1
|
Zhong Y, Zhang M, Xu H, Yu X, Hu Y, Xu Y, Xiao X, Yang C. Bacillus licheniformis Alleviates Clostridium perfringens-Induced Intestinal Injury in Mice Model by Modulating Inflammation, Apoptosis, and Cecal Microbial-Metabolic Responses. Animals (Basel) 2025; 15:1409. [PMID: 40427288 DOI: 10.3390/ani15101409] [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: 04/18/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Bacillus licheniformis (B. licheniformis) is a probiotic known for its ability to enhance host resistance against pathogenic infections. This study aimed to evaluate the protective effects and underlying mechanisms of B. licheniformis in a mouse model challenged with Clostridium perfringens (C. perfringens). C57BL/6J mice were pretreated with B. licheniformis for 21 days before oral infection with C. perfringens. The probiotic administration significantly prevented infection-induced weight loss and immune organ enlargement. Serum cytokine analysis revealed that B. licheniformis increased anti-inflammatory IL-4 and IL-10 levels while reducing pro-inflammatory IL-1β, IL-6, and TNF-α levels. Histological analysis showed that B. licheniformis preserved intestinal morphology and inhibited epithelial cell apoptosis. Moreover, the probiotic mitigated the infection-induced decline in volatile fatty acid (VFA) production. 16S rRNA gene sequencing revealed that B. licheniformis reshaped the cecal microbiota, characterized by the increased abundance of Lachnospiraceae_NK4A136_group, Muribaculaceae, and Parabacteroides, and reduced abundance of Alistipes. Untargeted metabolomic profiling identified differential metabolites-including D-glucono-1,5-lactone, D-erythrose 4-phosphate, and D-sedoheptulose 7-phosphate-enriched in the pentose phosphate pathway, suggesting a regulatory role in redox homeostasis and host response. Collectively, these results indicate that B. licheniformis exerts protective effects against C. perfringens infection by modulating inflammation, apoptosis, microbial composition, and metabolic pathways. This work provides new insights into the application of B. licheniformis as a functional microbial feed additive in livestock disease prevention.
Collapse
Affiliation(s)
- Yifan Zhong
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Meiting Zhang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Haocheng Xu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Xiaorong Yu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Yashi Hu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Yangyi Xu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Xiao Xiao
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| | - Caimei Yang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China
| |
Collapse
|
2
|
Shen Y, Wang J, Li Y, Kang X, Gu L. Intestinal injury and changes of the gut microbiota after ischemic stroke. Front Cell Neurosci 2025; 19:1557746. [PMID: 40313590 PMCID: PMC12043883 DOI: 10.3389/fncel.2025.1557746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025] Open
Abstract
Stroke is the second leading cause of death and the third leading cause of disability worldwide, with ischemic stroke (IS) accounting for the vast majority of cases. This paper reviews the latest research on intestinal damage, changes in the gut microbiota, and related therapeutic strategies after IS. Following IS, the integrity of the intestinal mucosal barrier is compromised, leading to increased intestinal permeability. The gut microbiota can translocate to other organs, triggering systemic immune responses that inhibit recovery after IS. Moreover, the composition and proportion of the gut microbiota change after IS. The number of beneficial bacteria decreases, whereas the number of harmful bacteria increases. The production of beneficial metabolites, such as short-chain fatty acids (SCFAs), is reduced, and the levels of harmful metabolites, such as trimethylamine N-oxide (TMAO), increase. Antibiotics after IS not only help prevent infection but also have neuroprotective effects. Although poststroke reperfusion therapy can effectively restore cerebral blood flow, it may also cause intestinal mucosal damage and gastrointestinal dysfunction. Nutritional support after IS can alter the gut microbiota structure and promote neurological recovery. Therefore, individualized treatment for IS patients is crucial. In summary, IS affects not only the brain but the entire body system, especially the gut. Intestinal damage and dysbiosis are critical in IS occurrence, development, and prognosis. By protecting the intestinal mucosa and modulating the structure of the gut microbiota, intestinal damage and related infections can be reduced, improving patient prognosis. Future research is needed to explore therapeutic methods targeting the gut microbiota, providing more comprehensive and effective treatment strategies for IS patients.
Collapse
Affiliation(s)
- Yang Shen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yina Li
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lijuan Gu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Xu Q, Xue L, Wu Z, Kang S, Li J, Wu Y, Wu Y, Zhao J, Wu R, Lv H, Wang J, Han D. Dietary Qiwenghuangbo powder-enriched Limosilactobacillus reuteri protects the intestinal epithelium and alleviates inflammation via a strain-specific mechanism. Animal Model Exp Med 2025. [PMID: 40109036 DOI: 10.1002/ame2.70016] [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: 01/11/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Qiwenghuangbo powder (QP), composed of Astragalus, Phellodendron chinensis, and Radix pulsatilla, is a traditional Chinese herbal formula, but its effects on weaned piglets remained unclear. METHODS Weaned piglets fed with 0.5 kg/t QP (QP1), 1 kg/t QP (QP2), low-zinc oxide (ZnO; negative control), and high-ZnO (positive control) diets in two phases, respectively, and the growth performance, intestinal morphology, cytokines, and microbial communities were profiled. The mouse models of colitis induced by Citrobacter rodentium and dextran sulfate sodium (DSS) were employed to elucidate the potential role of QP-fed enriched key species. RESULTS Dietary 1.0 kg/t QP alleviated diarrhea and inflammation and improved intestinal development and growth performance of weaned piglets. Moreover, this dietary intervention notably altered microbiota composition, characterized by the enrichment of Limosilactobacillus reuteri. Furthermore, out of three isolated L. reuteri, two strains could alleviate pathogen infection and intestinal inflammation, respectively. Specifically, the anti-inflammatory effect of one strain was achieved by promoting the colonization resistance of C. rodentium as significantly reduced pathogen loads. The other strain mitigated DSS-induced colitis by enhancing the goblet cell function and inhibiting the secretion of pro-inflammatory cytokines, particularly interleukin-1β (IL-1ß) and tumor necrosis factor-α (TNF-α). CONCLUSIONS Dietary QP improved the growth performance and intestinal health of weaned piglets by promoting the colonization of L. reuteri. The isolated commensal L. reuteri control colitis in a strain-specific mechanism, highlighting the potential of QP and L. reuteri in providing evidence for gut health promotion.
Collapse
Affiliation(s)
- Qian Xu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lei Xue
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuaishuai Kang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jia Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifan Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rujuan Wu
- Peking Centre Technology Co., Ltd., Beijing, China
| | - Huiyuan Lv
- Peking Centre Technology Co., Ltd., Beijing, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
4
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
5
|
JanssenDuijghuijsen L, Fransen K, Deng R, Perenboom C, de Wit N, Hooiveld G, van Trijp M. How to Study the Effects of Dietary Lipids on the Small Intestinal Microbiome? Methodological Design and Evaluation of the Human HealThy fAt, haPpy mIcRobiome (TAPIR) Proof-of-Concept Study. Curr Dev Nutr 2025; 9:104564. [PMID: 40092654 PMCID: PMC11908603 DOI: 10.1016/j.cdnut.2025.104564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/19/2025] Open
Abstract
Background Emerging evidence highlights the importance of the small intestinal microbiota in digestion and metabolism, underscoring the challenging need for human studies beyond fecal analyses. Objective The TAPIR (acronym of "healthy fat, happy microbiome") proof-of-concept study was primarily designed to confirm the interaction between the small intestinal microbiota and dietary lipids in healthy adults with a challenge test. We also aimed to assess the impact of a plant-based mild-ketogenic preconditioning diet on microbiome composition and function. Here, we comprehensively describe our extensive study protocol and evaluate the study execution. Methods Participants consumed an 8-day preconditioning diet, followed by a high-fat shake challenge test on day 9. During this test, fasting and postprandial small intestinal aspirates were collected every 20 min via a naso-intestinal catheter, and blood samples were collected hourly. Participants ingested small intestine aspiration capsules before (day 0), on day 6 of the preconditioning diet, and during the challenge test. Dietary compliance, capsule retrieval, sample collection, stool pattern, and gastrointestinal complaints were monitored to evaluate study execution. Results Twenty adults with a mean age of 48 y (19-88 y) and a mean body mass index (BMI) of 24.3 kg/m2 (19.5-30 kg/m2) consumed a preconditioning diet with a 96% compliance. There were no significant changes in gastrointestinal complaints and stool patterns during the study. Mean aspiration capsule retrieval rate was 94.7%, with mean sample weights per timepoint between 84.2 and 95.4 mg and median transit times between 32.8 and 49.3 h. The average success rate of aspirate collection by catheter was 49%, varying significantly between time points. Conclusion The dietary intervention was successful and well-tolerated. We sampled in the small intestine with capsules and catheters, each with its own (dis)advantages. The comprehensive description and evaluation of our study execution offer practical insights supporting future study designs in food-microbe interactions in the small intestine.The trial is registered at clinicaltrials.gov as NCT06064266.
Collapse
Affiliation(s)
| | - Karen Fransen
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Ruolei Deng
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Corine Perenboom
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Nicole de Wit
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, the Netherlands
| | - Guido Hooiveld
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| | - Mara van Trijp
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen University, Wageningen, the Netherlands
| |
Collapse
|
6
|
Fu W, Xie Q, Yu P, Liu S, Xu L, Ye X, Zhao W, Wang Q, Pan Y, Zhang Z, Wang Z. Pig jejunal single-cell RNA landscapes revealing breed-specific immunology differentiation at various domestication stages. Front Immunol 2025; 16:1530214. [PMID: 40151618 PMCID: PMC11947726 DOI: 10.3389/fimmu.2025.1530214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Background Domestication of wild boars into local and intensive pig breeds has driven adaptive genomic changes, resulting in significant phenotypic differences in intestinal immune function. The intestine relies on diverse immune cells, but their evolutionary changes during domestication remain poorly understood at single-cell resolution. Methods We performed single-cell RNA sequencing (scRNA-seq) and marker gene analysis on jejunal tissues from wild boars, a Chinese local breed (Jinhua), and an intensive breed (Duroc). Then, we developed an immune cell evaluation system that includes immune scoring, gene identification, and cell communication analysis. Additionally, we mapped domestication-related clustering relationships, highlighting changes in gene expression and immune function. Results We generated a single-cell atlas of jejunal tissues, analyzing 26,246 cells and identifying 11 distinct cell lineages, including epithelial and plasma cells, and discovered shared and unique patterns in intestinal nutrition and immunity across breeds. Immune cell evaluation analysis confirmed the conservation and heterogeneity of immune cells, manifested by highly conserved functions of immune cell subgroups, but wild boars possess stronger immune capabilities than domesticated breeds. We also discovered four patterns of domestication-related breed-specific genes related to metabolism, immune surveillance, and cytotoxic functions. Lastly, we identified a unique population of plasma cells with distinctive antibody production in Jinhua pig population. Conclusions Our findings provide valuable single-cell insights into the cellular heterogeneity and immune function evolution in the jejunum during pig at various domestication stages. The single-cell atlas also serves as a resource for comparative studies and supports breeding programs aimed at enhancing immune traits in pigs.
Collapse
Affiliation(s)
- Wenyu Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qinqin Xie
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Pengfei Yu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shuang Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lingyao Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaowei Ye
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei Zhao
- SciGene Biotechnology Co., Ltd, Hefei, China
| | - Qishan Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Building 11, Yongyou Industrial Park, Yazhou Bay Science and Technology City, Yazhou District, Sanya, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - Yuchun Pan
- Hainan Institute of Zhejiang University, Building 11, Yongyou Industrial Park, Yazhou Bay Science and Technology City, Yazhou District, Sanya, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Hainan Yazhou Bay Seed Lab, Yongyou Industrial Park, Yazhou Bay Sci-Tech City, Sanya, China
| | - Zhe Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| | - Zhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou, China
| |
Collapse
|
7
|
Escoda T, Retornaz F, Plauzolles A, Halfon P. [SIBO, from myth to reality]. Rev Med Interne 2025; 46:40-48. [PMID: 39237384 DOI: 10.1016/j.revmed.2024.08.002] [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: 05/23/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024]
Abstract
Digestive functional disorders are among the most frequent reasons for medical consultation and a significant source of medical wandering. Therapeutic management of these patients is difficult, particularly due to the absence of specific treatment linked to an incomplete understanding of the pathophysiological mechanisms. In a certain number of these patients, the symptoms are accompanied by a small intestinal bacterial overgrowth (SIBO). This entity, historically identified in specific post-surgical situations, seems finally very common and associated with very diverse pathologies. The diagnosis of SIBO is currently being made more accessible through the development of breathing tests. Therapeutic management, based mainly on antibiotic therapy and diet, remains to date largely empirical because it is based on few studies but the growing interest in SIBO should make it possible to identify effective treatments during robust clinical trials.
