1
|
Zhang Y, Wang A, Zhao W, Qin J, Zhang Y, Liu B, Yao C, Long J, Yuan M, Yan D. Microbial succinate promotes the response to metformin by upregulating secretory immunoglobulin a in intestinal immunity. Gut Microbes 2025; 17:2450871. [PMID: 39812329 PMCID: PMC11740685 DOI: 10.1080/19490976.2025.2450871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Metformin is the first-line pharmacotherapy for type 2 diabetes mellitus; however, many patients respond poorly to this drug in clinical practice. The potential involvement of microbiota-mediated intestinal immunity and related signals in metformin responsiveness has not been previously investigated. In this study, we successfully constructed a humanized mouse model by fecal transplantation of the gut microbiota from clinical metformin-treated - responders and non-responders, and reproduced the difference in clinical phenotypes of responsiveness to metformin. The abundance of Bacteroides thetaiotaomicron, considered a representative differential bacterium of metformin responsiveness, and the level of secretory immunoglobulin A (SIgA) in intestinal immunity increased significantly in responder recipient mice following metformin treatment. In contrast, no significant alterations in B. thetaiotaomicron and SIgA were observed in non-responder recipient mice. The study of IgA-/- mice confirmed that downregulated expression or deficiency of SIgA resulted in non-response to metformin, meaning that metformin was unable to improve dysfunctional glucose metabolism and reduce intestinal and adipose tissue inflammation, ultimately leading to systemic insulin resistance. Furthermore, supplementation with succinate, a microbial product of B. thetaiotaomicron, potentially reversed the non-response to metformin by inducing the production of SIgA. In conclusion, we demonstrated that upregulated SIgA, which could be regulated by succinate, was functionally involved in metformin response through its influence on immune cell-mediated inflammation and insulin resistance. Conversely, an inability to regulate SIgA may result in a lack of response to metformin.
Collapse
Affiliation(s)
- Ying Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiting Wang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zhao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia’an Qin
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bing Liu
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengcheng Yao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingxia Yuan
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Chikazawa M, Minato KI. Intestinal antibody repertoire is altered by diabetes and varies depending on the pathogenesis. Biochem Biophys Rep 2025; 42:101964. [PMID: 40114672 PMCID: PMC11923831 DOI: 10.1016/j.bbrep.2025.101964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/16/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Intestinal immunity is an important system for host defense and it is influenced by various factors such as diet and diseases. To elucidate the relationship between intestinal immunity and type 2 diabetes, we analyzed the effects of diabetes on intestinal antibody production and IgA repertoire using high-fat diet-fed mice and genetically diabetic KK-Ay mice model. The antibody level in the small intestine increased only in KK-Ay mice. We also confirmed that the IgA repertoire in both models experienced significant changes when compared to that in control mice, and no shared characteristics were observed between the two diabetic models. Antibody production in the intestine is influenced by stimuli associated with the onset of diabetes, and the types of induced IgA would differ depending on the process of disease onset.
Collapse
Affiliation(s)
- Miho Chikazawa
- Department of Applied Biological Chemistry, Faculty of Agriculture, Meijo University, 1-501 Shiogamaguchi, Nagoya, Japan
| | - Ken-Ichiro Minato
- Department of Applied Biological Chemistry, Faculty of Agriculture, Meijo University, 1-501 Shiogamaguchi, Nagoya, Japan
| |
Collapse
|
3
|
Lu J, Wang Y, Wu J, Duan Y, Zhang H, Du H. Linking microbial communities to rheumatoid arthritis: focus on gut, oral microbiome and their extracellular vesicles. Front Immunol 2025; 16:1503474. [PMID: 40308573 PMCID: PMC12040682 DOI: 10.3389/fimmu.2025.1503474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Rheumatoid arthritis (RA) is a severe, chronic autoimmune disease affecting approximately 1% of the global population. Research has demonstrated that microorganisms play a crucial role in the onset and progression of RA. This indicates that the disruption of immune homeostasis may originate from mucosal sites, such as the gut and oral cavity. In the intestines of patients in the preclinical stage of RA, an increased abundance of Prevotella species with a strong association to the disease was observed. In the oral cavity, infections by Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans can mediate the production of anti-citrullinated protein antibodies (ACPAs), potentially contributing to RA pathogenesis. Nevertheless, no single bacterial species has been consistently identified as the primary driver of RA. This review will discuss the connection between gut and oral bacteria in the development of arthritis. Additionally, it explores the role of bacterial extracellular vesicles (bEVs) in inducing inflammation and their potential pathogenic roles in RA.
Collapse
Affiliation(s)
- Jian Lu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yi Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing Wu
- Department of Laboratory Medicine, The Affiliated Guangji Hospital of Soochow University, Suzhou Mental Health Center, Suzhou, Jiangsu, China
| | - Yusi Duan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Brum ISC, Cardozo LFMF, Alvarenga L, Kemp JA, Baptista BG, Britto IK, Borges NA, Renouf D, Wong MMY, Fouque D, Mafra D. Fad diets for non-dialysis chronic kidney disease patients: Can "the miracle diet" be a threat? Nutrition 2025; 132:112668. [PMID: 39827647 DOI: 10.1016/j.nut.2024.112668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/29/2024] [Accepted: 12/01/2024] [Indexed: 01/22/2025]
Abstract
In the pursuit of an effortless "miracle cure", there has been a significant increase in the proliferation of fad diets. These diets generally exclude a food group or macronutrients and may also restrict energy intake; they become popular quickly but often lack substantial scientific evidence to support their efficacy and safety. They only show short-term results rather than promoting a lifestyle change. Fad diets are nutritionally unbalanced and can be dangerous for some individuals. Most fad diets are generally restrictive in carbohydrates, high in protein, or unbalanced and have low energy intake, which can harm patients with early stages of chronic kidney damage (CKD) who need a low-protein diet with adequate energy intake. This narrative review discusses the risk of fad diet prescriptions for non-dialysis CKD patients.
Collapse
Affiliation(s)
- Isabela S C Brum
- Graduate Program in Medical Sciences, Federal Fluminense University (UFF), Niteroi, Rio de Janeiro, Brazil
| | - Ludmila F M F Cardozo
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil; Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Livia Alvarenga
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil; Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julie A Kemp
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Beatriz G Baptista
- Graduate Program in Medical Sciences, Federal Fluminense University (UFF), Niteroi, Rio de Janeiro, Brazil
| | - Isadora K Britto
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natália A Borges
- Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dani Renouf
- St. Paul's Hospital, Providence Health Care, Vancouver, Rio de Janeiro, Canada
| | - Michelle M Y Wong
- Division of Nephrology, Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Denis Fouque
- Department of Nephrology, Centre Hopitalier Lyon Sud, Lyon, France
| | - Denise Mafra
- Graduate Program in Medical Sciences, Federal Fluminense University (UFF), Niteroi, Rio de Janeiro, Brazil; Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil; Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
5
|
Hong MG, Song EJ, Yoon HJ, Chung WH, Seo HY, Kim D, Lee D, Seo JG, Lee H, Kim SI, Kim GJ, Kim KN, Lee SN, Kim KS, Nam YD. Clade-specific extracellular vesicles from Akkermansia muciniphila mediate competitive colonization via direct inhibition and immune stimulation. Nat Commun 2025; 16:2708. [PMID: 40108178 PMCID: PMC11923206 DOI: 10.1038/s41467-025-57631-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Akkermansia muciniphila, a promising candidate for next-generation probiotics, exhibits significant genomic diversity, classified into several distinct clades (AmI to AmIV). Notably, a single Akkermansia clade tends to predominate within individual hosts, with co-occurrence of different clades being rare. The mechanisms driving such clade-specific exclusion remain unclear. Here, we show that extracellular vesicles (EVs) derived from AmII clade inhibit the growth of clade I (AmI), conferring a competitive advantage to AmII. Moreover, we observe clade-specific immunoglobulin A (IgA) responses, where AmII clade-specific IgAs, induced by EVs from AmII, facilitate niche occupancy and competitive exclusion of AmI. These findings provide insights into the competitive dynamics of A. muciniphila clades and suggest that future personalized microbiome interventions could be optimized by considering the clade composition of A. muciniphila in individual hosts.
Collapse
Affiliation(s)
- Moon-Gi Hong
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Eun-Ji Song
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Hye Jin Yoon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Won-Hyong Chung
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Hae Yeong Seo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dohak Kim
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Dokyung Lee
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Jae-Gu Seo
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Hayoung Lee
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, Republic of Korea
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seung Il Kim
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Gwang Joong Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
| | - Kil-Nam Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
| | - Sang-Nam Lee
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea.
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Young-Do Nam
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea.
| |
Collapse
|
6
|
Marquez-Paradas E, Torrecillas-Lopez M, Barrera-Chamorro L, del Rio-Vazquez JL, Gonzalez-de la Rosa T, Montserrat-de la Paz S. Microbiota-derived extracellular vesicles: current knowledge, gaps, and challenges in precision nutrition. Front Immunol 2025; 16:1514726. [PMID: 40051622 PMCID: PMC11882860 DOI: 10.3389/fimmu.2025.1514726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
The gut microbiota has co-evolved with its host, profoundly shaping the development and functioning of the immune system. This co-evolution has led to a dynamic relationship where microbial metabolites and molecular signals influence immune maturation, tolerance, and defense mechanisms, highlighting its essential role in maintaining host health. Recently, bacterial extracellular vesicles (BEVs), membrane nanoparticles produced by bacteria, have emerged as important players in gut balance and as potent immune modulators. These vesicles reflect the characteristics of the bacterial membrane and contain nucleic acids, proteins, lipids, and metabolites. They can regulate immune processes and are involved in neurological and metabolic diseases due to their ability to distribute both locally in the gut and systemically, affecting immune responses at both levels. This review provides a comprehensive overview of the characteristics and functional profile of BEVs, detailing how nutrition influences the production and function of these vesicles, how antibiotics can disrupt or alter their composition, and how these factors collectively impact immunity and disease development. It also highlights the potential of BEVs in the development of precision nutritional strategies through dietary modulation, such as incorporating prebiotic fibers to enhance beneficial BEV production, reducing intake of processed foods that may promote harmful BEVs, and tailoring probiotic interventions to influence specific microbial communities and their vesicular outputs.
