1
|
Nguyen N, Hoang TM, Huang TY, Nguyen LDH, Chang HH, Chang Y, Thi Nguyen MT, Lin KJ, Chen CC, Sung HW. Macrophage-hitchhiked, effervescence-induced nanoemulsions for enhanced oral chemotherapy and immunotherapy: Impact on absorption route. Biomaterials 2024; 316:123019. [PMID: 39700534 DOI: 10.1016/j.biomaterials.2024.123019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/08/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer. Paclitaxel (PTX), typically administered intravenously (IV) as chemotherapy, shows promise for triggering immunogenic cell death (ICD) and may serve as a potential immunotherapy. This study introduces an oral PTX delivery method using an enteric-coated gelatin capsule containing capric acid oil and an effervescent agent, optionally with decylamine-conjugated β-glucans (DA-βGlus). Upon dissolving in the small intestine, the capsule undergoes an effervescence reaction that produces emulsified oil droplets (ODs) by bile salts, forming either Bared/ODs/PTX or DA-βGlus/ODs/PTX, with the latter featuring surface-attached DA-βGlus. The study evaluates the oral absorption, pharmacokinetics, and therapeutic efficacy of these formulations, comparing them to IV administration. IV PTX causes rapid spikes in plasma concentration, quick metabolism, and elimination, which can be unsafe. In contrast, the oral delivery system maintains consistent drug levels in the bloodstream for longer periods, improving overall effectiveness. Bared/ODs/PTX follows conventional fat absorption pathways, limiting tumor targeting. On the other hand, DA-βGlus/ODs/PTX uses DA-βGlus to enhance specificity for tumors through endogenous macrophage-mediated transport, effectively acting as "cellular tumor-seeking vehicles". This method reduces tumor stroma fibrosis, delivers PTX precisely, induces apoptosis, triggers PTX-induced ICD, and enhances cytotoxic T cell responses, augmenting targeted anti-PDAC strategies.
Collapse
Affiliation(s)
- Nhien Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tuyet-Mai Hoang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tun-Yu Huang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Lam-Duc-Huy Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiao-Huan Chang
- Department of Surgery, School of Medicine, Taipei Medical University, and Division of Cardiovascular Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen Chang
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Mai Thanh Thi Nguyen
- Faculty of Chemistry, University of Science, Vietnam National University-Ho Chi Minh City, Ho Chi Minh, Viet Nam
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Chieh Chen
- Department of Orthopedic Surgery, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei, Taiwan.
| | - Hsing-Wen Sung
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
2
|
Villablanca EJ. Organismal mucosal immunology: A perspective through the eyes of game theory. Mucosal Immunol 2024:S1933-0219(24)00126-0. [PMID: 39672543 DOI: 10.1016/j.mucimm.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
In complex organisms, functional units must interact cohesively to maintain homeostasis, especially within mucosal barriers that house diverse, specialized cell exposed to constant environmental challenges. Understanding how homeostasis at mucosal barriers is maintained and how its disruption can lead to autoimmune diseases or cancer, requires a holistic view. Although omics approaches and systems immunology have become powerful tools, they are not without limitations; interpretations may reflect researchers' assumptions, even if other explanations exist. In this perspective, I propose that applying game theory concepts to mucosal immunology could help interpret complex data, offering fresh perspectives and supporting the exploration of alternative scenarios. By framing the mucosal immune system as a network of strategic interactions with multiple possible outcomes, game theory, which analyzes strategic interactions and decision-making processes, could illuminate novel cell types and functions, cell interactions, and responses to pathogens and commensals, leading to a more comprehensive understanding of immune homeostasis and diseases. In addition, game theory might encourage researchers to consider a broader range of possibilities, reduce the risk of myopic thinking, and ultimately enable a more refined and comprehensive understanding of the complexity of the immune system at mucosal barriers. This perspective aims to introduce game theory as a complementary framework for mucosal immunologists, encouraging them to incorporate these concepts into data interpretation and system modeling.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden; Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
3
|
Takahashi M, Nagata K, Watanuki Y, Yamaguchi M, Ishii K, Harada T, Minamikawa N, Katagiri M, Zhao W, Ito N, Yashiro T, Nishiyama C. Kaempferol Exerts Anti-Inflammatory Effects by Accelerating Treg Development via Aryl Hydrocarbon Receptor-Mediated and PU.1/IRF4-Dependent Transactivation of the Aldh1a2/Raldh2 Gene in Dendritic Cells. Allergy 2024. [PMID: 39660951 DOI: 10.1111/all.16410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/21/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024]
Affiliation(s)
- Miki Takahashi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Kazuki Nagata
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Yumi Watanuki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Masaki Yamaguchi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Kenta Ishii
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Tomohiro Harada
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Natsuki Minamikawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Mayuka Katagiri
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Weiting Zhao
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Naoto Ito
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Takuya Yashiro
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Chiharu Nishiyama
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| |
Collapse
|
4
|
Montasser A, Dakrory AE, Ibrahim MIM, El Zayyat E, Tallima H, El Ridi R. Differential murine responses to Schistosoma mansoni eggs in the liver and small intestine lead to downmodulation of hepatic but not intestinal periovular granulomas. Infect Immun 2024; 92:e0036224. [PMID: 39560403 PMCID: PMC11629614 DOI: 10.1128/iai.00362-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024] Open
Abstract
To control schistosomiasis mansoni, it is important to attempt preventing the worms' egg-induced pathology in the liver and limiting pathogen transmission following egg exit from the intestines to the exterior. Therefore, the present study aimed to clarify the reasons behind the decades-long riddle of periovular granulomas downmodulation in the liver, but not the small intestine, with the progression of murine schistosomiasis mansoni. Outbred female CD-1 mice were percutaneously exposed to 15 Schistosoma mansoni cercariae. The liver and small intestine were collected from mice harboring a minimum of a worm couple at 8, 12, 16, and 20 weeks post-infection, assessed for egg counts/g and histopathological changes, and used to prepare Triton X-100 extracts. Content of cytokines, saturated and unsaturated fatty acids, triglycerides, cholesterol, reactive oxygen species, and uric acid per mg tissue extract proteins were evaluated using capture enzyme-linked immunosorbent assays, gas chromatography-flame ionization detector, and standard commercially available reagents, respectively. Examination of hematoxylin-eosin-stained tissue sections confirmed the decrease in size and changes in cellular composition of periovular granulomas in the liver but not the small intestine, associated with wide differences in released cytokines types and amounts, and content of the bioactive lipids, arachidonic and docosahexaenoic acids, reactive oxygen species, and uric acid. The results together disclosed that the downmodulation of hepatic, but not the small intestine, circumoval granulomas with the progression of murine S. mansoni naturally results from site- and tissue- specific immunological and biochemical responses to the egg-derived antigens and molecules and suggested that the intestines appear to harbor immune-privileged sites.
Collapse
Affiliation(s)
- Ashgan Montasser
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmad E. Dakrory
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Emad El Zayyat
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Hatem Tallima
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
5
|
McFee RB. Lactose intolerance - A practical approach, Part 1. Dis Mon 2024; 70:101823. [PMID: 39627100 DOI: 10.1016/j.disamonth.2024.101823] [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: 12/16/2024]
Abstract
The toxicology, immunology and gastroenterology aspects of food remain important in clinical practice, as anyone treating food related anaphylaxis, or chronic intolerance syndromes can attest. Although entire editions of journals could focus on any one aspect of adverse effects from food, the following review addresses the important and prevalent gastrointestinal condition lactose intolerance and gastrointestinal (GI) conditions that have similar presentation but require different treatment.
Collapse
Affiliation(s)
- R B McFee
- Medical Director, Ellis Medical Toxicology, USA.