Collapse
Affiliation(s)
- Thomas Escoda
- Service de médecine interne et maladie infectieuse, hôpital européen, 6, rue Désirée-Clary, 13003 Marseille, France.
| | - Frédérique Retornaz
- Service de médecine interne et maladie infectieuse, hôpital européen, 6, rue Désirée-Clary, 13003 Marseille, France
| | - Anne Plauzolles
- Service de recherche clinique, laboratoire européen Alphabio Biogroup, Marseille, France
| | - Philippe Halfon
- Service de médecine interne et maladie infectieuse, hôpital européen, 6, rue Désirée-Clary, 13003 Marseille, France; Service de recherche clinique, laboratoire européen Alphabio Biogroup, Marseille, France
| |
Collapse
|
8
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
9
|
Giacosa A, Barrile GC, Perna S, Rondanelli M. Positive Outcomes of Supplementation with Lecithin-Based Delivery Formulation of Curcuma longa and Boswellia serrata in IBS Subjects with Small Bowel Dysbiosis. Life (Basel) 2024; 14:1410. [PMID: 39598208 PMCID: PMC11595465 DOI: 10.3390/life14111410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Small bowel dysbiosis (SBD) is a frequent finding in subjects with irritable bowel syndrome (IBS). The sunflower lecithin (phytosome) formulation of Curcuma longa and Boswellia serrata demonstrated beneficial effects on intestinal microbiota. This study aimed to evaluate the impact of a lecithin-based delivery formulation of Curcuma longa and Boswellia serrata extracts (CUBO) on SBD in IBS subjects. SUBJECTS Forty-nine adult subjects with IBS and SBD were randomly supplemented for 30 days with CUBO and a low-FODMAP diet (LFD) (intervention) or with LFD and placebo (control group). RESULTS The intervention group showed a significant reduction in urinary indican (p < 0.001), which was the marker of SBD, and of abdominal bloating (p < 0.001) and abdominal pain (p < 0.001). The pre-post control group analysis did not evidence significant differences. The comparison between the two groups (net effect in intervention minus control subjects) showed that the changes differ significantly for urinary indican p < 0.001 (-42.88; 95% CI: -62.04 to -23.72), abdominal bloating p < 0.001 (-1.50; 95% CI: -1.93 to -1.07), and abdominal pain p < 0.001 (-2.37; 95% CI: -3.61 to -1.13) and for the global assessment of efficacy (p < 0.001). The efficacy was 20% greater in males than in females. CONCLUSIONS In IBS subjects, the intervention with CUBO and LFD shows a significantly higher reduction in SBD, abdominal pain, and bloating compared to LFD and placebo. Additional research is needed to confirm these data.
Collapse
Affiliation(s)
| | - Gaetan Claude Barrile
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Simone Perna
- Department of Food, Environmental and Nutritional Sciences, Division of Human Nutrition, University of Milan, 20133 Milan, Italy;
| | - Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
10
|
Choi J, Lee J, Kim WK. Alterations in the gut microbiota of Eimeria infected broiler chickens fed diets supplemented with varying levels of dietary calcium and phosphorus, along with 25-hydroxycholecalciferol. Poult Sci 2024; 103:104223. [PMID: 39216268 PMCID: PMC11402547 DOI: 10.1016/j.psj.2024.104223] [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: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
The purpose of the study was to investigate the effects of the reduced dietary calcium (Ca) and phosphorus (P) level and supplementation of 25-hydroxycholecalciferol (25-OHD3) on the expression of vitamin D receptor (VDR) and antimicrobial peptides and gut microbiota of broiler chickens with/without Eimeria challenge. A total of 576 fourteen-day-old broiler chicks were randomly allocated according to a 2 × 2 × 2 factorial design with main effects including Eimeria challenging (125,000 Eimeria acervulina, 25,000 Eimeria maxima, and 25,000 Eimeria tenella), dietary Ca and P levels (0.84% Ca and 0.42% available P or 0.64% Ca and 0.22% available P), and supplementation of 25-OHD3 (3,000 IU/kg) of 6 replicates. Three-way ANOVA was performed, and the effects of 3 main factors and their interactions were investigated. The reduced dietary Ca and P level downregulated cathelicidins 3 (CATH3) in the upper jejunum in the Eimeria challenging condition (interaction; P < 0.05). The reduced dietary Ca and P level decreased the relative mRNA expression of jejunal avian beta defensin 5 (AvBD5) in the Eimeria challenging condition (interaction; P < 0.05). The reduced dietary Ca and P level tended to decrease the relative mRNA expression of jejunal AvBD9 in the Eimeria challenging condition (interaction; P = 0.051). The reduced dietary Ca and P level decreased observed features (alpha diversity parameter for richness) in the upper jejunal microbiota in the Eimeria challenging condition (interaction; P < 0.05). The supplementation of 25-OHD3 decreased the relative abundance of the phylum Bacteroidetes (P < 0.05) and increased the relative abundance of the family Ruminococcaceae (P < 0.05) in the cecal digesta. The supplementation of 25-OHD3 decreased the serum endotoxin level in the Eimeria challenging condition (interaction; P < 0.05). Therefore, the reduced dietary Ca and P level modulated the upper jejunal microbiota via modulating the expression of antimicrobial peptides, and the supplementation of 25-OHD3 favorably modulated the cecal microbiota in broiler chickens with/without Eimeria challenge.
Collapse
Affiliation(s)
- Janghan Choi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA; US National Poultry Research Center, USDA-ARS, Athens, GA 30605, USA
| | - Jihwan Lee
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
11
|
Tang Y, Liu Y, Jin Y, Zhang F, Zhang W, Luo S, Zang J, Yang W, Chen Y. Toxicological risk assessment of triadimenol for human exposure, broiler health, and food safety. Food Chem Toxicol 2024; 194:115071. [PMID: 39486609 DOI: 10.1016/j.fct.2024.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Triadimenol, a widely used triazole fungicide, leaves residues that pose risks to broiler health, food safety, and human health. Current studies focus on lab animals, leaving limited data regarding its impact on non-target organisms in agricultural ecosystems. Moreover, the doses in current studies often exceed typical agricultural pollution levels of triadimenol. Therefore, this study evaluates the toxic effects of triadimenol by exposing broilers to different concentrations (0.05-20 mg/kg) in their feed for 42 days, assessing growth performance, organ index, hematological parameters, histopathology, jejunum morphology, and tissue residues. The results show that triadimenol contamination at 0.05-20 mg/kg in feed does not significantly affect broiler growth performance. However, the significant changes in hematological parameters suggest the potential hematological toxicity of triadimenol in broilers. Triadimenol at 1 mg/kg or higher induces hepatotoxic and nephrotoxic effects, and significantly alters kidney organ index and histopathology in broilers. Additionally, when the triadimenol contamination level in feed exceeds 1 mg/kg, the residues in edible tissues of broilers exceed the limits set by the EU and China. Overall, our study indicates that even low-dose exposure to triadimenol poses potential risks, highlighting the need for strict regulation of its use in agriculture to protect food safety and human health.
Collapse
Affiliation(s)
- Yutong Tang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ying Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yongpeng Jin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Fude Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wanjun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Sunlin Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jianjun Zang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wenjun Yang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yiqiang Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
12
|
Han K, Xie F, Animasahun O, Nenwani M, Kitamoto S, Kim Y, Phoo MT, Xu J, Wuchu F, Omoloja K, Achreja A, Choppara S, Li Z, Gong W, Cho YS, Dobson H, Ahn J, Zhou X, Huang X, An X, Kim A, Xu Y, Wu Q, Lee SH, O'Konek JJ, Xie Y, Lei YL, Kamada N, Nagrath D, Moon JJ. Inulin-gel-based oral immunotherapy remodels the small intestinal microbiome and suppresses food allergy. NATURE MATERIALS 2024; 23:1444-1455. [PMID: 38977883 PMCID: PMC11442122 DOI: 10.1038/s41563-024-01909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/30/2024] [Indexed: 07/10/2024]
Abstract
Despite the potential of oral immunotherapy against food allergy, adverse reactions and loss of desensitization hinder its clinical uptake. Dysbiosis of the gut microbiota is implicated in the increasing prevalence of food allergy, which will need to be regulated to enable for an effective oral immunotherapy against food allergy. Here we report an inulin gel formulated with an allergen that normalizes the dysregulated ileal microbiota and metabolites in allergic mice, establishes allergen-specific oral tolerance and achieves robust oral immunotherapy efficacy with sustained unresponsiveness in food allergy models. These positive outcomes are associated with enhanced allergen uptake by antigen-sampling dendritic cells in the small intestine, suppressed pathogenic type 2 immune responses, increased interferon-γ+ and interleukin-10+ regulatory T cell populations, and restored ileal abundances of Eggerthellaceae and Enterorhabdus in allergic mice. Overall, our findings underscore the therapeutic potential of the engineered allergen gel as a suitable microbiome-modulating platform for food allergy and other allergic diseases.
Collapse
Affiliation(s)
- Kai Han
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Fang Xie
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olamide Animasahun
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Minal Nenwani
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sho Kitamoto
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yeji Kim
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - May Thazin Phoo
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fulei Wuchu
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kehinde Omoloja
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Abhinav Achreja
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Srinadh Choppara
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Zhaoheng Li
- Graduate Program in Biostatistics, University of Washington, Seattle, WA, USA
| | - Wang Gong
- Departments of Head and Neck Surgery and of Cancer Biology, the University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hannah Dobson
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jinsung Ahn
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xuehui Huang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xinran An
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yao Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Qi Wu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | - Yuying Xie
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Statistics and Probability, Michigan State University, East Lansang, MI, USA
| | - Yu Leo Lei
- Departments of Head and Neck Surgery and of Cancer Biology, the University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Deepak Nagrath
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Kong Y, Zhao S, Ou W, Mai K, Zhang Y. The Combination of Host-Associated Bacillus megaterium R32 and Stachyose Promotes the Intestinal Health of Turbot ( Scophthalmus maximus. L). AQUACULTURE NUTRITION 2024; 2024:8658386. [PMID: 39555546 PMCID: PMC11458268 DOI: 10.1155/2024/8658386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 06/25/2024] [Accepted: 09/06/2024] [Indexed: 11/19/2024]
Abstract
An 8-week feeding trial was conducted to investigate the effects of host-associated Bacillus megaterium R32 and stachyose on the intestinal mucosal defense system of turbot (Scophthalmus maximus. L). Three isonitrogenous and isolipidic diets were formulated: control diet (C), control diet with 1.0 × 108 CFU/g B. megaterium R32 (RC), and 1.0 × 108 CFU/g B. megaterium R32 + 1.5% stachyose (RS) supplementation separately. The results showed that diets RS and RC significantly inhibited the expression of cell development and apoptosis-related genes (β-catenin, CyclinD1, BAX, Bid); diets RS and RC significantly increased the expression of intestinal tight junction protein claudin-4, while RS group significantly decreased the expression of myosin light chain kinase; diets RS and RC significantly decreased the expression of proinflammatory factors (IL-13, IL-15, IFN- γ), diet RS also significantly decreased the expression of TNF-α and AP-1, and increased the expression of TGF-β. 16s rRNA gene sequencing results showed that diets RS and RC significantly decreased the abundance of conditional pathogenic bacteria (Corynebacterium, Desulfovibrio, Escherichia-Shigella). Among them, the abundance of Bacillus in the RS group was the highest. It is concluded that the combination of stachyose and B. megaterium R32 had a more positive effect on intestinal cell development and barrier function and strengthened the intestinal mucosal defense system of turbot.