Collapse
Affiliation(s)
- Elvira Marquez-Paradas
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Maria Torrecillas-Lopez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Luna Barrera-Chamorro
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Jose L. del Rio-Vazquez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Teresa Gonzalez-de la Rosa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| |
Collapse
|
7
|
Al-Herz W, Azizieh F, Raghupathy R. High protein diet increases the risk of allergic sensitization but not asthma in mice through modulation of the cytokine milieu toward Th2 bias. World Allergy Organ J 2025; 18:101031. [PMID: 39995506 PMCID: PMC11848459 DOI: 10.1016/j.waojou.2025.101031] [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: 05/06/2024] [Revised: 12/08/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Introduction The role of different nutrients in allergic sensitization is not clear. In this study we aimed to determine the effect of high protein (HP) diet on allergic sensitization, cytokine profile, and asthma in mice. Methods Seven- to eight-week old female BALB/c mice were fed either normal (ND) or HP diet and were sensitized with ovalbumin intraperitoneally followed by intranasal challenge. Allergic sensitization was tested by measuring anti-ovalbumin (OVA) IgE, IgG1, and IgG2a antibodies. Cytokine levels were tested by multiplex ELISA in splenocyte supernatants after stimulation. Airway inflammation was tested by measuring total and differential cell counts in bronchoalveolar lavage fluid and by measuring bronchial mucus production, goblet cell hyperplasia and perivascular and peribronchial inflammation severity scores by histologic examination. Results Mice fed HP diet had a significant increase in weight and higher levels of OVA-specific IgE and IgG1 antibodies compared to the ND group (P-values 0.002, 0.007 and <0.001, respectively). In addition, they showed a selective Th2 bias in cultured splenocyte supernatants compared to the ND group as demonstrated by higher IL-4 and IL-6 levels (P-values <0.001 and 0.011, respectively) and higher ratios of Th2 to Th1 cytokines. However, the level of airway inflammation was comparable between both groups. Conclusions HP diet increases the risk of allergic sensitization though increase in Th2 cytokines. Efforts should be made to define the upper limit of protein in the diet that does not predispose to allergic sensitization. The effect of diet on health should remain a focus of research for the establishment of optimal health and resilience.
Collapse
Affiliation(s)
- Waleed Al-Herz
- Department of Pediatrics, College of Medicine, Kuwait University, AND Allergy and Clinical Immunology Unit, Pediatric Department, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Fawaz Azizieh
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, And College of Integrative Studies, Abdullah Al Salem University, Kuwait
| | - Raj Raghupathy
- Department of Microbiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
8
|
Huang J, Qin TS, Bo Y, Li YJ, Liu RS, Yu Y, Li XD, He JC, Ma AX, Tao DP, Ren WJ, Peng J. The Role of the Intestinal Flora and Its Derivatives in Neurocognitive Disorders: A Narrative Review from Surgical Perspective. Mol Neurobiol 2025; 62:1404-1414. [PMID: 38985257 PMCID: PMC11772545 DOI: 10.1007/s12035-024-04322-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Perioperative neurocognitive dysfunction is a significant concern for population health, impacting postoperative recovery and increasing the financial burden on patients. With an increasing number of surgical procedures being performed, the prevention and management of perioperative neurocognitive dysfunction have garnered significant attention. While factors such as age, lifestyle, genetics, and education are known to influence the development of cognitive dysfunction, recent research has highlighted the role of the gut microbiota in neurological health. An increased abundance of pro-inflammatory gut microbiota can trigger and worsen neuroinflammation, neuronal cell damage, and impaired cellular autophagy. Moreover, the inflammation-promoting gut microbiota can disrupt immune function, impair neuroautophagy, and affect the production and circulation of extracellular vesicles and neurotransmitters. These factors collectively play a role in the onset and advancement of cognitive impairment. This narrative review delves into the molecular mechanisms through which gut microbiota and their derivatives contribute to cognitive impairment, focusing on the impact of anesthesia surgery, changes in gut microbial populations, and perioperative cognitive impairment associations. The study suggests that alterations in the abundance of various bacterial species and their metabolites pre- and post-surgery may be linked to postoperative cognitive impairment. Furthermore, the potential of probiotics or prebiotics in addressing cognitive impairment is discussed, offering a promising avenue for investigating the treatment of perioperative neurocognitive disorders.
Collapse
Affiliation(s)
- Jian Huang
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Tian-Shou Qin
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Yun Bo
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yu-Jin Li
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Rong-Sheng Liu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Yang Yu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Xiao-Dong Li
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Jin-Can He
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Ai-Xin Ma
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, People's Republic of China
| | - Da-Peng Tao
- School of Information Science and Engineering, Yunnan University, Kunming, 650504, China
| | - Wen-Jun Ren
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China.
| | - Jun Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| |
Collapse
|
9
|
Senior AM, Raubenheimer D, Couteur DGL, Simpson SJ. The Geometric Framework for Nutrition and Its Application to Rodent Models. Annu Rev Anim Biosci 2025; 13:389-410. [PMID: 39546416 DOI: 10.1146/annurev-animal-111523-102327] [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: 11/17/2024]
Abstract
Rodents have been the primary model for mammalian nutritional physiology for decades. Despite an extensive body of literature, controversies remain around the effects of specific nutrients and total energy intake on several aspects of nutritional biology, even in this well-studied model. One approach that is helping to bring clarity to the field is the geometric framework for nutrition (GFN). The GFN is a multidimensional paradigm that can be used to conceptualize nutrition and nutritional effects, design experiments, and interpret results. To date, more than 30 publications have applied the GFN to data from rodent models of nutrition. Here we review the major conclusions from these studies. We pay particular attention to the effects of macronutrients on satiety, glucose metabolism, lifespan and the biology of aging, reproductive function, immune function, and the microbiome. We finish by highlighting several knowledge gaps that became evident upon reviewing this literature.
Collapse
Affiliation(s)
- Alistair M Senior
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, New South Wales, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David Raubenheimer
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - David G Le Couteur
- ANZAC Research Institute, The Concord Hospital, Concord, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| | - Stephen J Simpson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; , , ,
| |
Collapse
|
10
|
Taitz JJ, Tan J, Ni D, Potier-Villette C, Grau G, Nanan R, Macia L. Antibiotic-mediated dysbiosis leads to activation of inflammatory pathways. Front Immunol 2025; 15:1493991. [PMID: 39850904 PMCID: PMC11754057 DOI: 10.3389/fimmu.2024.1493991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction The gut microbiota plays a pivotal role in influencing host health, through the production of metabolites and other key signalling molecules. While the impact of specific metabolites or taxa on host cells is well-documented, the broader impact of a disrupted microbiota on immune homeostasis is less understood, which is particularly important in the context of the increasing overuse of antibiotics. Methods Female C57BL/6 mice were gavaged twice daily for four weeks with Vancomycin, Polymyxin B, or PBS (control). Caecal microbiota composition was assessed via 16S rRNA sequencing and caecal metabolites were quantified with NMR spectroscopy. Immune profiles of spleen and mesenteric lymph nodes (MLNs) were assessed by flow cytometry, and splenocytes assessed for ex vivo cytokine production. A generalised additive model approach was used to examine the relationship between global antibiotic consumption and IBD incidence. Results Antibiotics significantly altered gut microbiota composition, reducing alpha-diversity. Acetate and butyrate were significantly reduced in antibiotic groups, while propionate and succinate increased in Vancomycin and PmB-treated mice, respectively. The MLNs and spleen showed changes only to DC numbers. Splenocytes from antibiotic-treated mice stimulated ex vivo exhibited increased production of TNF. Epidemiological analysis revealed a positive correlation between global antibiotic consumption and IBD incidence. Discussion Our findings demonstrate that antibiotic-mediated dysbiosis results in significantly altered short-chain fatty acid levels but immune homeostasis in spleen and MLNs at steady state is mostly preserved. Non-specific activation of splenocytes ex vivo, however, revealed mice with perturbed microbiota had significantly elevated production of TNF. Thus, this highlights antibiotic-mediated disruption of the gut microbiota may program the host towards dysregulated immune responses, predisposing to the development of TNF-associated autoimmune or chronic inflammatory disease.
Collapse
Affiliation(s)
- Jemma J. Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Georges Grau
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
11
|
Wang X, Zhou XJ, Qiao X, Falchi M, Liu J, Zhang H. The evolving understanding of systemic mechanisms in organ-specific IgA nephropathy: a focus on gut-kidney crosstalk. Theranostics 2025; 15:656-681. [PMID: 39744688 PMCID: PMC11671385 DOI: 10.7150/thno.104631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
The interplay between multiple organs, known as inter-organ crosstalk, represents a complex and essential research domain in understanding the mechanisms and therapies for kidney diseases. The kidneys not only interact pathologically with many other organs but also communicate with other systems through various signaling pathways. It is of paramount importance to comprehend these mechanisms for the development of more efficient therapeutic strategies. Despite extensive research in IgA nephropathy (IgAN), the most common kidney disease, the elaboration mechanism of IgAN remains challenging. Numerous studies suggest that alterations in the intestinal microbiome and its metabolites are pivotal in the progression of IgAN, opening new avenues for understanding its mechanisms. Interestingly, certain presumed probiotics, such as Akkermansia muciniphila, have been implicated in the onset of IgAN, making the exploration of gut microbiota in the context of IgAN pathogenesis even more intriguing. In this review, we summarize the status of gut microbiology studies of IgAN and explore the possible mechanisms and intervention prospects. Future research and treatment directions may increasingly emphasize systemic, multi-organ combined interventions to decelerate the advancement of kidney disease and enhance the overall prognosis of patients.
Collapse
Affiliation(s)
- Xin Wang
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xue Qiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Science, Beijing, 100190, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
12
|
Wu J, Zhang J, Huang G, Zhong Y, Yang Y, Deng P. Evidence from mendelian randomization identifies several causal relationships between primary membranous nephropathy and gut microbiota. Ren Fail 2024; 46:2349136. [PMID: 38770992 PMCID: PMC11110878 DOI: 10.1080/0886022x.2024.2349136] [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: 12/08/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Research has showcased a correlation between disruptions in gut microbiota and primary membranous nephropathy (pMN), giving rise to the concept of the 'gut-kidney axis'. However, the precise relationship between gut microbiota and pMN remains elusive. Hence, this study endeavors to investigate whether a causal relationship exists between gut microbiota and pMN utilizing Mendelian randomization (MR) analysis. METHODS The primary method employed for MR analysis is the inverse variance weighting method, supplemented by MR-Egger and the weighted median method, to infer causality. This approach was validated within the pMN cohort across two distinct populations. RESULTS At the species level, the abundance of Bifidobacterium bifidum and Alistipes indistinctus was negatively correlated with the risk of pMN. Conversely, pMN was positively associated with Bacilli abundance at the class level, Lachnospiraceae abundance at the family level, and Dialister abundance at the genus level. Specifically, at the species level, pMN was positively correlated with the abundance of Ruminococcus lactaris, Dialister invisus, and Coprococcus_sp_ART55_1. CONCLUSION These findings lay the groundwork for future research exploring the interplay between pMN and the gut microbiota, with substantial implications for the prevention and treatment of pMN and its associated complications.
