1
|
Moore M, Whittington HD, Knickmeyer R, Azcarate-Peril MA, Bruno-Bárcena JM. Non-stochastic reassembly of a metabolically cohesive gut consortium shaped by N-acetyl-lactosamine-enriched fibers. Gut Microbes 2025; 17:2440120. [PMID: 39695352 DOI: 10.1080/19490976.2024.2440120] [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: 04/30/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Diet is one of the main factors shaping the human microbiome, yet our understanding of how specific dietary components influence microbial consortia assembly and subsequent stability in response to press disturbances - such as increasing resource availability (feeding rate) - is still incomplete. This study explores the reproducible re-assembly, metabolic interplay, and compositional stability within microbial consortia derived from pooled stool samples of three healthy infants. Using a single-step packed-bed reactor (PBR) system, we assessed the reassembly and metabolic output of consortia exposed to lactose, glucose, galacto-oligosaccharides (GOS), and humanized GOS (hGOS). Our findings reveal that complex carbohydrates, especially those containing low inclusion (~1.25 gL-1) components present in human milk, such as N-acetyl-lactosamine (LacNAc), promote taxonomic, and metabolic stability under varying feeding rates, as shown by diversity metrics and network analysis. Targeted metabolomics highlighted distinct metabolic responses to different carbohydrates: GOS was linked to increased lactate, lactose to propionate, sucrose to butyrate, and CO2, and the introduction of bile salts with GOS or hGOS resulted in butyrate reduction and increased hydrogen production. This study validates the use of single-step PBRs for reliably studying microbial consortium stability and functionality in response to nutritional press disturbances, offering insights into the dietary modulation of microbial consortia and their ecological dynamics.
Collapse
Affiliation(s)
- Madison Moore
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Hunter D Whittington
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Rebecca Knickmeyer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose M Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
2
|
Zhang K, Zhang H, Sun Y, Zhou P, Xue L, Wang Y, Fan M, Qian H, Li Y, Wang L. Aging-associated intestinal dysfunction impairs 5-heptadecylresorcinol absorption: Mechanistic insights into transporter-mediated uptake, barrier integrity and inflammatory regulation. Food Res Int 2025; 211:116499. [PMID: 40356190 DOI: 10.1016/j.foodres.2025.116499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/29/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025]
Abstract
5-heptadecylresorcinol (AR-C17), a key bioactive component of whole grain (WG) wheat and rye, has been shown to exhibit anti-aging properties. However, the intestinal absorption dynamics of AR-C17 across different intestinal segments, as well as the aging-dependent differences in absorption between young and aging mice and their underlying mechanisms, remain poorly understood. In this study, we systematically evaluated AR-C17 absorption in various intestinal regions using an Ussing chamber system and elucidated potential contributing factors. Our results revealed that the jejunum and ileum served as the principal sites for AR-C17 absorption, with total absorption rates (TAR) reaching approximately 60 % in young mice. Conversely, aging mice exhibited significantly diminished AR-C17 absorption, with TAR values reduced to 20-45 % in these segments. Aging-associated alterations in key absorption parameters-including apparent permeability coefficient, transmucosal resistance, and short-circuit current-were observed during AR-C17 absorption, ultimately leading to reduced uptake in aging mice. Histopathological analysis demonstrated aging-associated structural deterioration, characterized by villus damage with irregular crypt architecture in the jejunum and villus vacuolization in the ileum. These morphological changes were accompanied by downregulated expression of Oct1 in the jejunum and Octn2 in the ileum, which collectively contributed to impaired AR-C17 absorption in aging mice. Further mechanistic investigations indicated that the differential absorption of AR-C17 between young and aging mice was closely associated with alterations in transmembrane transporters, inflammation, and tight junction. This study provided compelling evidence that aging-associated intestinal dysfunction significantly attenuated AR-C17 absorption, providing novel insights for optimizing the bioavailability of WG-derived bioactive compounds in the elderly.
Collapse
Affiliation(s)
- Kuiliang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yujie Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Peng Zhou
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lamei Xue
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yu Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
3
|
Babakhani K, Kucinskas AL, Ye X, Giles ED, Sun Y. Aging immunity: unraveling the complex nexus of diet, gut microbiome, and immune function. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00061. [PMID: 40352822 PMCID: PMC12063687 DOI: 10.1097/in9.0000000000000061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is associated with immune senescence and gut dysbiosis, both of which are heavily influenced by the diet. In this review, we summarize current knowledge regarding the impact of diets high in fiber, protein, or fat, as well as different dietary components (tryptophan, omega-3 fatty acids, and galacto-oligosaccharides) on the immune system and the gut microbiome in aging. Additionally, this review discusses how aging alters tryptophan metabolism, contributing to changes in immune function and the gut microbiome. Understanding the relationship between diet, the gut microbiome, and immune function in the context of aging is critical to formulate sound dietary recommendations for older individuals, and these personalized nutritional practices will ultimately improve the health and longevity of the elderly.
Collapse
Affiliation(s)
| | - Amanda L. Kucinskas
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xiangcang Ye
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
4
|
Xiang M, Wu S, Liu M, Zhang B, Xia X, Tan W, Xiang S. Iota-carrageenan oligosaccharide ameliorates DSS-induced colitis in mice by mediating gut microbiota dysbiosis and modulating SCFAs-PI3K-AKT pathway. Inflammopharmacology 2025:10.1007/s10787-025-01718-w. [PMID: 40167852 DOI: 10.1007/s10787-025-01718-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Iota-carrageenan oligosaccharides (iCOs), derived from marine red algae, are traditionally used as antithrombotic and anti-inflammatory agents in folk medicinal practice. Despite the prevailing emphasis on these aspects in their applications, the potential of iCOs as a prebiotic agent for gut health and its subsequent impact on intestinal disorders such as colitis remains largely unexplored. A DSS-induced colitis model was employed in C57BL/6 male mice to analyze the gut microbiota via 16S rRNA sequencing. Fecal microbiota transplantation (FMT) was used to assess the therapeutic effects of iCOs on colitis. RNA sequencing (RNA-Seq) identified pathways and genes affected by iCOs. ELISA measured inflammatory cytokines, while western blot and RT-qPCR evaluated protein and gene expressions, respectively. The iCOs increased beneficial bacteria, such as Lactobacillus, Bifidobacterium, and Akkermansia. They enhanced short-chain fatty acid production and upregulated GPR41, GPR43, and GPR109A mRNA, influencing cytokine secretion. The iCOs reduced mRNA of SPHK1, BDKRB1, LCN2, and so on, potentially through PI3K-Akt pathway inhibition, and promoted tight junction protein expression. Our findings highlight the novel therapeutic potential of iCOs in colitis, indicating a multifaceted approach to treatment that includes gut microbiota modulation, intestinal barrier restoration, and the suppression of inflammatory responses.
Collapse
Affiliation(s)
- Meixian Xiang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China.
| | - Songtao Wu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Hongshan District, No. 16, Huangjiahu West Road, Wuhan, 430065, People's Republic of China
| | - Minxin Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Bin Zhang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, People's Republic of China
| | - Xiankun Xia
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Wenjing Tan
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, Hubei, People's Republic of China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
5
|
Liao J, Wang M, Li H, Li T, Deng Z, Li J, Zheng L, Yan Y, Duan S, Zhang B. Human Milk Oligosaccharide LNnT Promotes Intestinal Epithelial Growth and Maturation During the Early Life of Infant Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6678-6690. [PMID: 40048505 DOI: 10.1021/acs.jafc.4c10055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Lacto-N-neotetraose (LNnT) is a prevalent neutral core human milk oligosaccharides (HMOs) recognized for its numerous benefits to infant health. In infant formula, galactooligosaccharide (GOS) are frequently used as substitutes for HMOs. However, the regulatory roles of LNnT and GOS in early intestinal development are not yet fully understood. This study aims to elucidate the effects of LNnT and GOS on intestinal development during early life. Our findings show that administering LNnT or GOS significantly increased the spleen and liver indices of infant mice at postnatal day 21. Immunofluorescence and qPCR analysis showed that feeding LNnT significantly promoted the proliferation and differentiation of intestinal stem cells (ISCs) in the colon of infant mice at postnatal day 21, and increased the expression of differentiation markers of goblet cells, intestinal epithelial cells, Paneth cells, and intestinal endocrine cells. Conversely, feeding GOS had no significant effect on the proliferation and differentiation of ISCs. Furthermore, intestinal microbiota analysis showed that LNnT increased the microbiota associated with intestinal regeneration and ISCs proliferation and differentiation in infant mice at postnatal day 21. In conclusion, LNnT promoted ISCs proliferation and differentiation in the colon and alters the composition and function of the intestinal microbiota to support intestinal development in infant mice.
Collapse
Affiliation(s)
- Jinqiang Liao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Minghui Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Ting Li
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Yalu Yan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Sufang Duan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| |
Collapse
|
6
|
Xiao Y, Feng Y, Zhao J, Chen W, Lu W. Achieving healthy aging through gut microbiota-directed dietary intervention: Focusing on microbial biomarkers and host mechanisms. J Adv Res 2025; 68:179-200. [PMID: 38462039 PMCID: PMC11785574 DOI: 10.1016/j.jare.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Population aging has become a primary global public health issue, and the prevention of age-associated diseases and prolonging healthy life expectancies are of particular importance. Gut microbiota has emerged as a novel target in various host physiological disorders including aging. Comprehensive understanding on changes of gut microbiota during aging, in particular gut microbiota characteristics of centenarians, can provide us possibility to achieving healthy aging or intervene pathological aging through gut microbiota-directed strategies. AIM OF REVIEW This review aims to summarize the characteristics of the gut microbiota associated with aging, explore potential biomarkers of aging and address microbiota-associated mechanisms of host aging focusing on intestinal barrier and immune status. By summarizing the existing effective dietary strategies in aging interventions, the probability of developing a diet targeting the gut microbiota in future is provided. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key notions: Firstly, gut microbiota has become a new target for regulating health status and lifespan, and its changes are closely related to age. Thus, we summarized aging-associated gut microbiota features at the levels of key genus/species and important metabolites through comparing the microbiota differences among centenarians, elderly people and younger people. Secondly, exploring microbiota biomarkers related to aging and discussing future possibility using dietary regime/components targeted to aging-related microbiota biomarkers promote human healthy lifespan. Thirdly, dietary intervention can effectively improve the imbalance of gut microbiota related to aging, such as probiotics, prebiotics, and postbiotics, but their effects vary among.