Collapse
Affiliation(s)
- Yaoyao Kong
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Ministry of Education)Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, China
| | - Sifan Zhao
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Ministry of Education)Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, China
| | - Weihao Ou
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Ministry of Education)Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, China
| | - Kangsen Mai
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Ministry of Education)Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, China
- Laboratory for Marine Fisheries Science and Food Production ProcessesQingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, China
| | - Yanjiao Zhang
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture) and the Key Laboratory of Mariculture (Ministry of Education)Ocean University of China, 5 Yushan Road, Qingdao, Shandong 266003, China
- Laboratory for Marine Fisheries Science and Food Production ProcessesQingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, China
| |
Collapse
|
14
|
Soldán M, Argalášová Ľ, Hadvinová L, Galileo B, Babjaková J. The Effect of Dietary Types on Gut Microbiota Composition and Development of Non-Communicable Diseases: A Narrative Review. Nutrients 2024; 16:3134. [PMID: 39339734 PMCID: PMC11434870 DOI: 10.3390/nu16183134] [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: 07/26/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION The importance of diet in shaping the gut microbiota is well established and may help improve an individual's overall health. Many other factors, such as genetics, age, exercise, antibiotic therapy, or tobacco use, also play a role in influencing gut microbiota. AIM This narrative review summarizes how three distinct dietary types (plant-based, Mediterranean, and Western) affect the composition of gut microbiota and the development of non-communicable diseases (NCDs). METHODS A comprehensive literature search was conducted using the PubMed, Web of Science, and Scopus databases, focusing on the keywords "dietary pattern", "gut microbiota" and "dysbiosis". RESULTS Both plant-based and Mediterranean diets have been shown to promote the production of beneficial bacterial metabolites, such as short-chain fatty acids (SCFAs), while simultaneously lowering concentrations of trimethylamine-N-oxide (TMAO), a molecule associated with negative health outcomes. Additionally, they have a positive impact on microbial diversity and therefore are generally considered healthy dietary types. On the other hand, the Western diet is a typical example of an unhealthy nutritional approach leading to an overgrowth of pathogenic bacteria, where TMAO levels rise and SCFA production drops due to gut dysbiosis. CONCLUSION The current scientific literature consistently highlights the superiority of plant-based and Mediterranean dietary types over the Western diet in promoting gut health and preventing NCDs. Understanding the influence of diet on gut microbiota modulation may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Ľubica Argalášová
- Institute of Hygiene, Faculty of Medicine, Comenius University in Bratislava, Špitálska 24, 813 72 Bratislava, Slovakia; (M.S.); (L.H.); (B.G.); (J.B.)
| | | | | | | |
Collapse
|
15
|
Gong J, Xu F, Li Y, He Y, Liang Z, Chen X, Zhang X, Liu L, Zhou L, Huang X. Metagenomic analysis of intestinal microbial function and key genes responsive to acute high-salinity stress in Nile tilapia (Oreochromis niloticus). Gene 2024; 913:148371. [PMID: 38485034 DOI: 10.1016/j.gene.2024.148371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
The intestinal microbiota is increasingly recognized as playing an important role in aquatic animals. To investigate the functional roles and mechanisms of the intestinal microbial genes/enzymes responding to salinity stress or osmotic pressure in fish, metagenomic analysis was carried out to evaluate the response of intestinal microbiota and especially their functional genes/enzymes from freshwater (the control group) to acute high salinity stress (the treatment group) in Nile tilapia. Our results showed that at the microbial community level, the intestinal microbiota in Nile tilapia generally underwent significant changes in diversity after acute high salinity stress. Among them, the shift in the bacterial community (mainly from Actinobacteria to Proteobacteria) dominated and had a large impact, the fungal community showed a very limited response, and other microbiota, such as phages, likely had a negligible response. At the functional level, the intestinal bacteriadecreased the normal physiological demand and processes, such as those of the digestive system and nervous system, but enhanced energy metabolism. Furthermore, at the gene level, some gene biomarkers, such as glutathione S-transferase, myo-inositol-1(or 4)-monophosphatase, glycine betaine/proline transport system permease protein, and some families of carbohydrate-active enzymes (GT4, GT2), were significantly enriched. However, GH15, GH23 and so on were significantly reduced. Exploring the functional details of the intestinal microbial genes/enzymes that respond to salinity stress in Nile tilapia sheds light on the mechanism of action of the intestinal microbiota with respect to the salinity adaptation of fish.
Collapse
Affiliation(s)
- Jiayi Gong
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Fengmeng Xu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China; Guangzhou Fishtech Biotechnology Co., Ltd., Guangzhou 510640, China
| | - Yao Li
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Yiyong He
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Zhizheng Liang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiao Chen
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Li Liu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Lei Zhou
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.
| | - Xiande Huang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
16
|
Firrman J, Friedman ES, Hecht A, Strange WC, Narrowe AB, Mahalak K, Wu GD, Liu L. Preservation of conjugated primary bile acids by oxygenation of the small intestinal microbiota in vitro. mBio 2024; 15:e0094324. [PMID: 38727244 PMCID: PMC11237543 DOI: 10.1128/mbio.00943-24] [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: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 06/13/2024] Open
Abstract
Bile acids play a critical role in the emulsification of dietary lipids, a critical step in the primary function of the small intestine, which is the digestion and absorption of food. Primary bile acids delivered into the small intestine are conjugated to enhance functionality, in part, by increasing aqueous solubility and preventing passive diffusion of bile acids out of the gut lumen. Bile acid function can be disrupted by the gut microbiota via the deconjugation of primary bile acids by bile salt hydrolases (BSHs), leading to their conversion into secondary bile acids through the expression of bacterial bile acid-inducible genes, a process often observed in malabsorption due to small intestinal bacterial overgrowth. By modeling the small intestinal microbiota in vitro using human small intestinal ileostomy effluent as the inocula, we show here that the infusion of physiologically relevant levels of oxygen, normally found in the proximal small intestine, reduced deconjugation of primary bile acids, in part, through the expansion of bacterial taxa known to have a low abundance of BSHs. Further recapitulating the small intestinal bile acid composition of the small intestine, limited conversion of primary into secondary bile acids was observed. Remarkably, these effects were preserved among four separate communities, each inoculated with a different small intestinal microbiota, despite a high degree of taxonomic variability under both anoxic and aerobic conditions. In total, these results provide evidence for a previously unrecognized role that the oxygenated environment of the small intestine plays in the maintenance of normal digestive physiology. IMPORTANCE Conjugated primary bile acids are produced by the liver and exist at high concentrations in the proximal small intestine, where they are critical for proper digestion. Deconjugation of these bile acids with subsequent transformation via dehydroxylation into secondary bile acids is regulated by the colonic gut microbiota and reduces their digestive function. Using an in vitro platform modeling the small intestinal microbiota, we analyzed the ability of this community to transform primary bile acids and studied the effect of physiological levels of oxygen normally found in the proximal small intestine (5%) on this metabolic process. We found that oxygenation of the small intestinal microbiota inhibited the deconjugation of primary bile acids in vitro. These findings suggest that luminal oxygen levels normally found in the small intestine may maintain the optimal role of bile acids in the digestive process by regulating bile acid conversion by the gut microbiota.
Collapse
Affiliation(s)
- Jenni Firrman
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Elliot S. Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aaron Hecht
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William C. Strange
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adrienne B. Narrowe
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Karley Mahalak
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, US Department of Agriculture, Wyndmoor, Pennsylvania, USA
| |
Collapse
|
17
|
Pironi L, D'Amico F, Guidetti M, Brigidi P, Sasdelli AS, Turroni S. The gut microbiota in adults with chronic intestinal failure. Clin Nutr 2024; 43:1331-1342. [PMID: 38677044 DOI: 10.1016/j.clnu.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVE Fecal microbiota was investigated in adult patients with chronic intestinal failure (CIF) due to short bowel syndrome (SBS) with jejunocolonic anastomosis (SBS-2). Few or no data are available on SBS with jejunostomy (SBS-1) and CIF due to intestinal dysmotility (DYS) or mucosal disease (MD). We profiled the fecal microbiota of various pathophysiological mechanisms of CIF. METHODS Cross-sectional study on 61 adults with CIF (SBS-1 30, SBS-2 17, DYS 8, MD 6). Fecal samples were collected and profiled by 16S rRNA amplicon sequencing. Healthy controls (HC) were selected from pre-existing cohorts, matched with patients by sex and age. RESULTS Compared to HC, SBS-1, SBS-2 and MD patients showed lower alpha diversity; no difference was found for DYS. In beta diversity analysis, SBS-1, SBS-2 and DYS groups segregated from HC and from each other. Taxonomically, the CIF groups differed from HC even at the phylum level. In particular, CIF patients' microbiota was dominated by Lactobacillaceae and Enterobacteriaceae, while depleted in typical health-associated taxa belonging to Lachnospiraceae and Ruminococcaceae. Notably, compositional peculiarities of the CIF groups emerged. Furthermore, in the SBS groups, the microbiota profile differed according to the amount of parenteral nutrition required and the duration of CIF. CONCLUSIONS CIF patients showed marked intestinal dysbiosis with microbial signatures specific to the pathophysiological mechanism of CIF as well as to the severity and duration of SBS.
Collapse
Affiliation(s)
- Loris Pironi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; Centre for Chronic Intestinal Failure, IRCCS AOUBO, Bologna, Italy.
| | - Federica D'Amico
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
18
|
Leite G, Barlow GM, Rashid M, Hosseini A, Cohrs D, Parodi G, Morales W, Weitsman S, Rezaie A, Pimentel M, Mathur R. Characterization of the Small Bowel Microbiome Reveals Different Profiles in Human Subjects Who Are Overweight or Have Obesity. Am J Gastroenterol 2024; 119:1141-1153. [PMID: 38578969 PMCID: PMC11142649 DOI: 10.14309/ajg.0000000000002790] [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: 11/02/2023] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
INTRODUCTION Gut microbiome changes are linked to obesity, but findings are based on stool data. In this article, we analyzed the duodenal microbiome and serum biomarkers in subjects with normal weight, overweight, and obesity. METHODS Duodenal aspirates and serum samples were obtained from subjects undergoing standard-of-care esophagogastroduodenoscopy without colon preparation. Aspirate DNAs were analyzed by 16S rRNA and shotgun sequencing. Predicted microbial metabolic functions and serum levels of metabolic and inflammatory biomarkers were also assessed. RESULTS Subjects with normal weight (N = 105), overweight (N = 67), and obesity (N = 42) were identified. Overweight-specific duodenal microbial features include lower relative abundance (RA) of Bifidobacterium species and Escherichia coli strain K-12 and higher Lactobacillus intestinalis , L. johnsonii , and Prevotella loescheii RA. Obesity-specific features include higher Lactobacillus gasseri RA and lower L. reuteri (subspecies rodentium ), Alloprevotella rava , and Leptotrichia spp RA. Escalation features (progressive changes from normal weight through obesity) include decreasing Bacteroides pyogenes , Staphylococcus hominis , and unknown Faecalibacterium species RA, increasing RA of unknown Lactobacillus and Mycobacterium species, and decreasing microbial potential for biogenic amines metabolism. De-escalation features (direction of change altered in normal to overweight and overweight to obesity) include Lactobacillus acidophilus , L. hominis , L. iners , and Bifidobacterium dentium . An unknown Lactobacillus species is associated with type IIa dyslipidemia and overweight, whereas Alloprevotella rava is associated with type IIb and IV dyslipidemias. DISCUSSION Direct analysis of the duodenal microbiome has identified key genera associated with overweight and obesity, including some previously identified in stool, e.g., Bifidobacterium and Lactobacillus . Specific species and strains exhibit differing associations with overweight and obesity, including escalation and de-escalation features that may represent targets for future study and therapeutics.
Collapse
Affiliation(s)
- Gabriela Leite
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Gillian M. Barlow
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Mohamad Rashid
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Ava Hosseini
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Daniel Cohrs
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Gonzalo Parodi
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Walter Morales
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Stacy Weitsman
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
| | - Ali Rezaie
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai, Los Angeles, California, USA
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai, Los Angeles, California, USA
| | - Ruchi Mathur
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, California, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai, Los Angeles, California, USA
| |
Collapse
|
19
|
van Trijp MPH, Rios-Morales M, Witteman B, Abegaz F, Gerding A, An R, Koehorst M, Evers B, van Dongen KCV, Zoetendal EG, Schols H, Afman LA, Reijngoud DJ, Bakker BM, Hooiveld GJ. Intraintestinal fermentation of fructo- and galacto-oligosaccharides and the fate of short-chain fatty acids in humans. iScience 2024; 27:109208. [PMID: 38420581 PMCID: PMC10901090 DOI: 10.1016/j.isci.2024.109208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
Consumption of fructo- (FOS) and galacto-oligosaccharides (GOS) has health benefits which have been linked in part to short-chain fatty acids (SCFA) production by the gut microbiota. However, detailed knowledge of this process in the human intestine is lacking. We aimed to determine the acute fermentation kinetics of a FOS:GOS mixture in healthy males using a naso-intestinal catheter for sampling directly in the ileum or colon. We studied the fate of SCFA as substrates for glucose and lipid metabolism by the host after infusion of 13C-SCFA. In the human distal ileum, no fermentation of FOS:GOS, nor SCFA production, or bacterial cross-feeding was observed. The relative composition of intestinal microbiota changed rapidly during the test day, which demonstrates the relevance of postprandial intestinal sampling to track acute responses of the microbial community toward interventions. SCFA were vividly taken up and metabolized by the host as shown by incorporation of 13C in various host metabolites.