| |
Collapse
|
6
|
Zhang S, Li G, Qian K, Zou Y, Zheng X, Ai H, Lin F, Lei C, Hu S. Exosomes derived from cancer cells relieve inflammatory bowel disease in mice. J Drug Target 2024; 32:1073-1085. [PMID: 38958251 DOI: 10.1080/1061186x.2024.2369876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Exosome therapy has garnered significant attention due to its natural delivery capabilities, low toxicity, high biocompatibility, and potential for personalised treatment through engineering modifications. Recent studies have highlighted the ability of tumour cell-derived exosomes (TDEs) to interact with immune cells or modify the immune microenvironment to suppress host immune responses, as well as their unique homing ability to parental cells. The core question of this study is whether this immunomodulatory property of TDEs can be utilised for the immunotherapy of inflammatory diseases. In our experiments, we prepared exosomes derived from murine colon cancer cells CT26 (CT26 exo) using ultracentrifugation, characterised them, and conducted proteomic analysis. The therapeutic potential of CT26 exo was evaluated in our dextran sulphate sodium salt (DSS)-induced inflammatory bowel disease (IBD) mouse model. Compared to the control and 293 T exo treatment groups, mice treated with CT26 exo showed a reduction in the disease activity index (DAI) and colon shortening rate, with no noticeable weight loss. Haematoxylin and eosin (H&E) staining of colon paraffin sections revealed reduced inflammatory infiltration and increased epithelial goblet cells in the colons of CT26 exo-treated group. Furthermore, we conducted preliminary mechanistic explorations by examining the phenotyping and function of CD4+ T cells and dendritic cells (DCs) in the colonic lamina propria of mice. The results indicated that the ameliorative effect of CT26 exosomes might be due to their inhibition of pro-inflammatory cytokine secretion by colonic DCs and selective suppression of Th17 cell differentiation in the colon. Additionally, CT26 exo exhibited good biosafety. Our findings propose a novel exosome-based therapeutic approach for IBD and suggest the potential application of TDEs in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Shuyi Zhang
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Guangyao Li
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Kewen Qian
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Yitan Zou
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xinya Zheng
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Hongru Ai
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Fangxing Lin
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| |
Collapse
|
7
|
Chen Q, Shao B, Xu YN, Li X, Ren SH, Wang HD, Zhang JY, Sun CL, Liu T, Xiao YY, Zhao PY, Yang GM, Liu X, Wang H. IGF2 contributes to the immunomodulatory effects of exosomes from endometrial regenerative cells on experimental colitis. Int Immunopharmacol 2024; 140:112825. [PMID: 39079347 DOI: 10.1016/j.intimp.2024.112825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Exosomes derived from endometrial regenerative cells (ERC-Exos) can inherit the immunomodulatory function from ERCs, however, whether ERC-Exos exhibit such effect on inflammatory bowel diseases with mucosal immune dysregulation has not been explored. Insulin-like growth factor-Ⅱ (IGF2) is considered to possess the potential to induce an anti-inflammatory phenotype in immune cells. In this study, the contribution of IGF2 in mediating the protective efficacy of ERC-Exos on colitis was investigated. METHODS Lentiviral transfection was employed to obtain IGF2-specific knockout ERC-Exos (IGF2-/--ERC-Exos). Experimental colitis mice induced by dextran sulfate sodium (DSS) were divided into the phosphate-buffered saline (untreated), ERC-Exos-treated and IGF2-/--ERC-Exos-treated groups. Colonic histopathological analysis and intestinal barrier function were explored. The infiltration of CD4+ T cells and dendritic cells (DCs) were analyzed by immunofluorescence staining and flow cytometry. The maturation and function of bone marrow-derived dendritic cells (BMDCs) in different exosome administrations were evaluated by flow cytometry, ELISA and the coculture system, respectively. RESULTS Compared with the untreated group, ERC-Exos treatment significantly attenuated DSS-induced weight loss, bloody stools, shortened colon length, pathological damage, as well as repaired the weakened intestinal mucosal barrier, including promoting the goblet cells retention, restoring the intestinal barrier integrity and enhancing the expression of tight junction proteins, while the protective effect of exosomes was impaired with the knockout of IGF2 in ERC-Exos. Additionally, IGF2-expressing ERC-Exos decreased the proportions of Th1 and Th17, increased the proportions of Treg, as well as attenuated DC infiltration and maturation in mesenteric lymph nodes and lamina propria of the colitis mice. ERC-Exos were also observed to be phagocytosed by BMDCs and IGF2 is responsible for the modulating effect of ERC-Exos on BMDCs in vitro. CONCLUSIONS Exosomes derived from ERCs can exert a therapeutic effect on experimental colitis with remarkable alleviation of the intestinal barrier damage and the abnormal mucosal immune responses. We emphasized that IGF2 plays a critical role for ERC-Exos mediated immunomodulatory function and protection against colitis.
Collapse
Affiliation(s)
- Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Yi-Ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Jing-Yi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Cheng-Lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Yi-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Peng-Yu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Guang-Mei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China; Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China.
| |
Collapse
|
8
|
Mei L, Wang J, Hao Y, Zeng X, Yang Y, Wu Z, Ji Y. A comprehensive update on the immunoregulatory mechanisms of Akkermansia muciniphila: insights into active ingredients, metabolites, and nutrient-driven modulation. Crit Rev Food Sci Nutr 2024:1-18. [PMID: 39413040 DOI: 10.1080/10408398.2024.2416481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Akkermansia muciniphila (A. muciniphila) has gained recognition as a pioneering probiotic, exhibiting considerable potential to enhance immune conditions across both humans and animals. The health benefits of A. muciniphila are attributed to its various components, including outer membrane proteins (PilQ and Amuc_1100), secreted proteins (P9 and AmTARS), extracellular vesicles, and metabolites such as SCFAs, ornithine lipids, γ-aminobutyric acid, cobalamin, and inosine. The dynamic control of the mucus layer by A. muciniphila plays a crucial role in regulating intestinal mucosal immunity. Furthermore, A. muciniphila modulates immune function by interacting with macrophages, dendritic cells, T lymphocytes, and Paneth cells. Increasing the abundance of A. muciniphila in the gut through nutritional strategies represents a safe and effective means to augment immune function. Various polyphenols, oligosaccharides, and polysaccharides have been shown to elevate the levels of this bacterium, thereby contributing to favorable immunoregulatory outcomes. This paper delves into the latest research advancements related to the probiotic mechanisms of A. muciniphila and provides an overview of the current understanding of how its abundance responds to nutrients. These insights offer a theoretical foundation for the utilization of A. muciniphila in immunoregulation.
Collapse
Affiliation(s)
- Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
9
|
Sha C, Jin Z, Ku SY, Kogosov AS, Yu S, Bergese SD, Hsieh H. Necrotizing Enterocolitis and Neurodevelopmental Impairments: Microbiome, Gut, and Brain Entanglements. Biomolecules 2024; 14:1254. [PMID: 39456187 PMCID: PMC11505939 DOI: 10.3390/biom14101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
There is significant communication and interdependence among the gut, the microbiome, and the brain during development. Diseases, such as necrotizing enterocolitis (NEC), highlight how injury to the immature gastrointestinal tract leads to long-term neurological consequences, due to vulnerabilities of the brain in the early stages of life. A better understanding of the developing gut-microbiota-brain axis is needed to both prevent and treat the devastating consequences of these disease processes. The gut-microbiota-brain axis is a bidirectional communication pathway that includes metabolic, nervous, endocrine, and immune components. In this review, we discuss gut development, microbiome colonization and maturation, and the interactions that influence neurodevelopment in the context of NEC. We describe the components of the gut-brain axis and how the microbiome is an integral member of this relationship. Finally, we explore how derangements within the microbiome and gut-microbiota-brain axis affect the normal development and function of the other systems and long-term neurodevelopmental consequences for patients.
Collapse
Affiliation(s)
- Cuilee Sha
- Department of Pharmacological Sciences, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA;
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Zhaosheng Jin
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Stella Y. Ku
- Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Ann S. Kogosov
- Renaissance School of Medicine, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sun Yu
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| | - Sergio D. Bergese
- Department of Anesthesiology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA;
| | - Helen Hsieh
- Center for Nervous System Disorders, Stony Brook University, 100 Nicolls Road, Stony Brook, NY 11794, USA
- Department of Surgery, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY 11794, USA
| |
Collapse
|
10
|
Khaksari M, Pourali M, Rezaei Talabon S, Gholizadeh Navashenaq J, Bashiri H, Amiresmaili S. Protective effects of 17-β-estradiol on liver injury: The role of TLR4 signaling pathway and inflammatory response. Cytokine 2024; 181:156686. [PMID: 38991382 DOI: 10.1016/j.cyto.2024.156686] [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: 04/20/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Liver injury, a major global health issue, stems from various causes such as alcohol consumption, nonalcoholic steatohepatitis, obesity, diabetes, metabolic syndrome, hepatitis, and certain medications. The liver's unique susceptibility to ischemia and hypoxia, coupled with the critical role of the gut-liver axis in inflammation, underscores the need for effective therapeutic interventions. The study highlights E2's interaction with estrogen receptors (ERs) and its modulation of the Toll-like receptor 4 (TLR4) signaling pathway as key mechanisms in mitigating liver injury. Activation of TLR4 leads to the release of pro-inflammatory cytokines and chemokines, exacerbating liver inflammation and injury. E2 down-regulates TLR4 expression, reduces oxidative stress, and inhibits pro-inflammatory cytokines, thereby protecting the liver. Both classic (ERα and ERβ) and non-classic [G protein-coupled estrogen receptor (GPER)] receptors are influenced by E2. ERα is particularly crucial for liver regeneration, preventing liver failure by promoting hepatocyte proliferation. Furthermore, E2 exerts anti-inflammatory, antioxidant, and anti-apoptotic effects by inhibiting cytokines such as IL-6, IL-1β, TNF-α, and IL-17, and by reducing lipid peroxidation and free radical damage. The article calls for further clinical research to validate these findings and to develop estrogen-based treatments for liver injuries. Overall, the research emphasizes the significant potential of E2 as a therapeutic agent for liver injuries. It advocates for extensive clinical studies to validate E2 hepatoprotective properties and develop effective estrogen-based treatments.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Neuroscince and Endocrinology and Metabolism Research Centers, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | | | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Ira
| | | |
Collapse
|
11
|
Meng EX, Verne GN, Zhou Q. Macrophages and Gut Barrier Function: Guardians of Gastrointestinal Health in Post-Inflammatory and Post-Infection Responses. Int J Mol Sci 2024; 25:9422. [PMID: 39273369 PMCID: PMC11395020 DOI: 10.3390/ijms25179422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
The gut barrier is essential for protection against pathogens and maintaining homeostasis. Macrophages are key players in the immune system, are indispensable for intestinal health, and contribute to immune defense and repair mechanisms. Understanding the multifaceted roles of macrophages can provide critical insights into maintaining and restoring gastrointestinal (GI) health. This review explores the essential role of macrophages in maintaining the gut barrier function and their contribution to post-inflammatory and post-infectious responses in the gut. Macrophages significantly contribute to gut barrier integrity through epithelial repair, immune modulation, and interactions with gut microbiota. They demonstrate active plasticity by switching phenotypes to resolve inflammation, facilitate tissue repair, and regulate microbial populations following an infection or inflammation. In addition, tissue-resident (M2) and infiltration (M1) macrophages convert to each other in gut problems such as IBS and IBD via major signaling pathways mediated by NF-κB, JAK/STAT, PI3K/AKT, MAPK, Toll-like receptors, and specific microRNAs such as miR-155, miR-29, miR-146a, and miR-199, which may be good targets for new therapeutic approaches. Future research should focus on elucidating the detailed molecular mechanisms and developing personalized therapeutic approaches to fully harness the potential of macrophages to maintain and restore intestinal permeability and gut health.