Collapse
Affiliation(s)
- Jianwei Wu
- Department of Medical Technology, Gannan Healthcare Vocational College, Ganzhou, China
| | - Jing Zhang
- Department of Medical Technology, Gannan Healthcare Vocational College, Ganzhou, China
| | - Gang Huang
- Department of Laboratory, GanZhou Cancer Hospital, Ganzhou, China
| | - Yinglian Zhong
- Department of Blood Transfusion, Ganzhou Fifth People’s Hospital, Ganzhou, China
| | - Yi Yang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Deng
- Department of Endocrinology, Department of Nephrology, Ganzhou Fifth People’s Hospital, Ganzhou, China
| |
Collapse
|
13
|
Lu J, Petri R, McCart D, Baxendell-Young A, Collins SA. Distinct fecal microbiome between wild and habitat-housed captive polar bears (Ursus maritimus): Impacts of captivity and dietary shifts. PLoS One 2024; 19:e0311518. [PMID: 39565828 PMCID: PMC11578516 DOI: 10.1371/journal.pone.0311518] [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: 04/05/2024] [Accepted: 09/19/2024] [Indexed: 11/22/2024] Open
Abstract
Understanding the gut microbiome of polar bears can shed light on the effects of climate change-induced prolonged ice-free seasons on their health and nutritional status as a sentinel species. The fecal microbiome of habitat-housed captive polar bears who had consumed a high protein diet long-term was compared with that of the wild population. Individual differences, season, year and dietary inclusion of a brown seaweed (Fucus spiralis; part of the natural diet of wild polar bears), as a representation for nutritional change, were investigated for their effects on the fecal microbiome of captive polar bears. Microbial variations among fecal samples from wild and captive polar bears were investigated using 16s rRNA gene based metataxonomic profiling. The captive bears exhibited more diverse fecal microbiota than wild bears (p<0.05). The difference was due to significantly increased Firmicutes, Campilobacterota and Fusobacteriota, decreased Actinobacteriota (p<0.05), and absent Bdellovibrionota and Verrucomicrobiota in the captive bears. Compared with other factors, individual variation was the main driver of differences in fecal microbial composition in the captive bears. Seaweed consumption did not alter microbial diversity or composition, but this did not rule out dietary influences on the hosts. This is the first study, to the best of our knowledge, comparing the fecal microbiota of captive and wild polar bears and it reveals distinct differences between the two groups, which could result from many factors, including available food sources and the ratio of dietary macronutrients. Our findings provide preliminary insights into climate-change induced dietary shifts in polar bears related to climate-associated habitat change.
Collapse
Affiliation(s)
- Jing Lu
- Faculty of Agriculture, Department Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Renee Petri
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrook, QC, Canada
| | - Dylan McCart
- Churchill Northern Studies Centre, Churchill, MB, Canada
| | | | - Stephanie Anne Collins
- Faculty of Agriculture, Department Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| |
Collapse
|
14
|
Qiu M, Ye C, Zhao X, Zou C, Tang R, Xie J, Liu Y, Hu Y, Hu X, Zhang N, Fu Y, Wang J, Zhao C. Succinate exacerbates mastitis in mice via extracellular vesicles derived from the gut microbiota: a potential new mechanism for mastitis. J Nanobiotechnology 2024; 22:712. [PMID: 39543623 PMCID: PMC11566393 DOI: 10.1186/s12951-024-02997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND A high grain diet causes an ecological imbalance in the gut microbiota and serves as an important endogenous trigger of mastitis in dairy cows, but the underlying mechanisms are unclear. Our previous study revealed that subacute rumen acidosis (SARA)-associated mastitis has distinct metabolic profiles in the rumen, especially a significant increase in succinate, but the role of succinate in the pathogenesis of mastitis remains unclear. RESULTS Succinate treatment exacerbates low-grade endotoxemia-induced mastitis in mice. Specifically, succinate increased the production of gut microbiota-extracellular vehicles (mEVs) containing lipopolysaccharides, which can diffuse across the damaged intestinal barrier into the mammary glands. Administration of mEVs promotes mammary inflammation via activation of the TLR4/NF-κB pathway. CONCLUSIONS Our findings suggest that succinate promotes mastitis through the proliferation of enteric pathogens and mEVs production, suggesting a potential strategy for mastitis intervention on the basis of intestinal metabolic regulation and pathogen inhibition. The role of mEVs in interspecific communication has also been elucidated.
Collapse
Affiliation(s)
- Min Qiu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Cong Ye
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Xiaotong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Chenyu Zou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Ruibo Tang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Jiaxin Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yiheng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, China.
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
15
|
Wu Q, Kan J, Fu C, Liu X, Cui Z, Wang S, Le Y, Li Z, Liu Q, Zhang Y, Du J. Insights into the unique roles of extracellular vesicles for gut health modulation: Mechanisms, challenges, and perspectives. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100301. [PMID: 39525958 PMCID: PMC11550031 DOI: 10.1016/j.crmicr.2024.100301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs), which play significant regulatory roles in maintaining homeostasis and influencing immune responses, significantly impact gut microbiota composition and function, affecting overall gut health. Despite considerable progress, there are still knowledge gaps regarding the mechanisms by which EVs, including plant-derived EVs (PDEVs), animal-derived EVs (ADEVs), and microbiota-derived EVs (MDEVs), modulate gut health. This review delves into the roles and mechanisms of EVs from diverse sources in regulating gut health, focusing on their contributions to maintaining epithelial barrier integrity, facilitating tissue healing, eliciting immune responses, controlling pathogens, and shaping microbiota. We emphasize open challenges and future perspectives for harnessing EVs in the modulation of gut health to gain a deeper understanding of their roles and impact. Importantly, a comprehensive research framework is presented to steer future investigations into the roles and implications of EVs on gut health, facilitating a more profound comprehension of this emerging field.
Collapse
Affiliation(s)
- Qiming Wu
- Nutrilite Health Institute, Shanghai 200031, China
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai 200031, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Xin Liu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhengying Cui
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Sixu Wang
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Yi Le
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhanming Li
- Department of Food Quality and Safety, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Qin Liu
- Centre for Chinese Medicine Drug Development Limited, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai 200031, China
| |
Collapse
|
16
|
Zhi W, Li A, Wang Q, Yuan X, Qing J, Zhang C, Wang Y, Li Y. Safety and efficacy assessment of fecal microbiota transplantation as an adjunctive treatment for IgA nephropathy: an exploratory clinical trial. Sci Rep 2024; 14:22935. [PMID: 39358432 PMCID: PMC11446926 DOI: 10.1038/s41598-024-74171-4] [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: 06/21/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
To assess the safety and efficacy of fecal microbiota transplantation (FMT) as an adjunctive therapeutic intervention for IgA nephropathy (IgAN). Fifteen patients with IgA nephropathy were recruited based on inclusion and exclusion criteria and underwent FMT using enteric microbial capsules. Clinical indicators, intestinal microbiota and metabolomic profiles, as well as changes in serum immune cells and cytokines, were monitored before and after FMT. No severe adverse reactions were observed in the subjects. After FMT, there was a reduction in the 24-h urinary protein quantification in subjects. The relative abundances of Phocaeicola_vulgatus, Bacteroides_uniformis, Prevotella_copri, Phocaeicola_dorei, Bacteroides_ovatus, Bacteroides_xylanisolvens, Parabacteroides _distasonis, Bifidobacterium_pseudocatenulatum, Bacteroides_sp._HF-162, and Bifidobacterium_longum changed after FMT. In terms of intestinal metabolites, the levels of acylcarnitine18:0 (ACar.18:0), cotinine, N-arachidonoyl-L-serine, phosphatidylcholine (PC. (18:3e/22:6)), serotonin, and fumagillin showed significant changes. Flow cytometry analysis showed the absolute count of plasma B cells decreased in subjects, and this change correlated with alterations in the intestinal microbiota and metabolites. This study preliminarily evaluates the safety and efficacy of FMT in patients with IgAN. No significant adverse reactions were observed, and the administration of FMT alongside ACEI/ARB therapy was effective in reducing urinary protein levels in patients with IgAN, a process that may be associated with B-cell immunity.
Collapse
Affiliation(s)
- Wenqiang Zhi
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Aizhong Li
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Qian Wang
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaoli Yuan
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Jianbo Qing
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Caixiang Zhang
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuxin Wang
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Yafeng Li
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030001, China.
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital), Shanxi Medical University, Taiyuan, 030001, China.
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, 030001, China.
- Hejin Municipal People's Hospital, Hejin, 043300, China.
| |
Collapse
|
17
|
Tan J, Potier-Villette C, Ni D, Hoeckh M, Taitz J, Simpson SJ, Nanan R, Macia L. Succinate induces a Th2 environment in the small intestine but does not exacerbate food allergy. Allergy 2024; 79:2853-2855. [PMID: 38747065 DOI: 10.1111/all.16155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/27/2024] [Accepted: 05/01/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, New South Wales, Australia
| | - Maike Hoeckh
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jemma Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School Nepean, The University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
| |
Collapse
|
18
|
Kou Y, Zhang S, Chen J, Shen Y, Zhang Z, Huang H, Ma Y, Xiang Y, Liao L, Zhou J, Cheng W, Zhou Y, Yang H, Liu Z, Wei Y, Wang H, Wang Y. A mouse protozoan boosts antigen-specific mucosal IgA responses in a specific lipid metabolism- and signaling-dependent manner. Nat Commun 2024; 15:7914. [PMID: 39256385 PMCID: PMC11387640 DOI: 10.1038/s41467-024-52336-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
IgA antibodies play an important role in mucosal immunity. However, there is still no effective way to consistently boost mucosal IgA responses, and the factors influencing these responses are not fully understood. We observed that colonization with the murine intestinal symbiotic protozoan Tritrichomonas musculis (T.mu) boosted antigen-specific mucosal IgA responses in wild-type C57BL/6 mice. This enhancement was attributed to the accumulation of free arachidonic acid (ARA) in the intestinal lumen, which served as a signal to stimulate the production of antigen-specific mucosal IgA. When ARA was prevented from undergoing its downstream metabolic transformation using the 5-lipoxygenase inhibitor zileuton or by blocking its downstream biological signaling through genetic deletion of the Leukotriene B4 receptor 1 (Blt1), the T.mu-mediated enhancement of antigen-specific mucosal IgA production was suppressed. Moreover, both T.mu transfer and dietary supplementation of ARA augmented the efficacy of an oral vaccine against Salmonella infection, with this effect being dependent on Blt1. Our findings elucidate a tripartite circuit linking nutrients from the diet or intestinal microbiota, host lipid metabolism, and the mucosal humoral immune response.