Collapse
Affiliation(s)
- Mara P H van Trijp
- Division of Human Nutrition and Health, Wageningen University, Wageningen 6708 WE, the Netherlands
| | - Melany Rios-Morales
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Ben Witteman
- Division of Human Nutrition and Health, Wageningen University, Wageningen 6708 WE, the Netherlands
- Hospital Gelderse Vallei, Department of Gastroenterology and Hepatology, Ede 6716 RP, the Netherlands
| | - Fentaw Abegaz
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
- Statistics and Probability Unit, University of Groningen, Groningen 9747 AG, the Netherlands
| | - Albert Gerding
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Ran An
- Laboratory of Microbiology, Wageningen University, Wageningen 6708 WE, the Netherlands
| | - Martijn Koehorst
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Bernard Evers
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Katja C V van Dongen
- Division of Toxicology, Wageningen University, Wageningen 6708 WE, the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University, Wageningen 6708 WE, the Netherlands
| | - Henk Schols
- Laboratory of Food Chemistry, Wageningen University, Wageningen 6708 WG, the Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen 6708 WE, the Netherlands
| | - Dirk-Jan Reijngoud
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Barbara M Bakker
- Laboratory of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Guido J Hooiveld
- Division of Human Nutrition and Health, Wageningen University, Wageningen 6708 WE, the Netherlands
| |
Collapse
|
20
|
Álvarez-Herms J, González-Benito A, Corbi F, Odriozola A. What if gastrointestinal complications in endurance athletes were gut injuries in response to a high consumption of ultra-processed foods? Please take care of your bugs if you want to improve endurance performance: a narrative review. Eur J Appl Physiol 2024; 124:383-402. [PMID: 37839038 DOI: 10.1007/s00421-023-05331-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
To improve performance and recovery faster, athletes are advised to eat more often than usual and consume higher doses of simple carbohydrates, during and after exercise. Sports energetic supplements contain food additives, such as artificial sweeteners, emulsifiers, acidity regulators, preservatives, and salts, which could be harmful to the gut microbiota and impair the intestinal barrier function. The intestinal barrier plays a critical function in bidirectionally regulation of the selective transfer of nutrients, water, and electrolytes, while preventing at the same time, the entrance of harmful substances (selective permeability). The gut microbiota helps to the host to regulate intestinal homeostasis through metabolic, protective, and immune functions. Globally, the gut health is essential to maintain systemic homeostasis in athletes, and to ensure proper digestion, metabolization, and substrate absorption. Gastrointestinal complaints are an important cause of underperformance and dropout during endurance events. These complications are directly related to the loss of gut equilibrium, mainly linked to microbiota dysbiosis and leaky gut. In summary, athletes must be cautious with the elevated intake of ultra-processed foods and specifically those contained on sports nutrition supplements. This review points out the specific nutritional interventions that should be implemented and/or discontinued depending on individual gut functionality.
Collapse
Affiliation(s)
- Jesús Álvarez-Herms
- Phymolab (Physiology and Molecular Laboratory), Collado Hermoso, Segovia, Spain.
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - A González-Benito
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - F Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), University of Lleida (UdL), Lleida, Spain
| | - A Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
21
|
An R, Wilms E, Gerritsen J, Kim HK, Pérez CS, Besseling-van der Vaart I, Jonkers DM, Rijkers GT, de Vos WM, Masclee AA, Zoetendal EG, Troost FJ, Smidt H. Spatio-temporal dynamics of the human small intestinal microbiome and its response to a synbiotic. Gut Microbes 2024; 16:2350173. [PMID: 38738780 PMCID: PMC11093041 DOI: 10.1080/19490976.2024.2350173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024] Open
Abstract
Although fecal microbiota composition is considered to preserve relevant and representative information for distal colonic content, it is evident that it does not represent microbial communities inhabiting the small intestine. Nevertheless, studies investigating the human small intestinal microbiome and its response to dietary intervention are still scarce. The current study investigated the spatio-temporal dynamics of the small intestinal microbiome within a day and over 20 days, as well as its responses to a 14-day synbiotic or placebo control supplementation in 20 healthy subjects. Microbial composition and metabolome of luminal content of duodenum, jejunum, proximal ileum and feces differed significantly from each other. Additionally, differences in microbiota composition along the small intestine were most pronounced in the morning after overnight fasting, whereas differences in composition were not always measurable around noon or in the afternoon. Although overall small intestinal microbiota composition did not change significantly within 1 day and during 20 days, remarkable, individual-specific temporal dynamics were observed in individual subjects. In response to the synbiotic supplementation, only the microbial diversity in jejunum changed significantly. Increased metabolic activity of probiotic strains during intestinal passage, as assessed by metatranscriptome analysis, was not observed. Nevertheless, synbiotic supplementation led to a short-term spike in the relative abundance of genera included in the product in the small intestine approximately 2 hours post-ingestion. Collectively, small intestinal microbiota are highly dynamic. Ingested probiotic bacteria could lead to a transient spike in the relative abundance of corresponding genera and ASVs, suggesting their passage through the entire gastrointestinal tract. This study was registered to http://www.clinicaltrials.gov, NCT02018900.
Collapse
Affiliation(s)
- Ran An
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Department of Food science and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Ellen Wilms
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jacoline Gerritsen
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Winclove Probiotics, Amsterdam, The Netherlands
| | - Hye Kyong Kim
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Celia Seguí Pérez
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Winclove Probiotics, Amsterdam, The Netherlands
- Infectious Diseases & Immunology, University of Utrecht, Utrecht, The Netherland
| | | | - Daisy M.A.E. Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ger T. Rijkers
- Science Department, University College Roosevelt, Middelburg, The Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Human Microbiomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ad A.M. Masclee
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Erwin G. Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Freddy J. Troost
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Food Innovation and Health, Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Venlo, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
22
|
Macedo MH, Dias Neto M, Pastrana L, Gonçalves C, Xavier M. Recent Advances in Cell-Based In Vitro Models to Recreate Human Intestinal Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301391. [PMID: 37736674 PMCID: PMC10625086 DOI: 10.1002/advs.202301391] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/03/2023] [Indexed: 09/23/2023]
Abstract
Inflammatory bowel disease causes a major burden to patients and healthcare systems, raising the need to develop effective therapies. Technological advances in cell culture, allied with ethical issues, have propelled in vitro models as essential tools to study disease aetiology, its progression, and possible therapies. Several cell-based in vitro models of intestinal inflammation have been used, varying in their complexity and methodology to induce inflammation. Immortalized cell lines are extensively used due to their long-term survival, in contrast to primary cultures that are short-lived but patient-specific. Recently, organoids and organ-chips have demonstrated great potential by being physiologically more relevant. This review aims to shed light on the intricate nature of intestinal inflammation and cover recent works that report cell-based in vitro models of human intestinal inflammation, encompassing diverse approaches and outcomes.
Collapse
Affiliation(s)
- Maria Helena Macedo
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Mafalda Dias Neto
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Lorenzo Pastrana
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Catarina Gonçalves
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Miguel Xavier
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| |
Collapse
|
23
|
Hosseini A, Rashid M, Leite G, Barlow GM, Parodi G, Sanchez M, Ayyad S, Pimentel ML, Morales W, Weitsman S, Pimentel M, Mathur R. Coronavirus Disease 2019 (COVID-19) Pandemic Lifestyle Changes May Have Influenced Small Bowel Microbial Composition and Microbial Resistance. Dig Dis Sci 2023; 68:3902-3912. [PMID: 37578565 PMCID: PMC10516792 DOI: 10.1007/s10620-023-08061-6] [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: 10/27/2022] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) global pandemic necessitated many severe lifestyle changes, including lockdowns, social distancing, altered food consumption and exercise patterns, and extensive hygiene practices. These extensive changes may have affected the human gut microbiome, which is highly influenced by lifestyle. AIMS To examine the potential effects of pandemic-related lifestyle changes on the metabolically relevant small bowel microbiome. METHODS Adult subjects presenting for upper endoscopy without colonoscopy were identified and divided into two matched groups: pre-pandemic (February 2019-March 2020) and intra-pandemic (April 2021-September 2021, all COVID-19 negative). Duodenal aspirates and blood samples were collected. Duodenal microbiomes were analyzed by 16S rRNA sequencing. Serum cytokine levels were analyzed by Luminex FlexMap3D. RESULTS Fifty-six pre-pandemic and 38 COVID-negative intra-pandemic subjects were included. There were no significant changes in duodenal microbial alpha diversity in the intra-pandemic vs. pre-pandemic group, but beta diversity was significantly different. The relative abundance (RA) of phylum Deinococcus-Thermus and family Thermaceae, which are resistant extremophiles, was significantly higher in the intra-pandemic vs. pre-pandemic group. The RA of several Gram-negative taxa including Bacteroidaceae (phylum Bacteroidetes) and the Proteobacteria families Enterobacteriaceae and Pseudomonadaceae, and the RA of potential disruptor genera Escherichia-Shigella and Rothia, were significantly lower in the intra-pandemic vs. pre-pandemic group. Circulating levels of interleukin-18 were also lower in the intra-pandemic group. CONCLUSIONS These findings suggest the small bowel microbiome underwent significant changes during the pandemic, in COVID-19-negative individuals. Given the key roles of the small bowel microbiota in host physiology, this may have implications for human health.
Collapse
Affiliation(s)
- Ava Hosseini
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Mohamad Rashid
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Gabriela Leite
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Gillian M Barlow
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Gonzalo Parodi
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Maritza Sanchez
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Sarah Ayyad
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Maya L Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Walter Morales
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Stacy Weitsman
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai, Los Angeles, CA, USA
| | - Ruchi Mathur
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Feng Y, Wu X, Hu D, Wang C, Chen Q, Ni Y. Comparison of the Effects of Feeding Compound Probiotics and Antibiotics on Growth Performance, Gut Microbiota, and Small Intestine Morphology in Yellow-Feather Broilers. Microorganisms 2023; 11:2308. [PMID: 37764152 PMCID: PMC10535644 DOI: 10.3390/microorganisms11092308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
This study was devoted to the comparison of the probiotic effect of compound probiotics to antibiotics as a feed additive for chicken. Two hundred and seventy newly hatched yellow-feather broilers were randomly divided into three groups: the control group (Con), probiotics (Pb), and antibiotics group (Ab). The Pb group received compound probiotics (Bifidobacterium, Lactobacillus acidophilus, Streptococcus faecalis, and yeast) via drinking water for 24 days. The Ab group received antibiotics (zinc bacitracin and colistin sulfate) in their diet for 24 days. All broilers were slaughtered on day 42. Compared with the Con group, the body weight was significantly increased on days 13, 28, and 42 in the Pb group (p < 0.05), and markedly increased on day 28 in the Ab group (p < 0.05). Compared with the Ab group, the body weight of the broilers in the Pb group increased significantly on day 13 (p < 0.05). Compared to the Con and Pb groups, the antibiotics treatment reduced the feed intake (p < 0.05), but there was no significant difference in the feed conversion ratio between the Ab and Pb groups (p > 0.05). The feed conversion ratio of the broilers treated with antibiotics or probiotics significantly decreased compared to the Con group (p < 0.05). The depth of duodenum, jejunum, and ileum crypts in the Pb group decreased significantly compared to the Con and Ab group (p < 0.05). The ratio of the villi length to crypt depth of duodenum, jejunum, and ileum epithelium was significantly increased in the Pb group compared to the Con group (p < 0.05). The genera Bacteroides and Barnesiella were the most significantly enriched bacteria in the Ab and Pb groups, respectively (p < 0.05). The expression of the genes related to antibiotic resistance was significantly decreased in the Pb group compared to the Ab group (p < 0.05). Although both compound probiotics and antibiotics can improve growth performance, antibiotics increased the abundance of harmful bacteria and drug-resistant genes, while probiotics increased Barnesiella abundance, which is related to a decrease in the drug-resistant gene expression. Moreover, the probiotics treatment improved small intestinal morphology and fecal emissions, while antibiotics have no significant effect on these indicators, indicating a bright future for probiotics as an alternative to feed antibiotics in the yellow-feather broiler industry.
