gut dysbiosis with cardiovascular risk factors such as hypertension, atherosclerosis, and metabolic syndrome offer a novel perspective on heart health. Excitingly, interventions targeting the microbiome, such as dietary adjustments, probiotics, and prebiotics, hold promise in reducing CVD risks. Adopting personalized approaches to microbiome therapy, tailored to individuals’ unique microbial profiles, could usher in more effective treatments for CVD. As research continues to unveil the intricate role of the gut microbiome, the future of CVD treatment and prevention appears poised for significant transformation. Therefore, this burgeoning field promises to bring about a paradigm shift in cardiovascular health management, with a strong emphasis on personalized, microbiome-based therapeutic strategies, and preventive measures through diet and lifestyle modifications. This review aims to emphasize the potential of integrating the gut microbial insights into clinical practice and how it can be exploited to revolutionize the prevention and management of CVD in future.
Collapse
Affiliation(s)
| | - Safia Mohamed
- University of Massachusetts Chan Medical School, Baystate Medical Center, Springfield, MA, USA
| | - Zeynep Yukselen
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA, USA
| | - Juniali Hatwal
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Aida Metri
- Department of Gastroenterology, Johns Hopkins University, Baltimore, MD, United States
| | - Arshia Bhardwaj
- Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Nikhil Bush
- Department of Internal Medicine, Trinity Health Oakland/Wayne State University, Pontiac, MI, USA
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| |
Collapse
|
153
|
Guidi L, Martinez-Tellez B, Ortega Santos CP. Obesity, gut bacteria, and the epigenetic control of metabolic disease. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:333-368. [DOI: 10.1016/b978-0-443-18979-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
154
|
Saha S, Schnabl B. Modulating the microbiome in chronic liver diseases - current evidence on the role of fecal microbiota transplantation. Expert Rev Gastroenterol Hepatol 2025; 19:53-64. [PMID: 39760535 PMCID: PMC11882407 DOI: 10.1080/17474124.2025.2450707] [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: 10/29/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/07/2025]
Abstract
INTRODUCTION The gut microbiota has a complex relationship with the human host and is key to maintaining health. Disruption of the healthy diverse gut microbial milieu plays an important role in the pathogenesis of several diseases including Clostridioides difficile infection (CDI), inflammatory bowel disease, irritable bowel syndrome, alcohol-related liver disease and metabolic-dysfunction associated steatotic liver disease (MASLD). Fecal microbiota transplantation (FMT) is highly effective in treating CDI, though its utility in other diseases is still being explored. AREAS COVERED In this narrative review, we explore the role of gut microbiota in liver diseases, focusing on key changes in the microbial composition and function. We summarize current evidence on the role of FMT, identifying gaps in current research and outlining future directions for investigation. We comprehensively searched PubMed through 15 October 2024 to identify relevant studies. EXPERT OPINION While data from available studies shows promise, more research is necessary before we can use FMT for liver diseases. Key areas that require further study are - determining the optimal FMT regimen for each disease, establishing efficacy and safety with larger clinical trials, ensuring safe and equitable access to the FMT product and mechanistic insights into the reasons for success or failure of FMT.
Collapse
Affiliation(s)
- Srishti Saha
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California San Diego, San Diego, CA
| | - Bernd Schnabl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California San Diego, San Diego, CA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
155
|
Sin HCL, Haifer C. Faecal transplantation: the good, the bad and the ugly. Intern Med J 2025; 55:35-40. [PMID: 39629909 DOI: 10.1111/imj.16559] [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: 07/02/2024] [Accepted: 10/13/2024] [Indexed: 01/18/2025]
Abstract
There continues to be significant interest from both clinicians and patients in using faecal transplantation, as the integral role of the gut microbiome is increasingly recognised in various disease conditions, both within and beyond the gut. This Clinical Perspectives article provides an overview of existing literature, factors limiting the use of faecal microbial transplantation in clinical practice and exciting new advancements on the horizon.
Collapse
Affiliation(s)
- Hiu C L Sin
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney, New South Wales, Australia
| | - Craig Haifer
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney, New South Wales, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
156
|
Yang B, Zhang X, Zhu J, Wu Q, Yang B, Chitrakar B, Sang Y. Effect of extraction methods of polysaccharides from Tricholoma mongolicum Imai on digestion and fecal fermentation in vitro. Food Chem X 2024; 24:101725. [PMID: 39431207 PMCID: PMC11488415 DOI: 10.1016/j.fochx.2024.101725] [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: 06/04/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 10/22/2024] Open
Abstract
This study employed extraction methods, namely acid, alkaline, ultrasonic-assisted, hot-water, and dual enzyme-assisted extraction to extract polysaccharides from Tricholoma mongolicum Imai (TMIPs), and investigated them for intestinal digestion and fecal fermentation in vitro. Furthermore, using fructo-oligosaccharide as a positive prebiotic control, the impact of these TMIPs as carbon sources on the growth of Lactobacillus and Bifidobacterium in liquid culture was assessed. The results showed that all fractions transit through the gastrointestinal tract without degradation. Additionally, compared to the control group, the five polysaccharides significantly promoted the growth of probiotics, with a significant increase in short-chain fatty acid production after 48 h of fermentation. Furthermore, all five polysaccharides modulated the composition of gut microbiota. This offers theoretical guidance in the rational advancement of functional products derived from edible mushrooms, aiming to enhance gastrointestinal health in humans.
Collapse
Affiliation(s)
- Bing Yang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Xinyu Zhang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Jingbo Zhu
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Qunjun Wu
- Taijiyuan Biotechnology co., Ltd., Hi-tech Development Zone, 725700, Xunyang, PR China
| | - Boxiang Yang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Bimal Chitrakar
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| | - Yaxin Sang
- College of Food Science and Technology, Hebei Agricultural University, 289 Lingyusi Road, Baoding, Hebei 071001, PR China
| |
Collapse
|
157
|
Hu J, Li G, He X, Gao X, Pan D, Dong X, Huang W, Qiu F, Chen LF, Hu X. Brd4 modulates metabolic endotoxemia-induced inflammation by regulating colonic macrophage infiltration in high-fat diet-fed mice. Commun Biol 2024; 7:1708. [PMID: 39733044 DOI: 10.1038/s42003-024-07437-2] [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: 07/30/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
High-fat diet (HFD) induces low-grade chronic inflammation, contributing to obesity and insulin resistance. However, the precise mechanisms triggering obesity-associated metabolic inflammation remain elusive. In this study, we identified epigenetic factor Brd4 as a key player in this process by regulating the expression of Ccr2/Ccr5 in colonic macrophage. Upon 4-week HFD, myeloid-lineage-specific Brd4 deletion (Brd4-CKO) mice showed reduced colonic inflammation and macrophage infiltration with decreased expression of Ccr2 and Ccr5. Mechanistically, Brd4 was recruited by NF-κB to the enhancer regions of Ccr2 and Ccr5, promoting enhancer RNA expression, which facilitated Ccr2/Ccr5 expression and macrophage migration. Furthermore, decreased infiltration of Ccr2/Ccr5-positive colonic macrophages in Brd4-CKO mice altered gut microbiota composition and reduced intestinal permeability, thereby lowering metabolic endotoxemia. Finally, Brd4-CKO mice subjected to a 4-week LPS infusion exhibited restored susceptibility to HFD-induced obesity and insulin resistance. This study identifies Brd4 as a critical initiator of colonic macrophage-mediated inflammation and metabolic endotoxemia upon HFD, suggesting Brd4 as a potential target for mitigating HFD-induced inflammation, obesity, and its metabolic complications.
Collapse
Affiliation(s)
- Jinfeng Hu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Basic Medical Sciences, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Guo Li
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Basic Medical Sciences, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaoxin He
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Basic Medical Sciences, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xuming Gao
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Basic Medical Sciences, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Dun Pan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xingchen Dong
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Wentao Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University; Department of Hepato-Pancreato-Biliary Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Funan Qiu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University; Department of Hepato-Pancreato-Biliary Surgery, Fujian Provincial Hospital, Fuzhou, China.
| | - Lin-Feng Chen
- Department of Biochemistry, College of Liberal Arts & Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Xiangming Hu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Basic Medical Sciences, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
158
|
Stachelska MA, Karpiński P, Kruszewski B. Health-Promoting and Functional Properties of Fermented Milk Beverages with Probiotic Bacteria in the Prevention of Civilization Diseases. Nutrients 2024; 17:9. [PMID: 39796443 PMCID: PMC11722897 DOI: 10.3390/nu17010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES There is scattered information in the scientific literature regarding the characterization of probiotic bacteria found in fermented milk beverages and the beneficial effects of probiotic bacteria on human health. Our objective was to gather the available information on the use of probiotic bacteria in the prevention of civilization diseases, with a special focus on the prevention of obesity, diabetes, and cancer. METHODS We carried out a literature review including the following keywords, either individually or collectively: lactic acid bacteria; probiotic bacteria; obesity; lactose intolerance; diabetes; cancer protection; civilization diseases; intestinal microbiota; intestinal pathogens. RESULTS This review summarizes the current state of knowledge on the use of probiotic bacteria in the prevention of civilization diseases. Probiotic bacteria are a set of living microorganisms that, when administered in adequate amounts, exert a beneficial effect on the health of the host and allow for the renewal of the correct quantitative and qualitative composition of the microbiota. Probiotic bacteria favorably modify the composition of the intestinal microbiota, inhibit the development of intestinal pathogens, prevent constipation, strengthen the immune system, and reduce symptoms of lactose intolerance. As fermented milk beverages are an excellent source of probiotic bacteria, their regular consumption can be a strong point in the prevention of various types of civilization diseases. CONCLUSIONS The presence of lactic acid bacteria, including probiotic bacteria in fermented milk beverages, reduces the incidence of obesity and diabetes and serves as a tool in the prevention of cancer diseases.
Collapse
Affiliation(s)
| | - Piotr Karpiński
- Faculty of Health Sciences, University of Lomza, Akademicka 14, 18-400 Łomża, Poland;
| | - Bartosz Kruszewski
- Department of Food Technology and Assessment, Institute of Food Sciences, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland
| |
Collapse
|
159
|
Li X, Li X, Xiao H, Xu J, He J, Xiao C, Zhang B, Cao M, Hong W. Meta-analysis of gut microbiota alterations in patients with irritable bowel syndrome. Front Microbiol 2024; 15:1492349. [PMID: 39777150 PMCID: PMC11703917 DOI: 10.3389/fmicb.2024.1492349] [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: 09/06/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction Irritable bowel syndrome (IBS) is a common chronic disorder of gastrointestinal function with a high prevalence worldwide. Due to its complex pathogenesis and heterogeneity, there is urrently no consensus in IBS research. Methods We collected and uniformly reanalyzed 1167 fecal 16S rRNA gene sequencing samples (623 from IBS patients and 544 from healthy subjects) from 9 studies. Using both a random effects (RE) model and a fixed effects (FE) model, we calculated the odds ratios for metrics including bacterial alpha diversity, beta diversity, common genera and pathways between the IBS and control groups. Results Significantly lower alpha-diversity indexes were observed in IBS patients by random effects model. Twenty-six bacterial genera and twelve predicted pathways were identified with significant odds ratios and classification potentials for IBS patients. Based on these feature, we used transfer learning to enhance the predictive capabilities of our model, which improved model performance by approximately 10%. Moreover, through correlation network analysis, we found that Ruminococcaceae and Christensenellaceae were negatively correlated with vitamin B6 metabolism, which was decreased in the patients with IBS. Ruminococcaceae was also negatively correlated with tyrosine metabolism, which was decreased in the patients with IBS. Discussion This study revealed the dysbiosis of fecal bacterial diversity, composition, and predicted pathways of patients with IBS by meta-analysis and identified universal biomarkers for IBS prediction and therapeutic targets.