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| | - Yingxuan Feng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
7
|
Marcari AB, Paiva AD, Simon CR, Dos Santos MESM. Leaky Gut Syndrome: An Interplay Between Nutrients and Dysbiosis. Curr Nutr Rep 2025; 14:25. [PMID: 39890659 DOI: 10.1007/s13668-025-00614-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE OF REVIEW The gut microbiota (GM) is directly related to health and disease. In this context, disturbances resulting from excessive stress, unbalanced diet, alcohol abuse, and antibiotic use, among other factors, can contribute to microbiota imbalance, with significant impacts on host health. This review provides a comprehensive examination of the literature on the influence of diet on dysbiosis and increased intestinal permeability over the past five years. RECENT FINDINGS Diet can be considered one of the main modulating factors of GM, impacting its composition and functionality. Excessive consumption of simple carbohydrates, saturated fats, and processed foods appears to be directly linked to dysbiosis, which can lead to intestinal hyperpermeability and leaky gut syndrome. On the other hand, diets primarily composed of food groups such as nuts, vegetables, fruits, fish, and poultry in moderate quantities, along with limited consumption of red and processed meats, are associated with a more diverse, healthier, and beneficial GM for the host. It is worth noticing that the use of prebiotics and probiotics, omega-3 supplementation, polyunsaturated fatty acids, and vitamins A, B, C, D, and E can positively modulate the intestinal microbiota by altering its metabolic activity, microbial composition, and improve intestinal barrier function. This review points to a new perspective regarding individualized dietary intervention and the need to integrate it into several aspects of cellular biology, biochemistry, and microbiology to prescribe more effective diets and thus contribute to patients' comprehensive health.
Collapse
Affiliation(s)
- Ana Beatriz Marcari
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Claudio Roberto Simon
- Department of Structural Biology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Maria Emilia Soares Martins Dos Santos
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil.
| |
Collapse
|
8
|
Reis FN, Câmara JVF, Abuna G, Moraes SM, da Silva NDG, Ventura TMO, Araujo TT, Rodrigues CMVBF, Pardi V, Murata RM, Buzalaf MAR. Resveratrol alters oral biofilm in vitro and in vivo. J Dent 2025; 152:105466. [PMID: 39557280 DOI: 10.1016/j.jdent.2024.105466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024] Open
Abstract
OBJECTIVE To evaluate the ability of resveratrol to reduce dental caries in vitro and in vivo. METHODS In part 1, a microcosm biofilm protocol was employed. One hundred twenty-six bovine enamel specimens were treated with: Resveratrol (50, 100, 200, 400 µg/mL), Phosphate buffered saline (negative control), Dimethyl sulfoxide (negative control) and 0.12% Chlorhexidine (positive control). The biofilm was produced from the saliva of 10 volunteers, under 0.2% sucrose exposure for 5 days, and daily treated with the solutions (1 min). At the end of the experimental period, resazurin and viable plate count assays were performed. Enamel demineralization was evaluated by transverse microrradiography (TMR). In part 2, 12 volunteers participated in a triple-blind crossover protocol for 7 weeks, according to the following treatments: 1) 100 mg/L resveratrol; 2) 0.05% NaF (226 mg/L F); 3) 100 mg/L resveratrol + 0.05% NaF; 4) Deionized water (negative control). Biofilm samples were collected from both sides of the mouth 12 h after the use of the solutions. RESULTS Resveratrol at 50 and 200 µg/mL significantly reduced biofilm metabolic activity and mutans streptococci, respectively. Chlorhexidine was an effective treatment to significantly reduce all parameters, being an important antimicrobial and anticaries agent in vitro. Resveratrol alone or associated with NaF modulated several caries-associated bacteria in vivo. CONCLUSION The present study represents the first step regarding the use of resveratrol within the concept of acquired enamel pellicle and biofilm engineering to prevent dental caries.
Collapse
Affiliation(s)
- Fernanda Navas Reis
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil
| | - João Victor Frazão Câmara
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil
| | - Gabriel Abuna
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, United States.
| | - Samanta Mascarenhas Moraes
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil.
| | - Natara Dias Gomes da Silva
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil.
| | - Talita Mendes Oliveira Ventura
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil.
| | - Tamara Teodoro Araujo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil.
| | | | - Vanessa Pardi
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, United States.
| | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, United States.
| | - Marília Afonso Rabelo Buzalaf
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, São Paulo 17012-901, Brazil.
| |
Collapse
|
9
|
Chenafa A, Ji N, Gu Y, Zhao B, Xu L, Zhu Y. Isolation, characterization, and immobilization of β-galactosidase from Klebsiella michiganensis B5582Y for enhanced transgalactosylation. Int J Biol Macromol 2025; 287:138582. [PMID: 39662551 DOI: 10.1016/j.ijbiomac.2024.138582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
β-Galactosidases are highly desirable in various biotechnological applications. However, research on those obtained from Klebsiella strains has been noticeably restricted. The present investigation centers on the isolation, purification, and characterization of a β-galactosidase enzyme derived from Klebsiella michiganensis (GALB5582Y). Additionally, the study aims to immobilize GALB5582Y onto functionalized graphene oxide (GO)-based polystyrene electrospun nanofibrous membranes (ENMs). The ultimate goal is to enhance the enzyme's transgalactosylation and catalytic efficiency, thereby expanding its range of potential applications. The GALB5582Y gene was sequenced, revealing a 3354 bp sequence that encodes 1024 amino acids. This discovery provides vital information about the gene's structural arrangement. The effectiveness of functionalized graphene oxide (GO)-based engineered nanomaterials (ENMs) in immobilising GALB5582Y was confirmed using SEM, FTIR, and XRD investigations. Significant stability was reported during assessments, with the enzyme activity remaining extended. Additionally, it was shown that the enzyme was efficiently distributed across the surface of the ENM. Although there have been breakthroughs in enzyme production and immobilisation techniques, there is still room for improvement in maximizing the effectiveness of GALB5582Y immobilisation and increasing the yield of galactooligosaccharides (GOS). This calls for additional investigation and refinement.
Collapse
Affiliation(s)
- Aicha Chenafa
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Nairu Ji
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Yangyang Gu
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Bingyu Zhao
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Liya Xu
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Yunping Zhu
- Key Laboratory of Green Manufacturing and Biosynthesis of Food Bioactive Substances, China General Chamber of Commerce, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
10
|
Tsuji K, Uchida N, Nakanoh H, Fukushima K, Haraguchi S, Kitamura S, Wada J. The Gut-Kidney Axis in Chronic Kidney Diseases. Diagnostics (Basel) 2024; 15:21. [PMID: 39795549 PMCID: PMC11719742 DOI: 10.3390/diagnostics15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
The gut-kidney axis represents the complex interactions between the gut microbiota and kidney, which significantly impact the progression of chronic kidney disease (CKD) and overall patient health. In CKD patients, imbalances in the gut microbiota promote the production of uremic toxins, such as indoxyl sulfate and p-cresyl sulfate, which impair renal function and contribute to systemic inflammation. Mechanisms like endotoxemia, immune activation and oxidative stress worsen renal damage by activating pro-inflammatory and oxidative pathways. Insights into these mechanisms highlight the impact of gut-derived metabolites, bacterial translocation, and immune response changes on kidney health, suggesting new potential approaches for CKD treatment. Clinical applications, such as dietary interventions, prebiotics, probiotics and fecal microbiota transplantation, are promising in adjusting the gut microbiota to alleviate CKD symptoms and slow disease progression. Current research highlights the clinical relevance of the gut-kidney axis, but further study is essential to clarify these mechanisms' diagnostic biomarkers and optimize therapeutic interventions. This review emphasizes the importance of an integrated approach to CKD management, focusing on the gut microbiota as a therapeutic target to limit kidney injury.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Naruhiko Uchida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | - Soichiro Haraguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nephrology, Aoe Clinic, Okayama 700-8607, Japan
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| |
Collapse
|
11
|
Yoshida K, Kokubo E, Morita S, Sonoki H, Miyaji K. Combination of Inulin and Resistant Dextrin Has Superior Prebiotic Effects and Reduces Gas Production During In Vitro Fermentation of Fecal Samples from Older People. Nutrients 2024; 16:4262. [PMID: 39770884 PMCID: PMC11678394 DOI: 10.3390/nu16244262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Older people are more susceptible to deterioration of the gut microbiota. Prebiotics help improve the gut microbiota. Inulin, a major prebiotic, stimulates the growth of Bifidobacterium; however, it produces a large amount of gas, which leads to abdominal symptoms. METHODS In this study, in vitro fecal fermentation was performed using fecal samples from seven older people (mean subject age, 73.4 years; five men and two women) to examine whether combining inulin with another prebiotic material, resistant dextrin, could lead to decreased gas production and show prebiotic effects. RESULTS The Bifidobacterium counts and short-chain fatty acid production did not differ significantly between the inulin 0.5% group and the inulin 0.25% plus resistant dextrin 0.25% combination group. However, the inulin 0.25% plus resistant dextrin 0.25% combination group had lower gas production than the inulin 0.5% group (p < 0.10). Furthermore, compared with the inulin 0.5% group, the 0.25% combination group showed significantly greater gut microbiota diversity and tended toward a lower pH in the fermentation medium at the end of fermentation (p = 0.09). These effects are believed to be due to the combination of inulin, which is highly selective for Bifidobacterium and rapidly utilized by the gut microbiota, and resistant dextrin, which is slowly utilized by various bacterial genera. CONCLUSIONS These findings suggest that the inulin plus resistant dextrin combination has superior prebiotic effects in older people and causes less gas production than inulin alone.
Collapse
Affiliation(s)
- Kazuma Yoshida
- Health Care & Nutritional Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Kanagawa, Japan; (E.K.); (S.M.); (H.S.); (K.M.)
| | | | | | | | | |
Collapse
|
12
|
Xu L, Li X, Han S, Mu C, Zhu W. Galacto-oligosaccharides regulate intestinal mucosal sialylation to counteract antibiotic-induced mucin dysbiosis. Food Funct 2024; 15:12016-12032. [PMID: 39563647 DOI: 10.1039/d4fo04626a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Intestinal mucin offers a physical barrier to maintain host-commensal homeostasis. Glycosylation is essential for the appropriate functioning of mucin. Galacto-oligosaccharides (GOS) have been used as a prebiotic with proven intestinal benefits, while their regulatory mechanism on mucin remains unclear. This study employed an antibiotic-treated rat model to mimic gut dysbiosis and attempted to restore gut dysbiosis using GOS. The gut microbiome and intestinal mucus O-glycosylations (O-glycans) in the small intestine were profiled by high-throughput sequencing and glycomics. The sialic acid phenotype at the end of O-glycans was further validated with lectin staining. Expressions of key enzymes in sialic acid metabolism and epithelial morphology were determined as well. Antibiotics significantly increased the relative abundance of Escherichia/Shigella and decreased the relative abundance of Lactobacillus. This was accompanied by decreased microbial sialidase activity and increased sialic acid in the digesta, as well as an increase in epithelial sialidase activity. Analysis of key sialylation enzymes showed the upregulation of α 2,6 sialylation (e.g. ST6GALNACs) and downregulation of α 2,3 sialylation (e.g. ST3GALs) after antibiotic treatment. The glycomics results revealed that antibiotics increased core 4 and α 2,6 sialylated O-glycans and decreased core 1, core 3 and α 2,3 sialylated O-glycans in the intestinal mucus of rats, which was further confirmed by lectin staining. Intestinal histology results demonstrated that antibiotic treatment led to the dysbiosis of intestinal mucus homeostasis. To further test the role of microbiota in regulating intestinal mucus sialylation, we supplemented GOS with antibiotics. The results showed that GOS reversed the effects of antibiotics on the gut microbiota and intestinal mucus O-glycans (especially sialylated O-glycans), characterized by an increase of Lactobacillus and α 2,3 sialylated O-glycans and a decrease of Escherichia/Shigella and α 2,6 sialylated O-glycans. What's more, GOS reduced the stimulation of the intestinal mucosa by lipopolysaccharide (LPS) by increasing α 2,3 sialylated intestinal alkaline phosphatase (IAP) to enhance IAP activity, thereby restoring intestinal mucus homeostasis. Overall, GOS counteracts antibiotic-induced mucin deficiency by remedying the gut ecology and changing the mucin sialylation pattern, as reflected by the increase of α 2,3 sialylated O-glycans and the decrease of α 2,6 sialylated O-glycans.