Collapse
Affiliation(s)
- Yuyan Feng
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoting Wu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, China
| | - Dan Hu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, China
| | - Canyang Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, China
| | - Qu Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
25
|
Xiang J, Guo J, Zhang S, Wu H, Chen YG, Wang J, Li B, Liu H. A stromal lineage maintains crypt structure and villus homeostasis in the intestinal stem cell niche. BMC Biol 2023; 21:169. [PMID: 37553612 PMCID: PMC10408166 DOI: 10.1186/s12915-023-01667-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/24/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND The nutrient-absorbing villi of small intestines are renewed and repaired by intestinal stem cells (ISCs), which reside in a well-organized crypt structure. Genetic studies have shown that Wnt molecules secreted by telocytes, Gli1+ stromal cells, and epithelial cells are required for ISC proliferation and villus homeostasis. Intestinal stromal cells are heterogeneous and single-cell profiling has divided them into telocytes/subepithelial myofibroblasts, myocytes, pericytes, trophocytes, and Pdgfralow stromal cells. Yet, the niche function of these stromal populations remains incompletely understood. RESULTS We show here that a Twist2 stromal lineage, which constitutes the Pdgfralow stromal cell and trophocyte subpopulations, maintains the crypt structure to provide an inflammation-restricting niche for regenerating ISCs. Ablating Twist2 lineage cells or deletion of one Wntless allele in these cells disturbs the crypt structure and impairs villus homeostasis. Upon radiation, Wntless haplo-deficiency caused decreased production of anti-microbial peptides and increased inflammation, leading to defective ISC proliferation and crypt regeneration, which were partially rescued by eradication of commensal bacteria. In addition, we show that Wnts secreted by Acta2+ subpopulations also play a role in crypt regeneration but not homeostasis. CONCLUSIONS These findings suggest that ISCs may require different niches for villus homeostasis and regeneration and that the Twist2 lineage cells may help to maintain a microbe-restricted environment to allow ISC-mediated crypt regeneration.
Collapse
Affiliation(s)
- Jinnan Xiang
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China
| | - Jigang Guo
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China
| | - Shaoyang Zhang
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China
| | - Hongguang Wu
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junping Wang
- Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Baojie Li
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China.
| | - Huijuan Liu
- The Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200024, China.
| |
Collapse
|
26
|
Gao Y, Tian T. mTOR Signaling Pathway and Gut Microbiota in Various Disorders: Mechanisms and Potential Drugs in Pharmacotherapy. Int J Mol Sci 2023; 24:11811. [PMID: 37511569 PMCID: PMC10380532 DOI: 10.3390/ijms241411811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) integrates multiple intracellular and extracellular upstream signals involved in the regulation of anabolic and catabolic processes in cells and plays a key regulatory role in cell growth and metabolism. The activation of the mTOR signaling pathway has been reported to be associated with a wide range of human diseases. A growing number of in vivo and in vitro studies have demonstrated that gut microbes and their complex metabolites can regulate host metabolic and immune responses through the mTOR pathway and result in disorders of host physiological functions. In this review, we summarize the regulatory mechanisms of gut microbes and mTOR in different diseases and discuss the crosstalk between gut microbes and their metabolites and mTOR in disorders in the gastrointestinal tract, liver, heart, and other organs. We also discuss the promising application of multiple potential drugs that can adjust the gut microbiota and mTOR signaling pathways. Despite the limited findings between gut microbes and mTOR, elucidating their relationship may provide new clues for the prevention and treatment of various diseases.
Collapse
Affiliation(s)
- Yuan Gao
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
27
|
Kerem G, Yu X, Ismayi A, Teng B, Udduttula A, Liu C, Yu Z, Tohty D, Zhang JV, Ren PG. Small intestinal microbiota composition altered in obesity-T2DM mice with high salt fed. Sci Rep 2023; 13:8256. [PMID: 37217529 DOI: 10.1038/s41598-023-33909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Obesity has become a global concern because of increasing the risk of many diseases. Alterations in human gut microbiota have been proven to be associated with obesity, yet the mechanism of how the microbiota are altered by high salt diet (HSD) remains obscure. In this study, the changes of Small Intestinal Microbiota (SIM) in obesity-T2DM mice were investigated. High-throughput sequencing was applied for the jejunum microbiota analysis. Results revealed that high salt intake (HS) could suppress the body weight (B.W.) in some extent. In addition, significant T2DM pathological features were revealed in high salt-high food diet (HS-HFD) group, despite of relatively lower food intake. High-throughput sequencing analysis indicated that the F/B ratio in HS intake groups increased significantly (P < 0.001), whereas beneficial bacteria, such as lactic acid or short chain fatty acid producing bacteria, were significantly decreased in HS-HFD group (P < 0.01 or P < 0.05). Furthermore, Halorubrum luteum were observed in small intestine for the first time. Above results preliminary suggested that in obesity-T2DM mice, high dietary salt could aggravate the imbalance of composition of SIM to unhealthy direction.
Collapse
Affiliation(s)
- Goher Kerem
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiangfang Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Aynur Ismayi
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bin Teng
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Anjaneyulu Udduttula
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chang Liu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Zhongjia Yu
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Dilbar Tohty
- Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Jian V Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Key Laboratory of Nanomedicine, Shenzhen, China
| | - Pei-Gen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
28
|
García Mendez D, Sanabria J, Wist J, Holmes E. Effect of Operational Parameters on the Cultivation of the Gut Microbiome in Continuous Bioreactors Inoculated with Feces: A Systematic Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6213-6225. [PMID: 37070710 PMCID: PMC10143624 DOI: 10.1021/acs.jafc.2c08146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 05/03/2023]
Abstract
Since the early 1980s, multiple researchers have contributed to the development of in vitro models of the human gastrointestinal system for the mechanistic interrogation of the gut microbiome ecology. Using a bioreactor for simulating all the features and conditions of the gastrointestinal system is a massive challenge. Some conditions, such as temperature and pH, are readily controlled, but a more challenging feature to simulate is that both may vary in different regions of the gastrointestinal tract. Promising solutions have been developed for simulating other functionalities, such as dialysis capabilities, peristaltic movements, and biofilm growth. This research field is under constant development, and further efforts are needed to drive these models closer to in vivo conditions, thereby increasing their usefulness for studying the gut microbiome impact on human health. Therefore, understanding the influence of key operational parameters is fundamental for the refinement of the current bioreactors and for guiding the development of more complex models. In this review, we performed a systematic search for operational parameters in 229 papers that used continuous bioreactors seeded with human feces. Despite the reporting of operational parameters for the various bioreactor models being variable, as a result of a lack of standardization, the impact of specific operational parameters on gut microbial ecology is discussed, highlighting the advantages and limitations of the current bioreactor systems.
Collapse
Affiliation(s)
- David
Felipe García Mendez
- Australian
National Phenome Centre and Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Harry Perkins Building, Perth, Australia WA6150
| | - Janeth Sanabria
- Australian
National Phenome Centre and Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Harry Perkins Building, Perth, Australia WA6150
- Environmental
Microbiology and Biotechnology Laboratory, Engineering School of Environmental
& Natural Resources, Engineering Faculty, Universidad del Valle—Sede Meléndez, Cali, Colombia 76001
| | - Julien Wist
- Australian
National Phenome Centre and Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Harry Perkins Building, Perth, Australia WA6150
- Chemistry
Department, Universidad del Valle, 76001, Cali, Colombia
| | - Elaine Holmes
- Australian
National Phenome Centre and Computational and Systems Medicine, Health
Futures Institute, Murdoch University, Harry Perkins Building, Perth, Australia WA6150
| |
Collapse
|
29
|
Deyaert S, Moens F, Pirovano W, van den Bogert B, Klaassens ES, Marzorati M, Van de Wiele T, Kleerebezem M, Van den Abbeele P. Development of a reproducible small intestinal microbiota model and its integration into the SHIME®-system, a dynamic in vitro gut model. Front Microbiol 2023; 13:1054061. [PMID: 37008301 PMCID: PMC10063983 DOI: 10.3389/fmicb.2022.1054061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/14/2022] [Indexed: 03/19/2023] Open
Abstract
The human gastrointestinal tract consists of different regions, each characterized by a distinct physiology, anatomy, and microbial community. While the colonic microbiota has received a lot of attention in recent research projects, little is known about the small intestinal microbiota and its interactions with ingested compounds, primarily due to the inaccessibility of this region in vivo. This study therefore aimed to develop and validate a dynamic, long-term simulation of the ileal microbiota using the SHIME®-technology. Essential parameters were identified and optimized from a screening experiment testing different inoculation strategies, nutritional media, and environmental parameters over an 18-day period. Subjecting a synthetic bacterial consortium to the selected conditions resulted in a stable microbiota that was representative in terms of abundance [8.81 ± 0.12 log (cells/ml)], composition and function. Indeed, the observed community mainly consisted of the genera Streptococcus, Veillonella, Enterococcus, Lactobacillus, and Clostridium (qPCR and 16S rRNA gene targeted Illumina sequencing), while nutrient administration boosted lactate production followed by cross-feeding interactions towards acetate and propionate. Furthermore, similarly as in vivo, bile salts were only partially deconjugated and only marginally converted into secondary bile salts. After confirming reproducibility of the small intestinal microbiota model, it was integrated into the established M-SHIME® where it further increased the compositional relevance of the colonic community. This long-term in vitro model provides a representative simulation of the ileal bacterial community, facilitating research of the ileum microbiota dynamics and activity when, for example, supplemented with microbial or diet components. Furthermore, integration of this present in vitro simulation increases the biological relevance of the current M-SHIME® technology.
Collapse
Affiliation(s)
| | | | | | | | | | - Massimo Marzorati
- ProDigest BV, Gent, Belgium
- Center of Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
- *Correspondence: Massimo Marzorati,
| | - Tom Van de Wiele
- ProDigest BV, Gent, Belgium
- Center of Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Michiel Kleerebezem
- Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | | |
Collapse
|
30
|
Zaccaria E, Klaassen T, Alleleyn AME, Boekhorst J, Smokvina T, Kleerebezem M, Troost FJ. Endogenous small intestinal microbiome determinants of transient colonisation efficiency by bacteria from fermented dairy products: a randomised controlled trial. MICROBIOME 2023; 11:43. [PMID: 36879297 PMCID: PMC9990280 DOI: 10.1186/s40168-023-01491-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The effects of fermented food consumption on the small intestine microbiome and its role on host homeostasis are largely uncharacterised as our knowledge on intestinal microbiota relies mainly on faecal samples analysis. We investigated changes in small intestinal microbial composition and functionality, short chain fatty acid (SCFA) profiles, and on gastro-intestinal (GI) permeability in ileostomy subjects upon the consumption of fermented milk products. RESULTS We report the results from a randomised, cross-over, explorative study where 16 ileostomy subjects underwent 3, 2-week intervention periods. In each period, they consumed either milk fermented by Lacticaseibacillus rhamnosus CNCM I-3690, or milk fermented by Streptococcus thermophilus CNCM I-1630 and Lactobacillus delbrueckii subsp. bulgaricus CNCM I-1519, or a chemically acidified milk (placebo) daily. We performed metataxonomic, metatranscriptomic analysis, and SCFA profiling of ileostomy effluents as well as a sugar permeability test to investigate the microbiome impact of these interventions and their potential effect on mucosal barrier function. Consumption of the intervention products impacted the overall small intestinal microbiome composition and functionality, mainly due to the introduction of the product-derived bacteria that reach in several samples 50% of the total microbial community. The interventions did not affect the SCFA levels in ileostoma effluent, or gastro-intestinal permeability and the effects on the endogenous microbial community were negligible. The impact on microbiome composition was highly personalised, and we identified the poorly characterised bacterial family, Peptostreptococcaceae, to be positively associated with a low abundance of the ingested bacteria. Activity profiling of the microbiota revealed that carbon- versus amino acid-derived energy metabolism of the endogenous microbiome could be responsible for the individual-specific intervention effects on the small intestine microbiome composition and function, reflected also on urine microbial metabolites generated through proteolytic fermentation. CONCLUSIONS The ingested bacteria are the main drivers of the intervention effect on the small intestinal microbiota composition. Their transient abundance level is highly personalised and influenced by the energy metabolism of the ecosystem that is reflected by its microbial composition ( http://www. CLINICALTRIALS gov , ID NCT NCT02920294). Video Abstract.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
| | - Tim Klaassen
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Annick M E Alleleyn
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| | - Jos Boekhorst
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands
| | - Tamara Smokvina
- Danone Nutricia Research, Av. De la Vauve, 91767, Palaiseau, France
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, De Elst 1, 6708WD, Wageningen, The Netherlands.
| | - Freddy J Troost
- Food Innovation and Health, Center for Healthy Eating and Food Innovation, Maastricht University, Venlo, 5911AA, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 5800, 6202AZ, Maastricht, The Netherlands
| |
Collapse
|
31
|
Shi M, Song R, Gu L. Different Regulatory Effects of Heated Products and Maillard Reaction Products of Half-Fin Anchovy Hydrolysates on Intestinal Antioxidant Defense in Healthy Animals. Int J Mol Sci 2023; 24:ijms24032355. [PMID: 36768685 PMCID: PMC9917108 DOI: 10.3390/ijms24032355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The oxidative state of intestinal tracts of healthy animals were investigated after short-term intake of half-fin anchovy hydrolysates (HAHp) and their thermal or Maillard reaction products (MRPs). After one month of continuous oral gavage of HAHp, HAHp-heated products (HAHp-H), the MRPs of HAHp with 3% of glucose (HAHp-3%G MRPs), and the MRPs of HAHp with 3% of fructose (HAHp-3%F MRPs) at a dose of 1.0 g/kg of body weight per day into healthy ICR male mice, the concentrations of serum low-density and high-density lipoprotein cholesterol did not significantly change compared to the control group (CK, gavage with saline). Similar results were found for the interleukin-6 concentrations of all groups. By comparison, HAHp-H, HAHp-3%G MRPs, and HAHp-3%F MRPs administration decreased serum tumor necrosis factor-α concentration as compared to the CK group (p < 0.05). No histological damage was observed in the jejunum, ileum, and colonic tissues of all groups. However, HAHp-H treatment induced higher upregulation of Kelch-like ECH-associated protein 1, transcription factors Nrf-2, associated protective phase-II enzymes of NAD(P)H: quinine oxidoreductase-1, and hemoxygenase-1 in colon tissue, as well as higher upregulation of endogenous antioxidant enzymes, including copper/zinc superoxide dismutase, manganese superoxide dismutase, catalase, and glutathione peroxidase 2 than other groups (p < 0.05). Additionally, increases in Nε-carboxymethyllysine expression in the colonic tissues of all groups were consistent with their increased oligopeptide transporter 1 expressions. Our results suggest that the thermal products of HAHp might have a broad application prospect in improving antioxidant defense in vivo in healthy animals.
