1
|
Kim SM, Shin HY, Sim SH, Rho Y, Yu KW, Shin KS. Effects of pectin-type polysaccharides derived from Houttuynia cordata on the intestinal immune system in native C3H/HeN mice. Int J Biol Macromol 2025; 309:142995. [PMID: 40210073 DOI: 10.1016/j.ijbiomac.2025.142995] [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: 12/26/2024] [Revised: 03/18/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Dietary polysaccharides have attracted considerable attention because of their species-specific chemical composition and diverse biological activities. Polysaccharides derived from Houttuynia cordata (HC) exhibit broad pharmacological activities, emphasizing their diverse roles in biological processes. This study investigated the effects of HC-derived polysaccharides on Peyer's patch (PP)-mediated intestinal immune response. The crude polysaccharide (HCP) isolated from HC extract (HCE) was identified as a pectin-type polysaccharide rich in the homogalacturonan (HG) domain, which was further hydrolyzed by endo-polygalacturonase to produce HCPE. In vitro, HCPE significantly enhanced PP-mediated cytokine secretion and bone marrow cell (BMC) proliferation compared with HCP. In vivo, a 4-week treatment with oral HCPE stimulated PP-mediated secretion of hematopoietic growth factors and promoted BMC proliferation. Additionally, HCPE upregulated IgA-associated factors, leading to increased IgA levels in the small intestine, serum, and feces, while also significantly elevating short-chain fatty acid levels, potentially improving the gut environment. To the best of our knowledge, this is the first study to systematically analyze PP-mediated intestinal immunostimulatory activity of HC-derived polysaccharides both in vitro and in vivo. These findings provide valuable insights into the potential of HCPE as an intestinal immunostimulatory agent and establish a foundation for future research on the immunological effects of HC-derived polysaccharides.
Collapse
Affiliation(s)
- So Min Kim
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Se Hyeon Sim
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - YangKook Rho
- PharmacoBio Co. Ltd., Seongnam 13219, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| |
Collapse
|
2
|
Ahn SY, Joo HG, Ko EJ. Lactobacillus johnsonii JERA01 activates macrophages and increases Th-1 T cell population in mouse small intestine. PLoS One 2025; 20:e0320946. [PMID: 40273138 PMCID: PMC12021164 DOI: 10.1371/journal.pone.0320946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/26/2025] Open
Abstract
Lactobacillus johnsonii is a commensal bacterium isolated from the vaginal and gastrointestinal tracts of vertebrate hosts, including humans. It is a potential anti-inflammatory bacterium. As reported in many animal studies, L. johnsonii supplementation reduces inflammation in the intestine and enhances the epithelial barrier. However, in this study, we observed immunostimulatory effects of heat-killed L. johnsonii JERA01 (LJ) supplementation on antigen-presenting cells, such as dendritic cells and macrophages, in mice. LJ pretreatment increased the expression of maturation markers and TNF-α, IL-6, IL-12p40 production in bone marrow-derived dendritic cells and macrophages (BMDCs and BMDMs). Co-culture of LJ-pretreated BMDCs or BMDMs with lymphocytes enhanced IFN-γ production in vitro. Oral LJ (108 CFU) supplementation induced macrophage infiltration into the peritoneal cavity and Peyer's patch at 12-h after administration, resulting in an increase in the population of IFN-γ-producing T cells in the Peyer's patch. Our investigation revealed the effects of LJ, which activates macrophages and increases the Th-1 T cell population in the intestine, implying the possibility of using L. johnsonii as an immune stimulator.
Collapse
Affiliation(s)
- So Yeon Ahn
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Hong-Gu Joo
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| | - Eun-Ju Ko
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
3
|
Zhang J, Tang C, Liu Y, Sun J, Li X, Long K, Kong F, Chen S, Liang H, Ding Y, Li M, Shen L, Ge J, Ma J, Ge L. Single cell transcriptome profiling of immune tissues from germ-free and specific pathogen-free piglet. Sci Data 2025; 12:652. [PMID: 40251240 PMCID: PMC12008294 DOI: 10.1038/s41597-025-04957-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/04/2025] [Indexed: 04/20/2025] Open
Abstract
The commensal microbiota provides immunomodulatory signals during the development, differentiation and activation of immune cells, and is crucial for maintaining host immune homeostasis. However, the systematic effects of commensal microbiota on host immunity based on large animal model at the single-cell level remain to be resolved. Here, we utilized single-cell RNA sequencing (scRNA-seq) to analyze the transcriptome profiling containing 57,720 cells from three important immune tissues [Peyer's patches (PP), mesenteric lymph node (MLN), and spleen] of germ-free (GF) and specific pathogen-free (SPF) piglet. We presented detailed description of the dataset and preliminarily identified the major cell types including immune and non-immune cells, and further annotated the immune cell subsets. This dataset provides a data mining resource for researchers involved in microbe-host interactions, and enables in-depth analysis of cell map alterations caused by the microbiota colonization during early immune development. As the first single-cell transcriptomics dataset for immune tissue of GF and SPF piglet, this provides a valuable data resource for the study of commensal microbe-host immunity regulation.