Collapse
Affiliation(s)
- Laipeng Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuibing Han
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Chunlong Mu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB., Canada.
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
13
|
Azcarate-Peril MA. Has the two decades of research on the gut microbiome resulted in making healthier choices? GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e10. [PMID: 39703542 PMCID: PMC11658936 DOI: 10.1017/gmb.2024.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/30/2024] [Accepted: 09/03/2024] [Indexed: 12/21/2024]
Abstract
The gut microbiome is widely recognized for its significant contribution to maintaining human health across all life stages, from infancy to adulthood and beyond. This perspective article focuses on the impacts of well-supported microbiome research on global caesarean delivery rates, breastfeeding practices, and antimicrobial use. The article also explores the impact of dietary choices, particularly those involving ultra-processed foods, on the gut microbiota and their potential contribution to conditions like obesity, metabolic syndrome, and inflammatory diseases. This perspective aims to emphasize the need for updated guidelines and policy interventions to address the increasing global trends of caesarean deliveries, reduced breastfeeding, overuse of antibiotics, and consumption of highly processed foods to counter their adverse effects on gut health.
Collapse
Affiliation(s)
- M. Andrea Azcarate-Peril
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Fan S, Zhang Q, She J, Dai X. Agar oligosaccharides improve the intestinal health of induced-aging mice by maintaining intestinal homeostasis via balancing the ISCs proliferation and differentiation. Eur J Nutr 2024; 64:9. [PMID: 39546038 DOI: 10.1007/s00394-024-03512-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE Aging is a process that accompanies a decline in the function of various tissues and organs, especially affecting intestinal health. Agarose oligosaccharide (AOS) can prolong the lifespan of organisms and protect the intestine in the previous study. It was examined to evaluate the effects of AOS on intestinal health, and the potential associations between intestinal homeostasis and health status were further validated. METHODS D-galactose-induced aging mice were used to investigate the role of AOS in promoting intestinal health by determining intestinal physiology, microbiota and stem cells. RESULTS AOS supplementation decreased the clinical frailty index of aging mice with increasing intestinal length and crypt depth; moreover, it decreased the average flatulence index and PCNA protein content in the intestine. Besides, AOS contributed to the diversity of the gut microbiota by increasing the relative abundance of Bacteroidetes and other bacteria that could produce short-chain fatty acids. Furthermore, AOS affected the expression of proinflammatory factors in aging mice, promoting the proliferative equilibrium of intestinal stem cells. CONCLUSION These findings confirmed that AOS could improve intestinal health in aging mice by maintaining intestinal homeostasis, which provides new insights into the potential application of AOS as a prebiotic.
Collapse
Affiliation(s)
- Shuhang Fan
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Qianyi Zhang
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Jianyi She
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China
| | - Xianjun Dai
- College of Life Sciences, China Jiliang University, Hangzhou, 310018, China.
- Key Laboratory of Specialty Agri-Product Quality and Hazard Controlling Technology of Zhejiang Province, Hangzhou, 310018, China.
| |
Collapse
|
15
|
Cappio Barazzone E, Diard M, Hug I, Larsson L, Slack E. Diagnosing and engineering gut microbiomes. EMBO Mol Med 2024; 16:2660-2677. [PMID: 39468301 PMCID: PMC11554810 DOI: 10.1038/s44321-024-00149-4] [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: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
The microbes, nutrients and toxins that we are exposed to can have a profound effect on the composition and function of the gut microbiome. Thousands of peer-reviewed publications link microbiome composition and function to health from the moment of birth, right through to centenarians, generating a tantalizing glimpse of what might be possible if we could intervene rationally. Nevertheless, there remain relatively few real-world examples where successful microbiome engineering leads to beneficial health effects. Here we aim to provide a framework for the progress needed to turn gut microbiome engineering from a trial-and-error approach to a rational medical intervention. The workflow starts with truly understanding and accurately diagnosing the problems that we are trying to fix, before moving on to developing technologies that can achieve the desired changes.
Collapse
Affiliation(s)
- Elisa Cappio Barazzone
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Médéric Diard
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Isabelle Hug
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Louise Larsson
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland.
- Basel Research Centre for Child Health, Basel, Switzerland.
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
16
|
Liu S, Yin J, Wan D, Yin Y. The Role of Iron in Intestinal Mucus: Perspectives from Both the Host and Gut Microbiota. Adv Nutr 2024; 15:100307. [PMID: 39341502 PMCID: PMC11533511 DOI: 10.1016/j.advnut.2024.100307] [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: 06/02/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
Although research on the role of iron in host immunity has a history spanning decades, it is only relatively recently that attention has been directed toward the biological effects of iron on the intestinal mucus layer, prompted by an evolving understanding of the role of this material in immune defense. The mucus layer, secreted by intestinal goblet cells, covers the intestinal epithelium, and given its unique location, interactions between the host and gut microbiota, as well as among constituent microbiota, occur frequently within the mucus layer. Iron, as an essential nutrient for the vast majority of life forms, regulates immune responses from both the host and microbial perspectives. In this review, we summarize the iron metabolism of both the host and gut microbiota and describe how iron contributes to intestinal mucosal homeostasis via the intestinal mucus layer with respect to both host and constituent gut microbiota. The findings described herein offer a new perspective on iron-mediated intestinal mucosal barrier function.
Collapse
Affiliation(s)
- Shuan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
17
|
Zhang X, Wang L, Xu C, Wang H, Yan A, Zheng Q, Wang K, Qiao X. Intestinal dysbiosis causes spatial memory impairment in alcohol-exposed male mice by inducing neuroinflammation. Exp Neurol 2024; 383:115028. [PMID: 39490622 DOI: 10.1016/j.expneurol.2024.115028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Alcohol abuse damages the brain and triggers cognitive impairment. Intestinal dysbiosis has recently been shown to be involved in psychiatric disorders, which suggests the possibility of intestine-to-brain interactions in the development of alcohol abuse. In this study, chronic intermittent alcohol exposure (CIAE) model was established in C57BL/6 male mice and the spatial memory were detected by Barnes maze (n = 16/group). The fecal microbiota and its metabolites were detected by 16S rDNA sequencing and non-target liquid chromatograph mass spectrometer (LC-MS) (n = 8/group). Effects of alcohol on intestinal barrier and blood-brain barrier (BBB) permeability were detected by Evens blue leakage assay (n = 4/group), and the activation state of microglia and TLR4 expression were conducted by immunofluorescence co-localization (n = 4/group). The morphological changes of microglia were analyzed with Image J Analyze Skeleton software, and the protein levels of TLR4 and inflammatory factors were detected by Western Blot (n = 8/group). Results indicated that alcohol alters the components of fecal microbiota and metabolites, and damages the intestinal barrier and BBB, leading to spatial memory impairment in mice. By giving mice specific prebiotics (n = 16/group), we pointed out that increased endotoxin coming from Gram negative bacteria such as lipopolysaccharides (LPS) cross the BBB to activate microglia and inflammatory pathways in the prefrontal cortical (PFC) and hippocampus (HIP), releasing inflammatory factors and resulting in neuroinflammation. Thus, the fecal microbiota seems to be a potential target in the management of alcoholic brain disease.
Collapse
Affiliation(s)
- Xinlei Zhang
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Lulu Wang
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Chen Xu
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Heng Wang
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - An Yan
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Qingmeng Zheng
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Ke Wang
- School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China
| | - Xiaomeng Qiao
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China.
| |
Collapse
|
18
|
Chaudhary S, Kaur P, Singh TA, Bano KS, Vyas A, Mishra AK, Singh P, Mehdi MM. The dynamic crosslinking between gut microbiota and inflammation during aging: reviewing the nutritional and hormetic approaches against dysbiosis and inflammaging. Biogerontology 2024; 26:1. [PMID: 39441393 DOI: 10.1007/s10522-024-10146-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The early-life gut microbiota (GM) is increasingly recognized for its contributions to human health and disease over time. Microbiota composition, influenced by factors like race, geography, lifestyle, and individual differences, is subject to change. The GM serves dual roles, defending against pathogens and shaping the host immune system. Disruptions in microbial composition can lead to immune dysregulation, impacting defense mechanisms. Additionally, GM aids digestion, releasing nutrients and influencing physiological systems like the liver, brain, and endocrine system through microbial metabolites. Dysbiosis disrupts intestinal homeostasis, contributing to age-related diseases. Recent studies are elucidating the bacterial species that characterize a healthy microbiota, defining what constitutes a 'healthy' colonic microbiota. The present review article focuses on the importance of microbiome composition for the development of homeostasis and the roles of GM during aging and the age-related diseases caused by the alteration in gut microbial communities. This article might also help the readers to find treatments targeting GM for the prevention of various diseases linked to it effectively.
Collapse
Affiliation(s)
- Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Pardeep Kaur
- Department of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Thokchom Arjun Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Kaniz Shahar Bano
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
19
|
Mei L, Wang J, Hao Y, Zeng X, Yang Y, Wu Z, Ji Y. A comprehensive update on the immunoregulatory mechanisms of Akkermansia muciniphila: insights into active ingredients, metabolites, and nutrient-driven modulation. Crit Rev Food Sci Nutr 2024:1-18. [PMID: 39413040 DOI: 10.1080/10408398.2024.2416481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Akkermansia muciniphila (A. muciniphila) has gained recognition as a pioneering probiotic, exhibiting considerable potential to enhance immune conditions across both humans and animals. The health benefits of A. muciniphila are attributed to its various components, including outer membrane proteins (PilQ and Amuc_1100), secreted proteins (P9 and AmTARS), extracellular vesicles, and metabolites such as SCFAs, ornithine lipids, γ-aminobutyric acid, cobalamin, and inosine. The dynamic control of the mucus layer by A. muciniphila plays a crucial role in regulating intestinal mucosal immunity. Furthermore, A. muciniphila modulates immune function by interacting with macrophages, dendritic cells, T lymphocytes, and Paneth cells. Increasing the abundance of A. muciniphila in the gut through nutritional strategies represents a safe and effective means to augment immune function. Various polyphenols, oligosaccharides, and polysaccharides have been shown to elevate the levels of this bacterium, thereby contributing to favorable immunoregulatory outcomes. This paper delves into the latest research advancements related to the probiotic mechanisms of A. muciniphila and provides an overview of the current understanding of how its abundance responds to nutrients. These insights offer a theoretical foundation for the utilization of A. muciniphila in immunoregulation.