Collapse
Affiliation(s)
| | - George Nicholas Verne
- College of Medicine, University of Tennessee, Memphis, TN 38103, USA
- Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Qiqi Zhou
- College of Medicine, University of Tennessee, Memphis, TN 38103, USA
- Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| |
Collapse
|
12
|
Keever-Keigher MR, Harvey L, Williams V, Vyhlidal CA, Ahmed AA, Johnston JJ, Louiselle DA, Grundberg E, Pastinen T, Friesen CA, Chevalier R, Smail C, Shakhnovich V. Genomic insights into pediatric intestinal inflammatory and eosinophilic disorders using single-cell RNA-sequencing. Front Immunol 2024; 15:1420208. [PMID: 39192974 PMCID: PMC11347318 DOI: 10.3389/fimmu.2024.1420208] [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: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Chronic inflammation of the gastrointestinal tissues underlies gastrointestinal inflammatory disorders, leading to tissue damage and a constellation of painful and debilitating symptoms. These disorders include inflammatory bowel diseases (Crohn's disease and ulcerative colitis), and eosinophilic disorders (eosinophilic esophagitis and eosinophilic duodenitis). Gastrointestinal inflammatory disorders can often present with overlapping symptoms necessitating the use of invasive procedures to give an accurate diagnosis. Methods This study used peripheral blood mononuclear cells from individuals with Crohn's disease, ulcerative colitis, eosinophilic esophagitis, and eosinophilic duodenitis to better understand the alterations to the transcriptome of individuals with these diseases and identify potential markers of active inflammation within the peripheral blood of patients that may be useful in diagnosis. Single-cell RNA-sequencing was performed on peripheral blood mononuclear cells isolated from the blood samples of pediatric patients diagnosed with gastrointestinal disorders, including Crohn's disease, ulcerative colitis, eosinophilic esophagitis, eosinophilic duodenitis, and controls with histologically healthy gastrointestinal tracts. Results We identified 730 (FDR < 0.05) differentially expressed genes between individuals with gastrointestinal disorders and controls across eight immune cell types. Discussion There were common patterns among GI disorders, such as the widespread upregulation of MTRNR2L8 across cell types, and many differentially expressed genes showed distinct patterns of dysregulation among the different gastrointestinal diseases compared to controls, including upregulation of XIST across cell types among individuals with ulcerative colitis and upregulation of Th2-associated genes in eosinophilic disorders. These findings indicate both overlapping and distinct alterations to the transcriptome of individuals with gastrointestinal disorders compared to controls, which provide insight as to which genes may be useful as markers for disease in the peripheral blood of patients.
Collapse
Affiliation(s)
| | - Lisa Harvey
- Children’s Mercy Kansas City, Kansas, MO, United States
| | | | | | - Atif A. Ahmed
- Seattle Children’s Hospitals, University of Washington, Seattle, WA, United States
| | | | | | - Elin Grundberg
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
| | - Tomi Pastinen
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
| | - Craig A. Friesen
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
| | - Rachel Chevalier
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
| | - Craig Smail
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
| | - Valentina Shakhnovich
- Children’s Mercy Kansas City, Kansas, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas, MO, United States
- Ironwood Pharmaceuticals, Boston, MA, United States
| |
Collapse
|
13
|
Asgari F, Khodadoust M, Nikzamir A, Jahani-Sherafat S, Rezaei Tavirani M, Rostami-Nejad M. The role of tryptophan metabolism and tolerogenic dendritic cells in maintaining immune tolerance: Insights into celiac disease pathogenesis. Immun Inflamm Dis 2024; 12:e1354. [PMID: 39150219 PMCID: PMC11328117 DOI: 10.1002/iid3.1354] [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: 01/07/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND In mammals, amino acid metabolism has evolved to control immune responses. Tryptophan (Trp) is the rarest essential amino acid found in food and its metabolism has evolved to be a primary regulatory node in the control of immune responses. Celiac disease (CeD) is a developed immunological condition caused by gluten intolerance and is linked to chronic small intestine enteropathy in genetically predisposed individuals. Dendritic cells (DCs), serving as the bridge between innate and adaptive immunities, can influence immunological responses in CeD through phenotypic alterations. OBJECTIVE This review aims to highlight the connection between Trp metabolism and tolerogenic DCs, and the significance of this interaction in the pathogenesis of CeD. RESULTS It is been recognized that various DC subtypes contribute to the pathogenesis of CeD. Tolerogenic DCs, in particular, are instrumental in inducing immune tolerance, leading to T-reg differentiation that helps maintain intestinal immune tolerance against inflammatory responses in CeD patients and those with other autoimmune disorders. T-regs, a subset of T-cells, play a crucial role in maintaining intestinal immunological homeostasis by regulating the activities of other immune cells. Notably, Trp metabolism, essential for T-reg function, facilitates T-reg differentiation through microbiota-mediated degradation and the kynurenine pathway. CONCLUSION Therefore, alterations in Trp metabolism could potentially influence the immune response in CeD, affecting both the development of the disease and the persistence of symptoms despite adherence to a gluten-free diet.
Collapse
Affiliation(s)
- Fatemeh Asgari
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Khodadoust
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolrahim Nikzamir
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Jahani-Sherafat
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Rostami-Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Wu Y, Huang JY, Conlon MT, Shenoy MK, Chao JL, Chooi MY, Koch MA, Gerner MY. Distal Immunization and Systemic Cytokines Establish a Transient Immune Alert State in the Intestine. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:373-383. [PMID: 38884660 PMCID: PMC11250722 DOI: 10.4049/jimmunol.2400209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024]
Abstract
Conventionally, immune responses are studied in the context of inflamed tissues and their corresponding draining lymph nodes (LNs). However, little is known about the effects of systemic inflammatory signals generated during local inflammation on distal tissues and nondraining LNs. Using a mouse model of cutaneous immunization, we found that systemic inflammatory stimuli triggered a rapid and selective distal response in the small intestine and the mesenteric LN (mesLN). This consisted of increased permeability of intestinal blood vessels and lymphatic drainage of bloodborne solutes into the mesLN, enhanced activation and migration of intestinal dendritic cells, as well as amplified T cell responses in the mesLNs to systemic but not orally derived Ags. Mechanistically, we found that the small intestine endothelial cells preferentially expressed molecules involved in TNF-α signaling and that TNF-α blockade markedly diminished distal intestinal responses to cutaneous immunization. Together, these findings reveal that the intestinal immune system is rapidly and selectively activated in response to inflammatory cues regardless of their origin, thus identifying an additional layer of defense and enhanced surveillance of a key barrier organ at constant risk of pathogen encounter.
Collapse
Affiliation(s)
- Yixuan Wu
- Department of Immunology, University of Washington, Seattle, WA
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Jessica Y Huang
- Department of Immunology, University of Washington, Seattle, WA
| | | | - Meera K Shenoy
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jaime L Chao
- Department of Immunology, University of Washington, Seattle, WA
| | - Ming Yao Chooi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore
| | - Meghan A Koch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | |
Collapse
|
15
|
Luo PK, Ho HM, Chiang MC, Chu LA, Chuang YH, Lyu PC, Hu IC, Chang WA, Peng SY, Jayakumar J, Chen HL, Huang MH, Sung HW. pH-Responsive β-Glucans-Complexed mRNA in LNPs as an Oral Vaccine for Enhancing Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404830. [PMID: 38895941 DOI: 10.1002/adma.202404830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/07/2024] [Indexed: 06/21/2024]
Abstract
mRNA vaccines for cancer immunotherapy are commonly delivered using lipid nanoparticles (LNPs), which, when administered intravenously, may accumulate in the liver, potentially limiting their therapeutic efficacy. To overcome this challenge, the study introduces an oral mRNA vaccine formulation tailored for efficient uptake by immune cells in the gastrointestinal (GI) tract, known for its high concentration of immune cells, including dendritic cells (DCs). This formulation comprises mRNA complexed with β-glucans (βGlus), a potential adjuvant for vaccines, encapsulated within LNPs (βGlus/mRNA@LNPs). The βGlus/mRNA complexes within the small compartments of LNPs demonstrate a distinctive ability to partially dissociate and reassociate, responding to pH changes, effectively shielding mRNA from degradation in the harsh GI environment. Upon oral administration to tumor-bearing mice, βGlus/mRNA@LNPs are effectively taken up by intestinal DCs and local nonimmune cells, bypassing potential liver accumulation. This initiates antigen-specific immune responses through successful mRNA translation, followed by drainage into the mesenteric lymph nodes to stimulate T cells and trigger specific adaptive immune responses, ultimately enhancing antitumor effects. Importantly, the vaccine demonstrates safety, with no significant inflammatory reactions observed. In conclusion, the potential of oral βGlus/mRNA@LNPs delivery presents a promising avenue in cancer immunotherapy, offering needle-free and user-friendly administration for widespread adoption and self-administration.
Collapse
Affiliation(s)
- Po-Kai Luo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Hui-Min Ho
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 350401, Taiwan ROC
| | - Min-Chun Chiang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Ya-Han Chuang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Ping-Chiang Lyu
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - I-Chen Hu
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Wan-An Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Sheng-Yao Peng
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Jayachandran Jayakumar
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Hsin-Lung Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 350401, Taiwan ROC
| | - Hsing-Wen Sung
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| |
Collapse
|
16
|
Alamri A. Sema-3E/PlexinD1 axis modulates dendritic cell phenotypes and functions: Current status and future implications. Hum Immunol 2024; 85:110815. [PMID: 38772051 DOI: 10.1016/j.humimm.2024.110815] [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: 01/27/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
This comprehensive research review explores the complex interplay between the Sema-3E/PlexinD1 axis and dendritic cells (DCs), highlighting its critical role in immune modulation with implications for clinical application Critical regulators of immune responses Dendritic cells are central to adaptive immunity, and the Sema-3E /PlexinD1 axis emerges as a key modulator affecting their phenotypes and functions Review delineates the impact of this signaling axis on DC maturation, migration, antigen presentation, and cytokine production, unravels its multifaceted role in shaping the immune response. Recognizing the limitations and gaps in current knowledge, the study highlights the need for further studies to condition downstream signaling events and related information experienced by the Sema-3E/PlexinD1 axis emphasizes the clarity of the immune system. The review concludes by identifying opportunities for translation, focusing on therapeutic and diagnostic potential. It highlights the importance of collaborative, interdisciplinary efforts to address the challenges and harness the therapeutic and pathological potential of targeting the Sema-3E/PlexinD1 axis, thus opening the way for transformative advances in immunology and clinical medicine.