Collapse
Affiliation(s)
- Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
- Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chuang Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Liu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- Sichuan Animal Science Academy, Chengdu, 610066, Sichuan, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Xiaokai Li
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Keren Long
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Fanli Kong
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Shuangshuang Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hao Liang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Yuchun Ding
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Li Shen
- Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Junbo Ge
- Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jideng Ma
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.
- Ministry of Agriculture Key Laboratory of Pig Industry Sciences, Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China.
| |
Collapse
|
4
|
Colombo APV, Lourenço TGB, de Oliveira AM, da Costa ALA. Link Between Oral and Gut Microbiomes: The Oral-Gut Axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:71-87. [PMID: 40111686 DOI: 10.1007/978-3-031-79146-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
In the last decades, groundbreaking research on the human microbiome has changed our reductionist conception of the etiology and pathogenesis of several chronic diseases. As a result, we have come to appreciate the significance of a balanced microbiome in maintaining human health. In this context, the upper and lower gastrointestinal tracts (GITs) comprise the most abundant and diverse microbiotas of the human body. In addition to its diversity, functional redundancy, and temporal stability, a healthy GIT microbiome is characterized by its body site specificity. In fact, current evidence has indicated that the translocation of oral species to the gut environment through the oral-gut axis is increased in an array of illnesses, including chronic inflammatory and metabolic diseases, neurological disorders, and cancer. Oral pathogens have also been shown to promote gut dysbiosis and systemic inflammation in animal models. Yet, some level of overlapping between oral and gut microbiomes may occur without disruption of these microbial communities and loss of site specificity. The uniqueness of each host-microbiome entity may hinder our ability to define a "universal" normal GIT microbiome. Despite that, this chapter summarizes the predominant health-related taxa along the human GIT, as well as their role in the physiology and immunity of the digestive system. Some mechanisms that may lead to disturbances and relevant shifts in the oral and gut microbiomes of major inflammatory chronic diseases are also pointed out. Lastly, oral-fecal microbial signatures are presented as potential biomarkers for several oral and systemic disorders. The recognition of such symbiotic/dysbiotic microbial profiles may provide insights into the development of more accurate early diagnosis and therapeutic ecological approaches to restore the balance of the GIT microbiome.
Collapse
Affiliation(s)
- Ana Paula Vieira Colombo
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil.
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | - Adriana Miranda de Oliveira
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
5
|
Cerovic V, Pabst O, Mowat AM. The renaissance of oral tolerance: merging tradition and new insights. Nat Rev Immunol 2025; 25:42-56. [PMID: 39242920 DOI: 10.1038/s41577-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Oral tolerance is the process by which feeding of soluble proteins induces antigen-specific systemic immune unresponsiveness. Oral tolerance is thought to have a central role in suppressing immune responses to 'harmless' food antigens, and its failure can lead to development of pathologies such as food allergies or coeliac disease. However, on the basis of long-standing experimental observations, the relevance of oral tolerance in human health has achieved new prominence recently following the discovery that oral administration of peanut proteins prevents the development of peanut allergy in at-risk human infants. In this Review, we summarize the new mechanistic insights into three key processes necessary for the induction of tolerance to oral antigens: antigen uptake and transport across the small intestinal epithelial barrier to the underlying immune cells; the processing, transport and presentation of fed antigen by different populations of antigen-presenting cells; and the development of immunosuppressive T cell populations that mediate antigen-specific tolerance. In addition, we consider how related but distinct processes maintain tolerance to bacterial antigens in the large intestine. Finally, we outline the molecular mechanisms and functional consequences of failure of oral tolerance and how these may be modulated to enhance clinical outcomes and prevent disease.