Collapse
Affiliation(s)
- Yanbo Kou
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Shenghan Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou, China
| | - Junru Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yusi Shen
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zhiwei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Haohan Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yulu Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yaoyao Xiang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Longxiang Liao
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Junyang Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yuan Zhou
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School, Xuzhou Medical University, Xuzhou, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School, Xuzhou Medical University, Xuzhou, China
| | - Zhuanzhuan Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yanxia Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yugang Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China.
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
19
|
Zeng C, Wan SR, Guo M, Tan XZ, Zeng Y, Wu Q, Xie JJ, Yan P, Long Y, Zheng L, Jiang ZZ, Teng FY, Xu Y. Fecal virome transplantation: A promising strategy for the treatment of metabolic diseases. Biomed Pharmacother 2024; 177:117065. [PMID: 38971010 DOI: 10.1016/j.biopha.2024.117065] [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: 04/06/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024] Open
Abstract
Metabolic diseases are a group of disorders caused by metabolic abnormalities, including obesity, diabetes, non-alcoholic fatty liver disease, and more. Increasing research indicates that, beyond inherent metabolic irregularities, the onset and progression of metabolic diseases are closely linked to alterations in the gut microbiota, particularly gut bacteria. Additionally, fecal microbiota transplantation (FMT) has demonstrated effectiveness in clinically treating metabolic diseases, notably diabetes. Recent attention has also focused on the role of gut viruses in disease onset. This review first introduces the characteristics and influencing factors of gut viruses, then summarizes their potential mechanisms in disease development, highlighting their impact on gut bacteria and regulation of host immunity. We also compare FMT, fecal filtrate transplantation (FFT), washed microbiota transplantation (WMT), and fecal virome transplantation (FVT). Finally, we review the current understanding of gut viruses in metabolic diseases and the application of FVT in treating these conditions. In conclusion, FVT may provide a novel and promising treatment approach for metabolic diseases, warranting further validation through basic and clinical research.
Collapse
Affiliation(s)
- Chen Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Sheng-Rong Wan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao-Zhen Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Qi Wu
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao 999078, China; Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jia-Jie Xie
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pijun Yan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Institute of Cardiovascular Research, Peking University, Beijing 100871, China
| | - Yang Long
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lemin Zheng
- Institute of Cardiovascular Research, Peking University, Beijing 100871, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
20
|
Neettiyath A, Chung K, Liu W, Lee LP. Nanoplasmonic sensors for extracellular vesicles and bacterial membrane vesicles. NANO CONVERGENCE 2024; 11:23. [PMID: 38918255 PMCID: PMC11199476 DOI: 10.1186/s40580-024-00431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Extracellular vesicles (EVs) are promising tools for the early diagnosis of diseases, and bacterial membrane vesicles (MVs) are especially important in health and environment monitoring. However, detecting EVs or bacterial MVs presents significant challenges for the clinical translation of EV-based diagnostics. In this Review, we provide a comprehensive discussion on the basics of nanoplasmonic sensing and emphasize recent developments in nanoplasmonics-based optical sensors to effectively identify EVs or bacterial MVs. We explore various nanoplasmonic sensors tailored for EV or bacterial MV detection, emphasizing the application of localized surface plasmon resonance through gold nanoparticles and their multimers. Additionally, we highlight advanced EV detection techniques based on surface plasmon polaritons using plasmonic thin film and nanopatterned structures. Furthermore, we evaluate the improved detection capability of surface-enhanced Raman spectroscopy in identifying and classifying these vesicles, aided by plasmonic nanostructures. Nanoplasmonic sensing techniques have remarkable precision and sensitivity, making them a potential tool for accurate EV detection in clinical applications, facilitating point-of-care molecular diagnostics. Finally, we summarize the challenges associated with nanoplasmonic EV or bacterial MV sensors and offer insights into potential future directions for this evolving field.
Collapse
Affiliation(s)
- Aparna Neettiyath
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Kyungwha Chung
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Wenpeng Liu
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Luke P Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Harvard University, Boston, MA 02115, USA.
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA 94720, USA.
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea.
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
21
|
Zhang Y, Zhang Y, Wang T. Can Non-farm Employment Improve Dietary Diversity of Left-Behind Family Members in Rural China? Foods 2024; 13:1818. [PMID: 38928760 PMCID: PMC11202984 DOI: 10.3390/foods13121818] [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: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Rural residents in China are still at risk of malnutrition, and increasing dietary diversity is crucial to improving their health. This study empirically analyzed the impact of non-farm employment on the dietary diversity of rural left-behind family members based on the China Land Economy Survey (CLES) 2020-2021 panel data at the farm and village levels. Dietary diversity was measured using the dietary diversity score (DDS) and the Chinese Food Guide Pagoda Score (CFGPS). The empirical results show that non-farm employment significantly enhances the dietary diversity of rural left-behind household members, including animal food diversity and plant food diversity. This result verifies the altruism phenomenon of non-farm employment in family diet. Mechanism analysis shows that non-farm employment enhances the dietary diversity of rural left-behind family members by increasing the level of family income, Internet accessibility, and family education. Heterogeneity analysis shows that non-farm employment does not enhance the dietary diversity of rural empty nesters and even has a negative impact. This reminds us that the nutritional health of rural empty nesters needs attention in the context of rapid urbanization and aging.
Collapse
Affiliation(s)
| | - Yifeng Zhang
- College of Economics and Management, Nanjing Forestry University, Nanjing 210037, China; (Y.Z.); (T.W.)
| | | |
Collapse
|
22
|
Xie Y, Liu F. The role of the gut microbiota in tumor, immunity, and immunotherapy. Front Immunol 2024; 15:1410928. [PMID: 38903520 PMCID: PMC11188355 DOI: 10.3389/fimmu.2024.1410928] [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: 04/02/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
In recent years, with the deepening understanding of the gut microbiota, it has been recognized to play a significant role in the development and progression of diseases. Particularly in gastrointestinal tumors, the gut microbiota influences tumor growth by dysbiosis, release of bacterial toxins, and modulation of host signaling pathways and immune status. Immune checkpoint inhibitors (ICIs) have greatly improved cancer treatment efficacy by enhancing immune cell responses. Current clinical and preclinical studies have demonstrated that the gut microbiota and its metabolites can enhance the effectiveness of immunotherapy. Furthermore, certain gut microbiota can serve as biomarkers for predicting immunotherapy responses. Interventions targeting the gut microbiota for the treatment of gastrointestinal diseases, especially colorectal cancer (CRC), include fecal microbiota transplantation, probiotics, prebiotics, engineered bacteria, and dietary interventions. These approaches not only improve the efficacy of ICIs but also hold promise for enhancing immunotherapy outcomes. In this review, we primarily discuss the role of the gut microbiota and its metabolites in tumors, host immunity, and immunotherapy.
Collapse
Affiliation(s)
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
23
|
Wang J, He M, Yang M, Ai X. Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci 2024; 345:122612. [PMID: 38588949 DOI: 10.1016/j.lfs.2024.122612] [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: 02/04/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Gut microbiota is a complex microbial community with the ability of maintaining intestinal health. Intestinal homeostasis largely depends on the mucosal immune system to defense external pathogens and promote tissue repair. In recent years, growing evidence revealed the importance of gut microbiota in shaping intestinal mucosal immunity. Therefore, according to the existing findings, this review first provided an overview of intestinal mucosal immune system before summarizing the regulatory roles of gut microbiota in intestinal innate and adaptive immunity. Specifically, this review delved into the gut microbial interactions with the cells such as intestinal epithelial cells (IECs), macrophages, dendritic cells (DCs), neutrophils, and innate lymphoid cells (ILCs) in innate immunity, and T and B lymphocytes in adaptive immunity. Furthermore, this review discussed the main effects of gut microbiota dysbiosis in intestinal diseases and offered future research prospects. The review highlighted the key regulatory roles of gut microbiota in intestinal mucosal immunity via various host-microbe interactions, providing valuable references for the development of microbial therapy in intestinal diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Mei He
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
24
|
Zhao M, Liang X, Meng Y, Lu H, Lin K, Gong P, Liu T, Yi H, Pan J, Zhang Y, Zhang Z, Zhang L. Probiotics induce intestinal IgA secretion in weanling mice potentially through promoting intestinal APRIL expression and modulating the gut microbiota composition. Food Funct 2024; 15:4862-4873. [PMID: 38587236 DOI: 10.1039/d4fo00962b] [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: 04/09/2024]
Abstract
Intestinal infections are strongly associated with infant mortality, and intestinal immunoglobulin A (IgA) is important to protect infants from intestinal infections after weaning. This study aims to screen probiotics that can promote the production of intestinal IgA after weaning and further explore their potential mechanisms of action. In this study, probiotics promoting intestinal IgA production were screened in weanling mouse models. The results showed that oral administration of Bifidobacterium bifidum (B. bifidum) FL228.1 and Bifidobacterium bifidum (B. bifidum) FL276.1 significantly enhanced IgA levels in the small intestine and upregulated the expression of a proliferation-inducing ligand (APRIL) and its upstream regulatory factor toll-like receptor 4 (TLR4). Furthermore, B. bifidum FL228.1 upregulated the relative abundance of Lactobacillus, while B. bifidum FL276.1 increased the relative abundance of Marvinbryantia and decreased Mucispirillum, further elevating intestinal IgA levels. In summary, B. bifidum FL228.1 and B. bifidum FL276.1 can induce IgA production in the intestinal tract of weanling mice by promoting intestinal APRIL expression and mediating changes in the gut microbiota, thus playing a significant role in enhancing local intestinal immunity in infants.