Collapse
Affiliation(s)
- Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
17
|
Gélineau A, Marcelin G, Ouhachi M, Dussaud S, Voland L, Manuel R, Baba I, Rouault C, Yvan-Charvet L, Clément K, Tussiwand R, Huby T, Gautier EL. Fructooligosaccharides benefits on glucose homeostasis upon high-fat diet feeding require type 2 conventional dendritic cells. Nat Commun 2024; 15:5413. [PMID: 38926424 PMCID: PMC11208547 DOI: 10.1038/s41467-024-49820-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.
Collapse
Affiliation(s)
- Adélaïde Gélineau
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Geneviève Marcelin
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Melissa Ouhachi
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Sébastien Dussaud
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lise Voland
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Raoul Manuel
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ines Baba
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christine Rouault
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale, Inserm, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Karine Clément
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, service de Nutrition, Paris, France
| | - Roxane Tussiwand
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Thierry Huby
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Emmanuel L Gautier
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
18
|
Barrett KA, Kassama FJ, Surks W, Mulholland AJ, Moulton KD, Dube DH. Helicobacter pylori glycan biosynthesis modulates host immune cell recognition and response. Front Cell Infect Microbiol 2024; 14:1377077. [PMID: 38572314 PMCID: PMC10987845 DOI: 10.3389/fcimb.2024.1377077] [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: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction The pathogenic bacterium Helicobacter pylori has evolved glycan-mediated mechanisms to evade host immune defenses. This study tests the hypothesis that genetic disruption of H. pylori glycan biosynthesis alters immune recognition and response by human gastric epithelial cells and monocyte-derived dendritic cells. Methods To test this hypothesis, human cell lines were challenged with wildtype H. pylori alongside an array of H. pylori glycosylation mutants. The relative levels of immune response were measured via immature dendritic cell maturation and cytokine secretion. Results Our findings indicate that disruption of lipopolysaccharide biosynthesis diminishes gastric cytokine production, without disrupting dendritic cell recognition and activation. In contrast, variable immune responses were observed in protein glycosylation mutants which prompted us to test the hypothesis that phase variation plays a role in regulating bacterial cell surface glycosylation and subsequent immune recognition. Lewis antigen presentation does not correlate with extent of immune response, while the extent of lipopolysaccharide O-antigen elaboration does. Discussion The outcomes of this study demonstrate that H. pylori glycans modulate the host immune response. This work provides a foundation to pursue immune-based tailoring of bacterial glycans towards modulating immunogenicity of microbial pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | - Danielle H. Dube
- Department of Chemistry & Biochemistry, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
19
|
Nguyen OTP, Misun PM, Hierlemann A, Lohasz C. A Versatile Intestine-on-Chip System for Deciphering the Immunopathogenesis of Inflammatory Bowel Disease. Adv Healthc Mater 2024; 13:e2302454. [PMID: 38253407 PMCID: PMC11468350 DOI: 10.1002/adhm.202302454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/21/2023] [Indexed: 01/24/2024]
Abstract
The multifactorial nature of inflammatory bowel disease (IBD) necessitates reliable and practical experimental models to elucidate its etiology and pathogenesis. To model the intestinal microenvironment at the onset of IBD in vitro, it is important to incorporate relevant cellular and noncellular components before inducing stepwise pathogenic developments. A novel intestine-on-chip system for investigating multiple aspects of IBD's immunopathogenesis is presented. The system includes an array of tight and polarized barrier models formed from intestinal epithelial cells on an in-vivo-like subepithelial matrix within one week. The dynamic remodeling of the subepithelial matrix by cells or their secretome demonstrates the physiological relevance of the on-chip barrier models. The system design enables introduction of various immune cell types and inflammatory stimuli at specific locations in the same barrier model, which facilitates investigations of the distinct roles of each cell type in intestinal inflammation development. It is showed that inflammatory behavior manifests in an upregulated expression of inflammatory markers and cytokines (TNF-α). The neutralizing effect of the anti-inflammatory antibody Infliximab on levels of TNF-α and its inducible cytokines could be explicitly shown. Overall, an innovative approach to systematically developing a microphysiological system to comprehend immune-system-mediated disorders of IBD and to identify new therapeutic strategies is presented.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Patrick M. Misun
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Andreas Hierlemann
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| | - Christian Lohasz
- Bio Engineering LaboratoryDepartment of Biosystems Science and EngineeringETH ZurichKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
20
|
McCoy R, Oldroyd S, Yang W, Wang K, Hoven D, Bulmer D, Zilbauer M, Owens RM. In Vitro Models for Investigating Intestinal Host-Pathogen Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306727. [PMID: 38155358 PMCID: PMC10885678 DOI: 10.1002/advs.202306727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Indexed: 12/30/2023]
Abstract
Infectious diseases are increasingly recognized as a major threat worldwide due to the rise of antimicrobial resistance and the emergence of novel pathogens. In vitro models that can adequately mimic in vivo gastrointestinal physiology are in high demand to elucidate mechanisms behind pathogen infectivity, and to aid the design of effective preventive and therapeutic interventions. There exists a trade-off between simple and high throughput models and those that are more complex and physiologically relevant. The complexity of the model used shall be guided by the biological question to be addressed. This review provides an overview of the structure and function of the intestine and the models that are developed to emulate this. Conventional models are discussed in addition to emerging models which employ engineering principles to equip them with necessary advanced monitoring capabilities for intestinal host-pathogen interrogation. Limitations of current models and future perspectives on the field are presented.
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sophie Oldroyd
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Woojin Yang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Kaixin Wang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Darius Hoven
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - David Bulmer
- Department of PharmacologyUniversity of CambridgeCambridgeCB2 1PDUK
| | - Matthias Zilbauer
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| |
Collapse
|
21
|
Zhao R, Hu Z, Zhang X, Huang S, Yu G, Wu Z, Yu W, Lu J, Ruan B. The oncogenic mechanisms of the Janus kinase-signal transducer and activator of transcription pathway in digestive tract tumors. Cell Commun Signal 2024; 22:68. [PMID: 38273295 PMCID: PMC10809652 DOI: 10.1186/s12964-023-01421-9] [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: 09/06/2023] [Accepted: 12/03/2023] [Indexed: 01/27/2024] Open
Abstract
Digestive tract tumors are heterogeneous and involve the dysregulation of multiple signaling pathways. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway plays a notable role in the oncogenesis of digestive tract tumors. Typically activated by pro-inflammatory cytokines, it regulates important biological processes, such as cell growth, differentiation, apoptosis, immune responses, and inflammation. The aberrant activation of this pathway manifests in different forms, including mutations in JAKs, overexpression of cytokine receptors, and sustained STAT activation, and contributes to promoting the malignant characteristics of cancer cells, including uncontrolled proliferation, resistance to apoptosis, enhanced invasion and metastasis, angiogenesis, acquisition of stem-like properties, and drug resistance. Numerous studies have shown that aberrant activation of the JAK-STAT pathway is closely related to the development and progression of digestive tract tumors, contributing to tumor survival, angiogenesis, changes in the tumor microenvironment, and even immune escape processes. In addition, this signaling pathway also affects the sensitivity of digestive tract tumors to chemotherapy and targeted therapy. Therefore, it is crucial to comprehensively understand the oncogenic mechanisms underlying the JAK-STAT pathway in order to develop effective therapeutic strategies against digestive tract tumors. Currently, several JAK-STAT inhibitors are undergoing clinical and preclinical trials as potential treatments for various human diseases. However, further investigation is required to determine the role of this pathway, as well as the effectiveness and safety of its inhibitors, especially in the context of digestive tract tumors. In this review, we provide an overview of the structure, classic activation, and negative regulation of the JAK-STAT pathway. Furthermore, we discuss the pathogenic mechanisms of JAK-STAT signaling in different digestive tract tumors, with the aim of identifying potential novel therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Ruihong Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhangmin Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Xiaoli Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Shujuan Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Guodong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| | - Bing Ruan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
22
|
Mahajan D, Kumar T, Rath PK, Sahoo AK, Mishra BP, Kumar S, Nayak NR, Jena MK. Dendritic Cells and the Establishment of Fetomaternal Tolerance for Successful Human Pregnancy. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0010. [PMID: 38782369 DOI: 10.2478/aite-2024-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 05/25/2024]
Abstract
Pregnancy is a remarkable event where the semi-allogeneic fetus develops in the mother's uterus, despite genetic and immunological differences. The antigen handling and processing at the maternal-fetal interface during pregnancy appear to be crucial for the adaptation of the maternal immune system and for tolerance to the developing fetus and placenta. Maternal antigen-presenting cells (APCs), such as macrophages (Mφs) and dendritic cells (DCs), are present at the maternal-fetal interface throughout pregnancy and are believed to play a crucial role in this process. Despite numerous studies focusing on the significance of Mφs, there is limited knowledge regarding the contribution of DCs in fetomaternal tolerance during pregnancy, making it a relatively new and growing field of research. This review focuses on how the behavior of DCs at the maternal-fetal interface adapts to pregnancy's unique demands. Moreover, it discusses how DCs interact with other cells in the decidual leukocyte network to regulate uterine and placental homeostasis and the local maternal immune responses to the fetus. The review particularly examines the different cell lineages of DCs with specific surface markers, which have not been critically reviewed in previous publications. Additionally, it emphasizes the impact that even minor disruptions in DC functions can have on pregnancy-related complications and proposes further research into the potential therapeutic benefits of targeting DCs to manage these complications.