Collapse
Affiliation(s)
| | - Ru Song
- Correspondence: or ; Tel.: +86-0580-2554-781
| | | |
Collapse
|
32
|
Peng Y, Wei J, Jia X, Luan F, Man M, Ma X, Luo Y, Li Y, Li N, Wang Q, Wang X, Zhou Y, Ji Y, Mu W, Wang J, Wang C, Zhang Q, Yu K, Zhao M, Wang C. Changes in the microbiota in different intestinal segments of mice with sepsis. Front Cell Infect Microbiol 2023; 12:954347. [PMID: 36704101 PMCID: PMC9871835 DOI: 10.3389/fcimb.2022.954347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction The small intestine, as the main digestion and absorption site of the gastrointestinal tract, is often overlooked in studies, and the overall microbiota does not reflect the makeup of the microbiota in different segments of the intestine. Therefore, we aimed to exclude the influence of routine ICU treatment measures on sepsis patients and observed changes in the diversity and abundance of gut microbiota in different intestinal segments of septic mice. Methods The mice were randomly divided into the CLP6h group and the sham group. The contents of the colon and small intestine of the experimental group and the control group were collected after 6 h. Results After CLP, the number and structure of the gut microbiota in the colon changed most obviously, among which Bacteroidetes had the most significant changes. Akkermansia, D.Firmicutes_bacterium_M10_2, Blautia, Bifidobacterium, Lactobacillus, Candidatus_Arthromitus, and Muribaculaceae were changed in the colon. Lactobacillus, Bifidobacterium, Akkermansia, Blautia, Candidatus_Arthromitus, and Lachnospiraceae_NK4A136_group were changed in the small intestine. Discussion Our experiment found that there were different numbers of unique and common gut microbiota in the small intestine and colon after sepsis, and the gut microbiota of the colon changed more drastically after sepsis than the small intestine. Thus, we should focus on protective gut microbiota and mucin-degrading microbes. We hope that these results will provide help for sepsis treatment in the future.
Collapse
Affiliation(s)
- Yahui Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Jieling Wei
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xiaonan Jia
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Feiyu Luan
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Mingyin Man
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xiaohui Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yinghao Luo
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Nana Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Qian Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xibo Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yang Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yuanyuan Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Wenjing Mu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Jun Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Chunying Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Qianqian Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Changsong Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
33
|
Zaccaria E, Klaassen T, Alleleyn AM, Boekhorst J, Chervaux C, Smokvina T, Troost FJ, Kleerebezem M. L. rhamnosus CNCM I-3690 survival, adaptation, and small bowel microbiome impact in human. Gut Microbes 2023; 15:2244720. [PMID: 37589280 PMCID: PMC10438856 DOI: 10.1080/19490976.2023.2244720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/05/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
Fermented foods and beverages are a significant source of dietary bacteria that enter the gastrointestinal (GI) tract. However, little is known about how these microbes survive and adapt to the small intestinal environment. Colony-forming units (CFU) enumeration and viability qPCR of Lacticaseibacillus rhamnosus CNCM I-3690 in the ileal effluent of 10 ileostomy subjects during 12-h post consumption of a dairy product fermented with this strain demonstrated the high level of survival of this strain during human small intestine passage. Metatranscriptome analyses revealed the in situ transcriptome of L. rhamnosus in the small intestine, which was contrasted with transcriptome data obtained from in vitro cultivation. These comparative analyses revealed substantial metabolic adaptations of L. rhamnosus during small intestine transit, including adjustments of carbohydrate metabolism, surface-protein expression, and translation machinery. The prominent presence of L. rhamnosus in the effluent samples did not elicit an appreciable effect on the composition of the endogenous small intestine microbiome, but significantly altered the ecosystem's overall activity profile, particularly of pathways associated with carbohydrate metabolism. Strikingly, two of the previously recognized gut-brain metabolic modules expressed in situ by L. rhamnosus (inositol degradation and glutamate synthesis II) are among the most dominantly enriched activities in the ecosystem's activity profile. This study establishes the survival capacity of L. rhamnosus in the human small intestine and highlights its functional adjustment in situ, which we postulate to play a role in the probiotic effects associated with this strain.
Collapse
Affiliation(s)
- Edoardo Zaccaria
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Tim Klaassen
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Venlo, The Netherlands
| | - Annick M.E. Alleleyn
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Venlo, The Netherlands
| | - Jos Boekhorst
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Tamara Smokvina
- Danone Nutricia Research, Centre Daniel Carasso, Palaiseau, France
| | - Freddy J. Troost
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Michiel Kleerebezem
- Host Microbe Interactomics Group, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
34
|
Nabeh OA. Gut microbiota and cardiac arrhythmia: a pharmacokinetic scope. Egypt Heart J 2022; 74:87. [PMID: 36583819 PMCID: PMC9803803 DOI: 10.1186/s43044-022-00325-2] [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: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dealing with cardiac arrhythmia is a difficult challenge. Choosing between different anti-arrhythmic drugs (AADs) while being cautious about the pro-arrhythmic characteristics of some of these drugs and their diverse interaction with other drugs is a real obstacle. MAIN BODY Gut microbiota (GM), in our bodies, are now being considered as a hidden organ which can regulate our immune system, digest complex food, and secrete bioactive compounds. Yet, GM are encountered in the pathophysiology of arrhythmia and can affect the pharmacokinetics of AADs, as well as some anti-thrombotics, resulting in altering their bioavailability, therapeutic function and may predispose to some of their unpleasant adverse effects. CONCLUSIONS Knowledge of the exact role of GM in the pharmacokinetics of these drugs is now essential for better understanding of the art of arrhythmia management. Also, it will help deciding when to consider probiotics as an adjunctive therapy while treating arrhythmia. This should be discovered in the near future.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- grid.7776.10000 0004 0639 9286Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Mukhtar I, Anwar H, Iftikhar A, Hashem HE, Ali Q, Siddique F. Human targeted phenobarbital presents a poor substrate of gut microbiome deciphering new drug targets beyond pharmacokinetic curbs. BMC Pharmacol Toxicol 2022; 23:85. [DOI: 10.1186/s40360-022-00618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Background
The gut microbiome, a new organ of the body, can potentially alter the pharmacokinetics of orally administered drugs through microbial enzymes. However, absorption of orally administered non-antibiotic drugs by the gut microbiome, during drug-microbiome interaction, is barely addressed. Structural homology studies confirm similar membrane transport proteins in gut epithelial cells and the gut microbiome of the host that may compete for drug substrates with the host itself for its absorbance. Therefore, it is hypothesized that orally administered human targeted phenobarbital may interact and/or be uptake by the gut microbiome during its transit through the small intestine.
Methods
In the current in vivo study, thirty-six male Wistar albino rats were divided into six groups including one control and 5 treatment groups, each having an equal number of rats (n = 6). Phenobarbital was administered orally (single dose of 15 mg/kg bw) to treatment groups. Animals were subsequently sacrificed to harvest microbial mass pallets residing in the small intestine after 2, 3, 4, 5, and 6 h of phenobarbital administration. Phenobarbital absorbance by the microbiome in the microbial lysate was estimated through RP-HPLC–UV at a wavelength of 207 nm.
Results
Maximum phenobarbital absorbance (149.0 ± 5.93 µg) and drug absorbance per milligram of microbial mass (1.19 ± 0.05 µg) were found significantly higher at 4 h of post-administration in comparison to other groups. Percent dose recovery of phenobarbital was 5.73 ± 0.19% at 4 h while the maximum intestinal transit time was 5 h till the drug was absorbed by the microbes. Such results pronounce the idea of the existence of structural homology between membrane transporters of the gut microbiome and intestinal enterocytes of the host that may competitively absorb orally administered phenobarbital during transit in the small intestine. The docking studies revealed that the phenobarbital is a poor substrate for the gut microbiome.
Conclusion
Gut microbiome may competitively absorb the non-antibiotics such as phenobarbital as novel substrates due to the presence of structurally homologous transporting proteins as in enterocytes. This phenomenon suggests the microbiome as a potential candidate that can significantly alter the pharmacokinetics of drugs.
Collapse
|
36
|
Zhang J, Xie Q, Huo X, Liu Z, Da M, Yuan M, Zhao Y, Shen G. Impact of intestinal dysbiosis on breast cancer metastasis and progression. Front Oncol 2022; 12:1037831. [PMID: 36419880 PMCID: PMC9678367 DOI: 10.3389/fonc.2022.1037831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/19/2022] [Indexed: 07/30/2023] Open
Abstract
Breast cancer has a high mortality rate among malignant tumors, with metastases identified as the main cause of the high mortality. Dysbiosis of the gut microbiota has become a key factor in the development, treatment, and prognosis of breast cancer. The many microorganisms that make up the gut flora have a symbiotic relationship with their host and, through the regulation of host immune responses and metabolic pathways, are involved in important physiologic activities in the human body, posing a significant risk to health. In this review, we build on the interactions between breast tissue (including tumor tissue, tissue adjacent to the tumor, and samples from healthy women) and the microbiota, then explore factors associated with metastatic breast cancer and dysbiosis of the gut flora from multiple perspectives, including enterotoxigenic Bacteroides fragilis, antibiotic use, changes in gut microbial metabolites, changes in the balance of the probiotic environment and diet. These factors highlight the existence of a complex relationship between host-breast cancer progression-gut flora. Suggesting that gut flora dysbiosis may be a host-intrinsic factor affecting breast cancer metastasis and progression not only informs our understanding of the role of microbiota dysbiosis in breast cancer development and metastasis, but also the importance of balancing gut flora dysbiosis and clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guoshuang Shen
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, China
| |
Collapse
|
37
|
Piccioni A, Rosa F, Manca F, Pignataro G, Zanza C, Savioli G, Covino M, Ojetti V, Gasbarrini A, Franceschi F, Candelli M. Gut Microbiota and Clostridium difficile: What We Know and the New Frontiers. Int J Mol Sci 2022; 23:13323. [PMID: 36362106 PMCID: PMC9657115 DOI: 10.3390/ijms232113323] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Our digestive system, particularly our intestines, harbors a vast amount of microorganisms, whose genetic makeup is referred to as the microbiome. Clostridium difficile is a spore-forming Gram-positive bacterium, which can cause an infection whose symptoms range from asymptomatic colonization to fearsome complications such as the onset of toxic megacolon. The relationship between gut microbiota and Clostridium difficile infection has been studied from different perspectives. One of the proposed strategies is to be able to specifically identify which types of microbiota alterations are most at risk for the onset of CDI. In this article, we understood once again how crucial the role of the human microbiota is in health and especially how crucial it becomes, in the case of its alteration, for the individual's disease. Clostridium difficile infection is an emblematic example of how a normal and physiological composition of the human microbiome can play a very important role in immune defense against such a fearsome disease.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Federico Rosa
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Manca
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giulia Pignataro
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Christian Zanza
- Foundation of Ospedale Alba-Bra, Department of Anesthesia, Critical Care and Emergency Medicine, Michele and Pietro Ferrero Hospital, 12060 Verduno, Italy
| | - Gabriele Savioli
- Emergency Department, Policlinico Universitario San Matteo, IRCCS, 27100 Pavia, Italy
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Veronica Ojetti
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
38
|
Ou J, Wang Z, Liu X, Song B, Chen J, Li R, Jia X, Huang R, Xiang W, Zhong S. Regulatory effects of marine polysaccharides on gut microbiota dysbiosis: A review. Food Chem X 2022; 15:100444. [PMID: 36211733 PMCID: PMC9532782 DOI: 10.1016/j.fochx.2022.100444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
The gut microbiota dysbiosis is a state which the physiological combinations of flora are transformed into pathological combinations caused by factors such as diets, pollution, and drugs. Increasing evidence shows that dysbiosis is closely related to many diseases. With the continuous development and utilization of marine resources, marine polysaccharides have been found to regulate dysbiosis in many studies. In this review, we introduce the types of dysbiosis and the degree of it caused by different factors. We highlight the regulating effects of marine polysaccharides on dysbiosis as a potential prebiotic. The mechanisms of marine polysaccharides to regulate dysbiosis including protection of intestinal barrier, regulatory effect on gut microbiota, alteration for related metabolites, and some other possible mechanisms were summarized. And we aim to provide some references for the high-value utilization of marine polysaccharides and new targets for the treatment of gut microbiota dysbiosis by this review.