Collapse
Affiliation(s)
- Xiaxi Li
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiaoling Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haowei Xiao
- Center for Research and Development, Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Jiaying Xu
- Center for Research and Development, Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
| | - Jianquan He
- Department of Rehabilitation, School of Medicine, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Chuanxing Xiao
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Bangzhou Zhang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Man Cao
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wenxin Hong
- Department of Rehabilitation, School of Medicine, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, China
| |
Collapse
|
160
|
Yarahmadi A, Afkhami H, Javadi A, Kashfi M. Understanding the complex function of gut microbiota: its impact on the pathogenesis of obesity and beyond: a comprehensive review. Diabetol Metab Syndr 2024; 16:308. [PMID: 39710683 PMCID: PMC11664868 DOI: 10.1186/s13098-024-01561-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
Obesity is a multifactorial condition influenced by genetic, environmental, and microbiome-related factors. The gut microbiome plays a vital role in maintaining intestinal health, increasing mucus creation, helping the intestinal epithelium mend, and regulating short-chain fatty acid (SCFA) production. These tasks are vital for managing metabolism and maintaining energy balance. Dysbiosis-an imbalance in the microbiome-leads to increased appetite and the rise of metabolic disorders, both fuel obesity and its issues. Furthermore, childhood obesity connects with unique shifts in gut microbiota makeup. For instance, there is a surge in pro-inflammatory bacteria compared to children who are not obese. Considering the intricate nature and variety of the gut microbiota, additional investigations are necessary to clarify its exact involvement in the beginnings and advancement of obesity and related metabolic dilemmas. Currently, therapeutic methods like probiotics, prebiotics, synbiotics, fecal microbiota transplantation (FMT), dietary interventions like Mediterranean and ketogenic diets, and physical activity show potential in adjusting the gut microbiome to fight obesity and aid weight loss. Furthermore, the review underscores the integration of microbial metabolites with pharmacological agents such as orlistat and semaglutide in restoring microbial homeostasis. However, more clinical tests are essential to refine the doses, frequency, and lasting effectiveness of these treatments. This narrative overview compiles the existing knowledge on the multifaceted role of gut microbiota in obesity and much more, showcasing possible treatment strategies for addressing these health challenges.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | - Ali Javadi
- Department of Medical Sciences, Faculty of Medicine, Qom Medical Sciences, Islamic Azad University, Qom, Iran.
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Fellowship in Clinical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
161
|
Mushraf S, Chawla K, Fayaz SMA, Mathew AJ, Reddy GPK, Kappettu Gadahad MR, Shenoy PA, Devi V, Adiga S, Nayak V. Exploring the effects of probiotics on olanzapine-induced metabolic syndrome through the gut microbiota. Gut Pathog 2024; 16:77. [PMID: 39709451 DOI: 10.1186/s13099-024-00664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/07/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Maintaining gut microbial homeostasis is crucial for human health, as imbalances in the gut microbiota (GM) can lead to various diseases, including metabolic syndrome (MS), exacerbated by the use of antipsychotic medications such as olanzapine (OLZ). Understanding the role of the GM in OLZ-induced MS could lead to new therapeutic strategies. This study used metagenomic analysis to explore the impact of OLZ on the GM composition and examined how probiotics can mitigate its adverse effects in a rat model. Changes in weight, blood pressure, and lipid levels, which are key parameters defining MS, were assessed. Additionally, this study investigated serotonin, dopamine, and histopathological changes to explore their possible link with the microbiota-gut-brain axis (MGBA). RESULTS OLZ had an antagonistic effect on serotonin and dopamine receptors, and it was consistently found to alter the composition of the GM, with an increase in the relative abundance (RA) of the Firmicutes/Bacteroidetes phyla ratio and TM7 genera, indicating that the anticommonsal action of OLZ affects appetite and energy expenditure, contributing to obesity, dyslipidemia and increased blood pressure, which are core components of MS. Hepatic steatosis and intestinal damage in OLZ-treated rat tissues further indicate its role in MS. Conversely, the administration of probiotics, either alone or in combination with OLZ, was found to mitigate these OLZ-induced symptoms of MS by altering the GM composition. These alterations included increases in the abundances of the taxa Bacteroidetes, Actinobacteria, Prevotella, Blautia, Bacteroides, Bacteroidales, and Ruminococcaceae and a decrease in Firmicute abundance. These changes helped maintain gut barrier integrity and modulated neurotransmitter levels, suggesting that probiotics can counteract the adverse metabolic effects of OLZ by restoring the GM balance. Moreover, this study highlights the modulation of the MGBA by OLZ as a potential mechanism through which probiotics modulate serotonin and dopamine levels, influencing metabolic health. CONCLUSION These findings emphasise the significant impact of OLZ on the GM and its contribution to MS. These findings suggest that interventions targeting the GM, such as probiotics, could mitigate the metabolic side effects of OLZ. Future research should focus on developing integrative treatment approaches that consider the health of the gut microbiome in managing antipsychotic-induced adverse effects.
Collapse
Affiliation(s)
- Syed Mushraf
- Division of Pharmacology, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kiran Chawla
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shaik Mohammed Abdul Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Aranjani Jesil Mathew
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gayam Prasanna Kumar Reddy
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mohandas Rao Kappettu Gadahad
- Division of Anatomy, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Padmaja A Shenoy
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vasudha Devi
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shalini Adiga
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Veena Nayak
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
162
|
Shang X, Fu Y, Wang Y, Yan S. Ramulus Mori (Sangzhi) alkaloids ameliorate high-fat diet induced obesity in rats by modulating gut microbiota and bile acid metabolism. Front Endocrinol (Lausanne) 2024; 15:1506430. [PMID: 39758340 PMCID: PMC11695234 DOI: 10.3389/fendo.2024.1506430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Objective The objective of this study is to investigate the ability of Ramulus Mori (Sangzhi) alkaloid tablets (SZ-A) to ameliorate obesity and lipid metabolism disorders in rats subjected to a high-fat diet (HFD) through metagenomics, untargeted lipidomics, targeted metabolism of bile acid (BA), and BA pathways, providing a novel perspective on the management of metabolic disorders. Methods In this research, HFD-fed rats were concurrently administered SZ-A orally. We measured changes in body weight (BW), blood lipid profiles, and liver function to assess therapeutic effects. Liver lipid status was visualized through H&E and Oil Red O. Gut microbiota composition was elucidated using metagenomics. The LC-MS-targeted metabolomics approach was utilized to define the fecal BA profiles. Furthermore, the lipid metabolomics of adipose tissue samples was investigated using an LC-MS analysis platform. The expression levels of the BA receptor were determined by western blotting. Additionally, serum insulin (INS), glucagon-like peptide-1 (GLP-1), and inflammatory cytokines were quantified using an ELISA kit. The integrity of the colonic epithelial barrier was assessed using immunofluorescence. Results SZ-A notably decreased BW and blood lipid levels in obese rats while also alleviating liver injury. Additionally, SZ-A reduced the serum levels of leptin (LEP), INS, and GLP-1, indicating its potential to modulate key metabolic hormones. Most notably, SZ-A substantially improved gut microbiota composition. Specifically, it reshaped the gut microbiota structure in HFD-fed rats by increasing the relative abundance of beneficial bacteria, such as Bacteroides, while decreasing the populations of potentially harmful bacteria, such as Dorea and Blautia. At the BA level, SZ-A decreased the levels of harmful BAs, including hyodeoxycholic acid (HDCA), deoxycholic acid (DCA), 12-keto lithocholic acid (12-KLCA), lithocholic acid (LCA), and muricholic acid (MDCA). Between the model group and SZ-A, 258 differentially abundant metabolites were detected, with 72 upregulated and 186 downregulated. Furthermore, these BAs are implicated in the activation of the FXR-FGF15 and TGR5-GLP-1 pathways in the intestine. This activation helps to alleviate HFD-fed intestinal inflammation and restore intestinal barrier damage by modulating inflammatory cytokines and bolstering the intestinal barrier's capabilities. Conclusions Our findings indicate that SZ-A effectively modulates BW, serum lipid profiles, and liver function in HFD-fed rats. Moreover, SZ-A exerts a positive influence on inflammatory cytokines, thereby mitigating inflammation and promoting the restoration of the intestinal barrier. Significantly, our research indicates that adjusting the gut microbiome and BA levels could serve as an effective approach for both preventing and treating obesity and related metabolic dyslipidemia.
Collapse
Affiliation(s)
- Xin Shang
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- School of First Clinical, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yu Fu
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ying Wang
- Department of Geriatrics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuxun Yan
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
163
|
Jiang Z, Mei L, Li Y, Guo Y, Yang B, Huang Z, Li Y. Enzymatic Regulation of the Gut Microbiota: Mechanisms and Implications for Host Health. Biomolecules 2024; 14:1638. [PMID: 39766345 PMCID: PMC11727233 DOI: 10.3390/biom14121638] [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: 10/19/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
The gut microbiota, a complex ecosystem, is vital to host health as it aids digestion, modulates the immune system, influences metabolism, and interacts with the brain-gut axis. Various factors influence the composition of this microbiota. Enzymes, as essential catalysts, actively participate in biochemical reactions that have an impact on the gut microbial community, affecting both the microorganisms and the gut environment. Enzymes play an important role in the regulation of the intestinal microbiota, but the interactions between enzymes and microbial communities, as well as the precise mechanisms of enzymes, remain a challenge in scientific research. Enzymes serve both traditional nutritional functions, such as the breakdown of complex substrates into absorbable small molecules, and non-nutritional roles, which encompass antibacterial function, immunomodulation, intestinal health maintenance, and stress reduction, among others. This study categorizes enzymes according to their source and explores the mechanistic principles by which enzymes drive gut microbial activity, including the promotion of microbial proliferation, the direct elimination of harmful microbes, the modulation of bacterial interaction networks, and the reduction in immune stress. A systematic understanding of enzymes in regulating the gut microbiota and the study of their associated molecular mechanisms will facilitate the application of enzymes to precisely regulate the gut microbiota in the future and suggest new therapeutic strategies and dietary recommendations. In conclusion, this review provides a comprehensive overview of the role of enzymes in modulating the gut microbiota. It explores the underlying molecular and cellular mechanisms and discusses the potential applications of enzyme-mediated microbiota regulation for host gut health.