Collapse
Affiliation(s)
- Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| | - Allan McI Mowat
- School of Infection and Immunity, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
6
|
Ren H, Dai L, Ma C, Zhou L, Wang L. Hydrolysates exhibited differential modulatory effects on macrophage compared to the raw polysaccharide (xyloglucomannan) isolated from Atractylodes macrocephala Koidz. Prep Biochem Biotechnol 2024; 55:620-633. [PMID: 40311656 DOI: 10.1080/10826068.2024.2444979] [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: 05/03/2025]
Abstract
It has been claimed that Atractylodes macrocephala applied as a traditional Chinese medicinal herb for centuries, owing to its excellent immunomodulatory and hepatic protective properties. This study aims to explore the immunomodulation of oligosaccharides obtained by gastrointestinal digestion after oral administration of A. macrocephala polysaccharide (xyloglucomannan, XGM). The analysis of chemistry showed that XGM consisted of D-glucose, D-mannose, and D-xylose in a molar ratio of 6.8:3.0:1.0, and the molecular weight was 5465 Da. Furthermore, HCl and pancreatic amylase were used to simulate digestive tract hydrolysis of polysaccharides, obtaining two XGM hydrolysates (XGM-AH and -EH). Oligosaccharide identification results indicated that both XGM-AH and XGM-EH contained glucans (degree of polymerization, DP = 2 ∼ 5). XGM-AH had a wider variety of oligosaccharides than XGM-EH, mainly glucomannans. In vitro immunostimulatory assay indicated that XGM could effectively stimulate the activation and enhance the phagocytosis of RAW264.7 cells. In contrast, XGM-AH and -EH exhibited anti-inflammatory effects, inhibited lipopolysaccharide (LPS)-induced aberrant activation of macrophages, reduced the release of cytokines of macrophages. Flow cytometry assay suggested that XGM-AH and -EH inhibited LPS-induced M1-type polarization of macrophages. In conclusion, XGM-derived oligosaccharides possess anti-inflammatory bioactivities and exhibit differential macrophage regulatory behaviors in contrast to the immune-activating effects exhibited by the prototype polysaccharides.
Collapse
Affiliation(s)
- Huanzhi Ren
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Longchao Dai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Chang Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Luyao Zhou
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| | - Lingchong Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
7
|
Wang Z, Zhou L, Zhong X, Jiang Y, Zhang Z, Li W. Liquid-liquid separation in gut immunity. Front Immunol 2024; 15:1505123. [PMID: 39720729 PMCID: PMC11666445 DOI: 10.3389/fimmu.2024.1505123] [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: 10/02/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Gut immunity is essential for maintaining intestinal health. Recent studies have identified that intracellular liquid-liquid phase separation (LLPS) may play a significant role in regulating gut immunity, however, the underlying mechanisms remain unclear. LLPS refers to droplet condensates formed through intracellular molecular interactions, which are crucial for the formation of membraneless organelles and biomolecules. LLPS can contribute to the formation of tight junctions between intestinal epithelial cells and influence the colonization of probiotics in the intestine, thereby protecting the intestinal immune system by maintaining the integrity of the intestinal barrier and the stability of the microbiota. Additionally, LLPS can affect the microclusters on the plasma membrane of T cells, resulting in increased density and reduced mobility, which in turn influences T cell functionality. The occurrence of intracellular LLPS is intricately associated with the initiation and progression of gut immunity. This review introduces the mechanism of LLPS in gut immunity and analyzes future research directions and potential applications of this phenomenon.