Collapse
Affiliation(s)
- Deviyani Mahajan
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Tarun Kumar
- Department of Veterinary Clinical Complex, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125001, India
| | - Prasana Kumar Rath
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Anjan Kumar Sahoo
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
- Department of Veterinary Surgery and Radiology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Bidyut Prava Mishra
- Department of Veterinary Pathology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
- Department of Livestock Products Technology, College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Sudarshan Kumar
- Proteomics and Structural Biology Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Nihar Ranjan Nayak
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| |
Collapse
|
23
|
Abstract
The remarkable diversity of lymphocytes, essential components of the immune system, serves as an ingenious mechanism for maximizing the efficient utilization of limited host defense resources. While cell adhesion molecules, notably in gut-tropic T cells, play a central role in this mechanism, the counterbalancing molecular details have remained elusive. Conversely, we've uncovered the molecular pathways enabling extracellular vesicles secreted by lymphocytes to reach the gut's mucosal tissues, facilitating immunological regulation. This discovery sheds light on immune fine-tuning, offering insights into immune regulation mechanisms.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
24
|
Monaco MH, Wang M, Hauser J, Yan J, Dilger RN, Donovan SM. Formula supplementation with human and bovine milk oligosaccharides modulates blood IgG and T-helper cell populations, and ex vivo LPS-stimulated cytokine production in a neonatal preclinical model. Front Immunol 2023; 14:1327853. [PMID: 38179055 PMCID: PMC10765566 DOI: 10.3389/fimmu.2023.1327853] [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: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction Human milk contains structurally diverse oligosaccharides (HMO), which are multifunctional modulators of neonatal immune development. Our objective was to investigate formula supplemented with fucosylated (2'FL) + neutral (lacto-N-neotetraose, LNnt) oligosaccharides and/or sialylated bovine milk oligosaccharides (BMOS) on immunological outcomes. Methods Pigs (n=46) were randomized at 48h of age to four diets: sow milk replacer formula (CON), BMOS (CON + 6.5 g/L BMOS), HMO (CON + 1.0 g/L 2'FL + 0.5 g/L LNnT), or BMOS+HMO (CON + 6.5 g/L BMOS + 1.0 g/L 2'FL + 0.5 g/L LNnT). Blood and tissues were collected on postnatal day 33 for measurement of cytokines and IgG, phenotypic identification of immune cells, and ex vivo lipopolysaccharide (LPS)-stimulation of immune cells. Results Serum IgG was significantly lower in the HMO group than BMOS+HMO but did not differ from CON or BMOS. The percentage of PBMC T-helper cells was lower in BMOS+HMO than the other groups. Splenocytes from the BMOS group secreted more IL-1β when stimulated ex vivo with LPS compared to CON or HMO groups. For PBMCs, a statistical interaction of BMOS*HMO was observed for IL-10 secretion (p=0.037), with BMOS+HMO and HMO groups differing at p=0.1. Discussion The addition of a mix of fucosylated and sialylated oligosaccharides to infant formula provides specific activities in the immune system that differ from formulations supplemented with one oligosaccharide structure.
Collapse
Affiliation(s)
- Marcia H. Monaco
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Jonas Hauser
- Brain Health Department, Nestlé Institute of Health Sciences, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Jian Yan
- Nestlé Product Technology Center Nutrition, Vevey, Switzerland
| | - Ryan N. Dilger
- Department of Animal Sciences, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana Champaign, Urbana, IL, United States
| |
Collapse
|
25
|
Mehandru S, Colombel JF, Juarez J, Bugni J, Lindsay JO. Understanding the molecular mechanisms of anti-trafficking therapies and their clinical relevance in inflammatory bowel disease. Mucosal Immunol 2023; 16:859-870. [PMID: 37574127 PMCID: PMC11141405 DOI: 10.1016/j.mucimm.2023.08.001] [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: 07/27/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
In patients with inflammatory bowel disease (IBD), a combination of dysbiosis, increased intestinal permeability, and insufficient regulatory responses facilitate the development of chronic inflammation, which is driven by a complex interplay between the mucosal immune system and the environment and sustained by immune priming and ongoing cellular recruitment to the gut. The localization of immune cells is mediated by their expression of chemokine receptors and integrins, which bind to chemokines and adhesion molecules, respectively. In this article, we review the mechanisms of action of anti-trafficking therapies for IBD and consider clinical observations in the context of the different mechanisms of action. Furthermore, we discuss the evolution of molecular resistance to anti-cytokines, in which the composition of immune cells in the gut changes in response to treatment, and the potential implications of this for treatment sequencing. Lastly, we discuss the relevance of mechanism of action to combination therapy for IBD.
Collapse
Affiliation(s)
- Saurabh Mehandru
- The Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jean-Frederic Colombel
- The Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julius Juarez
- Takeda Pharmaceuticals U.S.A., Inc., Lexington, MA, USA
| | - James Bugni
- Takeda Pharmaceuticals U.S.A., Inc., Lexington, MA, USA
| | - James O Lindsay
- Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK; Department of Gastroenterology, Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
26
|
Ustaoglu A, Daudali FA, D’afflitto M, Murtough S, Lee C, Moreno E, Blaydon DC, Kelsell DP, Sifrim D, Woodland P, Peiris M. Identification of novel immune cell signature in gastroesophageal reflux disease: altered mucosal mast cells and dendritic cell profile. Front Immunol 2023; 14:1282577. [PMID: 38098488 PMCID: PMC10720318 DOI: 10.3389/fimmu.2023.1282577] [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: 08/24/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Heartburn pathogenesis in GERD remains incompletely understood. We aimed to identify differences in the immune cell signature and sensory mucosal markers between reflux phenotypes and healthy asymptomatic subjects. Methods Thirty-seven patients with heartburn symptoms were phenotyped endoscopically and with objective reflux studies into erosive reflux disease (ERD) (N=10), nonerosive reflux disease (NERD) (N=9), functional heartburn (FH) (N=9), and Barrett's esophagus (BO) (N=9). Bulk mRNA-sequencing(RNA-seq) was conducted on RNA extracted from endoscopic biopsies, and immune cell deconvolution analysis was performed using CIBERSORT. RNA-seq findings were validated by immunofluorescent staining for CD1a, nerve growth factor (NGF), and mast cell tryptase in corresponding patient biopsies. Results Transcriptomic analysis detected higher mast cell abundance in BO, ERD, and NERD compared to healthy controls (p<0.05), with decreased dendritic cell infiltration in BO, ERD, and NERD patients compared to healthy controls and FH patients. CD1a-positive dendritic cell infiltration was significantly higher in the healthy esophageal mucosa at protein level compared to BO (p=0.0005), ERD (p=0.0004), and FH patients (p=0.0096). Moreover, NGF co-expression on mast cells in GERD patients was significantly higher than in healthy controls (p=0.0094). Discussion The mucosa in patients with GERD had a significant increase in NGF expression on mast cells, suggesting an upregulation of signalling for neuronal sprouting in GERD. Moreover, decreased dendritic cell abundance in GERD esophageal mucosa may play a role in reduced oral tolerance and development of subsequent immune responses which may participate in esophageal sensitivity.
Collapse
Affiliation(s)
- Ahsen Ustaoglu
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Fatema Arif Daudali
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Manfredi D’afflitto
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stephen Murtough
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Chung Lee
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Estefania Moreno
- Royal London Hospital, Barts National Health Service (NHS) Health, London, United Kingdom
| | - Diana C. Blaydon
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - David P. Kelsell
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Daniel Sifrim
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Philip Woodland
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Madusha Peiris
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
27
|
Liang Y, Liu D, Li Y, Hou H, Li P, Ma X, Li P, Zhan J, Wang P. Maternal polysorbate 80 exposure causes intestinal ILCs and CD4 + T cell developmental abnormalities in mouse offspring. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122392. [PMID: 37595736 DOI: 10.1016/j.envpol.2023.122392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/20/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
This study aimed to investigate the transgenerational impacts of maternal intake of polysorbate 80 (P80), an emulsifier widely used in modern society, on the development of offspring immunity. Our results revealed that maternal P80 treatment led to impaired differentiation of innate lymphoid cells (ILCs) and CD4+ T cells in the small intestinal lamina propria (SiLP), resulting in intestinal dyshomeostasis in female offspring. Furthermore, we found that SiLP ILCs abundances were significantly altered in 0-day-old fetuses from P80-treated mothers, indicating a prenatal impact of P80-treated mothers on offspring immunity. Additionally, cesarean section and foster-nursing studies demonstrated that P80-induced altered SiLP ILCs in 0-day-old fetuses could further induce dysregulation of ILCs and CD4+ T cells in the SiLP, thus promoting intestinal dysregulation in offspring later in life. Overall, our findings suggest that maternal P80 intake could prenatally program the development of offspring immunity, exerting a significant and long-lasting impact.