Collapse
Affiliation(s)
- Zipeng Jiang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Liang Mei
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuqi Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuguang Guo
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Bo Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Zhiyi Huang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yangyuan Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| |
Collapse
|
164
|
Jeyaraman N, Jeyaraman M, Dhanpal P, Ramasubramanian S, Ragavanandam L, Muthu S, Santos GS, da Fonseca LF, Lana JF. Gut microbiome and orthopaedic health: Bridging the divide between digestion and bone integrity. World J Orthop 2024; 15:1135-1145. [DOI: 10.5312/wjo.v15.i12.1135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/12/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
The gut microbiome, a complex ecosystem of microorganisms in the digestive tract, has emerged as a critical factor in human health, influencing metabolic, immune, and neurological functions. This review explores the connection between the gut microbiome and orthopedic health, examining how gut microbes impact bone density, joint integrity, and skeletal health. It highlights mechanisms linking gut dysbiosis to inflammation in conditions such as rheumatoid arthritis and osteoarthritis, suggesting microbiome modulation as a potential therapeutic strategy. Key findings include the microbiome’s role in bone metabolism through hormone regulation and production of short-chain fatty acids, crucial for mineral absorption. The review also considers the effects of diet, probiotics, and fecal microbiota transplantation on gut microbiome composition and their implications for orthopedic health. While promising, challenges in translating microbiome research into clinical practice persist, necessitating further exploration and ethical consideration of microbiome-based therapies. This interdisciplinary research aims to link digestive health with musculoskeletal integrity, offering new insights into the prevention and management of bone and joint diseases.
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Priya Dhanpal
- Department of General Medicine, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of General Medicine, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Lavanya Ragavanandam
- Department of Pharmacology, Faculty of Medicine - Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
165
|
Tiwari A, Ika Krisnawati D, Susilowati E, Mutalik C, Kuo TR. Next-Generation Probiotics and Chronic Diseases: A Review of Current Research and Future Directions. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27679-27700. [PMID: 39588716 DOI: 10.1021/acs.jafc.4c08702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The burgeoning field of microbiome research has profoundly reshaped our comprehension of human health, particularly highlighting the potential of probiotics and fecal microbiota transplantation (FMT) as therapeutic interventions. While the benefits of traditional probiotics are well-recognized, the efficacy and mechanisms remain ambiguous, and FMT's long-term effects are still being investigated. Recent advancements in high-throughput sequencing have identified gut microbes with significant health benefits, paving the way for next-generation probiotics (NGPs). These NGPs, engineered through synthetic biology and bioinformatics, are designed to address specific disease states with enhanced stability and viability. This review synthesizes current research on NGP stability, challenges in delivery, and their applications in preventing and treating chronic diseases such as diabetes, obesity, and cardiovascular diseases. We explore the physiological characteristics, safety profiles, and mechanisms of action of various NGP strains while also addressing the challenges and opportunities presented by their integration into clinical practice. The potential of NGPs to revolutionize microbiome-based therapies and improve clinical outcomes is immense, underscoring the need for further research to optimize their efficacy and ensure their safety.
Collapse
Affiliation(s)
- Ashutosh Tiwari
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Dyah Ika Krisnawati
- Department of Nursing, Faculty of Nursing and Midwifery, Universitas Nahdlatul Ulama Surabaya, Surabaya, 60237 East Java, Indonesia
| | - Erna Susilowati
- Akademi Kesehatan Dharma Husada Kediri, Kediri, 64118 East Java, Indonesia
| | - Chinmaya Mutalik
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsung-Rong Kuo
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
166
|
Moon H, Lee K, Ha JH, Kim NY, Shin HR, Cho TJ, Oh NS, Park J, Tang J, Kim JK, Kim M. Momoridica charantia and fermented Momoridica charantia with Leuconostoc mesenteroides MKSR change intestinal microbial diversity indices and compositions in high-fat and high-cholesterol diet-fed C57BL/6 male mice. Front Vet Sci 2024; 11:1496067. [PMID: 39742315 PMCID: PMC11686596 DOI: 10.3389/fvets.2024.1496067] [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: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction This study explores the impact of 4% Momordica charantia (MC) and 4% fermented Momordica charantia (FMC) on gut microbiota and obesity-related health outcomes in male C57BL/6 mice, a model relevant to veterinary sciences for understanding metabolic and gut health disorders in animals. Methods Mice were assigned to four dietary regimens, including control, high-fat and high-cholesterol diet (POS), POS with 4% MC, and POS with 4% FMC (fermented with Leuconostoc mesenteroides MKSR) over 12 weeks. Fecal samples were collected for 16S rRNA sequencing to evaluate microbial diversity and composition, key factors influencing animal health. Results Both MC and FMC groups exhibited significant alterations in gut microbial communities, with FMC inducing a distinct shift in beta diversity indices. Changes in microbial taxa such as Bacteroidetes, Verrucomicrobia, and Firmicutes were observed, along with enhancement in the 'L-glutamate and L-glutamine biosynthesis' pathway. These shifts were associated with reduced body weight gain and liver weights. Discussion The findings suggest that MC and FMC have potential benefits for managing diet-induced metabolic disorders and protecting against obesity by modulating gut microbiota and improving gut metabolism.
Collapse
Affiliation(s)
- Heewon Moon
- Department of Food Science and Nutrition, Dankook University, Cheonan, Republic of Korea
| | - Kangwook Lee
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan, Republic of Korea
| | - Na Yeun Kim
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Hyo Ri Shin
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Tae Jin Cho
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Nam Su Oh
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jingsi Tang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Development of Biology, Chinese Academy of Sciences, Beijing, China
| | - Jae Kyeom Kim
- Department of Food Biotechnology, Korea University, Sejong, Republic of Korea
| | - Misook Kim
- Department of Food Science and Nutrition, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
167
|
Gao Y, Zhang H, Zhu D, Guo L. Different artificial feeding strategies shape the diverse gut microbial communities and functions with the potential risk of pathogen transmission to captive Asian small-clawed otters ( Aonyx cinereus). mSystems 2024; 9:e0095424. [PMID: 39601555 DOI: 10.1128/msystems.00954-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Captive otters raised in zoos are fed different artificial diets, which may shape gut microbiota. The objective is to evaluate the impacts of two different artificial diets on microbial communities and function capabilities and short-chain fatty acid (SCFA) profiles in healthy otters' feces. A total of 16 Asian small-clawed otters in two groups (n = 8) were selected. Group A otters were fed raw loaches supplemented with commercial cat food (LSCF) diet, and group B otters were fed raw crucian diet. The communities and functional capabilities of microbiota in feces were assessed with metagenomic sequencing. Captive otters fed two kinds of diets possessed different gut microbial communities and functional capabilities. Various pathogenic bacteria, like Escherichia coli and Clostridium perfringens, were enriched in the samples from the two groups, respectively. Most of the differential pathways of nutrient metabolism were significantly enriched in group A, and the distributions of carbohydrate enzymes in the two groups significantly differed from each other. Multiple resistance genes markedly accumulated in fecal samples of the group A otters with LSCF diet. Higher concentrations of SCFAs were also observed in group A otters. Two feeding strategies were both likely to facilitate the colonization and expansion of various pathogenic bacteria and the accumulation of resistance genes in the intestines of captive otters, suggesting that risk of pathogen transmission existed in the current feeding process. Commercial cat food could supplement various nutrients and provide a substrate for the production of SCFAs, which might be beneficial for the otters' intestinal fermentation and metabolism. IMPORTANCE Captive otters fed with different diets possessed distinct gut microbial communities and functions, with the enrichment of several pathogens and multiple resistance genes in their gut microbiota. The current artificial feeding strategies had the possibility to accelerate the colonization and proliferation of various pathogenic bacteria in the intestines of otters and the spread of resistance genes, increasing the risk of diseases. In addition, supplementation with commercial cat food had benefits for otters' intestinal fermentation and the metabolism of gut microbiota.
Collapse
Affiliation(s)
- Yuanda Gao
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Hangyu Zhang
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Dapeng Zhu
- Foping National Nature Reserve, Hanzhong, China
| | - Long Guo
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
168
|
Wang Y, Li B, Xu B, Qin W. The Relationship Between Soil and Gut Microbiota Influences the Adaptive Strategies of Goitered Gazelles in the Qaidam Basin. Animals (Basel) 2024; 14:3621. [PMID: 39765525 PMCID: PMC11672517 DOI: 10.3390/ani14243621] [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: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiota is integral to the health and adaptability of wild herbivores. Interactions with soil microbiota can shape the composition and function of the gut microbiota, thereby influencing the hosts' adaptive strategies. As a result, soil microbiota plays a pivotal role in enabling wild herbivores to thrive in extreme environments. However, the influence of soil microbiota from distinct regions on host's gut microbiota has often been overlooked. We conducted the first comprehensive analysis of the composition and diversity of gut and soil microbiota in goitered gazelles across six regions in the Qaidam Basin, utilizing source tracking and ecological assembly process analyses. Significant differences were observed in the composition and diversity of soil and gut microbiota among the six groups. Source tracking analysis revealed that soil microbiota in the GangciGC (GC) group contributed the highest proportion to fecal microbiota (8.94%), while the Huaitoutala (HTTL) group contributed the lowest proportion (1.80%). The GC group also exhibited the lowest α-diversity in gut microbiota. The observed differences in gut microbial composition and diversity among goitered gazelles from six regions in the Qaidam Basin were closely tied to their adaptive strategies. Ecological assembly process analysis indicated that the gut microbiota were primarily influenced by stochastic processes, whereas deterministic processes dominated most soil microbial groups. Both the differences and commonalities in gut and soil microbiota play essential roles in enabling these gazelles to adapt to diverse environments. Notably, the utilization pattern of soil microbiota by gut microbiota did not align with regional trends in gut microbial α-diversity. This discrepancy may be attributed to variations in environmental pressures and the gut's filtering capacity, allowing gazelles to selectively acquire microbiota from soil to maintain homeostasis. This study highlights the significant regional variation in gut and soil microbiota diversity among goitered gazelle populations in the Qaidam Basin and underscores the critical role of soil-derived microbiota in their environmental adaptation.