Collapse
Affiliation(s)
- Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Dong B, Peng Y, Wang M, Peng C, Li X. Multi-omics integrated analyses indicated that non-polysaccharides of Sijunzi decoction ameliorated spleen deficiency syndrome via regulating microbiota-gut-metabolites axis and exerted synergistic compatibility. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118276. [PMID: 38697408 DOI: 10.1016/j.jep.2024.118276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a classical traditional Chinese medicine formula to invigorating spleen and replenishing qi, Sijunzi decoction (SJZD) is composed of four herbs, which is applied to cure spleen deficiency syndrome (SDS) clinically. The non-polysaccharides (NPSs) of SJZD (SJZD_NPS) are important pharmacodynamic material basis. However, the amelioration mechanism of SJZD_NPS on SDS has not been fully elaborated. Additionally, the contribution of herbs compatibility to efficacy of this formula remains unclear. AIM OF THE STUDY The aim was to explore the underlying mechanisms of SJZD_NPS on improving SDS, and uncover the scientific connotation in SJZD compatibility. MATERIALS AND METHODS A strategy integrating incomplete formulae (called "Chai-fang" in Chinese) comparison, pharmacodynamics, gut microbiome, and metabolome was employed to reveal the role of each herb to SJZD compatibility against SDS. Additionally, the underlying mechanism harbored by SJZD_NPS was further explored through targeted metabolomics, network pharmacology, molecular docking, pseudo-sterile model, and metagenomics. RESULTS SJZD_NPS significantly alleviated diarrhea, disordered secretion of gastrointestinal hormones and neurotransmitters, damage of ileal morphology and intestinal barrier in SDS rats, which was superior to the NPSs of Chai-fang. 16S rRNA gene sequencing and metabolomics analyses revealed that SJZD_NPS effectively restored the disturbed gut microbiota community and abnormal metabolism caused by SDS, showing the most evident recovery. Moreover, SJZD_NPS recalled the levels of partial amino acids, short chain fatty acids and bile acids, which possessed strong binding affinity towards potential targets. The depletion of gut microbiota confirmed that the SDS-amelioration efficacy of SJZD_NPS is dependent on the intact gut microbiome, with the relative abundance of potential probiotics such as Lactobacillus_johnsonii and Lactobacillus_taiwanensis been enriched. CONCLUSION NPSs in SJZD can improve SDS-induced gastrointestinal-nervous system dysfunction through regulating microbiota-gut-metabolites axis, with four herbs exerting synergistic effects, which indicated the compatibility rationality of SJZD.
Collapse
Affiliation(s)
- Bangjian Dong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mengyue Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chongsheng Peng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaobo Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
21
|
Sproten R, Nohr D, Guseva D. Nutritional strategies modulating the gut microbiome as a preventative and therapeutic approach in normal and pathological age-related cognitive decline: a systematic review of preclinical and clinical findings. Nutr Neurosci 2024; 27:1042-1057. [PMID: 38165747 DOI: 10.1080/1028415x.2023.2296727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
CONTEXT The proportion of the elderly population is on the rise across the globe, and with it the prevalence of age-related neurodegenerative diseases. The gut microbiota, whose composition is highly regulated by dietary intake, has emerged as an exciting research field in neurology due to its pivotal role in modulating brain functions via the gut-brain axis. OBJECTIVES We aimed at conducting a systematic review of preclinical and clinical studies investigating the effects of dietary interventions on cognitive ageing in conjunction with changes in gut microbiota composition and functionality. METHODS PubMed and Scopus were searched using terms related to ageing, cognition, gut microbiota and dietary interventions. Studies were screened, selected based on previously determined inclusion and exclusion criteria, and evaluated for methodological quality using recommended risk of bias assessment tools. RESULTS A total of 32 studies (18 preclinical and 14 clinical) were selected for inclusion. We found that most of the animal studies showed significant positive intervention effects on cognitive behavior, while outcomes on cognition, microbiome features, and health parameters in humans were less pronounced. The effectiveness of dietary interventions depended markedly on the age, gender, degree of cognitive decline and baseline microbiome composition of participants. CONCLUSION To harness the full potential of microbiome-inspired nutrition for cognitive health, one of the main challenges remains to better understand the interplay between host, his microbiome, dietary exposures, whilst also taking into account environmental influences. Future research should aim toward making use of host-specific microbiome data to guide the development of personalized therapies.
Collapse
Affiliation(s)
- Rieke Sproten
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Donatus Nohr
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Daria Guseva
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| |
Collapse
|
22
|
Dorsey AF, Roach J, Burten RB, Azcarate-Peril MA, Thompson AL. Intestinal microbiota composition and efficacy of iron supplementation in Peruvian children. Am J Hum Biol 2024; 36:e24058. [PMID: 38420749 DOI: 10.1002/ajhb.24058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Despite repeated public health interventions, anemia prevalence among children remains a concern. We use an evolutionary medicine perspective to examine the intestinal microbiome as a pathway underlying the efficacy of iron-sulfate treatment. This study explores whether gut microbiota composition differs between anemic children who respond and do not respond to treatment at baseline and posttreatment and if specific microbiota taxa remain associated with response to iron supplementation after controlling for relevant inflammatory and pathogenic variables. METHODS Data come from 49 pre-school-aged anemic children living in San Juan de Lurigancho, Lima, Peru. We tested for differences in alpha and beta diversity using QIIME 2 and performed differential abundance testing in DESeq2 in R. We ran multivariate regression models to assess associations between abundance of specific taxa and response while controlling for relevant variables in Stata 17. RESULTS While we found no evidence for gut microbiota diversity associated with child response to iron treatment, we observed several differential abundance patterns between responders and non-responders at both timepoints. Additionally, we present support for a nonzero relationship between lower relative abundance of Barnesiellaceae and response to iron supplementation in samples collected before and after treatment. CONCLUSION While larger studies and more specific approaches are needed to understand the relationship between microbes and anemia in an epidemiological context, this study suggests that investigating nutritional status and pathogen exposure is key to better understanding the gut microbiome and impact of iron fortification.
Collapse
Affiliation(s)
- Achsah F Dorsey
- Department of Anthropology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Jeff Roach
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Rachel B Burten
- Graduate Program in Organismic and Evolutionary Biology, University of Massachusetts, Amherst, Massachusetts, USA
| | - M Andrea Azcarate-Peril
- Center for Gastrointestinal Biology and Disease (CGIBD), Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, UNC Microbiome Core, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amanda L Thompson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Anthropology, University of North Carolina, Chapel Hill, North Carolina, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Chen YJ, Sui X, Wang Y, Zhao ZH, Han TH, Liu YJ, Zhang JN, Zhou P, Yang K, Ye ZH. Preparation, structural characterization, biological activity, and nutritional applications of oligosaccharides. Food Chem X 2024; 22:101289. [PMID: 38544933 PMCID: PMC10966145 DOI: 10.1016/j.fochx.2024.101289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
Oligosaccharides are low-molecular-weight carbohydrates between monosaccharides and polysaccharides. They can be extracted directly from natural products by physicochemical methods or obtained by chemical synthesis or enzymatic reaction. Oligosaccharides have important physicochemical and physiological properties. Their research and production involve many disciplines such as medicine, chemical industry, and biology. Functional oligosaccharides, as an excellent functional food base, can be used as dietary fibrer and prebiotics to enrich the diet; improve the microecology of the gut; exert antitumour, anti-inflammatory, antioxidant, and lipid-lowering properties. Therefore, the industrial applications of oligosaccharides have increased rapidly in the past few years. It has great prospects in the field of food and medicinal chemistry. This review summarized the preparation, structural features and biological activities of oligosaccharides, with particular emphasis on the application of functional oligosaccharides in the food industry and human nutritional health. It aims to inform further research and development of oligosaccharides and food chemistry.
Collapse
Affiliation(s)
- Ya-jing Chen
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Xin Sui
- Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yue Wang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Zhi-hui Zhao
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Tao-hong Han
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Yi-jun Liu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Jia-ning Zhang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| | - Ping Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing 100191, China
| | - Ke Yang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China
| | - Zhi-hong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine, College of Life Science, China Jiliang University, Hangzhou 310018, China
| |
Collapse
|
24
|
Wang K, Duan F, Sun T, Zhang Y, Lu L. Galactooligosaccharides: Synthesis, metabolism, bioactivities and food applications. Crit Rev Food Sci Nutr 2024; 64:6160-6176. [PMID: 36632761 DOI: 10.1080/10408398.2022.2164244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Prebiotics are non-digestible ingredients that exert significant health-promoting effects on hosts. Galactooligosaccharides (GOS) have remarkable prebiotic effects and structural similarity to human milk oligosaccharides. They generally comprise two to eight sugar units, including galactose and glucose, which are synthesized from substrate lactose by microbial β-galactosidase. Enzyme sources from probiotics have received particular interest because of their safety and potential to synthesize specific structures that are particularly metabolized by intestinal probiotics. Owing to advancements in modern analytical techniques, many GOS structures have been identified, which vary in degree of polymerization, glycosidic linkage, and branch location. After intake, GOS adjust gut microbiota which produce short chain fatty acids, and exhibit excellent biological activities. They selectively stimulate the proliferation of probiotics, inhibit the growth and adhesion of pathogenic bacteria, alleviate gastrointestinal, neurological, metabolic and allergic diseases, modulate metabolites production, and adjust ion storage and absorption. Additionally, GOS are safe and stable, with high solubility and clean taste, and thus are widely used as food additives. GOS can improve the appearance, flavor, taste, texture, viscosity, rheological properties, shelf life, and health benefits of food products. This review systemically covers GOS synthesis, structure identifications, metabolism mechanisms, prebiotic bioactivities and wide applications, focusing on recent advances.
Collapse
Affiliation(s)
- Ke Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyu Duan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Sun
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Lu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Stribling & Ibrahim 2023: Commentary to the Editor. Clin Nutr ESPEN 2024; 61:449-450. [PMID: 38777468 DOI: 10.1016/j.clnesp.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/16/2024] [Indexed: 05/25/2024]
|
26
|
Chen L, Zhao L, Zhang G, Li Z, Qu L, Luo L. Long-term administration of royal jelly regulates age-related disorders and improves gut function in naturally aging mice. Food Funct 2024; 15:5272-5286. [PMID: 38629388 DOI: 10.1039/d4fo00781f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
A natural aging mouse model can exhibit physiological characteristics that closely resemble those of human aging. Through long-term observation, it reflects the occurrence and development of the aging process more accurately. Although numerous beneficial effects of royal jelly (RJ) have been extensively demonstrated in multiple experimental models, the effects of RJ on naturally aging mice have not yet been investigated. In this study, middle-aged male C57BL/6J mice were given RJ for 9 consecutive months to investigate its impact on the intestinal barrier function, gut microbiota, short-chain fatty acids (SCFAs) content and possible mechanisms. The results confirmed that RJ modulated serum lipids by reducing the levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C). Additionally, it protected the liver by increasing antioxidant enzyme levels while decreasing inflammatory cytokines TNF-α (by 51.97%), IL-6 (by 29.73%), and IL-1β (by 43.89%). Furthermore, RJ inhibited the expression of cell cycle-dependent kinase inhibitors including p16, p21, and p53. Importantly, RJ ameliorated gut dysfunctions by inhibiting reduction of tight junction proteins and reducing inflammatory cytokines content in the colon. We also observed an alteration in gut microbiota characterized by an elevated ratio of Firmicutes to Bacteroides (F/B) along with increased abundance of beneficial bacteria, i.e., Lachnospiraceae NK4A136 and Akkermansia. Correlation analysis revealed positive associations between most bacterial genera and SCFAs production. Functional profiling of gut microbiota composition indicated that RJ intervention regulated amino acid metabolism, glycan biosynthesis, and cofactor/vitamin metabolism. Overall, our findings provide an effective dietary intervention strategy for modulating age-associated frailty through the modulation of the gut microbiota.