Collapse
Affiliation(s)
- Jieying Ou
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Zhuo Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Xiaofei Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Bingbing Song
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Jianping Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Rui Li
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Xuejing Jia
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
| | - Riming Huang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Wenzhou Xiang
- South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China
- Shenzhen Research Institute, Guangdong Ocean University, Shenzhen 518108, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
39
|
Gulnaz A, Chang JE, Maeng HJ, Shin KH, Lee KR, Chae YJ. A mechanism-based understanding of altered drug pharmacokinetics by gut microbiota. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00600-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
40
|
Hu D, Wang C, Ente M, Zhang K, Zhang D, Li X, Li K, Chu H. Assessment of Adaptation Status of Reintroduced Equus Przewalskii Based on Comparative Analysis of Fecal Bacteria with Those of Captive E. Przewalskii, Domestic Horse and Mongolian Wild Ass. Animals (Basel) 2022; 12:ani12202874. [PMID: 36290262 PMCID: PMC9598124 DOI: 10.3390/ani12202874] [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: 08/25/2022] [Revised: 09/25/2022] [Accepted: 10/17/2022] [Indexed: 11/20/2022] Open
Abstract
Intestinal microbiota play an important role in the survival of the host. However, no study to date has elucidated the adjustment of intestinal microbiota of the host during rewilding. Thus, this study aims to describe the intestinal bacterial community of reintroduced Przewalski’s horse (RPH) after being released into their original habitat for approximately 20 years in comparison with that of captive Przewalski’s horse (CPH), sympatric domestic horse (DH) and Mongolian wild ass (MWA) by sequencing the 16S rRNA gene. The results showed that the prevalent bacterial communities were different among CPHs, RPHs, DHs and MWAs at the family level. NMDS and ANOSIM analysis showed that the pattern of bacterial community composition in captive equines was distinct from that in the wild groups. It is shown that some bacteria had significant differences among different taxa (p < 0.001), such as Firmicutes, Bacteroidetes, Armatimonadetes, Clostrida, Bacteroidia, Clostridiales, Bacteroidales, Rikenellaceae and Bacteroidales_UCG-001. These bacteria were associated with the transition from in captive to in the wild (CPH and RPH), which reflected the change of environmental conditions. Meanwhile, Proteobacteria, Clostridia, Bacilli, Negativicutes, Gammaproteobacteria, Clostridiales, Bacillales, Selenomonadales, Pseudomonadales and Planococcaceae were the changed groups among RPHs, MWAs and DHs, which are related to feeding habits and diseases. Our results clearly showed the differences between intestinal microbiota in reintroduced animals and wild animals and led us to understand the survival state of reintroduced animals in the wild.
Collapse
Affiliation(s)
- Dini Hu
- School of Ecology and Nature Conservation, Beijing Forestry University, Qinghua East Road No.35, Beijing 100083, China
| | - Chen Wang
- Xinjiang Kalamaili Ungulate Nature Reserve Management Center, Changji 831100, China
| | - Make Ente
- Xinjiang Research Centre for Breeding Przewalski’s Horse, Urumqi 831700, China
| | - Ke Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Qinghua East Road No.35, Beijing 100083, China
| | - Dong Zhang
- School of Ecology and Nature Conservation, Beijing Forestry University, Qinghua East Road No.35, Beijing 100083, China
| | - Xuefeng Li
- Xinjiang Research Centre for Breeding Przewalski’s Horse, Urumqi 831700, China
| | - Kai Li
- School of Ecology and Nature Conservation, Beijing Forestry University, Qinghua East Road No.35, Beijing 100083, China
- Correspondence: (K.L.); (H.C.)
| | - Hongjun Chu
- Institute of Forest Ecology, Xinjiang Academy of Forestry, Urumqi 830063, China
- Correspondence: (K.L.); (H.C.)
| |
Collapse
|
41
|
Li Y, Zhang L, Wang Q, Deng Y. Acupuncture for gastrointestinal urticaria: A protocol for systematic review and network meta-analysis. Front Med (Lausanne) 2022; 9:998655. [PMID: 36314007 PMCID: PMC9614266 DOI: 10.3389/fmed.2022.998655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/30/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction The purpose of this review is to evaluate the effectiveness and safety of acupuncture in the treatment of patients with gastrointestinal urticaria (GU) and to provide a clinician's guide to GU treatment options. Methods and analysis We plan to search multiple databases (i.e., PubMed, EMBASE, Springer, Web of Science, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database and Wanfang Database) for studies published before September 1, 2022. We will electronically search for all relevant studies concerning clinical acupuncture treatments of GU, including unpublished conference articles and other gray literature. The language limit of this systematic review is Chinese and English. Any reports of clinical randomized controlled trials of acupuncture for the treatment of GU will be included in the study. Two researchers will perform independent data extraction to increase the quality of the data extraction. The primary outcome was the Urticaria Activity Score 7 (UAS7). Abdominal visual analog scale (VAS) for abdominal pain, dermatological life quality index (DLQI), the total effective rate, recurrence rate, and occurrence of adverse events were secondary outcomes. We will use RevMan V.5.3 statistical software for pairwise meta-analysis and ADDIS V.1.16.8 software for Bayesian network meta-analysis. If feasible, meta-regression and subgroup analyses will also be performed to address the potential causes of inconsistency and heterogeneity. We will conduct a GRADE assessment of the quality of evidence for the interventions included in this review. Discussion This study may validate acupuncture as an alternative therapy for the effective treatment of GU. Trial registration number PROSPERO CRD42022333977.
Collapse
Affiliation(s)
- Yiwei Li
- Sichuan Second Chinese Medicine Hospital, Chengdu, China
| | - Leixiao Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan University West China Hospital, Chengdu, China
| | - Qiong Wang
- Department of Pain Treatment, Suining Central Hospital, Suining, China
| | - Yanli Deng
- Sichuan Second Chinese Medicine Hospital, Chengdu, China
| |
Collapse
|
42
|
Kamakura R, Raza GS, Sodum N, Lehto V, Kovalainen M, Herzig K. Colonic Delivery of Nutrients for Sustained and Prolonged Release of Gut Peptides: A Novel Strategy for Appetite Management. Mol Nutr Food Res 2022; 66:e2200192. [PMID: 35938221 PMCID: PMC9787473 DOI: 10.1002/mnfr.202200192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/17/2022] [Indexed: 12/30/2022]
Abstract
Obesity is one of the major global threats to human health and risk factors for cardiometabolic diseases and certain cancers. Glucagon-like peptide-1 (GLP-1) plays a major role in appetite and glucose homeostasis and recently the USFDA approved GLP-1 agonists for the treatment of obesity and type 2 diabetes. GLP-1 is secreted from enteroendocrine L-cells in the distal part of the gastrointestinal (GI) tract in response to nutrient ingestion. Endogenously released GLP-1 has a very short half-life of <2 min and most of the nutrients are absorbed before reaching the distal GI tract and colon, which hinders the use of nutritional compounds for appetite regulation. The review article focuses on nutrients that endogenously stimulate GLP-1 and peptide YY (PYY) secretion via their receptors in order to decrease appetite as preventive action. In addition, various delivery technologies such as pH-sensitive, mucoadhesive, time-dependent, and enzyme-sensitive systems for colonic targeting of nutrients delivery are described. Sustained colonic delivery of nutritional compounds could be one of the most promising approaches to prevent obesity and associated metabolic diseases by, e.g., sustained GLP-1 release.
Collapse
Affiliation(s)
- Remi Kamakura
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Ghulam Shere Raza
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Nalini Sodum
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsFaculty of Science and ForestryUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Miia Kovalainen
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Karl‐Heinz Herzig
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
- Department of Pediatric Gastroenterology and Metabolic DiseasesPediatric InstitutePoznan University of Medical SciencesPoznań60–572Poland
| |
Collapse
|
43
|
Codutti A, Cremer J, Alim K. Changing Flows Balance Nutrient Absorption and Bacterial Growth along the Gut. PHYSICAL REVIEW LETTERS 2022; 129:138101. [PMID: 36206418 DOI: 10.1103/physrevlett.129.138101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
Small intestine motility and its ensuing flow of luminal content impact both nutrient absorption and bacterial growth. To explore this interdependence we introduce a biophysical description of intestinal flow and absorption. Rooted in observations of mice we identify the average flow velocity as the key control of absorption efficiency and bacterial growth, independent of the exact contraction pattern. We uncover self-regulation of contraction and flow in response to nutrients and bacterial levels to promote efficient absorption while restraining detrimental bacterial overgrowth.
Collapse
Affiliation(s)
- Agnese Codutti
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| | - Jonas Cremer
- Biology Department, Stanford University, Stanford, 94305 California, USA
| | - Karen Alim
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
- Physics Department and CPA, Technische Universität München, 85748 Garching, Germany
| |
Collapse
|
44
|
Choukaife H, Seyam S, Alallam B, Doolaanea AA, Alfatama M. Current Advances in Chitosan Nanoparticles Based Oral Drug Delivery for Colorectal Cancer Treatment. Int J Nanomedicine 2022; 17:3933-3966. [PMID: 36105620 PMCID: PMC9465052 DOI: 10.2147/ijn.s375229] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
As per the WHO, colorectal cancer (CRC) caused around 935,173 deaths worldwide in 2020 in both sexes and at all ages. The available anticancer therapies including chemotherapy, radiotherapy and anticancer drugs are all associated with limited therapeutic efficacy, adverse effects and low chances. This has urged to emerge several novel therapeutic agents as potential therapies for CRC including synthetic and natural materials. Orally administrable and targeted drug delivery systems are attractive strategies for CRC therapy as they minimize the side effects, enhance the efficacy of anticancer drugs. Nevertheless, oral drug delivery till today faces several challenges like poor drug solubility, stability, and permeability. Various oral nano-based approaches and targeted drug delivery systems have been developed recently, as a result of the ability of nanoparticles to control the release of the encapsulant, drug targeting and reduce the number of dosages administered. The unique physicochemical properties of chitosan polymer assist to overcome oral drug delivery barriers and target the colon tumour cells. Chitosan-based nanocarriers offered additional improvements by enhancing the stability, targeting and bioavailability of several anti-colorectal cancer agents. Modified chitosan derivatives also facilitated CRC targeting through strengthening the protection of encapsulant against acidic and enzyme degradation of gastrointestinal track (GIT). This review aims to provide an overview of CRC pathology, therapy and the barriers against oral drug delivery. It also emphasizes the role of nanotechnology in oral drug targeted delivery system and the growing interest towards chitosan and its derivatives. The present review summarizes the relevant works to date that have studied the potential applications of chitosan-based nanocarrier towards CRC treatment.