Collapse
Affiliation(s)
- Yiran Liang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30, Xueyuan Road, Beijing, 100083, People's Republic of China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Yan Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Haonan Hou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Pengxi Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Xiaoran Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Peize Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
28
|
Martinelli S, Lamminpää I, Dübüş EN, Sarıkaya D, Niccolai E. Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation. Nutrients 2023; 15:4408. [PMID: 37892482 PMCID: PMC10610426 DOI: 10.3390/nu15204408] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Gastrointestinal (GI) cancers are a group of highly prevalent malignant tumors affecting the gastrointestinal tract. Globally, one in four cancer cases and one in three cancer deaths are estimated to be GI cancers. They can alter digestive and absorption functions, leading to severe malnutrition which may worsen the prognosis of the patients. Therefore, nutritional intervention and monitoring play a fundamental role in managing metabolic alterations and cancer symptoms, as well as minimizing side effects and increasing the effectiveness of chemotherapy. In this scenario, the use of immunonutrients that are able to modulate the immune system and the modification/regulation of the gut microbiota composition have gained attention as a possible strategy to improve the conditions of these patients. The complex interaction between nutrients and microbiota might contribute to maintaining the homeostasis of each individual's immune system; therefore, concurrent use of specific nutrients in combination with traditional cancer treatments may synergistically improve the overall care of GI cancer patients. This work aims to review and discuss the role of immunonutrition and microbiota modulation in improving nutritional status, postoperative recovery, and response to therapies in patients with GI cancer.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Ingrid Lamminpää
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Eda Nur Dübüş
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Dilara Sarıkaya
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| |
Collapse
|
29
|
Yin Z, Liu X, Guo L, Ren M, Kang W, Ma C, Waterhouse GIN, Sun-Waterhouse D. The potential of dietary fiber in building immunity against gastrointestinal and respiratory disorders. Crit Rev Food Sci Nutr 2023; 64:13318-13336. [PMID: 37837407 DOI: 10.1080/10408398.2023.2266462] [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: 10/16/2023]
Abstract
The numerous health benefits of dietary fibers (DFs) justify their inclusion in human diets and biomedical products. Given the short- and long-term human impacts of the COVID-19 virus on human health, the potential of DFs in building immunity against gastrointestinal and respiratory disorders is currently receiving high attention. This paper reviews the physicochemical properties of DFs, together with their immune functions and effects on the gastrointestinal tract and respiratory system mainly based on research in the last ten years. Possible modes of action of DFs in promoting health, especially building immunity, are explored. We seek to highlight the importance of understanding the exact physical and chemical characteristics and molecular behaviors of DFs in providing specific immune function. This review provides a perspective beyond the existing recognition of DFs' positive effects on human health, and offers a theoretical framework for the development of special DFs components and their application in functional foods and other therapeutic products against gastrointestinal and respiratory disorders. DFs enhance immunity from gastrointestinal and respiratory diseases to promote host health.
Collapse
Affiliation(s)
- Zhenhua Yin
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Xiaopeng Liu
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Lin Guo
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Mengjie Ren
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Wenyi Kang
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | | | | |
Collapse
|
30
|
Gray G, Scroggins DG, Wilson KT, Scroggins SM. Cellular Immunotherapy in Mice Prevents Maternal Hypertension and Restores Anti-Inflammatory Cytokine Balance in Maternal and Fetal Tissues. Int J Mol Sci 2023; 24:13594. [PMID: 37686399 PMCID: PMC10487605 DOI: 10.3390/ijms241713594] [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: 08/15/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Preeclampsia is the leading cause of maternal-fetal morbidity worldwide. The concept that persistent feto-placental intolerance is important in the pathogenesis of preeclampsia (PreE) has been demonstrated by our lab and others. Arginine vasopressin (AVP) infusion during pregnancy induces cardiovascular, renal, and T helper (TH) cell alterations in mice consistent with human PreE. In addition to their conventional immuno-stimulatory role, dendritic cells (DCs) also play a vital role in immune tolerance. In contrast to conventional DCs, regulatory DCs (DCregs) express low levels of co-stimulatory markers, produce anti-inflammatory cytokines, induce T regulatory (Treg) cells, and promote tolerance. In mice, DCregs prevent pro-inflammatory responses and induce antigen-specific tolerance. Given these known functions of DCregs, we hypothesize that DCregs will prevent the development of AVP-induced PreE in mice. C57BL/6J females were infused with AVP (24 ng/h) or saline throughout gestation via an osmotic minipump. Bone-marrow-derived DCregs were injected into AVP-infused dams at the time of the pump implantation or on gestational day (GD) 7. The blood pressure of the mice was taken throughout their pregnancy. The maternal urine proteins and TH-associated cytokines in maternal and fetal tissues were measured on GD 18. The treatment with DCregs effectively prevented the elevation of maternal blood pressure, proteinuria, and fetal growth restriction that were observed in AVP-infused dams. Furthermore, we noted a reduction in the pro-inflammatory TH-associated cytokines IFNγ and IL-17, while anti-inflammatory cytokines IL-4, IL-10, and TGFβ showed an increase following DCreg treatment. These outcomes provide strong evidence supporting the potential of DCregs as a valuable therapeutic approach in addressing PreE.
Collapse
Affiliation(s)
- Gabrielle Gray
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Douglas G. Scroggins
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Katlin T. Wilson
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Sabrina M. Scroggins
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| |
Collapse
|
31
|
Manoharan I, Shanmugam A, Ramalingam M, Patel N, Thangaraju M, Ande S, Pacholczyk R, Prasad PD, Manicassamy S. The Transcription Factor RXRα in CD11c+ APCs Regulates Intestinal Immune Homeostasis and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:853-861. [PMID: 37477694 PMCID: PMC10538854 DOI: 10.4049/jimmunol.2200909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
APCs such as dendritic cells and macrophages play a pivotal role in mediating immune tolerance and restoring intestinal immune homeostasis by limiting inflammatory responses against commensal bacteria. However, cell-intrinsic molecular regulators critical for programming intestinal APCs to a regulatory state rather than an inflammatory state are unknown. In this study, we report that the transcription factor retinoid X receptor α (RXRα) signaling in CD11c+ APCs is essential for suppressing intestinal inflammation by imparting an anti-inflammatory phenotype. Using a mouse model of ulcerative colitis, we demonstrated that targeted deletion of RXRα in CD11c+ APCs in mice resulted in the loss of T cell homeostasis with enhanced intestinal inflammation and increased histopathological severity of colonic tissue. This was due to the increased production of proinflammatory cytokines that drive Th1/Th17 responses and decreased expression of immune-regulatory factors that promote regulatory T cell differentiation in the colon. Consistent with these findings, pharmacological activation of the RXRα pathway alleviated colitis severity in mice by suppressing the expression of inflammatory cytokines and limiting Th1/Th17 cell differentiation. These findings identify an essential role for RXRα in APCs in regulating intestinal immune homeostasis and inflammation. Thus, manipulating the RXRα pathway could provide novel opportunities for enhancing regulatory responses and dampening colonic inflammation.
Collapse
Affiliation(s)
- Indumathi Manoharan
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | | | | | - Nikhil Patel
- Department of Pathology, Augusta University, Augusta, GA USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Satyanarayana Ande
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | | | - Puttur D. Prasad
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Santhakumar Manicassamy
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
- Department of Medicine, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
32
|
Meng F, Jiang X, Wang X, Zheng Q, Wang XN, Mei C, Yan S, He Y, Xue J, Zhang X, Fu W, You Y, Zhai J, Wang Y, Sun X. Tumor necrosis factor-like cytokine 1A plays a role in inflammatory bowel disease pathogenesis. Proc Natl Acad Sci U S A 2023; 120:e2120771120. [PMID: 37579137 PMCID: PMC10452302 DOI: 10.1073/pnas.2120771120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/03/2023] [Indexed: 08/16/2023] Open
Abstract
The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.
Collapse
Affiliation(s)
- Fanxiang Meng
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province110801, China
| | - Xuefeng Jiang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiao Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaonan N. Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Chenxue Mei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province110122, China
| | - Siqi Yan
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Yuting He
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Junxiu Xue
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaoqing Zhang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Teaching Center for Medical Experiment, China Medical University, Shenyang, Liaoning Province110122, China
| | - Wenda Fu
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Blood Transfusion, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province710032, China
| | - Yong You
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Immunology, Chengde medical university, Chengde, Hebei Province067000, China
| | - Jingbo Zhai
- Medical College, Inner Mongolia Minzu University, Tongliao028000, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao028000, China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province110032, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| |
Collapse
|
33
|
Hashemi P, Mahmoodi S, Ghasemian A. An updated review on oral protein-based antigen vaccines efficiency and delivery approaches: a special attention to infectious diseases. Arch Microbiol 2023; 205:289. [PMID: 37468763 DOI: 10.1007/s00203-023-03629-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
Various infectious agents affect human health via the oral entrance. The majority of pathogens lack approved vaccines. Oral vaccination is a convenient, safe and cost-effective approach with the potential of provoking mucosal and systemic immunity and maintaining individual satisfaction. However, vaccines should overcome the intricate environment of the gastrointestinal tract (GIT). Oral protein-based antigen vaccines (OPAVs) are easier to administer than injectable vaccines and do not require trained healthcare professionals. Additionally, the risk of needle-related injuries, pain, and discomfort is eliminated. However, OPAVs stability at environmental and GIT conditions should be considered to enhance their stability and facilitate their transport and storage. These vaccines elicit the local immunity, protecting GIT, genital tract and respiratory epithelial surfaces, where numerous pathogens penetrate the body. OPAVs can also be manipulated (such as using specific incorporated ligand and receptors) to elicit targeted immune response. However, low bioavailability of OPAVs necessitates development of proper protein carriers and formulations to enhance their stability and efficacy. There are several strategies to improve their efficacy or protective effects, such as incorporation of adjuvants, enzyme inhibitors, mucoadhesive or penetrating devices and permeation enhancers. Hence, efficient delivery of OPAVs into GIT require proper delivery systems mainly including smart target systems, probiotics, muco-adhesive carriers, lipid- and plant-based delivery systems and nano- and microparticles.