Collapse
Affiliation(s)
- Lili Chen
- Key Laboratory of Geriatric Nutrition and Health Ministry of Education, College of Food and Health, Beijing Technology and Business University, Beijing, 100048, China.
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| | - Li Zhao
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| | - Gaowei Zhang
- Key Laboratory of Geriatric Nutrition and Health Ministry of Education, College of Food and Health, Beijing Technology and Business University, Beijing, 100048, China.
| | - Zhuozhen Li
- Key Laboratory of Geriatric Nutrition and Health Ministry of Education, College of Food and Health, Beijing Technology and Business University, Beijing, 100048, China.
| | - Liangliang Qu
- Key Laboratory of Geriatric Nutrition and Health Ministry of Education, College of Food and Health, Beijing Technology and Business University, Beijing, 100048, China.
| | - Liping Luo
- Key Laboratory of Geriatric Nutrition and Health Ministry of Education, College of Food and Health, Beijing Technology and Business University, Beijing, 100048, China.
| |
Collapse
|
27
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
28
|
van de Put B, de Bruijn WJ, Schols HA. Structural Characterization of Disaccharides Using Cyclic Ion Mobility Spectrometry and Monosaccharide Standards. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1012-1020. [PMID: 38634722 PMCID: PMC11066964 DOI: 10.1021/jasms.4c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
To understand the mode of action of bioactive oligosaccharides, such as prebiotics, in-depth knowledge about all structural features, including monosaccharide composition, linkage type, and anomeric configuration, is necessary. Current analytical techniques provide limited information about structural features within complex mixtures unless preceded by extensive purification. In this study, we propose an approach employing cyclic ion mobility spectrometry (cIMS) for the in-depth characterization of oligosaccharides, here demonstrated for disaccharides. We were able to separate galactose and glucose anomers by exploiting the high ion mobility resolution of cIMS. Using the obtained monosaccharide mobilograms as references, we determined the composition and anomeric configuration of 4β-galactobiose by studying the monosaccharide fragments generated by collision-induced dissociation (CID) before the ion mobility separation. Drift times and individual MS2 spectra of partially resolved reducing-end anomers of 4β-galactobiose, 4β-galactosylglucose (lactose), and 4β-glucosylglucose (cellobiose) were obtained by deconvolution using CID fragmentation induced in the transfer region between the cIMS cell and TOF analyzer. The composition and anomeric configuration of the reducing end anomers of these disaccharides were identified using cIMS2 approaches, where first each anomer was isolated using cIMS and individually fragmented, and the monosaccharide fragments were again separated by cIMS for comparison with monosaccharide standards. With these results we demonstrate the promising application of cIMS for the structural characterization of isomeric oligosaccharides.
Collapse
Affiliation(s)
- Bram van de Put
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| | - Wouter J.C. de Bruijn
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| | - Henk A. Schols
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| |
Collapse
|
29
|
Deng L, Wei SL, Wang L, Huang JQ. Feruloylated Oligosaccharides Prevented Influenza-Induced Lung Inflammation via the RIG-I/MAVS/TRAF3 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9782-9794. [PMID: 38597360 DOI: 10.1021/acs.jafc.3c09390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-β, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.
Collapse
Affiliation(s)
- Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Shu-Lei Wei
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Lu Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jun-Qing Huang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
30
|
Cook CV, Lighty AM, Smith BJ, Ford Versypt AN. A review of mathematical modeling of bone remodeling from a systems biology perspective. FRONTIERS IN SYSTEMS BIOLOGY 2024; 4:1368555. [PMID: 40012834 PMCID: PMC11864782 DOI: 10.3389/fsysb.2024.1368555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Bone remodeling is an essential, delicately balanced physiological process of coordinated activity of bone cells that remove and deposit new bone tissue in the adult skeleton. Due to the complex nature of this process, many mathematical models of bone remodeling have been developed. Each of these models has unique features, but they have underlying patterns. In this review, the authors highlight the important aspects frequently found in mathematical models for bone remodeling and discuss how and why these aspects are included when considering the physiology of the bone basic multicellular unit, which is the term used for the collection of cells responsible for bone remodeling. The review also emphasizes the view of bone remodeling from a systems biology perspective. Understanding the systemic mechanisms involved in remodeling will help provide information on bone pathology associated with aging, endocrine disorders, cancers, and inflammatory conditions and enhance systems pharmacology. Furthermore, some features of the bone remodeling cycle and interactions with other organ systems that have not yet been modeled mathematically are discussed as promising future directions in the field.
Collapse
Affiliation(s)
- Carley V. Cook
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Ariel M. Lighty
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Brenda J. Smith
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, United States
- Department of Obstetrics and Gynecology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ashlee N. Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
31
|
Wang Y, Qu Z, Chu J, Han S. Aging Gut Microbiome in Healthy and Unhealthy Aging. Aging Dis 2024; 16:980-1002. [PMID: 38607737 PMCID: PMC11964416 DOI: 10.14336/ad.2024.0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
The characteristics of human aging manifest in tissue and organ function decline, heightening susceptibility to age-related ailments, thereby presenting novel challenges to fostering and sustaining healthy longevity. In recent years, an abundance of research on human aging has surfaced. Intriguingly, evidence suggests a pervasive correlation among gut microbiota, bodily functions, and chronic diseases. From infancy to later stages of adulthood, healthy individuals witness dynamic shifts in gut microbiota composition. This microbial community is associated with tissue and organ function deterioration (e.g., brain, bones, muscles, immune system, vascular system) and heightened risk of age-related diseases. Thus, we present a narrative review of the aging gut microbiome in both healthy and unhealthy aging contexts. Additionally, we explore the potential for adjustments to physical health based on gut microbiome analysis and how targeting the gut microbiome can potentially slow down the aging process.
Collapse
Affiliation(s)
- Yangyanqiu Wang
- Huzhou Central Hospital, Affiliated Central Hospital Zhejiang University, Huzhou, Zhejiang, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, Zhejiang, China.
| | - Zhanbo Qu
- Huzhou Central Hospital, Affiliated Central Hospital Zhejiang University, Huzhou, Zhejiang, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, Zhejiang, China.
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Zhejiang, China.
| | - Jian Chu
- Huzhou Central Hospital, Affiliated Central Hospital Zhejiang University, Huzhou, Zhejiang, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, Zhejiang, China.
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Zhejiang, China.
| | - Shuwen Han
- Huzhou Central Hospital, Affiliated Central Hospital Zhejiang University, Huzhou, Zhejiang, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, Zhejiang, China.
- Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Zhejiang, China.
| |
Collapse
|
32
|
Wu J, Liang C, Li Y, Zeng Y, Sun X, Jiang P, Chen W, Xiong D, Jin J, Tang S. Engineering and application of LacI mutants with stringent expressions. Microb Biotechnol 2024; 17:e14427. [PMID: 38465475 PMCID: PMC10926051 DOI: 10.1111/1751-7915.14427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Optimal transcriptional regulatory circuits are expected to exhibit stringent control, maintaining silence in the absence of inducers while exhibiting a broad induction dynamic range upon the addition of effectors. In the Plac /LacI pair, the promoter of the lac operon in Escherichia coli is characterized by its leakiness, attributed to the moderate affinity of LacI for its operator target. In response to this limitation, the LacI regulatory protein underwent engineering to enhance its regulatory properties. The M7 mutant, carrying I79T and N246S mutations, resulted in the lac promoter displaying approximately 95% less leaky expression and a broader induction dynamic range compared to the wild-type LacI. An in-depth analysis of each mutation revealed distinct regulatory profiles. In contrast to the wild-type LacI, the M7 mutant exhibited a tighter binding to the operator sequence, as evidenced by surface plasmon resonance studies. Leveraging the capabilities of the M7 mutant, a high-value sugar biosensor was constructed. This biosensor facilitated the selection of mutant galactosidases with approximately a seven-fold improvement in specific activity for transgalactosylation. Consequently, this advancement enabled enhanced biosynthesis of galacto-oligosaccharides (GOS).
Collapse
Affiliation(s)
- Jieyuan Wu
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chaoning Liang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Yufei Li
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yueting Zeng
- School of Life SciencesHebei UniversityBaodingChina
| | - Xu Sun
- Beijing Key Laboratory of Plant Resources Research and DevelopmentBeijing Technology and Business UniversityBeijingChina
| | - Peixia Jiang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Wei Chen
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Dandan Xiong
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jian‐Ming Jin
- Beijing Key Laboratory of Plant Resources Research and DevelopmentBeijing Technology and Business UniversityBeijingChina
| | - Shuang‐Yan Tang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, State Key Laboratory of Microbial ResourcesInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
33
|
Sun Y, Wang X, Li L, Zhong C, Zhang Y, Yang X, Li M, Yang C. The role of gut microbiota in intestinal disease: from an oxidative stress perspective. Front Microbiol 2024; 15:1328324. [PMID: 38419631 PMCID: PMC10899708 DOI: 10.3389/fmicb.2024.1328324] [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: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Recent studies have indicated that gut microbiota-mediated oxidative stress is significantly associated with intestinal diseases such as colorectal cancer, ulcerative colitis, and Crohn's disease. The level of reactive oxygen species (ROS) has been reported to increase when the gut microbiota is dysregulated, especially when several gut bacterial metabolites are present. Although healthy gut microbiota plays a vital role in defending against excessive oxidative stress, intestinal disease is significantly influenced by excessive ROS, and this process is controlled by gut microbiota-mediated immunological responses, DNA damage, and intestinal inflammation. In this review, we discuss the relationship between gut microbiota and intestinal disease from an oxidative stress perspective. In addition, we also provide a summary of the most recent therapeutic approaches for preventing or treating intestinal diseases by modifying gut microbiota.