Collapse
Affiliation(s)
- Yiran Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China;
- School of Ecological and Environmental Engineering, Qinghai University, Xining 810016, China
| | - Bin Li
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (B.L.); (B.X.)
| | - Bo Xu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China; (B.L.); (B.X.)
| | - Wen Qin
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China;
| |
Collapse
|
169
|
Veneruso I, Mennitti C, Gentile A, Di Bonito G, Ulisse J, Scarano C, Lombardo B, Terracciano D, Pero R, D'Alicandro G, Frisso G, D'Argenio V, Scudiero O. Effect of elite sport activity on salivary microbiota: The case of water polo. Heliyon 2024; 10:e40663. [PMID: 39654779 PMCID: PMC11626788 DOI: 10.1016/j.heliyon.2024.e40663] [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: 04/06/2024] [Revised: 10/30/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
It has been well established that the human gut microbiota plays a pivotal role in humans' health, since it is involved in nutrients' uptake, vitamins' synthesis, energy harvest, inflammatory modulation, and host immune responses. Moreover, gut microbiota alterations have been associated to an increasing number of diseases and its composition can be affected by several factors, including physical exercise. In particular, it has been reported that intense physical activity can induce metabolic changes which translate in alterations of specific biomarkers that can lead to the onset of infections, inflammation and hepatic or kidney disorders. Recently, the oral microbiota has shown its relevance not only for the health of oral cavity but also for human host's health, emerging as an ecological niche with a great potential for the study of gut microbiome alterations due also to its accessibility respect to other tracts that can be inferred through fecal samples analysis. Thus, the purpose of this study has been to assess the effect of intense physical activity, i.e., elite water polo, on the human salivary microbiota. Thirteen professional water polo players and nineteen sedentary controls were recruited for this study. The salivary microbiota analysis was performed in oral rinse collected from both controls and athletes three months after the beginning of the agonist season. Our results showed significant differences in the salivary microbiota between athletes and controls. In particular, three species, namely Oribacterium sinus, Oribacterium parvum and Oribacterium asaccharolyticum, were found to be significantly increased in the water polo players compared to controls. Even if these data have to be further validated, also to assess the role of these identified species, they strengthen the hypothesis that elite sports can influence and alter the status of the gut microbiota. Moreover, the saliva is confirmed as a suitable sample for microbiome evaluations that may improve athletes' status evaluation and monitoring.
Collapse
Affiliation(s)
- Iolanda Veneruso
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Alessandro Gentile
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Gennaro Di Bonito
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
| | - Jacopo Ulisse
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Carmela Scarano
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | - Raffaela Pero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100, Naples, Italy
| | - Giovanni D'Alicandro
- Department of Neuroscience and Rehabilitation, Center of Sports Medicine and Disability, AORN, Santobono-Pausillipon, 80122, Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
| | - Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100, Naples, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, via di Val Cannuta 247, 00166, Roma, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, via G. Salvatore 486, 80145, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80100, Naples, Italy
| |
Collapse
|
170
|
Mareș CR, Săsăran MO, Mărginean CO. Small Intestinal Bacterial Overgrowth and Childhood Malnutrition: A Comprehensive Review of Available Evidence. Nutrients 2024; 16:4319. [PMID: 39770940 PMCID: PMC11679674 DOI: 10.3390/nu16244319] [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: 11/10/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome is essential for children's normal growth and development, with its formation aligning closely with key stages of growth. Factors like birth method, feeding practices, and antibiotic exposure significantly shape the composition and functionality of the infant gut microbiome. Small intestinal bacterial overgrowth (SIBO) involves an abnormal increase in bacteria within the small intestine. This overgrowth can interfere with digestion, impair nutrient absorption, and lead to both local and systemic inflammation, potentially contributing to malnutrition. In this review, we provide a comprehensive overview of the current understanding of the relationship between SIBO and malnutrition, with a particular focus on the pediatric population. SIBO seems to play an important role in nutrient malabsorption through the gut microbiome imbalance, local inflammation, and disruption of the mucosal intestinal barrier. Additionally, SIBO is more prevalent in digestive disorders linked to malabsorption and malnutrition. Different therapeutic strategies for addressing malnutrition-related SIBO have been proposed. While antibiotics are the primary treatment for SIBO, their effectiveness in promoting weight gain among malnourished children remains uncertain. Hence, future research directed at the impact of microbiome imbalance on nutrient intake and absorption could bring to light new strategies for the effective prevention and treatment of malnutrition.
Collapse
Affiliation(s)
- Cristina Roxana Mareș
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| | - Maria Oana Săsăran
- Department of Pediatrics 3, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| |
Collapse
|
171
|
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
|
172
|
Phuong-Nguyen K, Mahmood M, Rivera L. Deleterious Effects of Yoyo Dieting and Resistant Starch on Gastrointestinal Morphology. Nutrients 2024; 16:4216. [PMID: 39683609 DOI: 10.3390/nu16234216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Obesity is associated with structural deterioration in the gut. Yoyo dieting, which refers to repeated phases of dieting and non-dieting periods leading to cyclic weight loss and regain, is a common occurrence in individuals with obesity. However, there is limited evidence on how gut structures are affected in yoyo dieting. There is good evidence suggesting that increased intake of resistant starch (RS) may be beneficial in promoting structural improvements in the gut. This investigation aimed to explore the effect of yoyo dieting on gastrointestinal structure and whether RS has beneficial effects in improving obesity-related gastrointestinal damage. METHOD In this study, male and female C57BL/6 mice were assigned to six different diets for 20 weeks: (1) control diet, (2) high fat diet (HF), (3) yoyo diet (alternating HF and control diets every 5 weeks), (4) control diet with RS, (5) HF with RS, and (6) yoyo diet with RS. Distal colon was collected for epithelial barrier integrity measurement. The small and large intestines were collected for histological assessment. RESULTS After 20 weeks, yoyo dieting resulted in increased colonic inflammation and exacerbated mucosal damage in comparison with continuous HF diet feeding. RS supplemented in HF and yoyo diets reduced mucosal damage in comparison to diets without RS. However, RS supplementation in a control diet significantly increased inflammation, crypt length, and goblet cell density. There were no significant differences in epithelial change and epithelial barrier integrity across diet groups. CONCLUSIONS This study suggests that yoyo dieting worsens gut damage, and incorporating high levels of RS may be detrimental in the absence of dietary challenge.
Collapse
Affiliation(s)
- Kate Phuong-Nguyen
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC 3220, Australia
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
| | - Malik Mahmood
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
| | - Leni Rivera
- School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC 3220, Australia
- School of Medicine, Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
173
|
Dias NW, Poole R, Soffa DR, Brown KJH. Dynamic principles of the microbiome and the bovine vagina: a review. Front Microbiol 2024; 15:1434498. [PMID: 39703711 PMCID: PMC11655496 DOI: 10.3389/fmicb.2024.1434498] [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: 05/17/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
The role of microbes inhabiting various body sites in supporting host physiology and health is substantial, and recent advancements in DNA sequencing technology have facilitated a more in-depth understanding of these microbial contributions. The influence of microbiota within a given organ can be broadly categorized as having two main functions: (1) promoting organ homeostasis and (2) creating conditions that inhibit the growth of pathogenic microorganisms, thereby protecting the host from diseases. In livestock production, numerous phenotypes critical to industry outcomes are affected by the microbiome, which has sparked considerable academic interest in recent years. This review aims to analyze the extensive data available on the microbiomes of humans and other mammalian species, examining microbiome ecology to elucidate principles that may assist in interpreting data on livestock microbiomes. Additionally, the review will discuss techniques available for investigating various microbiome aspects and will examine existing data on the reproductive microbiome, with a particular focus on the bovine vaginal microbiome.
Collapse
Affiliation(s)
- Nicholas Wege Dias
- Department of Animal Sciences & Industry, Kansas State University, Manhattan, KS, United States
| | - Rebecca Poole
- Department of Animal Science, Texas A and M University, College Station, TX, United States
| | - Dallas R. Soffa
- Department of Animal Science, Texas A and M University, College Station, TX, United States
| | | |
Collapse
|
174
|
Kurakawa T, Kani K, Chudan S, Nishikawa M, Tabuchi Y, Sakamoto K, Nagai Y, Ikushiro S, Furusawa Y. Rice Kefiran Ameliorates Obesity and Hepatic Steatosis Through the Change in Gut Microbiota. Microorganisms 2024; 12:2495. [PMID: 39770698 PMCID: PMC11728449 DOI: 10.3390/microorganisms12122495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity is a global epidemic and a significant risk factor for various diseases. Obesity and dysbiosis are associated, drawing attention to the mechanisms that regulate the gut microbiota. In this study, we focused on the postbiotic effects of rice kefiran (Kef), a functional product of Lactobacillus kefiranofaciens cultured in a rice-based medium, on obesity and its complications. Although Kef has the potential to improve obesity, the underlying mechanisms remain unknown. Therefore, we aimed to elucidate the mechanisms underlying changes in gut microbiota. The administration of Kef significantly suppressed diet-induced body weight gain, reduced liver fat accumulation, and modestly improved insulin resistance. Among the gut bacteria, Lachnospiraceae and Lachnoclostridium, which were positively correlated with obesity, decreased in mice administered Kef. In contrast, Bacteroides and Alistipes, both reported to ameliorate obesity, were increased. Consistent with the changes in the gut microbiota, Kef increased fecal acetate levels, which ameliorated obesity and hepatic steatosis. Predictive metagenomic analysis suggested that Kef administration increased the abundance of KEGG orthologs, associated with carbohydrate metabolism and improvements in insulin resistance. In conclusion, Kef improves diet-induced obesity, hepatic steatosis, and insulin resistance by regulating the gut microbiota's composition.
Collapse
Affiliation(s)
- Takuto Kurakawa
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Koudai Kani
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Seita Chudan
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Miyu Nishikawa
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Sugitani, Toyama 930-0194, Toyama, Japan
| | - Kazuichi Sakamoto
- College of Agriculture, Ibaraki University, 3-21-1. Chuo, Ami-cho, Ami-machi 300-0393, Ibaraki, Japan
| | - Yoshinori Nagai
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Shinichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu 939-0398, Toyama, Japan
| |
Collapse
|
175
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024; 22:791-805. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
176
|
Li X, Zhu R, Liu Q, Sun H, Sheng H, Zhu L. Effects of traditional Chinese medicine polysaccharides on chronic diseases by modulating gut microbiota: A review. Int J Biol Macromol 2024; 282:136691. [PMID: 39437951 DOI: 10.1016/j.ijbiomac.2024.136691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Intestinal tract is the largest immune system of human body. Gut microbiota (GM) can produce a large number of metabolites, such as short-chain fatty acids and bile acids, which regulate the physiological health of the host and affect the development of disease. In recent years, traditional Chinese medicine (TCM) polysaccharides have attracted extensive attention with multiple biological activities and low toxicity. TCM polysaccharides can promote the growth of intestinal beneficial bacteria and inhibit the growth of harmful bacteria by regulating the structure and function of GM, thus playing a crucial role in preventing or treating chronic diseases such as inflammatory bowel disease (IBD), obesity, type 2 diabetes mellitus (T2DM), liver diseases, cancer, etc. In this paper, the research progress of TCM polysaccharides in the treatment of chronic diseases such as inflammatory bowel disease, obesity, T2DM, liver diseases, cancer, etc. by modulating GM was reviewed. Meanwhile, this review makes an in-depth discussion on the shortcomings of the research of TCM polysaccharides on chronic diseases by modulating GM, and new valuable prospection for the future researches of TCM polysaccharides are proposed, which will provide new ideas for the further study of TCM polysaccharides.