Collapse
Affiliation(s)
- Maozhen Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Xi Liang
- College of Public Health, Qingdao University, Qingdao, 266000, China
| | - Yang Meng
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Haiyan Lu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Kai Lin
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Pimin Gong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Tongjie Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Jiancun Pan
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihar, 161000, China
| | - Yongjiu Zhang
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihar, 161000, China
| | - Zhe Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| |
Collapse
|
25
|
Selvaskandan H, Jhaveri KD, Rizk DV. Primary IgA Nephropathy: New Insights and Emerging Therapies. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:180-193. [PMID: 39004458 DOI: 10.1053/j.akdh.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 03/12/2024] [Accepted: 04/01/2024] [Indexed: 07/16/2024]
Abstract
Primary IgA nephropathy (IgAN) is a common glomerular disorder defined by predominant mesangial IgA deposition. Once thought to follow a progressive course in 10-20% of those diagnosed, emerging evidence now suggests most will progress to kidney failure over their lifetimes. Although the lack of safe and effective treatments to impede disease progression continues to present a challenge, the landscape of IgAN has dramatically evolved over the last 2 years. Driven by fundamental changes to accepted end points for IgAN clinical trials as well as fascinating new insights into the pathophysiology of IgAN, a swathe of novel and repurposed therapies are currently being evaluated. Already, two novel drugs, targeted-release formulation budesonide and sparsentan, have received conditional approvals for the treatment of IgAN, with sodium glucose co-transporter 2 inhibitors establishing themselves as further options. Soon to join this ensemble are likely to be treatments that modulate the complement system and B-cell activity; several are currently undergoing clinical trials in IgAN with promising interim results. In this review, we provide an overview of evolving epidemiological insights, disease mechanisms, emerging therapies, and contemporary challenges surrounding the management of IgAN.
Collapse
Affiliation(s)
- Haresh Selvaskandan
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Kenar D Jhaveri
- Northwell Health, New Hyde Park, NY; Glomerular Center at Northwell Health, Division of Kidney Diseases and Hypertension, Northwell Health, Great Neck, NY; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY.
| | - Dana V Rizk
- Division of Nephrology, Department of Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
26
|
Li F, Wang Z, Cao Y, Pei B, Luo X, Liu J, Ge P, Luo Y, Ma S, Chen H. Intestinal Mucosal Immune Barrier: A Powerful Firewall Against Severe Acute Pancreatitis-Associated Acute Lung Injury via the Gut-Lung Axis. J Inflamm Res 2024; 17:2173-2193. [PMID: 38617383 PMCID: PMC11016262 DOI: 10.2147/jir.s448819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/20/2024] [Indexed: 04/16/2024] Open
Abstract
The pathogenesis of severe acute pancreatitis-associated acute lung injury (SAP-ALI), which is the leading cause of mortality among hospitalized patients in the intensive care unit, remains incompletely elucidated. The intestinal mucosal immune barrier is a crucial component of the intestinal epithelial barrier, and its aberrant activation contributes to the induction of sustained pro-inflammatory immune responses, paradoxical intercellular communication, and bacterial translocation. In this review, we firstly provide a comprehensive overview of the composition of the intestinal mucosal immune barrier and its pivotal roles in the pathogenesis of SAP-ALI. Secondly, the mechanisms of its crosstalk with gut microbiota, which is called gut-lung axis, and its effect on SAP-ALI were summarized. Finally, a number of drugs that could enhance the intestinal mucosal immune barrier and exhibit potential anti-SAP-ALI activities were presented, including probiotics, glutamine, enteral nutrition, and traditional Chinese medicine (TCM). The aim is to offer a theoretical framework based on the perspective of the intestinal mucosal immune barrier to protect against SAP-ALI.
Collapse
Affiliation(s)
- Fan Li
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Zhengjian Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Yinan Cao
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Boliang Pei
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Xinyu Luo
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Jin Liu
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Peng Ge
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Yalan Luo
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Shurong Ma
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| | - Hailong Chen
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Laboratory of Integrative Medicine, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
| |
Collapse
|
27
|
Zhou X, Liang L, Sun B, Li K, Guo H, Zhang Y. The Effects of Yeast Protein on Gut Microbiota in Mice When Compared with Soybean Protein and Whey Protein Isolates. Nutrients 2024; 16:458. [PMID: 38337742 PMCID: PMC10857369 DOI: 10.3390/nu16030458] [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: 12/06/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Different protein sources can impact gut microbiota composition and abundance, and also participate in health regulation. In this study, mice were gavaged with yeast protein (YP), soybean protein isolate (SPI), and whey protein isolate (WPI) for 28 days. Body weights showed similar patterns across different protein administration groups. The ileum in YP-supplemented mice exhibited good morphology, and tight-junction (TJ) proteins were slightly upregulated. Immunoglobulin (Ig)A, IgM, and IgG levels in the ileum of different protein groups were significantly increased (p < 0.05). Interleukin (IL)-10 levels were significantly increased, whereas IL-6 levels were significantly reduced in the YP group when compared with the control (C) (p < 0.05). Glutathione peroxidase (GSH-Px) levels in the ileum were significantly increased in the YP group (p < 0.05). These results indicate that YP potentially improved intestinal immunity and inflammatory profiles. The relative abundances of Parabacteroides, Prevotella, and Pseudobutyrivibrio in the YP group were more enriched when compared with the C and SPI groups, and Parabacteroides was significantly upregulated when compared with the WPI group (p < 0.05). Overall, the results indicate that YP upregulates the beneficial bacteria and improves ileal immunity and anti-inflammatory capabilities.
Collapse
Affiliation(s)
- Xuewei Zhou
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (X.Z.); (L.L.); (B.S.)
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Li Liang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (X.Z.); (L.L.); (B.S.)
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (X.Z.); (L.L.); (B.S.)
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Ku Li
- National Key Laboratory of Agricultural Microbiology Core Facility, Angel Yeast Co., Ltd., Yichang 443003, China; (K.L.); (H.G.)
| | - Hui Guo
- National Key Laboratory of Agricultural Microbiology Core Facility, Angel Yeast Co., Ltd., Yichang 443003, China; (K.L.); (H.G.)
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Ministry of Education, Beijing 100048, China; (X.Z.); (L.L.); (B.S.)
- Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
28
|
Selvaskandan H, Barratt J, Cheung CK. Novel Treatment Paradigms: Primary IgA Nephropathy. Kidney Int Rep 2024; 9:203-213. [PMID: 38344739 PMCID: PMC10851020 DOI: 10.1016/j.ekir.2023.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 01/30/2025] Open
Abstract
IgA nephropathy (IgAN) is the most common primary glomerulonephritis worldwide. Approximately 30% to 45% of patients progress to kidney failure (KF) within 20 to 25 years of diagnosis, and there has long been a lack of effective treatments. The therapeutic landscape in IgAN is rapidly evolving, driven in large part by the acceptance of the surrogate clinical trial end point of proteinuria reduction by regulatory authorities for the accelerated approval of new therapies. Two drugs, targeted release formulation (TRF)-budesonide (nefecon) and sparsentan, have recently been approved under this scheme. Advancing insights into the pathophysiology of IgAN, including the roles of the mucosal immune system, B-cells, the complement system, and the endothelin system have driven development of therapies that target these factors. This review outlines current, recently approved, and emerging therapies for IgAN.
Collapse
Affiliation(s)
- Haresh Selvaskandan
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Jonathan Barratt
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Chee Kay Cheung
- Mayer IgA Nephropathy Laboratories, Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
29
|
Patel N, Dinesh S, Sharma S. From Gut to Glucose: A Comprehensive Review on Functional Foods and Dietary Interventions for Diabetes Management. Curr Diabetes Rev 2024; 20:e111023222081. [PMID: 37861021 DOI: 10.2174/0115733998266653231005072450] [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: 06/10/2023] [Revised: 07/17/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND In the realm of diabetes research, considerable attention has been directed toward elucidating the intricate interplay between the gastrointestinal tract and glucose regulation. The gastrointestinal tract, once exclusively considered for its role in digestion and nutrient assimilation, is presently acknowledged as a multifaceted ecosystem with regulatory supremacy over metabolic homeostasis and glucose metabolism. Recent studies indicate that alterations in the composition and functionality of the gut microbiota could potentially influence the regulation of glucose levels and glucose homeostasis in the body. Dysbiosis, characterized by perturbations in the equilibrium of gut microbial constituents, has been irrevocably linked to an augmented risk of diabetes mellitus (DM). Moreover, research has revealed the potential influence of the gut microbiota on important factors, like inflammation and insulin sensitivity, which are key contributors to the onset and progression of diabetes. The key protagonists implicated in the regulation of glucose encompass the gut bacteria, gut barrier integrity, and the gut-brain axis. A viable approach to enhance glycemic control while concurrently mitigating the burden of comorbidities associated with diabetes resides in the strategic manipulation of the gut environment through adapted dietary practices. OBJECTIVE This review aimed to provide a deep understanding of the complex relationship between gut health, glucose metabolism, and diabetes treatment. CONCLUSION This study has presented an exhaustive overview of dietary therapies and functional foods that have undergone extensive research to explore their potential advantages in the management of diabetes. It looks into the role of gut health in glucose regulation, discusses the impact of different dietary elements on the course of diabetes, and evaluates how well functional foods can help with glycemic control. Furthermore, it investigates the mechanistic aspects of these therapies, including their influence on insulin sensitivity, β-cell activity, and inflammation. It deliberates on the limitations and potential prospects associated with integrating functional foods into personalized approaches to diabetes care.
Collapse
Affiliation(s)
- Nirali Patel
- Department of Bioinformatics, BioNome, Bengaluru 560043, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru 560043, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru 560043, India
| |
Collapse
|
30
|
Abubaker S, Miri S, Mottawea W, Hammami R. Microbial Extracellular Vesicles in Host-Microbiota Interactions. Results Probl Cell Differ 2024; 73:475-520. [PMID: 39242390 DOI: 10.1007/978-3-031-62036-2_19] [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: 09/09/2024]
Abstract
Extracellular vesicles have emerged as key players in cellular communication, influencing various physiological processes and pathophysiological progression, including digestion, immune response, and tissue repairs. Recently, a class of EVs derived from microbial communities has gained significant attention due to their pivotal role in intercellular communication and their potential as biomarkers and biotherapeutic agents. Microbial EVs are membrane-bound molecules encapsulating bioactive metabolites that modulate host physiological and pathological processes. This chapter discusses the evolving history of microbiota-produced EVs, including their discovery, characterization, current research status, and their diverse mechanisms of interaction with other microbes and hosts. This review also highlights the importance of EVs in health and disease and discusses recent research that shows promising results for the therapeutic potential of EVs.