Collapse
Affiliation(s)
- Zhaoyang Wang
- Department of Gastrointestinal Surgery, Huadu District People’s Hospital, Guangzhou, China
- Biology, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Lili Zhou
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Xiaolan Zhong
- Department of Gastroenterology, Huadu District People’s Hospital, Guangzhou, China
| | - Yiguo Jiang
- Biology, School of Public Health, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Xinzao, Guangzhou, China
| | - Zhentao Zhang
- Obstetrics and Gynecology Department, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wanglin Li
- Department of Gastrointestinal Surgery, Huadu District People’s Hospital, Guangzhou, China
- Department of Gastroenterology, Huadu District People’s Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Fertitta V, Varano B, Del Cornò M, Fortini P, Aureli A, Conti L. Akkermansia muciniphila- and Pathogenic Bacteria-Derived Endotoxins Differently Regulate Human Dendritic Cell Generation and γδ T Lymphocyte Activation. Biomolecules 2024; 14:1571. [PMID: 39766278 PMCID: PMC11673428 DOI: 10.3390/biom14121571] [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: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Lipopolysaccharide (LPS) is a potent endotoxin released at high concentrations in acute infections, causing massive host inflammatory response. Accumulating evidence indicates that dysbiosis-associated chronic low levels of circulating LPS can sustain a prolonged sterile low-grade inflammation that increases the risk of several non-communicable diseases. Interventions aimed at increasing the abundance of beneficial/probiotic bacteria, including Akkermansia muciniphila, result in reduced inflammation, favoring metabolic and immune health. Immunosuppression is a common feature in conditions of chronic inflammation, and dendritic cells (DCs) represent key targets given their ability to shift the balance toward immunity or tolerance. In this study, the effects of low concentrations of LPS from pathogenic (Escherichia coli and Salmonella enterica) and probiotic (Akkermansia muciniphila) bacterial species on human DC generation and functions were compared. We report that monocyte precursor priming with Escherichia coli and Salmonella enterica LPS forces the differentiation of PD-L1-expressing DCs, releasing high levels of IL-6 and IL-10, and impairs their capacity to drive full TCR-Vδ2 T cell activation. Conversely, comparable concentrations of Akkermansia muciniphila promoted the generation of DCs with preserved activating potential and immunostimulatory properties. These results shed light on potential mechanisms underlying the impact of low endotoxemia on disease risk and pathogenesis, and increase our understanding of the immunomodulatory effects of Akkermansia muciniphila.
Collapse
Affiliation(s)
- Veronica Fertitta
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.F.); (P.F.)
| | - Barbara Varano
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| | - Manuela Del Cornò
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| | - Paola Fortini
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (V.F.); (P.F.)
| | - Anna Aureli
- Institute of Translational Pharmacology, National Research Council, 67100 L’Aquila, Italy;
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.V.); (M.D.C.)
| |
Collapse
|
9
|
Zhang B, Yang H, Cai G, Nie Q, Sun Y. The interactions between the host immunity and intestinal microorganisms in fish. Appl Microbiol Biotechnol 2024; 108:30. [PMID: 38170313 DOI: 10.1007/s00253-023-12934-1] [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: 08/03/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024]
Abstract
There is a huge quantity of microorganisms in the gut of fish, which exert pivotal roles in maintaining host intestinal and general health. The fish immunity can sense and shape the intestinal microbiota and maintain the intestinal homeostasis. In the meantime, the intestinal commensal microbes regulate the fish immunity, control the extravagant proliferation of pathogenic microorganisms, and ensure the intestinal health of the host. This review summarizes developments and progress on the known interactions between host immunity and intestinal microorganisms in fish, focusing on the recent advances in zebrafish (Danio rerio) showing the host immunity senses and shapes intestinal microbiota, and intestinal microorganisms tune host immunity. This review will offer theoretical references for the development, application, and commercialization of intestinal functional microorganisms in fish. KEY POINTS: • The interactions between the intestinal microorganisms and host immunity in zebrafish • Fish immunity senses and shapes the microbiota • Intestinal microbes tune host immunity in fish.
Collapse
Affiliation(s)
- Biyun Zhang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Hongling Yang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Guohe Cai
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Qingjie Nie
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China
| | - Yunzhang Sun
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, Fujian, China.
| |
Collapse
|
10
|
Jun YK, Kim N, Yoon H, Park JH, Kim HK, Choi Y, Lee JA, Shin CM, Park YS, Lee DH. Molecular Activity of Inflammation and Epithelial-Mesenchymal Transition in the Microenvironment of Ulcerative Colitis. Gut Liver 2024; 18:1037-1047. [PMID: 38384179 PMCID: PMC11565011 DOI: 10.5009/gnl230283] [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: 07/22/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 02/23/2024] Open
Abstract
Background/Aims : The genetic expression in the active inflammatory regions is increased in ulcerative colitis (UC) with endoscopic activity. The aim of this study was to investigate the molecular activity of inflammation and tissue remodeling markers in endoscopically inflamed and uninflamed regions of UC. Methods : Patients with UC (n=47) and controls (n=20) were prospectively enrolled at the Seoul National University Bundang Hospital. Inflamed tissue was obtained at the most active lesion, and uninflamed tissue was collected from approximately 15 cm above the upper end of the active lesion via colonoscopic biopsies. The messenger RNA expression levels of transforming growth factor β (TGF-β), interleukin (IL)-1β, IL-6, IL-17A, E-cadherin, olfactomedin-4 (OLFM4), leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), vimentin, fibroblast-specific protein-1 (FSP1), and α-smooth muscle actin (SMA) were evaluated. Mucosal healing (MH) was defined according to a Mayo endoscopic score of 0, 1 or non-MH (Mayo endoscopic score of 2 or 3). Results : The messenger RNA expressions of TGF-β, IL-1β, OLFM4, FSP1, vimentin, and α-SMA were significantly higher, and that of E-cadherin was significantly lower in inflamed and uninflamed regions of patients with UC than those in controls. In the inflamed regions, patients in the non-MH group had significantly increased genetic expression of TGF-β, FSP1, vimentin, and α-SMA compared to patients in the MH group. Similarly, the non-MH group had significantly higher genetic expression of TGF-β, IL-1β, IL-6, vimentin, and α-SMA than the MH group in the uninflamed regions. Conclusions : Endoscopic activity in UC suggests inflammation and tissue remodeling of uninflamed regions similar to inflamed regions (ClinicalTrials.gov, NCT05653011).