Collapse
Affiliation(s)
- Xinyu Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Riran Zhu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Henglai Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
177
|
Jiang C, Zhan Q, Zeng C. The 5-HT-related gut-brain axis in obesity. Life Sci 2024; 358:123171. [PMID: 39447731 DOI: 10.1016/j.lfs.2024.123171] [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: 05/02/2024] [Revised: 08/22/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
AIMS The incidence of obesity increases annually. It is closely related to the occurrence of cardiovascular diseases, malignant tumors, etc., and has become a major global health problem. 5-hydroxytryptamine (5-HT), a multifunctional monoamine neurotransmitter, is dispersed throughout the central nervous system and digestive tract. It is intimately related to the mechanism of obesity. MATERIALS AND METHODS PubMed, Web of Science and Embase were carefully searched. We collected articles that are closely related to 5-HT, the gut-brain axis, and obesity. KEY FINGDINGS The gut microbiota not only influences nutrient metabolism but also centrally meditates appetite and mood regulation. The gut-brain axis, a system connecting the gut and the brain, is known to participate in two-way communication between the gut flora and the central nervous system. SIGNIFICANCE There have been few reports on whether peripheral and central 5-HT interact bidirectionally via the gut-brain axis and jointly play a role in the pathogenesis of obesity. In this review, we summarize the rationale for the contribution of the 5-HT-related gut-brain axis to the development of obesity and explore feasible signaling pathways, which elucidates new targets for preventing and treating obesity.
Collapse
Affiliation(s)
- Chaoyong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiong Zhan
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chang Zeng
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
178
|
Gilbert JA, Hartmann EM. The indoors microbiome and human health. Nat Rev Microbiol 2024; 22:742-755. [PMID: 39030408 DOI: 10.1038/s41579-024-01077-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/21/2024]
Abstract
Indoor environments serve as habitat for humans and are replete with various reservoirs and niches for microorganisms. Microorganisms enter indoor spaces with their human and non-human hosts, as well as via exchange with outdoor sources, such as ventilation and plumbing. Once inside, many microorganisms do not survive, especially on dry, barren surfaces. Even reduced, this microbial biomass has critical implications for the health of human occupants. As urbanization escalates, exploring the intersection of the indoor environment with the human microbiome and health is increasingly vital. The indoor microbiome, a complex ecosystem of microorganisms influenced by human activities and environmental factors, plays a pivotal role in modulating infectious diseases and fostering healthy immune development. Recent advancements in microbiome research shed light on this unique ecological system, highlighting the need for innovative approaches in creating health-promoting living spaces. In this Review, we explore the microbial ecology of built environments - places where humans spend most of their lives - and its implications for immune, endocrine and neurological health. We further propose strategies to harness the indoor microbiome for better health outcomes.
Collapse
Affiliation(s)
- Jack A Gilbert
- Department of Paediatrics, University of California San Diego, La Jolla, CA, USA.
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Erica M Hartmann
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Medicine, Division of Pulmonary Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
| |
Collapse
|
179
|
Pinanga YD, Pyo KH, Shin EA, Lee H, Lee EH, Kim W, Kim S, Kim JE, Kim S, Lee JW. Association between hepatocyte TM4SF5 expression and gut microbiome dysbiosis during non-alcoholic fatty liver disease development. Life Sci 2024; 358:123164. [PMID: 39454995 DOI: 10.1016/j.lfs.2024.123164] [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: 05/25/2024] [Revised: 07/24/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Gut microbiome dysbiosis is involved in non-alcoholic fatty liver disease (NAFLD) development. Hepatic transmembrane 4 L six family member 5 (TM4SF5) overexpression promotes NAFLD. However, how gut microbiota are associated with TM4SF5-mediated NAFLD remains unexplored. We analyzed the gut microbiome using feces from hepatocyte-specific TM4SF5-overexpressing transgenic (Alb-TGTm4sf5-Flag, TG) or Tm4sf5-/- knock-out (KO) mice fed a normal chow diet (NCD), high-fat diet (HFD) for 2 weeks (HFD2W), or methionine-choline-deficient diet (MCD) for 4 weeks to investigate associations among Tm4sf5 expression, diet, and the gut microbiome. TG-NCD mice showed a higher Firmicutes-to-Bacteroidetes (F/B) ratio, with less enrichment of Akkermansia muciniphila and Lactobacillus reuteri. NASH-related microbiomes in feces were more abundant in TG-HFD2w mice than in KO-HFD2w mice. Further, TG-MCD showed a higher F/B ratio than TG-NCD or KO mice, with decreases or increases in microbiomes beneficial or detrimental to the liver, respectively. Such effects in TG-MCD animals were correlated with functional pathways producing short-chain fatty acids (SCFAs). Furthermore, potential functional pathways of the gut microbiome were metabolically parallel to NAFLD features in TG-MCD mice. These results suggest that hepatocyte Tm4sf5 supports gut microbiome dysbiosis and metabolic activity, leading to SCFA production and hepatic inflammation during NAFLD development.
Collapse
Affiliation(s)
- Yangie Dwi Pinanga
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung-Hee Pyo
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Ae Shin
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Haesong Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Hae Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Wonsik Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Soyeon Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Eon Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Semi Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejon 34141, Republic of Korea
| | - Jung Weon Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
180
|
Palomba S, Costanzi F, Caserta D, Vitagliano A. Pharmacological and non-pharmacological interventions for improving endometrial receptivity in infertile patients with polycystic ovary syndrome: a comprehensive review of the available evidence. Reprod Biomed Online 2024; 49:104381. [PMID: 39454320 DOI: 10.1016/j.rbmo.2024.104381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 10/28/2024]
Abstract
Direct and indirect evidence suggests that endometrial receptivity may play a crucial role in the reduced fertility rate of women with polycystic ovary syndrome (PCOS). Various pharmacological and non-pharmacological strategies with potential effects on endometrial receptivity in patients with PCOS have been proposed. The aim of this study was to summarize the rationale and the clinical and experimental evidence of interventions tested for improving endometrial receptivity in infertile patients with PCOS. A systematic review was conducted by consulting electronic databases. All interventions with a potential influence on endometrial receptivity in infertile patients with PCOS were evaluated, and their main biological mechanisms were analysed. In total, 24 interventions related to endometrial receptivity were identified. Notwithstanding a strong biological rationale, no intervention aimed at improving endometrial receptivity in women with PCOS is supported by an adequate body of evidence, limiting their use in clinical practice. Further high-quality research is needed in this field to limit potentially ineffective and unsafe add-on treatments in infertile patients with PCOS.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Flavia Costanzi
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy; University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Donatella Caserta
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Amerigo Vitagliano
- Unit of Obstetrics and Gynaecology, Department of Interdisciplinary Medicine, University of Bari, Bari, Italy
| |
Collapse
|
181
|
Goo D, Lee J, Paneru D, Sharma MK, Rafieian-Naeini HR, Mahdavi FS, Gyawali I, Gudidoddi SR, Han G, Kim WK. Effects of branched-chain amino acid imbalance and dietary valine and isoleucine supplementation in modified corn-soybean meal diets with corn distillers dried grains with solubles on growth performance, carcass quality, intestinal health, and cecal microbiome in Cobb 500. Poult Sci 2024; 103:104483. [PMID: 39510006 PMCID: PMC11577229 DOI: 10.1016/j.psj.2024.104483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
One important feature of corn distillers dried grains with solubles (DDGS) is its high leucine:lysine ratio, which can inhibit chicken growth by causing branched-chain amino acid (BCAA) antagonism. The current study was conducted to investigate the effects of BCAA imbalance of inclusion of DDGS and whether additional dietary valine and isoleucine could alleviate the negative effects in broilers. A total of 640 0-d-old male Cobb 500 broilers were allocated into 4 treatments with 8 replicates and reared until d 42. The four different dietary groups were as follows: 1) control (CON) group (corn-soybean meal-based diet); 2) 30% DDGS (30D) group (replacing soybean meal with 30% DDGS); 3) 30D + additional valine and isoleucine (30DB) group; and 4) the group of 30DB + additional valine and isoleucine to provide the same leucine:valine and leucine:isoleucine ratios as the CON group (30DBB). The analyzed leucine:lysine ratios of the CON group were 1.36/1.41/1.46 (starter/grower/finisher phase), whereas the average leucine:lysine ratios of the 30% DDGS groups were 1.61/1.70/1.78 (starter/grower/finisher phase). The 30% DDGS groups (30D, 30DB, and 30DBB) negatively affected body weight (BW) from d 7 to 42 and BW gain (BWG), feed intake, carcass weight, breast muscle weight, and jejunal and ileal villus height:crypt depth during the overall period (d 0 to 42) (P < 0.05). Furthermore, the 30% DDGS groups significantly altered expression levels of jejunal tight junction proteins, breast muscle mechanistic target of rapamycin (mTOR) pathway-related genes, BCAA catabolism genes, and AA transporters compared to the CON (P < 0.01). The 30% DDGS groups showed differences in beta-diversity indices compared to the CON group (P < 0.05). The 30DBB group showing the lowest d 21 and 42 BW and overall BWG had the largest differences compared to the CON group in most measurements. In conclusion, excessive replacement of soybean meal with DDGS can significantly increase leucine levels, which may negatively affect chicken growth. Additionally, inappropriate ratios of valine and isoleucine can further decrease growth performance.
Collapse
Affiliation(s)
- Doyun Goo
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Jihwan Lee
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Deependra Paneru
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Milan K Sharma
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | | | - Fatemeh S Mahdavi
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Ishwari Gyawali
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | | | - Gippeum Han
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA, United States.
| |
Collapse
|
182
|
Cao Z, Wang X, Liu H, Yang Z, Zeng Z. Gut microbiota mediate the alleviation effect of Xiehuo-Guzheng granules on β cell dedifferentiation in type 2 diabetes mellitus. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156151. [PMID: 39437686 DOI: 10.1016/j.phymed.2024.156151] [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: 05/30/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a worldwide public health problem characterized by a progressive decline in β cell function. In traditional Chinese medicine (TCM) theory, 'fire' and 'healthy qi deficiency' are important pathogeneses of T2DM, and purging 'fire' and reinforcing the 'healthy qi' (Pinyin name: Xiehuo-Guzheng, XHGZ) are important method of treatment. Over the years, we have observed its benefit for diabetes. However, the underlying mechanisms remain unclear. PURPOSE To investigate the mechanism of XHGZ granules against β cell dedifferentiation in T2DM based on gut microbiota. METHODS Rats with T2DM, induced by intraperitoneal injection of streptozotocin after eight weeks of high-fat diet, were randomly allocated to receive XHGZ granules, metformin, or distilled water for eight consecutive weeks. Changes in metabolic parameters, β cell dedifferentiation, inflammatory cytokines, gut microbiota, and microbial metabolites (lipopolysaccharide (LPS) and short-chain fatty acids (SCFAs)), were detected. Furthermore, faecal microbiota transplantation (FMT) was performed to confirm the anti-diabetic effect of XHGZ granule-regulated gut microbiota in pseudo-germ-free T2DM rats. RESULTS XHGZ granules significantly ameliorated hyperglycaemia, improved islet function and pathology, and reduced β cell dedifferentiation and pro-inflammatory cytokines in T2DM rats. 16S rRNA sequencing revealed that XHGZ granules decreased the LPS-containing microbiota (e.g., Colidextribacter, Desulfovibrionaceae, and Morganella) and increased the SCFAs-producing bacteria (e.g., Prevotella, Alloprevotella, and Muribaculaceae) and Lactobacillus_intestinalis. Correspondingly, it strengthened intestinal barrier, lowered LPS, and elevated acetic and butyric acids. Tax4Fun analysis indicated that XHGZ granules restored abnormal metabolism, lipopolysaccharide biosynthesis, and pantothenate and CoA biosynthesis. Moreover, the XHGZ granule-regulated microbiota also exhibited the effects of anti-diabetes, anti-β cell dedifferentiation, and anti-inflammation along with the reduction of LPS and the increase of SCFAs in pseudo-germ-free T2DM rats. CONCLUSION Our results show that XHGZ granules alleviate β cell dedifferentiation via regulating gut microbiota and their metabolites in T2DM, suggesting its potential as a promising complementary treatment for T2DM. As far as we know, there are very few studies on the alleviation of β cell dedifferentiation by TCM, and investigations into the mechanism from the perspective of intestinal flora and microbial metabolites are yet to be reported.