Collapse
Affiliation(s)
- Sarah Abubaker
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Saba Miri
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Walid Mottawea
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Riadh Hammami
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada.
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
31
|
Liu B, Fan L, Wang Y, Wang H, Yan Y, Chen S, Hung I, Liu C, Wei H, Ge L, Ren W. Gut microbiota regulates host melatonin production through epithelial cell MyD88. Gut Microbes 2024; 16:2313769. [PMID: 38353638 PMCID: PMC10868534 DOI: 10.1080/19490976.2024.2313769] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/30/2024] [Indexed: 02/16/2024] Open
Abstract
Melatonin has various physiological effects, such as the maintenance of circadian rhythms, anti-inflammatory functions, and regulation of intestinal barriers. The regulatory functions of melatonin in gut microbiota remodeling have also been well clarified; however, the role of gut microbiota in regulating host melatonin production remains poorly understood. To address this, we studied the contribution of gut microbiota to host melatonin production using gut microbiota-perturbed models. We demonstrated that antibiotic-treated and germ-free mice possessed diminished melatonin levels in the serum and elevated melatonin levels in the colon. The influence of the intestinal microbiota on host melatonin production was further confirmed by fecal microbiota transplantation. Notably, Lactobacillus reuteri (L. R) and Escherichia coli (E. coli) recapitulated the effects of gut microbiota on host melatonin production. Mechanistically, L. R and E. coli activated the TLR2/4/MyD88/NF-κB signaling pathway to promote expression of arylalkylamine N-acetyltransferase (AANAT, a rate-limiting enzyme for melatonin production), and MyD88 deficiency in colonic epithelial cells abolished the influence of intestinal microbiota on colonic melatonin production. Collectively, we revealed a specific underlying mechanism of gut microbiota to modulate host melatonin production, which might provide novel therapeutic ideas for melatonin-related diseases.
Collapse
Affiliation(s)
- Bingnan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Lijuan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Youxia Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Hao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Yuqi Yan
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Shuai Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Ifen Hung
- Anyou Biotechnology Group Co. LTD, Taicang, China
- Joint Laboratory of Functional Nutrition and Animal Health, Centree Bio-tech (Wuhan) Co., LTD, Wuhan, China
| | - Chunxue Liu
- Anyou Biotechnology Group Co. LTD, Taicang, China
| | - Hong Wei
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Liangpeng Ge
- National Center of Technology Innovation for Pigs, Chongqing, China
- Chongqing Academy of Animal Sciences, Key Laboratory of Pig Industry Science, Ministry of Agriculture, Chongqing, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| |
Collapse
|
32
|
Verbunt J, Jocken J, Blaak E, Savelkoul P, Stassen F. Gut-bacteria derived membrane vesicles and host metabolic health: a narrative review. Gut Microbes 2024; 16:2359515. [PMID: 38808455 PMCID: PMC11141482 DOI: 10.1080/19490976.2024.2359515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
The intestinal microbiota, consisting of an estimated 10^10-10^11 organisms, regulate physiological processes involved in digestion, metabolism, and immunity. Surprisingly, these intestinal microorganisms have been found to influence tissues that are not directly in contact with the gut, such as adipose tissue, the liver, skeletal muscle, and the brain. This interaction takes place even when intestinal barrier function is uncompromised. An increasing body of evidence suggests that bacterial membrane vesicles (bMVs), in addition to bacterial metabolites such as short-chain fatty acids, are able to mediate effects of the microbiota on these host tissues. The ability of bMVs to dissipate from the intestinal lumen into systemic circulation hereby facilitates the transport and presentation of bacterial components and metabolites to host organs. Importantly, there are indications that the interaction between bMVs and tissues or immune cells may play a role in the etiology of (chronic metabolic) disease. For example, the gut-derived bMV-mediated induction of insulin resistance in skeletal muscle cells and pro-inflammatory signaling by adipocytes possibly underlies diseases such as type 2 diabetes and obesity. Here, we review the current knowledge on bMVs in the microbiota's effects on host energy/substrate metabolism with a focus on etiological roles in the onset and progression of metabolic disease. We furthermore illustrate that vesicle production by bacterial microbiota could potentially be modulated through lifestyle intervention to improve host metabolism.
Collapse
Affiliation(s)
- Jari Verbunt
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Johan Jocken
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen Blaak
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paul Savelkoul
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Medical Microbiology and Infection Control, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Frank Stassen
- Department of Medical Microbiology, Infectious Diseases & Infection Prevention, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
33
|
Zhang J, Liang M, Wu L, Yang Y, Sun Y, Wang Q, Gao X. Bioconversion of feather waste into bioactive nutrients in water by Bacillus licheniformis WHU. Appl Microbiol Biotechnol 2023; 107:7055-7070. [PMID: 37750916 DOI: 10.1007/s00253-023-12795-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/23/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023]
Abstract
Feathers become hazardous pollutants when deposited directly into the environment. The rapid expansion of the poultry industry has significantly increased feather waste, necessitating the development of new ways to degrade and utilize feathers. This study investigated the ability of Bacillus licheniformis WHU to digest intact chicken feathers in water. The results indicated that yields of free amino acids, bioactive peptides, and keratin-derived nano-/micro-particles were improved in bacteria- versus purified keratinase-derived feather hydrolysate. Bacteria-derived feather hydrolysate supplementation induced health benefits in mice, including significantly increased intestinal villus height and zonula occludens-1 protein expression, as well as increased secretory immunoglobulin A levels in the intestinal mucosa and superoxide dismutase activity in serum. Additionally, feather hydrolysate supplementation modulated the mouse gut microbiota, reflected by increased relative abundance of probiotics such as Lactobacillus spp., decreased relative abundance of Proteobacteria at the phylum level and pathogens such as Staphylococcus spp., and increased Bacteroidota/Firmicutes ratio. This study developed a simple, cost-effective method to degrade feathers by B. licheniformis WHU digestion, yielding a hydrolysate that can be directly used as a bioactive nutrient resource. The study findings have applications in the livestock, poultry, and aquaculture industries, which have high demands for cheap protein. KEY POINTS: • Bacillus licheniformis could degrade intact feather in water. • The resulting feather hydrolysate shows prebiotic effects on mouse.
Collapse
Affiliation(s)
- Jing Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Manyu Liang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lijuan Wu
- Department of Endocrinology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yanhong Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yingjie Sun
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qin Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Dazhou Vocational College of Chinese Medicine, Tongchuan District, Luojiang Town, Dazhou, 635000, China.
| | - Xiaowei Gao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Dazhou Vocational College of Chinese Medicine, Tongchuan District, Luojiang Town, Dazhou, 635000, China.
- Green Pharmaceutical Technology Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
34
|
Xie J, Haesebrouck F, Van Hoecke L, Vandenbroucke RE. Bacterial extracellular vesicles: an emerging avenue to tackle diseases. Trends Microbiol 2023; 31:1206-1224. [PMID: 37330381 DOI: 10.1016/j.tim.2023.05.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/19/2023]
Abstract
A growing body of research, especially in recent years, has shown that bacterial extracellular vesicles (bEVs) are one of the key underlying mechanisms behind the pathogenesis of various diseases like pulmonary fibrosis, sepsis, systemic bone loss, and Alzheimer's disease. Given these new insights, bEVs are proposed as an emerging vehicle that can be used as a diagnostic tool or to tackle diseases when used as a therapeutic target. To further boost the understanding of bEVs in health and disease we thoroughly discuss the contribution of bEVs in disease pathogenesis and the underlying mechanisms. In addition, we speculate on their potential as novel diagnostic biomarkers and how bEV-related mechanisms can be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium; Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
35
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
36
|
Tan J, Taitz J, Nanan R, Grau G, Macia L. Dysbiotic Gut Microbiota-Derived Metabolites and Their Role in Non-Communicable Diseases. Int J Mol Sci 2023; 24:15256. [PMID: 37894934 PMCID: PMC10607102 DOI: 10.3390/ijms242015256] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Dysbiosis, generally defined as the disruption to gut microbiota composition or function, is observed in most diseases, including allergies, cancer, metabolic diseases, neurological disorders and diseases associated with autoimmunity. Dysbiosis is commonly associated with reduced levels of beneficial gut microbiota-derived metabolites such as short-chain fatty acids (SCFA) and indoles. Supplementation with these beneficial metabolites, or interventions to increase their microbial production, has been shown to ameliorate a variety of inflammatory diseases. Conversely, the production of gut 'dysbiotic' metabolites or by-products by the gut microbiota may contribute to disease development. This review summarizes the various 'dysbiotic' gut-derived products observed in cardiovascular diseases, cancer, inflammatory bowel disease, metabolic diseases including non-alcoholic steatohepatitis and autoimmune disorders such as multiple sclerosis. The increased production of dysbiotic gut microbial products, including trimethylamine, hydrogen sulphide, products of amino acid metabolism such as p-Cresyl sulphate and phenylacetic acid, and secondary bile acids such as deoxycholic acid, is commonly observed across multiple diseases. The simultaneous increased production of dysbiotic metabolites with the impaired production of beneficial metabolites, commonly associated with a modern lifestyle, may partially explain the high prevalence of inflammatory diseases in western countries.
Collapse
Affiliation(s)
- Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (J.T.); (J.T.); (R.N.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Jemma Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (J.T.); (J.T.); (R.N.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (J.T.); (J.T.); (R.N.)