Collapse
Affiliation(s)
- Yu Kyung Jun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung Kyung Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Pathology and Translational Genomics, Samsung Medical Center, Seoul, Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Ae Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Lee MJ, Shin S, Kim HW, Ko MK, Park SH, Kim SM, Park JH. Oral Administration of Zinc Sulfate with Intramuscular Foot-and-Mouth Disease Vaccine Enhances Mucosal and Systemic Immunity. Vaccines (Basel) 2024; 12:1268. [PMID: 39591171 PMCID: PMC11598382 DOI: 10.3390/vaccines12111268] [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: 10/09/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Foot-and-mouth disease (FMD) remains a significant global threat to livestock farming. Current commercial FMD vaccines present several challenges, including the risk of infection and adverse injection site reactions due to oil-based adjuvants. The complex immune environment of the gut-associated lymphoid tissue has the potential to induce broad and diverse immune responses. Therefore, we aimed to explore the potential of zinc sulfate as an oral adjuvant to enhance intestinal mucosal immunity and complement the effects of intramuscular (IM) FMD vaccination. Methods: We conducted serological analyses on mice and pigs, measuring secretory IgA (sIgA) levels and evaluating the expression of mucosal immunity-related genes in pigs. These assessments were used to investigate the systemic and mucosal immune responses induced by oral zinc sulfate administration in combination with an IM FMD vaccine. Results: This combination strategy significantly increased structural protein antibody titers and virus neutralization titers in experimental animals (mice) and target animals (pigs) across early, mid-, and long-term periods. Additionally, this approach enhanced the expression of key cytokines associated with mucosal immunity and increased sIgA levels, which are critical markers of mucosal immunity. Conclusions: Oral zinc sulfate administration may synergize with inactivated FMD vaccines, leading to sustained and enhanced long-term immune responses. This novel strategy could reduce the frequency of required vaccinations or allow for a lower antigen dose in vaccines, effectively stimulating the mucosal immune system and boosting systemic immunity. This approach has the potential to improve the overall efficacy of commercial FMD vaccines.
Collapse
|
12
|
Meskini M, Amanzadeh A, Salehi F, Bouzari S, Karimipoor M, Fuso A, Fateh A, Siadat SD. A protocol to isolate and characterize pure monocytes and generate monocyte-derived dendritic cells through FBS-Coated flasks. Sci Rep 2024; 14:23956. [PMID: 39397067 PMCID: PMC11471755 DOI: 10.1038/s41598-024-75376-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
This study explores methods to isolate high-pure monocytes and optimize the best growth factor concentration to generate monocytes-derived dendritic cells (mo-DCs), subset DC1, which is crucial in immune responses. Three protocols for monocyte isolation from peripheral blood mononuclear cells (PBMCs) were evaluated: three-hour incubation on FBS-coated flasks; an overnight incubation on FBS-coated flasks; and Magnetic Activated Cell Sorting (MACS). Additionally, five different concentrations of human recombinant granulocyte-macrophage colony-stimulating factor (hrGM-CSF) and human recombinant interleukin-4 (hrIL-4) were compared. We used Flow cytometry to assess the isolation, purification, and generation of pure monocytes characterized as CD14+, and expression of mo-DC classical markers (HLA-DR, CD80, CD83, and CD86). The obtained results show that monocytes isolated with the second method (overnight incubation) had the highest purity (P < 0.0001) but the lowest yield (P > 0.05), balancing purity and cost-effectiveness. A combination of hrGM-CSF and hrIL-4 at 400 U/mL produced the most favorable outcomes, leading to the highest rate of mo-DC generation (P < 0.05). Notably, this concentration resulted in increasing expression of HLA-DR, CD80, and CD86 surface markers in the generated DCs (P < 0.0001), with no changes in CD83 expression levels. In conclusion, this study offers valuable insights into selecting the optimal approach for monocyte isolation and mo-DC generation in various research contexts, providing a foundation for more effective immunological studies.