Collapse
Affiliation(s)
- Zebiao Cao
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Postdoctoral Research Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Xianzhe Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huijun Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China; Huangshi Hospital of Traditional Chinese Medicine, Huangshi, Hubei 435000, China
| | - Zhaojun Yang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Zhili Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China; Postdoctoral Research Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
183
|
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
|
184
|
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
|
185
|
Song H, Lu J, Chu Q. Polyphenols from Prunus salicina L. alleviate weight gain, obesity-related hyperlipidemia, hepatic steatosis, hyperglycemia, and modulate gut microbiota in mice fed a high-fat diet. Nutr Res 2024; 132:152-163. [PMID: 39580918 DOI: 10.1016/j.nutres.2024.11.001] [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: 07/04/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
Hyperlipidemia, hepatic steatosis, and hyperglycemia are prevalent metabolic disorders closely linked to obesity. The objective of this research was to examine the potential advantageous impacts of polyphenols extracted from Prunus salicina L. fruit (PSFP) on hyperlipidemia, hepatic steatosis, and hyperglycemia induced by a high-fat diet (HFD), as well as to elucidate the underlying mechanisms involved. Male C57BL/6J mice, free from specific pathogens, were assigned randomly into three groups. These groups were then subjected to a 14-week dietary intervention, including a low-fat diet, an HFD, or an HFD plus with PSFP via intragastric administration. The obesity-related biochemical indexes were evaluated. To assess alterations in gut microbiota resulting from PSFP treatment, 16S rRNA sequencing was performed. UPLC-ESI-MS/MS assay identified 162 distinct polyphenolic compounds in PSFP. The administration of PSFP significantly reduced both body weight gain and hyperlipidemia induced by HFD. In addition, PSFP ameliorated hepatic steatosis induced by HFD and enhanced liver function in mice. PSFP treatment also ameliorated HFD-induced insulin resistance and hyperglycemia, evidenced by the observed decrease in fasting serum concentrations of glucose and insulin, improved insulin sensitivity, and restored glucose tolerance. Moreover, PSFP modulated the composition and abundance of specific microbial genus, including Lachnospiraceae NK4A136 group, Akkermansia, Parabacteroides, Enterococcus, Adlercreutzia, and Roseburia. Correlation analysis indicated significant associations between gut microbiota and physiological indices associated with obesity. These findings suggested that PSFP supplementation ameliorated HFD-induced hyperlipidemia, hepatic steatosis, and hyperglycemia, potentially through modulating the gut microbiota composition and abundance of specific taxa.
Collapse
Affiliation(s)
- Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, PR China.
| | - Jing Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, PR China
| | - Qiang Chu
- College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
186
|
Wang Y, Ran L, Zhang F, Li H, Cha Q, Yang K, Wang H, Wu Y, Yu Z. Growth hormone attenuates obesity and reshapes gut microbiota in high-fat diet-fed mice. Metabol Open 2024; 24:100326. [PMID: 39513179 PMCID: PMC11541824 DOI: 10.1016/j.metop.2024.100326] [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: 09/13/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Growth hormone (GH) and gut microbiota are key regulators of metabolism and have been linked to the development and treatment of obesity. Although variations in GH levels are associated with changes in gut microbiota composition, the specific effects of GH on gut microbiota and its role in obesity remain unclear. This study explored the effects of various GH doses (0.25, 0.75 and 1.5 IU/kg) on adipose tissue mass and gut microbiota in high-fat diet-induced obese mice. Notably, high-dose GH (1.5 IU/kg) significantly reduced the adipose tissue mass. This dose also reversed high-fat diet-induced gut microbiota dysbiosis, restoring microbial diversity and increasing the abundance of beneficial genera such as Ruminococcaceae and Muribaculaceae. Additionally, high-dose GH normalized several obesity-related gut microbiota pathways, including starch and sucrose metabolism, galactose metabolism, and secondary bile acid biosynthesis. GH therapy also improved intestinal barrier function, a key determinant of gut microbial homeostasis. These findings underscore the therapeutic potential of GH in obesity management through its effects on gut microbiota, providing new avenues for obesity interventions.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Liyuan Ran
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Fang Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Haolin Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Qianqian Cha
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Kun Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| | - Haoan Wang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Province Key Lab of Genetically Engineered Animal Models, Dalian Medical University, Dalian, 116044, China
| | - Yingjie Wu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Province Key Lab of Genetically Engineered Animal Models, Dalian Medical University, Dalian, 116044, China
| | - Zichao Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250021, China
| |
Collapse
|
187
|
Yang Y, Shu X, Javed HU, Wu Q, Liu H, Han J, Zhou H. Dietary supplementation of poly-dihydromyricetin-fused zinc nanoparticles alleviates fatty liver hemorrhagic syndrome by improving antioxidant capacity, intestinal health and lipid metabolism of laying hens. Poult Sci 2024; 103:104301. [PMID: 39306955 PMCID: PMC11447411 DOI: 10.1016/j.psj.2024.104301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
Fatty liver hemorrhagic syndrome is the main cause of noninfectious death of laying hens and results in substantial economic losses to the poultry industry. This study focused on evaluating the effects of Poly-dihydromyricetin-fused zinc nanoparticles (PDMY-Zn NPs) on antioxidant capacity, liver lipid metabolism, and intestinal health in laying hens. A total of 288 Jingfen laying hens (52 wk old) with similar body weights were randomly divided into 4 dietary groups with 6 replicates in each group for 8 wk. The control group received a basal diet, while the treatment groups were supplemented with PDMY-Zn NPs at levels of 200, 400, and 600 mg/kg, respectively. The results indicate that PDMY-Zn NPs supplementation can enhance antioxidant parameters (P < 0.05) in the blood and liver of laying hens. Simultaneously, it can mitigate vacuolar degeneration and inflammatory necrosis in hepatocytes, improve the relative expression level of related parameters associated with liver lipid metabolism and key regulatory genes (P < 0.05). Furthermore, it has been observed to reshape the composition and diversity of cecum microbes by increasing beneficial probiotics such as Lactobacillus and Prevotella, while also enhancing villi height and villi/crypt ratio in the duodenum and ileum (P < 0.05). Additionally, it elevates liver bile acid content along with the relative expression of key genes involved in liver synthesis (P < 0.05). In summary, PDMY-Zn NPs showed potential to alleviate fatty liver hemorrhagic syndrome by enhancing antioxidant capacity, regulating liver lipid metabolism, and maintaining intestinal health.
Collapse
Affiliation(s)
- Yuanting Yang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Xugang Shu
- College of Chemistry and Chemical Engineering, Zhongkai University of Agricultural Engineering, Guangzhou 510225, China
| | - Hafiz Umer Javed
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, College of Food Engineering, Beibu Gulf University, Qinzhou 535011, China
| | - Qun Wu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Hu Liu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Jiancheng Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China.
| |
Collapse
|
188
|
Zha A, Qi M, Deng Y, Li H, Wang N, Wang C, Liao S, Wan D, Xiong X, Liao P, Wang J, Yin Y, Tan B. Gut Bifidobacterium pseudocatenulatum protects against fat deposition by enhancing secondary bile acid biosynthesis. IMETA 2024; 3:e261. [PMID: 39742294 PMCID: PMC11683477 DOI: 10.1002/imt2.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
Gut microbiome is crucial for lipid metabolism in humans and animals. However, how specific gut microbiota and their associated metabolites impact fat deposition remains unclear. In this study, we demonstrated that the colonic microbiome of lean and obese pigs differentially contributes to fat deposition, as evidenced by colonic microbiota transplantation experiments. Notably, the higher abundance of Bifidobacterium pseudocatenulatum was significantly associated with lower backfat thickness in lean pigs. Microbial-derived lithocholic acid (LCA) species were also significantly enriched in lean pigs and positively correlated with the abundance of B. pseudocatenulatum. In a high-fat diet (HFD)-fed mice model, administration of live B. pseudocatenulatum decreased fat deposition and enhances colonic secondary bile acid biosynthesis. Importantly, pharmacological inhibition of the bile salt hydrolase (BSH), which mediates secondary bile acid biosynthesis, impaired the anti-fat deposition effect of B. pseudocatenulatum in antibiotic-pretreated, HFD-fed mice. Furthermore, dietary LCA also decreased fat deposition in HFD-fed rats and obese pig models. These findings provide mechanistic insights into the anti-fat deposition role of B. pseudocatenulatum and identify BSH as a potential target for preventing excessive fat deposition in humans and animals.
Collapse
Affiliation(s)
- Andong Zha
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- School of Basic Medical Science, Central South UniversityChangshaChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Yuankun Deng
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Hao Li
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Nan Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Chengming Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Simeng Liao
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Xia Xiong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Peng Liao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Jing Wang
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| | - Yulong Yin
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro‐Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of SciencesChangshaChina
| | - Bi'e Tan
- Key Laboratory of Hunan Province for the Products Quality Regulation of Livestock and PoultryCollege of Animal Science and Technology, Hunan Agricultural UniversityChangshaChina
- Yuelushan LaboratoryHunanChina
| |
Collapse
|
189
|
Xiong S, Zhang K, Wang J, Bai S, Zeng Q, Liu Y, Peng H, Xuan Y, Mu Y, Tang X, Ding X. Effects of xylo-oligosaccharide supplementation on the production performance, intestinal morphology, cecal short-chain fatty acid levels, and gut microbiota of laying hens. Poult Sci 2024; 103:104371. [PMID: 39405830 PMCID: PMC11525217 DOI: 10.1016/j.psj.2024.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024] Open
Abstract
This study investigated the effects of xylo-oligosaccharide supplementation on the production performance, intestinal morphology, cecal short-chain fatty acid levels, and gut microbiota of laying hens. A total of 800 Lohmann pink layers, each 48 wk old, were randomly divided into 5 dietary treatment groups, namely XOS at 0 (CON), 100 (XOS1), 200 (XOS2), 300 (XOS3) and 400 (X0S4) mg/kg. The experimental period was 24 wk. The results revealed that the egg production rate and the number of eggs laid by each layer between 1 to 12 wk increased as the XOS concentration increased (Plinear < 0.05). The sand-shell egg percentage decreased significantly from 1 to 12 wk in the XOS1, XOS2, and XOS3 groups (PANOVA < 0.05). Compared with the CON group, the 4 XOS dosage groups presented significant increases in the villus height and the ratio of villus height to crypt depth in the jejunum (PANOVA < 0.05), whereas a linear decrease in jejunal crypt depth (Plinear < 0.05) was noted. In addition, XOS supplementation significantly increased the concentrations of butyric acid and isovaleric acid in the caeca (PANOVA < 0.05). High-throughput sequencing analysis of bacterial 16S rRNA revealed that dietary XOS supplementation influenced the cecal microbiota. The alpha diversity analysis indicated that the richness of cecal bacteria was greater in the laying hens fed XOS. The modulation of the cecal microbiota composition upon the addition of XOS was characterized by an increased abundance of Firmicutes and Bifidobacteriaceae, and decreased abundance of Bacteroidetes. At the genus level, dietary XOS supplementation resulted in decreases in the abundances of Bacteroides and Rikenellaceae_RC9_gut_group and an increase in the abundance of Lactobacillus. In conclusion, dietary XOS supplementation improved the production performance of laying hens by increasing the production of short-chain fatty acids and improving their intestinal morphology, which was achieved mainly through changes in the composition of the intestinal microbiota. The recommended level of XOS in the diet of laying hens is 200 mg/kg.