- Sydney Medical School and Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW 2006, Australia
| | - Georges Grau
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (J.T.); (J.T.); (R.N.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
- Sydney Cytometry, The Centenary Institute and The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
37
|
Taitz JJ, Tan JK, Potier-Villette C, Ni D, King NJ, Nanan R, Macia L. Diet, commensal microbiota-derived extracellular vesicles, and host immunity. Eur J Immunol 2023; 53:e2250163. [PMID: 37137164 DOI: 10.1002/eji.202250163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/04/2023] [Accepted: 05/02/2023] [Indexed: 05/05/2023]
Abstract
The gut microbiota has co-evolved with its host, and commensal bacteria can influence both the host's immune development and function. Recently, a role has emerged for bacterial extracellular vesicles (BEVs) as potent immune modulators. BEVs are nanosized membrane vesicles produced by all bacteria, possessing the membrane characteristics of the originating bacterium and carrying an internal cargo that may include nucleic acid, proteins, lipids, and metabolites. Thus, BEVs possess multiple avenues for regulating immune processes, and have been implicated in allergic, autoimmune, and metabolic diseases. BEVs are biodistributed locally in the gut, and also systemically, and thus have the potential to affect both the local and systemic immune responses. The production of gut microbiota-derived BEVs is regulated by host factors such as diet and antibiotic usage. Specifically, all aspects of nutrition, including macronutrients (protein, carbohydrates, and fat), micronutrients (vitamins and minerals), and food additives (the antimicrobial sodium benzoate), can regulate BEV production. This review summarizes current knowledge of the powerful links between nutrition, antibiotics, gut microbiota-derived BEV, and their effects on immunity and disease development. It highlights the potential of targeting or utilizing gut microbiota-derived BEV as a therapeutic intervention.
Collapse
Affiliation(s)
- Jemma J Taitz
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jian K Tan
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Nicholas Jc King
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Nepean Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry, University of Sydney and Centenary Institute, Sydney, NSW, Australia
| |
Collapse
|
38
|
Rathore APS, St John AL. Promises and challenges of mucosal COVID-19 vaccines. Vaccine 2023; 41:4042-4049. [PMID: 37045682 PMCID: PMC10083204 DOI: 10.1016/j.vaccine.2023.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Coronavirus disease-2019 (COVID-19) is an ongoing pandemic caused by the newly emerged virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Currently, COVID-19 vaccines are given intramuscularly and they have been shown to evoke systemic immune responses that are highly efficacious towards preventing severe disease and death. However, vaccine-induced immunity wanes within a short time, and booster doses are currently recommended. Furthermore, current vaccine formulations do not adequately restrict virus infection at the mucosal sites, such as in the nasopharyngeal tract and, therefore, have limited capacity to block virus transmission. With these challenges in mind, several mucosal vaccines are currently being developed with the aim of inducing long-lasting protective immune responses at the mucosal sites where SARS-COV-2 infection begins. Past successes in mucosal vaccinations underscore the potential of these developmental stage SARS-CoV-2 vaccines to reduce disease burden, if not eliminate it altogether. Here, we discuss immune responses that are triggered at the mucosal sites and recent advances in our understanding of mucosal responses induced by SARS-CoV-2 infection and current COVID-19 vaccines. We also highlight several mucosal SARS-COV-2 vaccine formulations that are currently being developed or tested for human use and discuss potential challenges to mucosal vaccination.
Collapse
Affiliation(s)
- Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA
| | - Ashley L St John
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore.
| |
Collapse
|
39
|
Doré E, Boilard E. Bacterial extracellular vesicles and their interplay with the immune system. Pharmacol Ther 2023; 247:108443. [PMID: 37210006 DOI: 10.1016/j.pharmthera.2023.108443] [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: 02/14/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
The mammalian intestinal tract harbors trillions of microorganisms confined within this space by mucosal barriers. Despite these barriers, bacterial components may still be found elsewhere in the body, even in healthy subjects. Bacteria can release small lipid-bound particles, also named bacterial extracellular vesicles (bEV). While bacteria themselves cannot normally penetrate the mucosal defense, bEVs may infiltrate the barrier and disseminate throughout the body. The extremely diverse cargo that bEVs can carry, depending on their parent species, strain, and growth conditions, grant them an equally broad potential to interact with host cells and influence immune functions. Herein, we review the current knowledge of processes underlying the uptake of bEVs by mammalian cells, and their effect on the immune system. Furthermore, we discuss how bEVs could be targeted and manipulated for diverse therapeutic purposes.
Collapse
Affiliation(s)
- Etienne Doré
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada; Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, QC, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada; Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, QC, Canada.
| |
Collapse
|
40
|
Matsumoto K, Sawano H, Otsubo M, Yui A. Comparison of the effects of three forms of soluble dietary fiber on the production of IgA in BALB/cAJcl and BALB/cAJcl-nu/nu mice. J Nutr 2023; 153:1618-1626. [PMID: 37011879 DOI: 10.1016/j.tjnut.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Their ability to induce the production of IgA, especially in the intestine, is one of the health benefits of soluble dietary fibers (SDFs), but the mechanism involved is unclear. OBJECTIVE This study was designed to identify the relationship between the induction of IgA by SDFs and the cecal short-chain fatty acids (SCFAs) content, and to evaluate the importance of T cell-independent IgA production for SDF-induced IgA production. METHODS We compared the SDFs fructooligosaccharides (FO), indigestible glucan (IG), and polydextrose (PD). Male BALB/cAJcl mice or T cell-deficient BALB/cAJcl-nu/nu (nude) mice were fed diets supplemented with one SDF (3% w/w) for 10 weeks and we measured IgA content in their feces, plasma, lung, and submandibular gland. RESULTS In BALB/cAJcl mice, the consumption of all three SDF diets induced fecal IgA production, but the response was stronger in the IG and PD groups than in the FO group. The IgA concentration of the plasma and lung were also higher in the FO and PD groups, and these groups showed significantly higher cecal acetic and n-butyric acid content. In contrast, in nude mice, the induction of IgA production was identified only in fecal samples of mice fed the three SDF diets, even though there were significant increases in cecal SCFAs content. CONCLUSIONS The induction of IgA production by SDFs is occurred T cell-independently in the intestine, but that in the plasma, lung, and submandibular gland is T cell-dependent. SCFAs generated in the large intestine might influence the systemic immune system, but there is no clear relationship between the generation of SCFAs and intestinal IgA production in response to SDF consumption.
Collapse
|
41
|
Niu G, Jian T, Gai Y, Chen J. Microbiota and plant-derived vesicles that serve as therapeutic agents and delivery carriers to regulate metabolic syndrome. Adv Drug Deliv Rev 2023; 196:114774. [PMID: 36906231 DOI: 10.1016/j.addr.2023.114774] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/13/2023]
Abstract
The gut is a fundamental organ in controlling human health. Recently, researches showed that substances in the intestine can alter the course of many diseases through the intestinal epithelium, especially intestinal flora and exogenously ingested plant vesicles that can be transported over long distances to various organs. This article reviews the current knowledge on extracellular vesicles in modulating gut homeostasis, inflammatory response and numerous metabolic disease that share obesity as a co-morbidity. These complex systemic diseases that are difficult to cure, but can be managed by some bacterial and plant vesicles. Vesicles, due to their digestive stability and modifiable properties, have emerged as novel and targeted drug delivery vehicles for effective treatment of metabolic diseases.
Collapse
Affiliation(s)
- Guanting Niu
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Tunyu Jian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yanan Gai
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| |
Collapse
|
42
|
Dietary fiber and SCFAs in the regulation of mucosal immunity. J Allergy Clin Immunol 2023; 151:361-370. [PMID: 36543697 DOI: 10.1016/j.jaci.2022.11.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
Gut bacterial metabolites such as short-chain fatty acids (SCFAs) have important effects on immune cells and the gut. SCFAs derive from the fermentation of dietary fiber by gut commensal bacteria. Insufficient fiber intake thus compromises SCFA production and, as a consequence, the host's physiology (particularly immune functions). We propose that many Western diseases, including those associated with impaired mucosal responses such as food allergy and asthma, may be affected by insufficient fiber intake and reduced SCFA levels in the gut and blood. Insufficient fiber intake is 1 alternative, or contributor, on top of the "hygiene hypothesis" to the rise of Western lifestyle diseases, and the 2 ideas need to be reconciled. The mechanisms by which SCFAs influence immunity and gut homeostasis are varied; they include stimulation of G protein-coupled receptors (GPCRs), such as GPR43 or GPR41; inhibition of histone deacetylases (and hence, gene transcription changes); and induction of intracellular metabolic changes. SCFAs modulate at many different levels to alter mucosal homeostasis, including changes to gut epithelial integrity, increases in regulatory T-cell numbers and function, and decreased expression of numerous inflammatory cytokines. There is scope for preventing and/or treating diseases by using diets that alter SCFA levels.
Collapse
|
43
|
Castaño C, Novials A, Párrizas M. An Overview of Inter-Tissue and Inter-Kingdom Communication Mediated by Extracellular Vesicles in the Regulation of Mammalian Metabolism. Int J Mol Sci 2023; 24:2071. [PMID: 36768391 PMCID: PMC9916451 DOI: 10.3390/ijms24032071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Obesity and type 2 diabetes are associated with defects of insulin action in different tissues or alterations in β-cell secretory capacity that may be triggered by environmental challenges, inadequate lifestyle choices, or an underlying genetic predisposition. In addition, recent data shows that obesity may also be caused by perturbations of the gut microbiota, which then affect metabolic function and energy homeostasis in the host. Maintenance of metabolic homeostasis in complex organisms such as mammals requires organismal-level communication, including between the different organs and the gut microbiota. Extracellular vesicles (EVs) have been identified in all domains of life and have emerged as crucial players in inter-organ and inter-kingdom crosstalk. Interestingly, EVs found in edible vegetables or in milk have been shown to influence gut microbiota or tissue function in mammals. Moreover, there is a multidirectional crosstalk mediated by EVs derived from gut microbiota and body organs that has implications for host health. Untangling this complex signaling network may help implement novel therapies for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Carlos Castaño
- Pathogenesis and Prevention of Diabetes Group, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Pathogenesis and Prevention of Diabetes Group, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 08036 Barcelona, Spain
| | - Anna Novials
- Pathogenesis and Prevention of Diabetes Group, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Pathogenesis and Prevention of Diabetes Group, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 08036 Barcelona, Spain
| | - Marcelina Párrizas
- Pathogenesis and Prevention of Diabetes Group, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 08036 Barcelona, Spain
| |
Collapse
|
44
|
Zhou YD, Liang FX, Tian HR, Luo D, Wang YY, Yang SR. Mechanisms of gut microbiota-immune-host interaction on glucose regulation in type 2 diabetes. Front Microbiol 2023; 14:1121695. [PMID: 36891383 PMCID: PMC9986296 DOI: 10.3389/fmicb.2023.1121695] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Intestinal absorption of food is one of the sources of glucose. Insulin resistance and impaired glucose tolerance caused by lifestyle and diet are the precursors of type 2 diabetes. Patients with type 2 diabetes have trouble controlling their blood sugar levels. For long-term health, strict glycemic management is necessary. Although it is thought to be well correlated with metabolic diseases like obesity, insulin resistance, and diabetes, its molecular mechanism is still not completely understood. Disturbed microbiota triggers the gut immune response to reshape the gut homeostasis. This interaction not only maintains the dynamic changes of intestinal flora, but also preserves the integrity of the intestinal barrier. Meanwhile, the microbiota establishes a systemic multiorgan dialog on the gut-brain and gut-liver axes, intestinal absorption of a high-fat diet affects the host's feeding preference and systemic metabolism. Intervention in the gut microbiota can combat the decreased glucose tolerance and insulin sensitivity linked to metabolic diseases both centrally and peripherally. Moreover, the pharmacokinetics of oral hypoglycemic medications are also influenced by gut microbiota. The accumulation of drugs in the gut microbiota not only affects the drug efficacy, but also changes the composition and function of them, thus may help to explain individual therapeutic variances in pharmacological efficacy. Regulating gut microbiota through healthy dietary patterns or supplementing pro/prebiotics can provide guidance for lifestyle interventions in people with poor glycemic control. Traditional Chinese medicine can also be used as complementary medicine to effectively regulate intestinal homeostasis. Intestinal microbiota is becoming a new target against metabolic diseases, so more evidence is needed to elucidate the intricate microbiota-immune-host relationship, and explore the therapeutic potential of targeting intestinal microbiota.