Collapse
Affiliation(s)
- Yiqi Sun
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xurui Wang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Zhong
- Traditional Chinese Medicine Department of Orthopaedic and Traumatic, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Xiangdong Yang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Mingyue Li
- Special Needs Outpatient Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Yang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
34
|
Bhatt AP, Arnold JW, Awoniyi M, Sun S, Santiago VF, Quintela PH, Walsh K, Ngobeni R, Hansen B, Gulati A, Carroll IM, Azcarate-Peril MA, Fodor AA, Swann J, Bartelt LA. Giardia Antagonizes Beneficial Functions of Indigenous and Therapeutic Intestinal Bacteria during Malnutrition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.575921. [PMID: 38328247 PMCID: PMC10849499 DOI: 10.1101/2024.01.22.575921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Undernutrition in children commonly disrupts the structure and function of the small intestinal microbial community, leading to enteropathies, compromised metabolic health, and impaired growth and development. The mechanisms by which diet and microbes mediate the balance between commensal and pathogenic intestinal flora remain elusive. In a murine model of undernutrition, we investigated the direct interactions Giardia lamblia, a prevalent small intestinal pathogen, on indigenous microbiota and specifically on Lactobacillus strains known for their mucosal and growth homeostatic properties. Our research reveals that Giardia colonization shifts the balance of lactic acid bacteria, causing a relative decrease in Lactobacillus spp . and an increase in Bifidobacterium spp . This alteration corresponds with a decrease in multiple indicators of mucosal and nutritional homeostasis. Additionally, protein-deficient conditions coupled with Giardia infection exacerbate the rise of primary bile acids and susceptibility to bile acid-induced intestinal barrier damage. In epithelial cell monolayers, Lactobacillus spp . mitigated bile acid-induced permeability, showing strain-dependent protective effects. In vivo, L. plantarum, either alone or within a Lactobacillus spp consortium, facilitated growth in protein-deficient mice, an effect attenuated by Giardia , despite not inhibiting Lactobacillus colonization. These results highlight Giardia's potential role as a disruptor of probiotic functional activity, underscoring the imperative for further research into the complex interactions between parasites and bacteria under conditions of nutritional deficiency.
Collapse
|
35
|
Huang S, Li F, Quan C, Jin D. Intestinal flora: a potential pathogenesis mechanism and treatment strategy for type 1 diabetes mellitus. Gut Microbes 2024; 16:2423024. [PMID: 39520706 PMCID: PMC11552262 DOI: 10.1080/19490976.2024.2423024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease characterized by destruction of pancreatic β-cells, leading to insulin deficiency and hyperglycemia, and its incidence is increasing year by year. The pathogenesis of T1DM is complex, mainly including genetic and environmental factors. Intestinal flora is the largest microbial community in the human body and plays a very important role in human health and disease. In recent years, more and more studies have shown that intestinal flora and its metabolites, as an environmental factor, regulate the development of T1DM through various mechanisms such as altering the intestinal mucosal barrier, influencing insulin secretion and body immune regulation. Intestinal flora transplantation, probiotic supplementation, and other approaches to modulate the intestinal flora appear to be potential therapeutic approaches for T1DM. This article reviews the dysbiosis of the intestinal flora in T1DM, the potential mechanisms by which the intestinal flora affects T1DM, as well as discusses potential approaches to treating T1DM by intervening in the intestinal flora.
Collapse
Affiliation(s)
- Shengnan Huang
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
- Department of Immunology and Pathogenic Biology, College of Medicine, Yanbian University, Yanji, China
| | - Fangfang Li
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
- Department of Immunology and Pathogenic Biology, College of Medicine, Yanbian University, Yanji, China
| | - Chunhua Quan
- Central Laboratory, The Affiliated Hospital of Yanbian University, Yanji, China
| | - Dan Jin
- Immunology Biology Key Laboratory, Yanbian University, Yanji, China
- Department of Immunology and Pathogenic Biology, College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
36
|
Bhatt AP, Arnold JW, Awoniyi M, Sun S, Feijoli Santiago V, Coskuner D, Henrique Quintela P, Walsh K, Xiao J, Ngobeni-Nyambi R, Hansen B, Gulati AS, Carroll IM, Azcarate-Peril MA, Fodor AA, Swann J, Bartelt LA. Giardia antagonizes beneficial functions of indigenous and therapeutic intestinal bacteria during protein deficiency. Gut Microbes 2024; 16:2421623. [PMID: 39501168 PMCID: PMC11542603 DOI: 10.1080/19490976.2024.2421623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/06/2024] [Accepted: 10/21/2024] [Indexed: 11/09/2024] Open
Abstract
Undernutrition in children commonly disrupts the structure and function of the small intestinal microbial community, leading to enteropathies, compromised metabolic health, and impaired growth and development. The mechanisms by which diet and microbes mediate the balance between commensal and pathogenic intestinal flora remain elusive. In a murine model of undernutrition, we investigated the direct interactions Giardia lamblia, a prevalent small intestinal pathogen, on indigenous microbiota and specifically on Lactobacillus strains known for their mucosal and growth homeostatic properties. Our research reveals that Giardia colonization shifts the balance of lactic acid bacteria, causing a relative decrease in Lactobacillus spp. and an increase in Bifidobacterium spp. This alteration corresponds with a decrease in multiple indicators of mucosal and nutritional homeostasis. Additionally, protein-deficient conditions coupled with Giardia infection exacerbate the rise of primary bile acids and susceptibility to bile acid-induced intestinal barrier damage. In epithelial cell monolayers, Lactobacillus spp. mitigated bile acid-induced permeability, showing strain-dependent protective effects. In vivo, L. plantarum, either alone or within a Lactobacillus spp consortium, facilitated growth in protein-deficient mice, an effect attenuated by Giardia, despite not inhibiting Lactobacillus colonization. These results highlight Giardia's potential role as a disruptor of probiotic functional activity, underscoring the imperative for further research into the complex interactions between parasites and bacteria under conditions of nutritional deficiency.
Collapse
Affiliation(s)
- Aadra P. Bhatt
- Division of Gastroenterology and Hepatology, Department of Medicine, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason W. Arnold
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Muyiwa Awoniyi
- Department of Gastroenterology Hepatology and Nutrition, Digestive Diseases and Surgery Institute of the Cleveland Clinic Foundation, and Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Shan Sun
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Verônica Feijoli Santiago
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Deniz Coskuner
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Kenneth Walsh
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jamie Xiao
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Brenna Hansen
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ajay S. Gulati
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ian M. Carroll
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M. Andrea Azcarate-Peril
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Microbiome Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Jonathan Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Luther A. Bartelt
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
37
|
Dagher SF, Vaishnav A, Stanley CB, Meilleur F, Edwards BFP, Bruno-Bárcena JM. Structural analysis and functional evaluation of the disordered ß-hexosyltransferase region from Hamamotoa (Sporobolomyces) singularis. Front Bioeng Biotechnol 2023; 11:1291245. [PMID: 38162180 PMCID: PMC10755861 DOI: 10.3389/fbioe.2023.1291245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
Hamamotoa (Sporobolomyces) singularis codes for an industrially important membrane bound ß-hexosyltransferase (BHT), (BglA, UniprotKB: Q564N5) that has applications in the production of natural fibers such as galacto-oligosaccharides (GOS) and natural sugars found in human milk. When heterologously expressed by Komagataella phaffii GS115, BHT is found both membrane bound and soluble secreted into the culture medium. In silico structural predictions and crystal structures support a glycosylated homodimeric enzyme and the presence of an intrinsically disordered region (IDR) with membrane binding potential within its novel N-terminal region (1-110 amino acids). Additional in silico analysis showed that the IDR may not be essential for stable homodimerization. Thus, we performed progressive deletion analyses targeting segments within the suspected disordered region, to determine the N-terminal disorder region's impact on the ratio of membrane-bound to secreted soluble enzyme and its contribution to enzyme activity. The ratio of the soluble secreted to membrane-bound enzyme shifted from 40% to 53% after the disordered N-terminal region was completely removed, while the specific activity was unaffected. Furthermore, functional analysis of each glycosylation site found within the C-terminal domain revealed reduced total secreted protein activity by 58%-97% in both the presence and absence of the IDR, indicating that glycosylation at all four locations is required by the host for the secretion of active enzyme and independent of the removed disordered N-terminal region. Overall, the data provides evidence that the disordered region only partially influences the secretion and membrane localization of BHT.
Collapse
Affiliation(s)
- Suzanne F. Dagher
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, United States
| | - Asmita Vaishnav
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, United States
| | | | - Flora Meilleur
- Neutron Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, United States
| | - Brian F. P. Edwards
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI, United States
| | - José M. Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
38
|
Li T, Yang S, Liu X, Li Y, Gu Z, Jiang Z. Dietary neoagarotetraose extends lifespan and impedes brain aging in mice via regulation of microbiota-gut-brain axis. J Adv Res 2023; 52:119-134. [PMID: 37085001 PMCID: PMC10555787 DOI: 10.1016/j.jare.2023.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 04/23/2023] Open
Abstract
INTRODUCTION Dietary oligosaccharides can impact the gut microbiota and confer tremendous health benefits. OBJECTIVES The aim of this study was to determine the impact of a novel functional oligosaccharide, neoagarotetraose (NAT), on aging in mice. METHODS 8-month-old C57BL/6J mice as the natural aging mice model were orally administered with NAT for 12 months. The preventive effect of NAT in Alzheimer's disease (AD) mice was further evaluated. Aging related indicators, neuropathology, gut microbiota and short-chain fatty acids (SCFAs) in cecal contents were analyzed. RESULTS NAT treatment extended the lifespan of these mice by up to 33.3 %. Furthermore, these mice showed the improved aging characteristics and decreased injuries in cerebral neurons. Dietary NAT significantly delayed DNA damage in the brain, and inhibited reduction of tight junction protein in the colon. A significant increase at gut bacterial genus level (such as Lactobacillus, Butyricimonas, and Akkermansia) accompanied by increasing concentrations of SCFAs in cecal contents was observed after NAT treatment. Functional profiling of gut microbiota composition indicated that NAT treatment regulated the glucolipid and bile acid-related metabolic pathways. Interestingly, NAT treatment ameliorated cognitive impairment, attenuated amyloid-β (Aβ) and Tau pathology, and regulated the gut microbiota composition and SCFAs receptor-related pathway of Alzheimer's disease (AD) mice. CONCLUSION NAT mitigated age-associated cerebral injury in mice through gut-brain axis. The findings provide novel evidence for the effect of NAT on anti-aging, and highlight the potential application of NAT as an effective intervention against age-related diseases.