Collapse
Affiliation(s)
- Parisa Hashemi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
34
|
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: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [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
|
35
|
Li S, Xu K, Cheng Y, Chen L, Yi A, Xiao Z, Zhao X, Chen M, Tian Y, Meng W, Tang Z, Zhou S, Ruan G, Wei Y. The role of complex interactions between the intestinal flora and host in regulating intestinal homeostasis and inflammatory bowel disease. Front Microbiol 2023; 14:1188455. [PMID: 37389342 PMCID: PMC10303177 DOI: 10.3389/fmicb.2023.1188455] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 07/01/2023] Open
Abstract
Pharmacological treatment of inflammatory bowel disease (IBD) is inefficient and difficult to discontinue appropriately, and enterobacterial interactions are expected to provide a new target for the treatment of IBD. We collected recent studies on the enterobacterial interactions among the host, enterobacteria, and their metabolite products and discuss potential therapeutic options. Intestinal flora interactions in IBD are affected in the reduced bacterial diversity, impact the immune system and are influenced by multiple factors such as host genetics and diet. Enterobacterial metabolites such as SCFAs, bile acids, and tryptophan also play important roles in enterobacterial interactions, especially in the progression of IBD. Therapeutically, a wide range of sources of probiotics and prebiotics exhibit potential therapeutic benefit in IBD through enterobacterial interactions, and some have gained wide recognition as adjuvant drugs. Different dietary patterns and foods, especially functional foods, are novel therapeutic modalities that distinguish pro-and prebiotics from traditional medications. Combined studies with food science may significantly improve the therapeutic experience of patients with IBD. In this review, we provide a brief overview of the role of enterobacteria and their metabolites in enterobacterial interactions, discuss the advantages and disadvantages of the potential therapeutic options derived from such metabolites, and postulate directions for further research.
Collapse
Affiliation(s)
- Siyu Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Basic Medicine College of Army Medical University, Army Medical University, Chongqing, China
| | - Kan Xu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Basic Medicine College of Army Medical University, Army Medical University, Chongqing, China
| | - Yi Cheng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lu Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ailin Yi
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhifeng Xiao
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xuefei Zhao
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Minjia Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuting Tian
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei Meng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zongyuan Tang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuhong Zhou
- Department of Laboratory Animal Center, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guangcong Ruan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Wei
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
36
|
Chen L, Ou Q, Kou X. Extracellular vesicles and their indispensable roles in pathogenesis and treatment of inflammatory bowel disease: A comprehensive review. Life Sci 2023; 327:121830. [PMID: 37286163 DOI: 10.1016/j.lfs.2023.121830] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Inflammatory bowel disease (IBD) is a global disease with rising incidence worldwide, and its debilitating symptoms and dissatisfactory therapies have brought heavy burdens for patients. Extracellular vesicles (EVs), a heterogeneous population of lipid bilayer membranes containing abundant bioactive molecules, have been indicated to play important roles in the pathogenesis and treatment of many diseases. However, to our knowledge, comprehensive reviews summarizing the various roles of diverse source-derived EVs in the pathogenesis and treatment of IBD are still lacking. This review, not only summarizes the EV characteristics, but also focuses on the multiple roles of diverse EVs in IBD pathogenesis and their treatment potential. In addition, hoping to push forward the research frontiers, we point out several challenges that the researchers are faced, about EVs in current IBD research and future therapeutic applications. We also put forward our prospects on future exploration regarding EVs in IBD treatment, including developing IBD vaccines and paying more attention on apoptotic vesicles. This review is aimed to enrich the knowledge on the indispensable roles of EVs in IBD pathogenesis and treatment, providing ideas and reference for future therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Linling Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong 510055, China.
| |
Collapse
|
37
|
Dahlgren D, Lennernäs H. Review on the effect of chemotherapy on the intestinal barrier: Epithelial permeability, mucus and bacterial translocation. Biomed Pharmacother 2023; 162:114644. [PMID: 37018992 DOI: 10.1016/j.biopha.2023.114644] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Chemotherapy kills fast-growing cells including gut stem cells. This affects all components of the physical and functional intestinal barrier, i.e., the mucus layer, epithelium, and immune system. This results in an altered intestinal permeability of toxic compounds (e.g., endotoxins) as well as luminal bacterial translocation into the mucosa and central circulation. However, there is uncertainty regarding the relative contributions of the different barrier components for the development of chemotherapy-induced gut toxicity. This review present an overview of the intestinal mucosal barrier determined with various types of molecular probes and methods, and how they are affected by chemotherapy based on reported rodent and human data. We conclude that there is overwhelming evidence that chemotherapy increases bacterial translocation, and that it affects the mucosal barrier by rendering the mucosa more permeable to large permeability probes. Chemotherapy also seems to impede the intestinal mucus barrier, even though this has been less clearly evaluated from a functional standpoint but certainly plays a role in bacteria translocation. Combined, it is however difficult to outline a clear temporal or succession between the different gastrointestinal events and barrier functions, especially as chemotherapy-induced neutropenia is also involved in intestinal immunological homeostasis and bacterial translocation. A thorough characterization of this would need to include a time dependent development of neutropenia, intestinal permeability, and bacterial translocation, ideally after a range of chemotherapeutics and dosing regimens.
Collapse
|
38
|
Zha C, Peng Z, Huang K, Tang K, Wang Q, Zhu L, Che B, Li W, Xu S, Huang T, Yu Y, Zhang W. Potential role of gut microbiota in prostate cancer: immunity, metabolites, pathways of action? Front Oncol 2023; 13:1196217. [PMID: 37265797 PMCID: PMC10231684 DOI: 10.3389/fonc.2023.1196217] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
The gut microbiota helps to reveal the relationship between diseases, but the role of gut microbiota in prostate cancer (PCa) is still unclear. Recent studies have found that the composition and abundance of specific gut microbiota are significantly different between PCa and non-PCa, and the gut microbiota may have common and unique characteristics between different diseases. Intestinal microorganisms are affected by various factors and interact with the host in a variety of ways. In the complex interaction model, the regulation of intestinal microbial metabolites and the host immune system is particularly important, and they play a key role in maintaining the ecological balance of intestinal microorganisms and metabolites. However, specific changes in the composition of intestinal microflora may promote intestinal mucosal immune imbalance, leading to the formation of tumors. Therefore, this review analyzes the immune regulation of intestinal flora and the production of metabolites, as well as their effects and mechanisms on tumors, and briefly summarizes that specific intestinal flora can play an indirect role in PCa through their metabolites, genes, immunity, and pharmacology, and directly participate in the occurrence, development, and treatment of tumors through bacterial and toxin translocation. We also discussed markers of high risk PCa for intestinal microbiota screening and the possibility of probiotic ingestion and fecal microbiota transplantation, in order to provide better treatment options for clinic patients. Finally, after summarizing a number of studies, we found that changes in immunity, metabolites.
Collapse
Affiliation(s)
- Cheng Zha
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zheng Peng
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Kunyuan Huang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Kaifa Tang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Urology & Andrology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiang Wang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lihua Zhu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bangwei Che
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Li
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shenghan Xu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tao Huang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ying Yu
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wenjun Zhang
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
39
|
Devi MB, Sarma HK, Mukherjee AK, Khan MR. Mechanistic Insights into Immune-Microbiota Interactions and Preventive Role of Probiotics Against Autoimmune Diabetes Mellitus. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10087-1. [PMID: 37171690 DOI: 10.1007/s12602-023-10087-1] [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] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Recent studies on genetically susceptible individuals and animal models revealed the potential role of the intestinal microbiota in the pathogenesis of type 1 diabetes (T1D) through complex interactions with the immune system. T1D incidence has been increasing exponentially with modern lifestyle altering normal microbiota composition, causing dysbiosis characterized by an imbalance in the gut microbial community. Dysbiosis has been suggested to be a potential contributing factor in T1D. Moreover, several studies have shown the potential role of probiotics in regulating T1D through various mechanisms. Current T1D therapies target curative measures; however, preventive therapeutics are yet to be proven. This review highlights immune microbiota interaction and the immense role of probiotics and postbiotics as important immunological interventions for reducing the risk of T1D.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | | | - Ashis K Mukherjee
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Mojibur R Khan
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India.
| |
Collapse
|
40
|
Yan X, Meng L, Zhang X, Deng Z, Gao B, Zhang Y, Yang M, Ma Y, Zhang Y, Tu K, Zhang M, Xu Q. Reactive oxygen species-responsive nanocarrier ameliorates murine colitis by intervening colonic innate and adaptive immune responses. Mol Ther 2023; 31:1383-1401. [PMID: 36855303 PMCID: PMC10188638 DOI: 10.1016/j.ymthe.2023.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/02/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic or relapsing inflammatory disease with limited therapeutic outcomes. Pterostilbene (PSB) is a polyphenol-based anti-oxidant that has received extensive interest for its intrinsic anti-inflammatory and anti-oxidative activities. This work aims to develop a reactive oxygen species (ROS)-responsive, folic acid (FA)-functionalized nanoparticle (NP) for efficient PSB delivery to treat UC. The resulting PSB@NP-FA had a nano-scaled diameter of 231 nm and a spherical shape. With ROS-responsive release and ROS-scavenging properties, PSB@NP could effectively scavenge H2O2, thereby protecting cells from H2O2-induced oxidative damage. After FA modification, the resulting PSB@NP-FA could be internalized by RAW 264.7 and Colon-26 cells efficiently and preferentially localized to the inflamed colon. In dextran sulfate sodium (DSS)-induced colitis models, PSB@NP-FA showed a prominent ROS-scavenging capacity and anti-inflammatory activity, therefore relieving murine colitis effectively. Mechanism results suggested that PSB@NP-FA ameliorated colitis by regulating dendritic cells (DCs), promoting macrophage polarization, and regulating T cell infiltration. Both innate and adaptive immunity were involved. More importantly, the combination of the PSB and dexamethasone (DEX) enhanced the therapeutic efficacy of colitis. This ROS-responsive and ROS-scavenging nanocarrier represents an alternative therapeutic approach to UC. It can also be used as an enhancer for classic anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiangji Yan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Lingzhang Meng
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, Guangxi 530021, China; Center for Systemic Inflammation Research (CSIR), Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Xingzhe Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi 710061, China
| | - Zhichao Deng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Bowen Gao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Yujie Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Mei Yang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Yana Ma
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Mingzhen Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China.