Collapse
Affiliation(s)
- Siyu Xiong
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Keying Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Shiping Bai
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Qiufeng Zeng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yan Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Huanwei Peng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yue Xuan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yadong Mu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Xiaobing Tang
- Yibin Yatai Biotechnology Co., Ltd, Yibin 644000, China
| | - Xuemei Ding
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
190
|
Deng X, Cheng L, Qiao Y, Liu X, Zhou Y, Liu H, Wang L. Rutin ameliorates HCD-induced cholesterol metabolism disorder in zebrafish larvae revealed by transcriptome and metabolome analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156058. [PMID: 39341124 DOI: 10.1016/j.phymed.2024.156058] [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: 01/18/2024] [Revised: 08/26/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024]
Abstract
Changes in modern lifestyles have led to an increase in obesity rates. Excessive lipid accumulation leads to abnormal cholesterol metabolism, and maintaining a balanced cholesterol metabolism is essential for the normal functioning of cells and the body. Rutin belongs to the group of flavonoids with hypolipidemic, anti-inflammatory and antioxidant effects. The aim of this study was to investigate the role of rutin in cholesterol metabolism disorders induced by a high cholesterol diet in zebrafish larvae. The trial was divided into five groups: Normal diet (ND), 5 % high cholesterol diet (HCD), 5 % high cholesterol diet with 80 μg/g ezetimibe diet (EZE), 5 % high cholesterol diet with 5 % rutin diet (RL-HCD), and 5 % high cholesterol diet with 10 % rutin diet (RH-HCD). Zebrafish larvae at 5 dpf were randomly divided into five groups and continuously fed different diets for 10 days, after 10 days zebrafish samples were collected for subsequent experiments. Body length, body width, oil red O, and Nile red staining were measured to detect biochemical indexes, analyze inflammatory response and lipid accumulation. Vascular endothelial injury was assessed by stereofluorescence microscopy and ELISA. In order to study the protective effect of rutin in zebrafish with cholesterol metabolism disorder induced by HCD, RNA-seq and LC-MS/MS nontargeted metabolomics were employed. The results indicate that HCD led to an increase in the body length and width of zebrafish. The HCD group induced an increase in body length and width, lipid accumulation, and exacerbated inflammation. Additionally, vascular damage and abnormal expression of endothelial cell markers were observed. Rutin lowered lipid levels in zebrafish fed an HCD, reduced inflammation, and protected endothelial cells. The RNA-seq and metabolomic analysis combined demonstrated that rutin effectively ameliorates the disorder of cholesterol metabolism in vivo by reducing cholesterol synthesis and promoting cholesterol transport.
Collapse
Affiliation(s)
- Xinxin Deng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Lin Cheng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Ying Qiao
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Xuan Liu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Yongbing Zhou
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Hui Liu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China.
| | - Li Wang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China.
| |
Collapse
|
191
|
Zhang T, Liu J, Liu X, Wang Q, Zhang H. The causal impact of gut microbiota on circulating adipokine concentrations: a two-sample Mendelian randomization study. Hormones (Athens) 2024; 23:789-799. [PMID: 38564143 DOI: 10.1007/s42000-024-00553-y] [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: 01/04/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Evidence from previous experimental and observational research demonstrates that the gut microbiota is related to circulating adipokine concentrations. Nevertheless, the debate as to whether gut microbiome composition causally influences circulating adipokine concentrations remains unresolved. This study aimed to take an essential step in elucidating this issue. METHODS We used two-sample Mendelian randomization (MR) to causally analyze genetic variation statistics for gut microbiota and four adipokines (including adiponectin, leptin, soluble leptin receptor [sOB-R], and plasminogen activator inhibitor-1 [PAI-1]) from large-scale genome-wide association studies (GWAS) datasets. A range of sensitivity analyses was also conducted to assess the stability and reliability of the results. RESULTS The composite results of the MR and sensitivity analyses revealed 22 significant causal associations. In particular, there is a suggestive causality between the family Clostridiaceae1 (IVW: β = 0.063, P = 0.034), the genus Butyrivibrio (IVW: β = 0.029, P = 0.031), and the family Alcaligenaceae (IVW: β=-0.070, P = 0.014) and adiponectin. Stronger causal effects with leptin were found for the genus Enterorhabdus (IVW: β=-0.073, P = 0.038) and the genus Lachnospiraceae (NK4A136 group) (IVW: β=-0.076, P = 0.01). Eight candidate bacterial groups were found to be associated with sOB-R, with the phylum Firmicutes (IVW: β = 0.235, P = 0.03) and the order Clostridiales (IVW: β = 0.267, P = 0.028) being of more interest. In addition, the genus Roseburia (IVW: β = 0.953, P = 0.022) and the order Lactobacillales (IVW: β=-0.806, P = 0.042) were suggestive of an association with PAI-1. CONCLUSION This study reveals a causal relationship between the gut microbiota and circulating adipokines and may help to offer novel insights into the prevention of abnormal concentrations of circulating adipokines and obesity-related diseases.
Collapse
Affiliation(s)
- Tongxin Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Jingyu Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xiao Liu
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Qian Wang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
192
|
Hewady S, Manuel CR, Pasquali C, Koya J, Reznik SE. OM-85 attenuates high-fat diet-induced obesity, insulin resistance, gut dysbiosis and nonalcoholic steatohepatitis in a murine model. Biomed Pharmacother 2024; 181:117710. [PMID: 39626377 DOI: 10.1016/j.biopha.2024.117710] [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: 08/25/2024] [Revised: 11/04/2024] [Accepted: 11/23/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Obesity is a global epidemic that is tied to a wide range of human disorders. Chronic consumption of a high-fat diet is linked to disruption of the intestinal microbiome, which drives obesity-related pathophysiology. Broncho-Vaxom® (OM-85), a bacterial lysate used for prophylaxis of recurrent respiratory tract infections, has both immunostimulatory and immunomodulatory functions. METHODS Male C57Bl/6 mice were maintained on normal control vs. high-fat diets for 8 weeks and treated or untreated with OM-85 or with the probiotic Lactobacillus plantarum, as a positive control. Mice were evaluated for weight gain, glucose tolerance, insulin tolerance, gut microbiome composition and non-alcoholic steatohepatitis (NASH). RESULTS High-fat diet mice developed obesity, insulin resistance, NASH and gut dysbiosis with a shift from the Bacteroidetes phylum, such as Bacteroidales order and Muribaculaceae family organisms to Firmicutes groups, such as the Clostridium and Blautia genuses. Treatment with OM-85 led to 1) prevention of obesity, 2) prevention of insulin resistance, 3) attenuation of NASH and 4) attenuation of gut dysbiosis, with decreased levels of the organisms mentioned above and increases in Verrucomicrobiae phylum organisms such as Akkermansia family microbes as well as Muribaculaceae organisms. These shifts in the gut microbiome predict favorable effects on the short chain fatty acid profile in the gut and increased integrity of the intestinal barrier. Pathway analysis showed that OM-85 decreases rates of carbohydrate metabolism, providing an additional mechanism whereby OM-85 prevents obesity. CONCLUSION Immune modulators such as OM-85 should be investigated for their potential therapeutic effects on metabolism.
Collapse
Affiliation(s)
- Sarah Hewady
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | | | | | | | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA; Departments of Pathology and Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
193
|
Härer A, Thompson KA, Schluter D, Rennison DJ. Associations Between Gut Microbiota Diversity and a Host Fitness Proxy in a Naturalistic Experiment Using Threespine Stickleback Fish. Mol Ecol 2024; 33:e17571. [PMID: 39466622 PMCID: PMC11589663 DOI: 10.1111/mec.17571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
The vertebrate gut microbiota is a critical determinant of organismal function, yet whether and how gut microbial communities affect host fitness under natural conditions remains largely unclear. We characterised associations between a fitness proxy-individual growth rate-and bacterial gut microbiota diversity and composition in threespine stickleback fish introduced to large semi-natural ponds. We detected a 63% higher richness of bacterial taxa (α-diversity) in the guts of high-fitness fish compared to low-fitness fish, which might be driven by stronger bacterial dispersal among high-fitness fish according to the fit of a neutral community model. Further, microbial communities of high-fitness fish were more similar to one another (i.e., exhibited lower β-diversity) than those of low-fitness fish. The lower β-diversity found to be associated with higher host fitness is consistent with the Anna Karenina principle-that there are fewer ways to have a functional microbiota than a dysfunctional microbiota. Our study links differences in α- and β-diversity to a fitness-related trait in a vertebrate species reared under naturalistic conditions and our findings provide a basis for functional tests of the fitness consequences of host-microbiota interactions.
Collapse
Affiliation(s)
- Andreas Härer
- Department of Ecology, Behavior, & Evolution, School of Biological SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Ken A. Thompson
- Department of Zoology, Biodiversity Research CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of BiologyStanford UniversityStanfordCaliforniaUSA
- Department of Plant BiologyCarnegie Institution for ScienceStanfordCaliforniaUSA
| | - Dolph Schluter
- Department of Zoology, Biodiversity Research CentreUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Diana J. Rennison
- Department of Ecology, Behavior, & Evolution, School of Biological SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
194
|
Du L, Ding X, Tian Y, Chen J, Li W. Effect of anthocyanins on metabolic syndrome through interacting with gut microbiota. Pharmacol Res 2024; 210:107511. [PMID: 39577753 DOI: 10.1016/j.phrs.2024.107511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
Metabolic syndrome, as a complex pathological condition, is caused by a series of pathogenic factors and has become a global public health challenge. Anthocyanins, a natural water-soluble flavonoid pigment, have attracted much attention due to their antioxidant, anti-inflammatory, and anticancer biological activities. After ingestion, a majority of anthocyanins is not directly absorbed but rather reaches the colon. Hence, the exertion of their biological benefits is closely intertwined with the role played by gut microbiota. In this review, we introduce the pathogenesis and intervention methods of metabolic syndrome, as well as the interaction between anthocyanins and gut microbiota. We also discuss the therapeutic potential of anthocyanins through gut microbiota in addressing a range of metabolic syndrome conditions, including obesity, type 2 diabetes mellitus, cardiovascular diseases, non-alcoholic fatty liver disease, inflammatory bowel disease, polycystic ovary syndrome, osteoporosis, and cancer.