Collapse
Affiliation(s)
- Maryam Meskini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Fahimeh Salehi
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Karimipoor
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, 00161, Italy
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
13
|
Guimarães-Pinto K, Leandro M, Corrêa A, Maia EP, Rodrigues L, da Costa ALA, Rafael Machado Ferreira J, Claudio-Etienne E, Siebenlist U, He J, Rigoni TDS, Ferreira TPT, Jannini-Sa YAP, Matos-Guedes HL, Costa-da-Silva AC, Lopes MF, Silva PMR, Kelsall BL, Filardy AA. Differential regulation of lung homeostasis and silicosis by the TAM receptors MerTk and Axl. Front Immunol 2024; 15:1380628. [PMID: 38774866 PMCID: PMC11106457 DOI: 10.3389/fimmu.2024.1380628] [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: 02/01/2024] [Accepted: 04/10/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 μL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-β and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.
Collapse
Affiliation(s)
- Kamila Guimarães-Pinto
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Monique Leandro
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonia Corrêa
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ester P. Maia
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia Rodrigues
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - André Luiz Amorim da Costa
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Estefannia Claudio-Etienne
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Ulrich Siebenlist
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Jianping He
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Thaís da Silva Rigoni
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | - Herbert Leonel Matos-Guedes
- Laboratório de Imunobiotecnologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Caroline Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Maryland, MD, United States
| | - Marcela Freitas Lopes
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Brian Lee Kelsall
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Alessandra Almeida Filardy
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
14
|
Guan Y, Zheng W, Bai Y, Wu B. Yupingfeng polysaccharide promote the growth of chickens via regulating gut microbiota. Front Vet Sci 2024; 11:1337698. [PMID: 38464700 PMCID: PMC10920335 DOI: 10.3389/fvets.2024.1337698] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Yupingfeng polysaccharide (YPF-P) is the main substance of alcohol deposition in Yupingfeng powder, which has many biological functions such as enhancing immunity, repairing intestinal barrier and enhancing antioxidant ability. This study employed in vitro growth-promoting drug feed additives and animal experiments to comprehensively evaluate the use of YPF-P in broiler production. Methods A total of 1,296 151 days-old Qingyuan Partridge chickens were randomly divided into four groups with six replicates and 54 hens per replicate: the control group was fed basal diet, and the experimental groups were fed diets supplemented with 4 g/kg, 8 g/kg, and 12 g/kg YPF-P for 14 days. Broilers were weighed before and at the end of the experiment to calculate total weight gain (GW), average daily gain (ADG), and feed compensation. At the end of the experiment, six chickens from each group were randomly selected for subwing vein blood sampling, which was used to measure serum biochemical indicators GHRH, GH, and IGF-1 by ELISA method. Randomly select chickens from control group and 8 g/kg group for slaughter, and cecal contents were collected for 16S high-throughput sequencing. Results Dietary supplementation of 8 g/kg YPF-P can significantly increase the final body weight, total weight gain, average daily gain and decrease the feed to gain ratio of chickens. During 151-165 days, serum IGF-1 concentrations increased significantly (p < 0.05). There were no significant changes in serum GH concentration (p > 0.05). In terms of gut microbiota, there was no significant difference between control group and test group in Shannon index and Simpson index. Compared with the control group,the addition of 8 g/kgYPF-P significantly increased the abundance of Firmicutes and significantly decreased the abundance of Bacteroides at the phylum level.At the genus level, the relative abundance of unclassified_Oscillospiraceae was significantly increased and the unclassified_Muribaculaceae, uncultured_Bacteroidales_bacterium, Lactobacillus, Alloprevotella, Ligilactobacillus, Prevotellaceae_UCG_001, and unclassified_Atopobiaceae was significantly decreased. Conclusion The above results showed that adding 8 mg/kg of YPF-P could increase the average daily gain of Qingyuan Partridge chickens, reduce the ratio of feed to meat, and affect the distribution proportion of intestinal microflora in chickens to some extent.
Collapse
Affiliation(s)
| | | | | | - Bo Wu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
15
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [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: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|