Collapse
Affiliation(s)
- Tao Li
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shaoqing Yang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoyan Liu
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanxiao Li
- Department of Nutrition and Health, College of Engineering, China Agricultural University, Beijing, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, NY, USA; Greater Bay Area Institute of Precision Medicine (Guangzhou), Nansha District, Guangzhou 511400, China; Institute of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Zhengqiang Jiang
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
| |
Collapse
|
39
|
Zou Y, Yan H, Li C, Wen F, Jize X, Zhang C, Liu S, Zhao Y, Fu Y, Li L, Liu F, Chen J, Li R, Chen X, Tian M. A Pectic Polysaccharide from Codonopsis pilosula Alleviates Inflammatory Response and Oxidative Stress of Aging Mice via Modulating Intestinal Microbiota-Related Gut-Liver Axis. Antioxidants (Basel) 2023; 12:1781. [PMID: 37760084 PMCID: PMC10525188 DOI: 10.3390/antiox12091781] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Aging is a biological process that leads to the progressive deterioration and loss of physiological functions in the human body and results in an increase in morbidity and mortality, and aging-related disease is a major global problem that poses a serious threat to public health. Polysaccharides have been shown to delay aging by reducing oxidative damage, suppressing inflammatory responses, and modulating intestinal microbiota. Our previous studies have shown that polysaccharide CPP-1 extracted from the root of Codonopsis pilosula possesses noticeable anti-oxidant activity in vitro. Thus, in our study, we tested the anti-aging effect of CPP-1 in naturally aging mice (in vivo). Eighteen C57/BL mice (48-week-old, male) were divided into a control group, high-dose CPP-1 group (20 mg/mL), and low-dose CPP-1 group (10 mg/mL). We discovered that CPP-1 can exert a reparative effect on aging stress in the intestine and liver, including alleviating inflammation and oxidative damage. We revealed that CPP-1 supplementation improved the intestinal microbiota composition and repaired the intestinal barrier in the gut. Furthermore, CPP-1 was proved to modulate lipid metabolism and repair hepatocyte injury in the liver by influencing the enterohepatic axis associated with the intestinal microbiota. Therefore, we concluded that CPP-1 prevents and alleviates oxidative stress and inflammatory responses in the intestine and liver of aging mice by modulating the intestinal microbiota-related gut-liver axis to delay aging.
Collapse
Affiliation(s)
- Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Hong Yan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Cenyu Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Fang Wen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Xiaoping Jize
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Chaowen Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Siqi Liu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Yuzhe Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Yuping Fu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China (L.L.)
| | - Fan Liu
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China; (F.L.)
| | - Ji Chen
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China; (F.L.)
| | - Rui Li
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China; (F.L.)
| | - Xingfu Chen
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China; (F.L.)
| | - Mengliang Tian
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China; (F.L.)
| |
Collapse
|
40
|
Zhang L, Yan J, Zhang C, Feng S, Zhan Z, Bao Y, Zhang S, Chao G. Improving intestinal inflammaging to delay aging? A new perspective. Mech Ageing Dev 2023; 214:111841. [PMID: 37393959 DOI: 10.1016/j.mad.2023.111841] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Greying population is becoming an increasingly critical issue for social development. In advanced aging context, organismal multiple tissues and organs experience a progressive deterioration, initially presenting with functional decline, followed by structural disruption and eventually organ failure. The aging of the gut is one of the key links. Decreased gut function leads to reduced nutrient absorption and can perturb systemic metabolic rates. The degeneration of the intestinal structure causes the migration of harmful components such as pathogens and toxins, inducing pathophysiological changes in other organs through the "brain-gut axis" and "liver-gut axis". There is no accepted singular underlying mechanism of aged gut. While the inflamm-aging theory was first proposed in 2000, the mutual promotion of chronic inflammation and aging has attracted much attention. Numerous studies have established that gut microbiome composition, gut immune function, and gut barrier integrity are involved in the formation of inflammaging in the aging gut. Remarkably, inflammaging additionally drives the development of aging-like phenotypes, such as microbiota dysbiosis and impaired intestinal barrier, via a broad array of inflammatory mediators. Here we demonstrate the mechanisms of inflammaging in the gut and explore whether aging-like phenotypes in the gut can be negated by improving gut inflammaging.
Collapse
Affiliation(s)
- Lan Zhang
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China
| | - Junbin Yan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, The Xin Hua Hospital of Zhejiang Province, Hangzhou 310000, China
| | - Chi Zhang
- Endoscopic Center, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China
| | - Shuyan Feng
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Zheli Zhan
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China
| | - Yang Bao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, The Xin Hua Hospital of Zhejiang Province, Hangzhou 310000, China.
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
41
|
Wu Y, Zhuang J, Zhang Q, Zhao X, Chen G, Han S, Hu B, Wu W, Han S. Aging characteristics of colorectal cancer based on gut microbiota. Cancer Med 2023; 12:17822-17834. [PMID: 37548332 PMCID: PMC10524056 DOI: 10.1002/cam4.6414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Aging is one of the factors leading to cancer. Gut microbiota is related to aging and colorectal cancer (CRC). METHODS A total of 11 metagenomic data sets related to CRC were collected from the R package curated Metagenomic Data. After batch effect correction, healthy individuals and CRC samples were divided into three age groups. Ggplot2 and Microbiota Process packages were used for visual description of species composition and PCA in healthy individuals and CRC samples. LEfSe analysis was performed for species relative abundance data in healthy/CRC groups according to age. Spearman correlation coefficient of age-differentiated bacteria in healthy individuals and CRC samples was calculated separately. Finally, the age prediction model and CRC risk prediction model were constructed based on the age-differentiated bacteria. RESULTS The structure and composition of the gut microbiota were significantly different among the three groups. For example, the abundance of Bacteroides vulgatus in the old group was lower than that in the other two groups, the abundance of Bacteroides fragilis increased with aging. In addition, seven species of bacteria whose abundance increases with aging were screened out. Furthermore, the abundance of pathogenic bacteria (Escherichia_coli, Butyricimonas_virosa, Ruminococcus_bicirculans, Bacteroides_fragilis and Streptococcus_vestibularis) increased with aging in CRCs. The abundance of probiotics (Eubacterium_eligens) decreased with aging in CRCs. The age prediction model for healthy individuals based on the 80 age-related differential bacteria and model of CRC patients based on the 58 age-related differential bacteria performed well, with AUC of 0.79 and 0.71, respectively. The AUC of CRC risk prediction model based on 45 disease differential bacteria was 0.83. After removing the intersection between the disease-differentiated bacteria and the age-differentiated bacteria from the healthy samples, the AUC of CRC risk prediction model based on remaining 31 bacteria was 0.8. CRC risk prediction models for each of the three age groups showed no significant difference in accuracy (young: AUC=0.82, middle: AUC=0.83, old: AUC=0.85). CONCLUSION Age as a factor affecting microbial composition should be considered in the application of gut microbiota to predict the risk of CRC.
Collapse
Affiliation(s)
- Yinhang Wu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| | - Jing Zhuang
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| | - Qi Zhang
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
| | - Xingming Zhao
- Institute of Science and Technology for Brain‐Inspired IntelligenceFudan UniversityShanghaiChina
| | - Gong Chen
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| | - Shugao Han
- Second Affiliated Hospital of School of MedicineZhejiang UniversityHangzhouChina
| | - Boyang Hu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| | - Wei Wu
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| | - Shuwen Han
- Huzhou Central HospitalAffiliated Central Hospital Huzhou UniversityHuzhouChina
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive CancerHuzhouChina
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central HospitalHuzhouChina
| |
Collapse
|
42
|
Stolfi C, Pacifico T, Monteleone G, Laudisi F. Impact of Western Diet and Ultra-Processed Food on the Intestinal Mucus Barrier. Biomedicines 2023; 11:2015. [PMID: 37509654 PMCID: PMC10377275 DOI: 10.3390/biomedicines11072015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/19/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The intestinal epithelial barrier plays a key role in the absorption of nutrients and water, in the regulation of the interactions between luminal contents and the underlying immune cells, and in the defense against enteric pathogens. Additionally, the intestinal mucus layer provides further protection due to mucin secretion and maturation by goblet cells, thus representing a crucial player in maintaining intestinal homeostasis. However, environmental factors, such as dietary products, can disrupt this equilibrium, leading to the development of inflammatory intestinal disorders. In particular, ultra-processed food, which is broadly present in the Western diet and includes dietary components containing food additives and/or undergoing multiple industrial processes (such as dry heating cooking), was shown to negatively impact intestinal health. In this review, we summarize and discuss current knowledge on the impact of a Western diet and, in particular, ultra-processed food on the mucus barrier and goblet cell function, as well as potential therapeutic approaches to maintain and restore the mucus layer under pathological conditions.
Collapse
Affiliation(s)
- Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
43
|
Chen Y, Wen Y, Zhu Y, Chen Z, Mu W, Zhao C. Synthesis of bioactive oligosaccharides and their potential health benefits. Crit Rev Food Sci Nutr 2023; 64:10319-10331. [PMID: 37341126 DOI: 10.1080/10408398.2023.2222805] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Oligosaccharides, a low polymerization degree of carbohydrate, possess various physiological activities, such as anti-diabetes, anti-obesity, anti-aging, anti-viral, and gut microbiota regulation, having a widely used in food and medical fields. However, due to the limited natural oligosaccharides, many un-natural oligosaccharides from complex polysaccharides are being studied for amplifying the available pool of oligosaccharides. More recently, various oligosaccharides were developed by using several artificial strategies, such as chemical degradation, enzyme catalysis, and biosynthesis, then they can be applied in various sectors. Moreover, it has gradually become a trend to use biosynthesis to realize the synthesis of oligosaccharides with clear structure. Emerging research has found that un-natural oligosaccharides exert more comprehensive effects against various human diseases through multiple mechanisms. However, these oligosaccharides from various routes have not been critical reviewed and summarized. Therefore, the purpose of this review is to present the various routes of oligosaccharides preparations and healthy effects, with a focus on diabetes, obesity, aging, virus, and gut microbiota. Additionally, the application of multi-omics for these natural and un-natural oligosaccharides has also been discussed. Especially, the multi-omics are needed to apply in various disease models to find out various biomarkers to respond to the dynamic change process of oligosaccharides.
Collapse
Affiliation(s)
- Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
| | - Yuxi Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, Ourense, Spain
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
| | - Zhengxin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
44
|
Zhu CH, Li YX, Xu YC, Wang NN, Yan QJ, Jiang ZQ. Tamarind Xyloglucan Oligosaccharides Attenuate Metabolic Disorders via the Gut-Liver Axis in Mice with High-Fat-Diet-Induced Obesity. Foods 2023; 12:foods12071382. [PMID: 37048202 PMCID: PMC10093524 DOI: 10.3390/foods12071382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
Functional oligosaccharides exert obesity-reducing effects by acting at various pathological sites responsible for the development of obesity. In this study, tamarind xyloglucan oligosaccharides (TXOS) were used to attenuate metabolic disorders via the gut-liver axis in mice with high-fat-diet (HFD)-induced obesity, as determined through LC/MS-MS and 16S rRNA sequencing technology. A TXOS dose equivalent to 0.39 g/kg/day in humans restored the gut microbiota in obese mice, which was in part supported by the key microflora, particularly Bifidobacterium pseudolongum. Moreover, TXOS reduced the abundance of opportunistic pathogen species, such as Klebsiella variicola and Romboutsia ilealis. The bodyweight and weight gain of TXOS-treated (4.8 g/kg per day) mice began to decrease at the 14th week, decreasing by 12.8% and 23.3%, respectively. Sixteen fatty acids were identified as potential biomarkers in the liver, and B. pseudolongum and caprylic acid were found to tightly regulate each other. This was associated with reduced inflammation in the liver, circulation, and adipose tissue and protection from metabolic disorders. The findings of this study indicate that TXOS can significantly increase the gut microbiota diversity of obese mice and restore the HFD-induced dysbiosis of gut microbiota.