Collapse
Affiliation(s)
- Lanlan Du
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yuwen Tian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Weilin Li
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
195
|
Lee JY, Kim Y, Kim J, Kim JK. Fecal Microbiota Transplantation: Indications, Methods, and Challenges. J Microbiol 2024; 62:1057-1074. [PMID: 39557804 DOI: 10.1007/s12275-024-00184-3] [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: 07/09/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
Over the past two decades, as the importance of gut microbiota to human health has become widely known, attempts have been made to treat diseases by correcting dysbiosis of gut microbiota through fecal microbiota transplantation (FMT). Apart from current knowledge of gut microbiota, FMT to treat disease has a long history, from the treatment of food poisoning in the fourth century to the treatment of Clostridioides difficile infections in the twentieth century. In 2013, FMT was recognized as a standard treatment for recurrent C. difficile because it consistently showed high efficacy. Though recurrent C. difficile is the only disease internationally recognized for FMT efficacy, FMT has been tested for other diseases and shown some promising preliminary results. Different FMT methods have been developed using various formulations and administration routes. Despite advances in FMT, some issues remain to be resolved, such as donor screening, manufacturing protocols, and unknown components in the fecal microbiota. In this review, we discuss the mechanisms, clinical indications, methods, and challenges of current FMT. We also discuss the development of alternative therapies to overcome the challenges of FMT.
Collapse
Affiliation(s)
- Jee Young Lee
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Yehwon Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyoun Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyeun Kate Kim
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea.
| |
Collapse
|
196
|
Bhatnagar K, Jha K, Dalal N, Patki N, Gupta G, Kumar A, Kumar A, Chaudhary S. Exploring micronutrients and microbiome synergy: pioneering new paths in cancer therapy. Front Immunol 2024; 15:1442788. [PMID: 39676876 PMCID: PMC11638209 DOI: 10.3389/fimmu.2024.1442788] [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: 06/02/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The human microbiome is the complex ecosystem consisting of trillions of microorganisms that play a key role in developing the immune system and nutrient metabolism. Alterations in the gut microbiome have been linked to cancer initiation, progression, metastasis, and response to treatment. Accumulating evidence suggests that levels of vitamins and minerals influence the gut environment and may have implications for cancer risk and progression. Bifidobacterium has been reported to reduce the colorectal cancer risk by binding to free iron. Additionally, zinc ions have been shown to activate the immune cells and enhance the effectiveness of immunotherapy. Higher selenium levels have been associated with a reduced risk of several cancers, including colorectal cancer. In contrast, enhanced copper uptake has been implicated in promoting cancer progression, including colon cancer. The interaction between cancer and gut bacteria, as well as dysbiosis impact has been studied in animal models. The interplay between prebiotics, probiotics, synbiotics, postbiotics and gut bacteria in cancer offers the diverse physiological benefits. We also explored the particular probiotic formulations like VSL#3, Prohep, Lactobacillus rhamnosus GG (LGG), etc., for their ability to modulate immune responses and reduce tumor burden in preclinical models. Targeting the gut microbiome through antibiotics, bacteriophage, microbiome transplantation-based therapies will offer a new perspective in cancer research. Hence, to understand this interplay, we outline the importance of micronutrients with an emphasis on the immunomodulatory function of the microbiome and highlight the microbiome's potential as a target for precision medicine in cancer treatment.
Collapse
Affiliation(s)
- Kartik Bhatnagar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Kanupriya Jha
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ninad Patki
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Garima Gupta
- Biological Engineering and Sciences, Indian Institute of Technology Gandhinagar Palaj, Gandhinagar, Gujarat, India
| | - Amit Kumar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Sarika Chaudhary
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
197
|
Liu X, Sun C, Zhou Q, Zheng X, Jiang S, Wang A, Han Y, Xu G, Liu B. Ferulic Acid Relieves the Oxidative Stress Induced by Oxidized Fish Oil in Oriental River Prawn ( Macrobrachium nipponense) with an Emphasis on Lipid Metabolism and Gut Microbiota. Antioxidants (Basel) 2024; 13:1463. [PMID: 39765792 PMCID: PMC11672775 DOI: 10.3390/antiox13121463] [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: 10/07/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
To investigate the potential of ferulic acid (FA) in attenuating the deleterious effects of oxidized fish oil (OF) on Macrobrachium nipponense, four experimental diets were formulated: 3% fresh fish oil (CT group, peroxide value: 2.2 mmol/kg), 3% oxidized fish oil (OF group, peroxide value: 318 mmol/kg), and 3% OF with an additional 160 and 320 mg/kg of FA (OF+FA160 group and OF+FA320 group, respectively). M. nipponense (initial weight: 0.140 ± 0.015 g) were randomly divided into four groups with six replicates (60 individuals per replicate) and reared for a period of 10 weeks. The results showed that the OF treatments significantly reduced the growth performance, the expression of antioxidant genes in the hepatopancreas, the levels of low-density lipoprotein cholesterol, and the gene expression levels of ACC, FAS, FABP10, ACBP, G6PDH, and SCD in the hepatopancreas (p < 0.05). OF supplementation significantly increased the levels of high-density lipoprotein cholesterol in hemolymph and the gene expression levels of CPT1 (p < 0.05). Addition of FA to the OF group significantly increased total bile acids (p < 0.05). In addition, it was found by Oil Red staining that the proportion of lipid droplets was significantly increased in the OF group (p < 0.05). However, the lipid droplets were alleviated by FA supplementation in the diet. OF was found to significantly reduce the diversity of intestinal microbiota by 16S rDNA sequencing and significantly increase the Firmicutes/Bacteroidetes (F/B) ratio (p < 0.05). Functional analysis of gut microbiota also showed that OF reduced lipolysis and led to fat deposition, which is related to gut microbiota. However, this study found that the composition of the gut microbiome of M. nipponense was changed by the addition of FA in the diet, including an increase in the abundance of Ruminococcaceae UCG-005 and Lachnospiraceae, a reduction in the F/B ratio, and an improvement in lipid metabolism. In conclusion, the OF induced oxidative stress, disturbed the balance of intestinal microbiota, promoted lipid accumulation, and caused disorders of lipid metabolism in M. nipponense by increasing lipid synthesis and reducing β-oxidation. However, the results of this study highlighted the potential of FA supplementation to modulate intestinal microbial composition, promote bile acid production, and activate genes related to lipid metabolism in the hepatopancreas, ultimately leading to a reduction in lipid deposition in M. nipponense.
Collapse
Affiliation(s)
- Xin Liu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
| | - Cunxin Sun
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Qunlan Zhou
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Xiaochuan Zheng
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Sufei Jiang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Aimin Wang
- Yancheng Academy of Fishery Science, Yancheng 224051, China; zam--
| | - Yongquan Han
- Guangzhou Cohoo Biotechnology Co., Ltd., Guangzhou 510663, China;
| | - Gangchun Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Bo Liu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (X.L.); (C.S.); (Q.Z.); (X.Z.); (S.J.)
- Key Laboratory of Aquatic Animal Nutrition and Health, Freshwater Fisheries Research Center, Chinese Academy of Fishery Science, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| |
Collapse
|
198
|
Esfandiari F, Bakhshi B, Shahbazi T, Derakhshan-nezhad E, Bahroudi M, Minaeeian S, Boustanshenas M, Alborzi F, Behboudi B, Fazeli MS. Significant difference in gut microbiota Bifidobacterium species but not Lactobacillus species in colorectal cancer patients in comparison with healthy volunteers using quantitative real-time PCR. PLoS One 2024; 19:e0294053. [PMID: 39602380 PMCID: PMC11602092 DOI: 10.1371/journal.pone.0294053] [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: 01/13/2023] [Accepted: 10/25/2023] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC), with a growing incidence trend, is one of the most diagnosed cancers and the second cause of cancer-related deaths worldwide. The literature has frequently focused attention on the correlation between the gut microbiota imbalance and CRC. The genera Lactobacillus and Bifidobacterium have recently received increasing attention because of their potential in restoring alterations in the gut microflora. Therefore, this study aimed to quantitatively evaluate the presence of lactobacilli and bifidobacterial strains in the fecal samples of CRC patients compared to healthy volunteers. METHODS From 2018 to 2019, 25 confirmed CRC patients and 25 age- and gender-matched control subjects were enrolled in the study. Bacterial DNA was extracted from the fecal samples and the presence of lactobacilli and bifidobacterial strains were quantitatively determined using quantitative real-time PCR using genus-specific 16S rDNA primers. RESULTS A significant decline in the abundance of bifidobacteria in CRC patients compared to healthy individuals (p value<0.003) was observed; however, no significant difference was observed between the two groups regarding the abundance of lactobacilli (p value<0.163). Correlation analysis showed a positive association between the lack of genetic history of CRC and the numbers of gut bifidobacteria and lactobacilli. CONCLUSION As a putative gut probiotic, depletion of bifidobacteria showed significant correlation to the development and progression of CRC; therefore, therapeutic use of these probiotic bacteria could be considered a possible adjuvant approach in disease management through modulation of the microbiota.
Collapse
Affiliation(s)
- Fahime Esfandiari
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tayebe Shahbazi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mahboube Bahroudi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Minaeeian
- Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Boustanshenas
- Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Forough Alborzi
- Division of Gastroenterology, Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Behboudi
- Division of Colon and Rectal Surgery, Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Sadegh Fazeli
- Division of Colon and Rectal Surgery, Department of Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
199
|
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
|
200
|
Mallick R, Basak S, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Roles of the gut microbiota in human neurodevelopment and adult brain disorders. Front Neurosci 2024; 18:1446700. [PMID: 39659882 PMCID: PMC11628544 DOI: 10.3389/fnins.2024.1446700] [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: 06/15/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Growing evidence demonstrates the connection between gut microbiota, neurodevelopment, and adult brain function. Microbial colonization occurs before the maturation of neural systems and its association with brain development. The early microbiome interactions with the gut-brain axis evolved to stimulate cognitive activities. Gut dysbiosis can lead to impaired brain development, growth, and function. Docosahexaenoic acid (DHA) is critically required for brain structure and function, modulates gut microbiota, and impacts brain activity. This review explores how gut microbiota influences early brain development and adult functions, encompassing the modulation of neurotransmitter activity, neuroinflammation, and blood-brain barrier integrity. In addition, it highlights processes of how the gut microbiome affects fetal neurodevelopment and discusses adult brain disorders.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ranjit K. Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Queretaro, Mexico
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|