Collapse
Affiliation(s)
- Hazem Choukaife
- Faculty of Pharmacy, Universiti Sultan Zainal Abidin, Besut Campus, Terengganu, 22200, Malaysia
| | - Salma Seyam
- Faculty of Pharmacy, Universiti Sultan Zainal Abidin, Besut Campus, Terengganu, 22200, Malaysia
| | - Batoul Alallam
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, 13200, Malaysia
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| | - Mulham Alfatama
- Faculty of Pharmacy, Universiti Sultan Zainal Abidin, Besut Campus, Terengganu, 22200, Malaysia
| |
Collapse
|
45
|
Salt-Sensitive Ileal Microbiota Plays a Role in Atrial Natriuretic Peptide Deficiency-Induced Cardiac Injury. Nutrients 2022; 14:nu14153129. [PMID: 35956306 PMCID: PMC9370783 DOI: 10.3390/nu14153129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Atrial natriuretic peptide (ANP) activity deficiency contributes to salt-sensitive hypertension in humans and mice. However, the role of ileal microbiota in salt sensitivity in ANP deficiency-related cardiac injury has not been investigated yet. This study used ANP−/− mice to analyze the role of the salt-sensitive ileal microbiome on cardiac injury. ANP−/− mice showed an increase in blood pressure (BP), the heart weight/body weight (HW/BW) ratio, and cardiac hypertrophy compared with wild-type (WT) mice. ANP deficiency did not impact the histological structure but reduced occludin expression in the ileum. Antibiotics significantly relieved BP and cardiac hypertrophy in ANP−/− mice. A high-salt diet (HSD) increased BP, the HW/BW ratio, and cardiac hypertrophy/fibrosis in WT and ANP−/− mice, and an HSD treatment in ANP−/− mice exacerbated these cardiac parameters. The HSD markedly decreased muscularis layer thickening, villus length, and numbers of Paneth and goblet cells in the ileum of WT and ANP−/− mice. Furthermore, the HSD increased the level of TLR4 and IL-1β in ANP−/− mice ileum compared with WT mice. Antibiotics reduced the HW/BW ratio, cardiac hypertrophy/fibrosis, and the level of TLR4 and IL-1β in the ileum, and rescued the muscularis layer thickening, villus length, and numbers of Paneth and goblet cells in the ileum of HSD-ANP−/− mice. Importantly, ANP deficiency induced the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum on the NSD diet, which was only observed in HSD-induced WT mice but not in WT mice on the NSD. Besides, the HSD significantly enhanced the sum of the percentage of the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum of ANP−/− mice. Ileal microbiota transfer (IMT) from ANP−/− mice to healthy C57BL/6J mice drove Lactobacillus johnsonii and Lactobacillus reuteri colonization in the ileum, which manifested an increase in BP, the HW/BW ratio, cardiac hypertrophy, and ileal pathology compared with IMT from WT mice. The HSD in C57BL/6J mice with IMT from ANP−/− mice drove the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum and further exacerbated the cardiac and ileal pathology. Our results suggest that salt-sensitive ileal microbiota is probably related to ANP deficiency-induced cardiac injury.
Collapse
|
46
|
Hu P, Mao J, Zeng Y, Sun Z, Deng H, Chen C, Sun W, Tang Z. Isolation, Identification, and Function of Rhodotorula mucilaginosa TZR2014 and Its Effects on the Growth and Health of Weaned Piglets. Front Microbiol 2022; 13:922136. [PMID: 35903473 PMCID: PMC9315203 DOI: 10.3389/fmicb.2022.922136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
A red yeast isolated from orange and grape soil and identified by the 26S rDNA sequence analysis revealed that it was Rhodotorula mucilaginosa and named TZR2014. Its biomass and carotenoid production reached a maximum when using the fermentation medium with pH 6.0, containing 5% glucose, 1% peptone, and 1.5% yeast powder. TZR2014 was resistant to 55°C for 15 min, 0.2% pig bile salts for 4 h, and artificial gastric and intestinal fluids. A total of thirty 28-day weaned pigs were divided into three groups, and the piglets were fed a basal diet (CON), a basal diet and orally administered 1 ml 1.0 × 1010 CFU/ml Candida utilis DSM 2361 three times (C. utilis), or a basal diet and orally administered 1 ml 1.0 × 1010 CFU/mL TZR2014 three times daily (R. mucilaginosa) for 4 weeks. Compared with the piglets in the CON group, those in the C. utilis or R. mucilaginosa group reported an increased average daily weight gain and average daily feed intake (P < 0.05) and a decreased feed/gain (P < 0.05). The diarrhea rate of piglets in the R. mucilaginosa group was lower than that in the CON and C. utilis groups (P < 0.05). Compared with that in the CON and C. utilis groups, the R. mucilaginosa group reported an increased ileum villus height (P < 0.05), serum concentration of total antioxidant content, total superoxide dismutase, and glutathione peroxidase and pepsin and lipase activities in the intestinal content, while it reported a decreased serum concentration of malondialdehyde and pH of the intestinal tract (P < 0.05). The relative abundances of Proteobacteria and Megasphaera of caecum in the R. mucilaginosa group were lower than those in the CON and C. utilis groups (P < 0.05). The relative abundances of Prevotella, Ruminococcaceae, Succinivibrio, Rikenellaceae RC9 gut group, and Roseburia of caecum in the R. mucilaginosa group were higher than those in the CON and C. utilis groups (P < 0.05). R. mucilaginosa TZR2014 can produce carotenoids and adapts to the animal's gastrointestinal environment. Oral R. mucilaginosa TZR2014 improved growth performance, enhanced antioxidant capacity, strengthened gastrointestinal digestion, and maintained the intestinal microbiological balance of piglets.
Collapse
Affiliation(s)
- Ping Hu
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Junxia Mao
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yan Zeng
- Fermentation Engineering Department, Hunan Institute of Microbiology, Changsha, China
| | - Zhihong Sun
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Huan Deng
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chen Chen
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Weizhong Sun
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhiru Tang
- Laboratory for Bio-Feed and Animal Nutrition, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
- *Correspondence: Zhiru Tang
| |
Collapse
|
47
|
Akritidou T, Smet C, Akkermans S, Tonti M, Williams J, Van de Wiele T, Van Impe JFM. A protocol for the cultivation and monitoring of ileal gut microbiota surrogates. J Appl Microbiol 2022; 133:1919-1939. [PMID: 35751580 DOI: 10.1111/jam.15684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 11/28/2022]
Abstract
AIMS This research aimed to develop and validate a cultivation and monitoring protocol that is suitable for a surrogate microbial community that accounts for the gut microbiota of the ileum of the small intestine. METHODS AND RESULTS Five bacterial species have been selected as representatives of the ileal gut microbiota and a general anaerobic medium (MS-BHI, as minimally supplemented BHI) has been constructed and validated against BCCM/LGM recommended and commercial media. Moreover, appropriate selective/differential media have been investigated for monitoring each ileal gut microbiota surrogate. Results showed that MS-BHI was highly efficient in displaying individual and collective behavior of the ileal gut microbiota species, when compared with other types of media. Likewise, the selective/differential media managed to identify and describe the behavior of their targeted species. CONCLUSIONS MS-BHI renders a highly efficient, inexpensive and easy-to-prepare cultivation and enumeration alternative for the surrogate ileal microbiota species. Additionally, the selective/differential media can identify and quantify the bacteria of the surrogate ileal microbial community. SIGNIFICANCE AND IMPACT OF STUDY The selected gut microbiota species can represent an in vitro ileal community, forming the basis for future studies on small intestinal microbiota. MS-BHI and the proposed monitoring protocol can be used as a standard for gut microbiota studies that utilize conventional microbiological techniques.
Collapse
Affiliation(s)
- Theodora Akritidou
- BioTeC+, Chemical and Biochemical Process Technology and Control, Department of Chemical Engineering, KU Leuven, Ghent, Belgium
| | - Cindy Smet
- BioTeC+, Chemical and Biochemical Process Technology and Control, Department of Chemical Engineering, KU Leuven, Ghent, Belgium
| | - Simen Akkermans
- BioTeC+, Chemical and Biochemical Process Technology and Control, Department of Chemical Engineering, KU Leuven, Ghent, Belgium
| | - Maria Tonti
- BioTeC+, Chemical and Biochemical Process Technology and Control, Department of Chemical Engineering, KU Leuven, Ghent, Belgium
| | - Jennifer Williams
- School of Biological Sciences, Faculty of Science, Dublin Institute of Technology, Dublin, Ireland
| | - Tom Van de Wiele
- Laboratory of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Jan F M Van Impe
- BioTeC+, Chemical and Biochemical Process Technology and Control, Department of Chemical Engineering, KU Leuven, Ghent, Belgium
| |
Collapse
|
48
|
Luo W, Guo S, Zhou Y, Zhao J, Wang M, Sang L, Chang B, Wang B. Hepatocellular Carcinoma: How the Gut Microbiota Contributes to Pathogenesis, Diagnosis, and Therapy. Front Microbiol 2022; 13:873160. [PMID: 35572649 PMCID: PMC9092458 DOI: 10.3389/fmicb.2022.873160] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/05/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is gaining increasing attention, and the concept of the "gut-liver axis" is gradually being recognized. Leaky gut resulting from injury and/or inflammation can cause the translocation of flora to the liver. Microbiota-associated metabolites and components mediate the activation of a series of signalling pathways, thereby playing an important role in the development of hepatocellular carcinoma (HCC). For this reason, targeting the gut microbiota in the diagnosis, prevention, and treatment of HCC holds great promise. In this review, we summarize the gut microbiota and the mechanisms by which it mediates HCC development, and the characteristic alterations in the gut microbiota during HCC pathogenesis. Furthermore, we propose several strategies to target the gut microbiota for the prevention and treatment of HCC, including antibiotics, probiotics, faecal microbiota transplantation, and immunotherapy.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
- The Second Clinical College, China Medical University, Shenyang, China
| | - Shiqi Guo
- The Second Clinical College, China Medical University, Shenyang, China
| | - Yang Zhou
- The Second Clinical College, China Medical University, Shenyang, China
| | - Jingwen Zhao
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mengyao Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lixuan Sang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Chang
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bingyuan Wang
- Department of Geriatric Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
49
|
Yang Y, Li Y, Xie Y, Qiao S, Yang L, Pan H. Comparative Study on Jejunal Immunity and Microbial Composition of Growing-Period Tibetan Pigs and Duroc × (Landrace × Yorkshire) Pigs. Front Vet Sci 2022; 9:890585. [PMID: 35548051 PMCID: PMC9085446 DOI: 10.3389/fvets.2022.890585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
The gut microbiota plays vital roles in metabolizing nutrient, maintaining the intestinal epithelial barrier but also in modulating immunity. Host genetics and the pig breed are implicated in shaping gut microbiota. Tibetan pig is a unique native Chinese breed and has evolved to manifest a strong disease resistance. However, the immunity and microbiota of growing Tibetan (TP) pigs were still rarely understood. The jejunal immunity phenotype and microbial composition of TP and Duroc × (Landrace × Yorkshire) (DLY) pigs were explored through immunohistochemistry and 16S rRNA sequencing. Higher scores of clusters of differentiation 4 (CD4+) and Toll-like receptor 9 (TLR9) were observed in TP pigs than those of DLY pigs (p < 0.05), as were Interleukin 10 (IL-10) and zonular occludens 1 (ZO-1) (p < 0.01). Similar levels of bacterial richness and diversity were found in the jejunal microbiota of the TP and DLY pigs. However, the TP pigs showed a significantly different microbiome compared to DLY pigs at the genus level (ANOSIM; p < 0.05). Pseudomonas, Stenotrophomonas, Phenylobacterium, and Sandaracinobacter were enriched in DLY pigs (p < 0.05), while the Lactobacillus and Solibacillus had higher abundances in TP pigs than DLY pigs (p < 0.05). Tibetan pigs have “healthier” intestinal microbial communities than DLY pigs. Close relationships were found between jejunal immune performance and the differential bacteria, Lactobacillus can enhance porcine jejunal immunity, while Stenotrophomonas will have a negative impact on porcine gut immunity.
Collapse
Affiliation(s)
- Yuting Yang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing, China
| | - Yongxiang Li
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yanggang Xie
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing, China
| | - Lijie Yang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Centre, China Agricultural University, Beijing, China
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- *Correspondence: Hongbin Pan
| |
Collapse
|
50
|
Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J, Chen ZS. Microbiota in health and diseases. Signal Transduct Target Ther 2022; 7:135. [PMID: 35461318 PMCID: PMC9034083 DOI: 10.1038/s41392-022-00974-4] [Citation(s) in RCA: 1218] [Impact Index Per Article: 406.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
The role of microbiota in health and diseases is being highlighted by numerous studies since its discovery. Depending on the localized regions, microbiota can be classified into gut, oral, respiratory, and skin microbiota. The microbial communities are in symbiosis with the host, contributing to homeostasis and regulating immune function. However, microbiota dysbiosis can lead to dysregulation of bodily functions and diseases including cardiovascular diseases (CVDs), cancers, respiratory diseases, etc. In this review, we discuss the current knowledge of how microbiota links to host health or pathogenesis. We first summarize the research of microbiota in healthy conditions, including the gut-brain axis, colonization resistance and immune modulation. Then, we highlight the pathogenesis of microbiota dysbiosis in disease development and progression, primarily associated with dysregulation of community composition, modulation of host immune response, and induction of chronic inflammation. Finally, we introduce the clinical approaches that utilize microbiota for disease treatment, such as microbiota modulation and fecal microbial transplantation.
Collapse
Affiliation(s)
- Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xuan-Yu Chen
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Dongya Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Chuanxing Xiao
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Jagadish B Koya
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Jilin Li
- Department of Cardiovascular, The Second Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| |
Collapse
|