Collapse
Affiliation(s)
- Yu-Dian Zhou
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hebei, China
| | - Feng-Xia Liang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hebei, China
| | - Hao-Ran Tian
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hebei, China
| | - Dan Luo
- Department of Respiratory Wuhan No.1 Hospital, Wuhan, Hebei, China
| | - Ya-Yuan Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hebei, China
| | - Shu-Rui Yang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hebei, China
| |
Collapse
|
45
|
Abstract
Succinate is a circulating metabolite, and the relationship between abnormal changes in the physiological concentration of succinate and inflammatory diseases caused by the overreaction of certain immune cells has become a research focus. Recent investigations have shown that succinate produced by the gut microbiota has the potential to regulate host homeostasis and treat diseases such as inflammation. Gut microbes are important for maintaining intestinal homeostasis. Microbial metabolites serve as nutrients in energy metabolism, and act as signal molecules that stimulate host cell and organ function and affect the structural balance between symbiotic gut microorganisms. This review focuses on succinate as a metabolite of both host cells and gut microbes and its involvement in regulating the gut - immune tissue axis by activating intestinal mucosal cells, including macrophages, dendritic cells, and intestinal epithelial cells. We also examined its role as the mediator of microbiota - host crosstalk and its potential function in regulating intestinal microbiota homeostasis. This review explores feasible ways to moderate succinate levels and provides new insights into succinate as a potential target for microbial therapeutics for humans.
Collapse
Affiliation(s)
- Yi-Han Wei
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiang-Chao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| |
Collapse
|
46
|
Attah FA, Lawal BA, Yusuf AB, Adedeji OJ, Folahan JT, Akhigbe KO, Roy T, Lawal AA, Ogah NB, Olorundare OE, Chamcheu JC. Nutritional and Pharmaceutical Applications of Under-Explored Knottin Peptide-Rich Phytomedicines. PLANTS (BASEL, SWITZERLAND) 2022; 11:3271. [PMID: 36501311 PMCID: PMC9737898 DOI: 10.3390/plants11233271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/02/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Phytomedicines reportedly rich in cystine knot peptides (Knottins) are found in several global diets, food/herbal supplements and functional foods. However, their knottin peptide content has largely been unexplored, notably for their emerging dual potentials at both the food and medicine space. The nutritional roles, biological targets and mechanism(s) of activity of these knotted peptides are largely unknown. Meanwhile, knottins have recently been unveiled as emerging peptide therapeutics and nutraceuticals of primary choice due to their broad spectrum of bioactivity, hyper stability, selective toxicity, impressive selectivity for biomolecular targets, and their bioengineering applications. In addition to their potential dietary benefits, some knottins have displayed desirable limited toxicity to human erythrocytes. In an effort to appraise what has been accomplished, unveil knowledge gaps and explore the future prospects of knottins, an elaborate review of the nutritional and pharmaceutical application of phytomedicines rich in knottins was carried out. Herein, we provide comprehensive data on common dietary and therapeutic knottins, the majority of which are poorly investigated in many food-grade phytomedicines used in different cultures and localities. Findings from this review should stimulate scientific interest to unveil novel dietary knottins and knottin-rich nutraceutical peptide drug candidates/leads with potential for future clinical application.
Collapse
Affiliation(s)
- Francis Alfred Attah
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Bilqis Abiola Lawal
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Abdulmalik Babatunde Yusuf
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Oluwakorede Joshua Adedeji
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Joy Temiloluwa Folahan
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana-Monroe, Monroe, LA 71209, USA
| | - Kelvin Oluwafemi Akhigbe
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Tithi Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana-Monroe, Monroe, LA 71209, USA
| | - Azeemat Adeola Lawal
- Department of Pharmacognosy and Drug Development, Faculty of Pharmaceutical Sciences, University of Ilorin, Ilorin 240272, Nigeria
| | - Ngozi Blessing Ogah
- Department of Biotechnology, Ebonyi State University, Abakaliki 480101, Nigeria
| | | | - Jean Christopher Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana-Monroe, Monroe, LA 71209, USA
| |
Collapse
|
47
|
Zou Y, Meng JX, Wei XY, Gu XY, Chen C, Geng HL, Yang LH, Zhang XX, Cao HW. CircRNA and miRNA expression analysis in livers of mice with Toxoplasma gondii infection. Front Cell Infect Microbiol 2022; 12:1037586. [PMID: 36389171 PMCID: PMC9646959 DOI: 10.3389/fcimb.2022.1037586] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/07/2022] [Indexed: 11/23/2022] Open
Abstract
Toxoplasmosis is an important zoonotic parasitic disease caused by Toxoplasma gondii (T. gondii). However, the functions of circRNAs and miRNAs in response to T. gondii infection in the livers of mice at acute and chronic stages remain unknown. Here, high-throughput RNA sequencing was performed for detecting the expression of circRNAs and miRNAs in livers of mice infected with 20 T. gondii cysts at the acute and chronic stages, in order to understand the potential molecular mechanisms underlying hepatic toxoplasmosis. Overall, 265 and 97 differentially expressed (DE) circRNAs were found in livers at the acute and chronic infection stages in comparison with controls, respectively. In addition, 171 and 77 DEmiRNAs were found in livers at the acute and chronic infection stages, respectively. Functional annotation showed that some immunity-related Gene ontology terms, such as “positive regulation of cytokine production”, “regulation of T cell activation”, and “immune receptor activity”, were enriched at the two infection stages. Moreover, the pathways “Valine, leucine, and isoleucine degradation”, “Fatty acid metabolism”, and “Glycine, serine, and threonine metabolism” were involved in liver disease. Remarkably, DEcircRNA 6:124519352|124575359 was significantly correlated with DEmiRNAs mmu-miR-146a-5p and mmu-miR-150-5p in the network that was associated with liver immunity and pathogenesis of disease. This study revealed that the expression profiling of circRNAs in the livers was changed after T. gondii infection, and improved our understanding of the transcriptomic landscape of hepatic toxoplasmosis in mice.
Collapse
Affiliation(s)
- Yang Zou
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
- College of Life Sciences, Changchun Sci-Tech University, Changchun, China
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jin-Xin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Xin-Yu Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiao-Yi Gu
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Chao Chen
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Hong-Li Geng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Li-Hua Yang
- College of Life Sciences, Changchun Sci-Tech University, Changchun, China
- *Correspondence: Li-Hua Yang, ; Xiao-Xuan Zhang, ; Hong-Wei Cao,
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
- *Correspondence: Li-Hua Yang, ; Xiao-Xuan Zhang, ; Hong-Wei Cao,
| | - Hong-Wei Cao
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
- *Correspondence: Li-Hua Yang, ; Xiao-Xuan Zhang, ; Hong-Wei Cao,
| |
Collapse
|
48
|
Pinget GV, Tan JK, Ni D, Taitz J, Daien CI, Mielle J, Moore RJ, Stanley D, Simpson S, King NJC, Macia L. Dysbiosis in imiquimod-induced psoriasis alters gut immunity and exacerbates colitis development. Cell Rep 2022; 40:111191. [PMID: 35977500 DOI: 10.1016/j.celrep.2022.111191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 05/30/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Psoriasis has long been associated with inflammatory bowel disease (IBD); however, a causal link is yet to be established. Here, we demonstrate that imiquimod-induced psoriasis (IMQ-pso) in mice disrupts gut homeostasis, characterized by increased proportions of colonic CX3CR1hi macrophages, altered cytokine production, and bacterial dysbiosis. Gut microbiota from these mice produce higher levels of succinate, which induce de novo proliferation of CX3CR1hi macrophages ex vivo, while disrupted gut homeostasis primes IMQ-pso mice for more severe colitis with dextran sulfate sodium (DSS) challenge. These results demonstrate that changes in the gut environment in psoriasis lead to greater susceptibility to IBD in mice, suggesting a two-hit requirement, that is, psoriasis-induced altered gut homeostasis and a secondary environmental challenge. This may explain the increased prevalence of IBD in patients with psoriasis.
Collapse
Affiliation(s)
- Gabriela Veronica Pinget
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jian Kai Tan
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Duan Ni
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jemma Taitz
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Claire Immediato Daien
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; CHRU Montpellier, University of Montpellier & INSERM U1046, CNRS UMR, PhyMedExp, 9214 Montpellier, France
| | - Julie Mielle
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; CHRU Montpellier, University of Montpellier & INSERM U1046, CNRS UMR, PhyMedExp, 9214 Montpellier, France
| | | | - Dragana Stanley
- School of Health, Medical and Applied Sciences, Central Queensland University, Kawana, QLD 4701, Australia
| | - Stephen Simpson
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, Faculty of Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Nicholas Jonathan Cole King
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Laurence Macia
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Sydney Cytometry, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|