Collapse
Affiliation(s)
- Chun-Hua Zhu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yan-Xiao Li
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Engineering, China Agricultural University, Beijing 100083, China
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Yun-Cong Xu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Nan-Nan Wang
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qiao-Juan Yan
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Engineering, China Agricultural University, Beijing 100083, China
| | - Zheng-Qiang Jiang
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
45
|
Mysonhimer AR, Cannavale CN, Bailey MA, Khan NA, Holscher HD. Prebiotic Consumption Alters Microbiota but Not Biological Markers of Stress and Inflammation or Mental Health Symptoms in Healthy Adults: A Randomized, Controlled, Crossover Trial. J Nutr 2023; 153:1283-1296. [PMID: 36841506 DOI: 10.1016/j.tjnut.2023.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/26/2023] [Accepted: 02/08/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Chronic stress contributes to systemic inflammation and diminished mental health. Although animal work suggests strong links with the microbiota-gut-brain axis, clinical trials investigating the effectiveness of prebiotics in improving mental health and reducing inflammation are lacking. OBJECTIVES We aimed to determine fructooligosaccharide (FOS) and galactooligosaccharide (GOS) effects on biological markers of stress and inflammation and mental health symptoms in adults. Secondary outcomes included fecal microbiota and metabolites, digestive function, emotion, and sleep. METHODS Twenty-four healthy adults (25-45 y; 14 females, 10 males; BMI, 29.3 ± 1.8 kg/m2) from central Illinois participated in a 2-period, randomized, controlled, single-blinded crossover trial. Interventions included the prebiotic (PRE) treatment (237 mL/d Lactaid low-fat 1% milk, 5 g/d FOS, 5 g/d GOS) and control (CON) (237 mL/d Lactaid), which were consumed in counterbalanced order for 4 wk each, separated by ≥4-wk washout. Inflammatory markers were measured in blood plasma (>10-h fast) and cortisol in urine. The Depression Anxiety Stress Scales-42 assessed mental health symptoms. Fecal samples were collected for 16S rRNA gene (V4 region) sequencing and analysis. Emotion was measured by rating images from a computer task. Sleep was assessed using 7-d records and accelerometers. Change scores were analyzed using linear mixed models with treatment and baseline covariate as fixed effects and participant ID as the random effect. RESULTS There were no differences in change scores between PRE and CON treatments on biological markers of stress and inflammation or mental health. PRE increased change in percent sequences (q = 0.01) of Actinobacteriota (CON: 0.46 ± 0.70%; PRE: 5.40 ± 1.67%) and Bifidobacterium (CON: -1.72 ± 0.43%; PRE: 4.92 ± 1.53%). There were also no differences in change scores between treatments for microbial metabolites, digestive function, emotion, or sleep quality. CONCLUSIONS FOS+GOS did not affect biological markers of stress and inflammation or mental health symptoms in healthy adults; however, it increased Bifidobacterium. CLINICAL TRIAL REGISTRY NCT04551937, www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
| | | | - Melisa A Bailey
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - Naiman A Khan
- Neuroscience Program, University of Illinois, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA; Department of Kinesiology and Community Health, University of Illinois, Urbana, IL, USA
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
46
|
Infant Fecal Fermentations with Galacto-Oligosaccharides and 2′-Fucosyllactose Show Differential Bifidobacterium longum Stimulation at Subspecies Level. CHILDREN 2023; 10:children10030430. [PMID: 36979988 PMCID: PMC10047592 DOI: 10.3390/children10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
The objective of the current study was to evaluate the potential of 2′-FL and GOS, individually and combined, in beneficially modulating the microbial composition of infant and toddler (12–18 months) feces using the micro-Matrix bioreactor. In addition, the impacts of GOS and 2′-FL, individually and combined, on the outgrowth of fecal bifidobacteria at (sub)species level was investigated using the baby M-SHIME® model. For young toddlers, significant increases in the genera Bifidobacterium, Veillonella, and Streptococcus, and decreases in Enterobacteriaceae, Clostridium XIVa, and Roseburia were observed in all supplemented fermentations. In addition, GOS, and combinations of GOS and 2′-FL, increased Collinsella and decreased Salmonella, whereas 2′-FL, and combined GOS and 2′-FL, decreased Dorea. Alpha diversity increased significantly in infants with GOS and/or 2′-FL, as well as the relative abundances of the genera Veillonella and Akkermansia with 2′-FL, and Lactobacillus with GOS. Combinations of GOS and 2′-FL significantly stimulated Veillonella, Lactobacillus, Bifidobacterium, and Streptococcus. In all supplemented fermentations, Proteobacteria decreased, with the most profound decreases accomplished by the combination of GOS and 2′-FL. When zooming in on the different (sub)species of Bifidobacterium, GOS and 2’-FL were shown to be complementary in stimulating breast-fed infant-associated subspecies of Bifidobacterium longum in a dose-dependent manner: GOS stimulated Bifidobacterium longum subsp. longum, whereas 2′-FL supported outgrowth of Bifidobacterium longum subsp. infantis.
Collapse
|
47
|
Gookin JL, Hartley AN, Aicher KM, Mathews KG, Cullen R, Cullen JM, Callahan BJ, Stowe DM, Seiler GS, Jacob ME, Arnold JW, Azcarate-Peril MA, Stauffer SH. Gallbladder microbiota in healthy dogs and dogs with mucocele formation. PLoS One 2023; 18:e0281432. [PMID: 36763596 PMCID: PMC9916591 DOI: 10.1371/journal.pone.0281432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
To date studies have not investigated the culture-independent microbiome of bile from dogs, a species where aseptic collection of bile under ultrasound guidance is somewhat routine. Despite frequent collection of bile for culture-based diagnosis of bacterial cholecystitis, it is unknown whether bile from healthy dogs harbors uncultivable bacteria or a core microbiota. The answer to this question is critical to understanding the pathogenesis of biliary infection and as a baseline to exploration of other biliary diseases in dogs where uncultivable bacteria could play a pathogenic role. A pressing example of such a disease would be gallbladder mucocele formation in dogs. This prevalent and deadly condition is characterized by excessive secretion of abnormal mucus by the gallbladder epithelium that can eventually lead to rupture of the gallbladder or obstruction of bile flow. The cause of mucocele formation is unknown as is whether uncultivable, and therefore unrecognized, bacteria play any systematic role in pathogenesis. In this study we applied next-generation 16S rRNA gene sequencing to identify the culture-negative bacterial community of gallbladder bile from healthy dogs and gallbladder mucus from dogs with mucocele formation. Integral to our study was the use of 2 separate DNA isolations on each sample using different extraction methods and sequencing of negative control samples enabling recognition and curation of contaminating sequences. Microbiota findings were validated by simultaneous culture-based identification, cytological examination of bile, and fluorescence in-situ hybridization (FISH) performed on gallbladder mucosa. Using culture-dependent, cytological, FISH, and 16S rRNA sequencing approaches, results of our study do not support existence of a core microbiome in the bile of healthy dogs or gallbladder mucus from dogs with mucocele formation. Our findings further document how contaminating sequences can significantly contribute to the results of sequencing analysis when performed on samples with low bacterial biomass.
Collapse
Affiliation(s)
- Jody L. Gookin
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| | - Ashley N. Hartley
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Kathleen M. Aicher
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Kyle G. Mathews
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Rachel Cullen
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| | - John M. Cullen
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Benjamin J. Callahan
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Devorah M. Stowe
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Gabriela S. Seiler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Megan E. Jacob
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jason W. Arnold
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - M. Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Stephen H. Stauffer
- Department of Clinical Sciences, College of Veterinary Medicine and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
48
|
Alam MZ, Maslanka JR, Abt MC. Immunological consequences of microbiome-based therapeutics. Front Immunol 2023; 13:1046472. [PMID: 36713364 PMCID: PMC9878555 DOI: 10.3389/fimmu.2022.1046472] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
The complex network of microscopic organisms living on and within humans, collectively referred to as the microbiome, produce wide array of biologically active molecules that shape our health. Disruption of the microbiome is associated with susceptibility to a range of diseases such as cancer, diabetes, allergy, obesity, and infection. A new series of next-generation microbiome-based therapies are being developed to treat these diseases by transplanting bacteria or bacterial-derived byproducts into a diseased individual to reset the recipient's microbiome and restore health. Microbiome transplantation therapy is still in its early stages of being a routine treatment option and, with a few notable exceptions, has had limited success in clinical trials. In this review, we highlight the successes and challenges of implementing these therapies to treat disease with a focus on interactions between the immune system and microbiome-based therapeutics. The immune activation status of the microbiome transplant recipient prior to transplantation has an important role in supporting bacterial engraftment. Following engraftment, microbiome transplant derived signals can modulate immune function to ameliorate disease. As novel microbiome-based therapeutics are developed, consideration of how the transplants will interact with the immune system will be a key factor in determining whether the microbiome-based transplant elicits its intended therapeutic effect.
Collapse
Affiliation(s)
| | | | - Michael C. Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
49
|
Hu J, Xue S, Xu Z, Wu Z, Xu X, Wang X, Liu G, Lu X, Li B, Liu X. Identification of core cuprotosis-correlated biomarkers in abdominal aortic aneurysm immune microenvironment based on bioinformatics. Front Immunol 2023; 14:1138126. [PMID: 37138870 PMCID: PMC10150024 DOI: 10.3389/fimmu.2023.1138126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Background The occurrence of abdominal aortic aneurysms (AAAs) is related to the disorder of immune microenvironment. Cuprotosis was reported to influence the immune microenvironment. The objective of this study is to identify cuprotosis-related genes involved in the pathogenesis and progression of AAA. Methods Differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) in mouse were identified following AAA through high-throughput RNA sequencing. The enrichment analyses of pathway were selected through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG). The validation of cuprotosis-related genes was conducted through immunofluorescence and western blot analyses. Results Totally, 27616 lncRNAs and 2189 mRNAs were observed to be differentially expressed (|Fold Change| ≥ 2 and q< 0.05) after AAA, including 10424 up-regulated and 17192 down-regulated lncRNAs, 1904 up-regulated and 285 down-regulated mRNAs. Gene ontology and KEGG pathway analysis showed that the DElncRNAs and DEmRNAs were implicated in many different biological processes and pathways. Furthermore, Cuprotosis-related genes (NLRP3, FDX1) were upregulated in the AAA samples compared with the normal one. Conclusion Cuprotosis-related genes (NLRP3,FDX1) involved in AAA immune environment might be critical for providing new insight into identification of potential targets for AAA therapy.
Collapse
Affiliation(s)
- Jiateng Hu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Song Xue
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xintong Xu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xin Wang
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Guang Liu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| | - Xiaobing Liu
- Department of Vascular Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xinwu Lu, ; Bo Li, ; Xiaobing Liu,
| |
Collapse
|
50
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [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: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|