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
41
|
Gao G, Shen S, Zhang T, Zhang J, Huang S, Sun Z, Zhang H. Lacticaseibacillus rhamnosus Probio-M9 enhanced the antitumor response to anti-PD-1 therapy by modulating intestinal metabolites. EBioMedicine 2023; 91:104533. [PMID: 37027929 PMCID: PMC10085781 DOI: 10.1016/j.ebiom.2023.104533] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Probiotics have been increasingly proposed for enhancing immune checkpoint blockade (ICB) treatments against cancer. However, its causal relationship with immunotherapeutic efficacy remains unclear, which promoted us to explore if and how probiotic Lacticaseibacillus rhamnosus Probio-M9 manipulates gut microbiome for expected outcomes. METHODS We evaluated the effects of Probio-M9 on the anti-PD-1 treatment against colorectal cancer in mice via a multi-omics approach. We defined the mechanisms of Probio-M9-mediated antitumor immunity by comprehensive analyses of metagenome and metabolites of commensal gut microbes as well as the immunologic factors and serum metabolome of the host. FINDINGS The results indicated that Probio-M9 intervention strengthened the anti-PD-1-based tumor inhibition. Both prophylactic and therapeutic administration of Probio-M9 showed conspicuous performance in controlling tumor growth with ICB treatment. The supplement of Probio-M9 modulated enhanced immunotherapy response through promoting beneficial microbes (e.g., Lactobacillus and Bifidobacterium animalis), producing beneficial metabolites including butyric acids in the gut, and accumulating blood-derived α-ketoglutaric acid, N-acetyl-l-glutamic acid and pyridoxine in particular, which promoted the infiltration and activation of cytotoxic T lymphocytes (CTLs) and suppressing the function of regulatory T cells (Tregs) in the tumor microenvironment (TME). Subsequently, we found that enhanced immunotherapeutic response was transmissible by transplanting either post-probiotic-treatment gut microbes or intestinal metabolites to new tumor-bearing mice. INTERPRETATION This study offered valuable insight into the causal role of Probio-M9 in correcting the defects in gut microbiota that compromised anti-PD-1 therapeutic efficacy, which can be used as an alternative synergetic agent with ICB for clinical cancer treatment. FUNDING This research was supported by Research Fund for the National Key R&D Program of China (2022YFD2100702), Inner Mongolia Science and Technology Major Projects (2021ZD0014), and China Agriculture Research System of MOF and MARA.
Collapse
Affiliation(s)
- Guangqi Gao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Siyuan Shen
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Tao Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, 010018, China.
| |
Collapse
|
42
|
Martínez-Puig D, Costa-Larrión E, Rubio-Rodríguez N, Gálvez-Martín P. Collagen Supplementation for Joint Health: The Link between Composition and Scientific Knowledge. Nutrients 2023; 15:nu15061332. [PMID: 36986062 PMCID: PMC10058045 DOI: 10.3390/nu15061332] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, generating pain, disability, and socioeconomic costs worldwide. Currently there are no approved disease-modifying drugs for OA, and safety concerns have been identified with the chronic use of symptomatic drugs. In this context, nutritional supplements and nutraceuticals have emerged as potential alternatives. Among them, collagen is being a focus of particular interest, but under the same term different types of collagens coexist with different structures, compositions, and origins, leading to different properties and potential effects. The aim of this narrative review is to generally describe the main types of collagens currently available in marketplace, focusing on those related to joint health, describing their mechanism of action, preclinical, and clinical evidence. Native and hydrolyzed collagen are the most studied collagen types for joint health. Native collagen has a specific immune-mediated mechanism that requires the recognition of its epitopes to inhibit inflammation and tissue catabolism at articular level. Hydrolyzed collagen may contain biologically active peptides that are able to reach joint tissues and exert chondroprotective effects. Although there are preclinical and clinical studies showing the safety and efficacy of food ingredients containing both types of collagens, available research suggests a clear link between collagen chemical structure and mechanism of action.
Collapse
|
43
|
Mattiola I, Diefenbach A. Regulation of innate immune system function by the microbiome: Consequences for tumor immunity and cancer immunotherapy. Semin Immunol 2023; 66:101724. [PMID: 36758379 DOI: 10.1016/j.smim.2023.101724] [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: 09/20/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Innate effector cells are immune cells endowed with host protective features and cytotoxic functions. By sensing the tissue environment, innate cells have an important role in regulating the transition from homeostasis to inflammation and the establishment of pathological states, including the onset and development of cancer. The tumor microenvironment induces molecular and functional modifications in innate cells, dampening their capability to initiate and sustain anti-tumor immune responses. Emerging studies clearly showed a contribution of the microbiota in modulating the functions of innate cells in cancer. Commensal microorganisms can not only directly interact with innate cells in the tumor microenvironment but can also exert immunomodulatory features from non-tumor sites through the release of microbial products. The microbiota can mediate the priming of innate cells at mucosal tissues and determine the strength of immune responses mediated by such cells when they migrate to non-mucosal tissues, having an impact on cancer. Finally, several evidences reported a strong contribution of the microbiota in promoting innate immune responses during anti-cancer therapies leading to enhanced therapeutic efficacy. In this review, we considered the current knowledge on the role of the microbiota in shaping host innate immune responses in cancer.
Collapse
Affiliation(s)
- Irene Mattiola
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany; Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
44
|
Complexification of In Vitro Models of Intestinal Barriers, A True Challenge for a More Accurate Alternative Approach. Int J Mol Sci 2023; 24:ijms24043595. [PMID: 36835003 PMCID: PMC9958734 DOI: 10.3390/ijms24043595] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
The use of cell models is common to mimic cellular and molecular events in interaction with their environment. In the case of the gut, the existing models are of particular interest to evaluate food, toxicants, or drug effects on the mucosa. To have the most accurate model, cell diversity and the complexity of the interactions must be considered. Existing models range from single-cell cultures of absorptive cells to more complex combinations of two or more cell types. This work describes the existing solutions and the challenges that remain to be solved.
Collapse
|
45
|
Cao M, Xue T, Huo H, Zhang X, Wang NN, Yan X, Li C. Spatial transcriptomes and microbiota reveal immune mechanism that respond to pathogen infection in the posterior intestine of Sebastes schlegelii. Open Biol 2023; 13:220302. [PMID: 36974664 PMCID: PMC9944294 DOI: 10.1098/rsob.220302] [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: 10/02/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023] Open
Abstract
The intestine is a site of immune cell priming at birth. Therefore, spatial transcriptomes were performed to define how the transcriptomic landscape was spatially organized in the posterior intestine of Sebastes schlegelii following Edwardsiella piscicida infection. In the healthy condition, we identified a previously unappreciated molecular regionalization of the posterior intestine. Following bacterial infection, most immune-related genes were identified in mucosa layer. Moreover, investigation of immune-related genes and genes in immune-related KEGG pathways based on spatial transcriptomes shed light on which sections of these genes are in the posterior intestine. Meanwhile, the high expression of genes related to regeneration also indicated that the posterior intestine was responding to the invasion of pathogens by constantly proliferating new cells. In addition, the increasing microbiota communities indicated that these bacteria maintained posterior intestine integrity and shaped the mucosal immune system. Taken together, spatial transcriptomes and microbiota compositions have significant implications for understanding the immune mechanism that responds to E. piscicida infection in the posterior intestine of S. schlegelii, which also provides a theoretical basis for the spatial distribution of immune genes and changes in bacterial flora in other teleosts in the process of resisting pathogens.
Collapse
Affiliation(s)
- Min Cao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ting Xue
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Huijun Huo
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xiaoyan Zhang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ning Ning Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xu Yan
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
46
|
Li X, Zhang M, Zhou G, Xie Z, Wang Y, Han J, Li L, Wu Q, Zhang S. Role of Rho GTPases in inflammatory bowel disease. Cell Death Dis 2023; 9:24. [PMID: 36690621 PMCID: PMC9871048 DOI: 10.1038/s41420-023-01329-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Rat sarcoma virus homolog (Rho) guanosine triphosphatases (GTPases) function as "molecular switch" in cellular signaling regulation processes and are associated with the pathogenesis of inflammatory bowel disease (IBD). This chronic intestinal tract inflammation primarily encompasses two diseases: Crohn's disease and ulcerative colitis. The pathogenesis of IBD is complex and considered to include four main factors and their interactions: genetics, intestinal microbiota, immune system, and environment. Recently, several novel pathogenic components have been identified. In addition, potential therapies for IBD targeting Rho GTPases have emerged and proven to be clinically effective. This review mainly focuses on Rho GTPases and their possible mechanisms in IBD pathogenesis. The therapeutic possibility of Rho GTPases is also discussed.
Collapse
Affiliation(s)
- Xiaoling Li
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Mudan Zhang
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Gaoshi Zhou
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Zhuo Xie
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Ying Wang
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jing Han
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Li Li
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Qirui Wu
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Shenghong Zhang
- grid.12981.330000 0001 2360 039XDivision of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| |
Collapse
|
47
|
Xie Y, Shao F, Duan X, Ding J, Ning Y, Sun X, Xia L, Pan J, Chen J, He S, Shen D, Qi C. Whole β-glucan particle attenuates AOM/DSS-induced colorectal tumorigenesis in mice via inhibition of intestinal inflammation. Front Pharmacol 2023; 14:1017475. [PMID: 36713833 PMCID: PMC9877317 DOI: 10.3389/fphar.2023.1017475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Yeast β-glucan is a polysaccharide purified from the Saccharomyces cerevisiae cell wall, and its multiple biological activities are essential for immune regulation. However, the effect of β-glucan on the intestinal immune response during colitis-associated colorectal cancer (CAC) is unclear. Here, we explore the possible role of β-glucan in the development of CAC. Wild type (WT) mice with CAC induced by azoxmethane (AOM) and dextran sodium sulfate (DSS) had fewer tumors than untreated mice after oral β-glucan because of increased antitumor dendritic cells (DCs) in the tumor microenvironment, resulting in more CD8+ T cells and the production of related cytokines. β-glucan also increased resistance to DSS-induced chronic colitis by reshaping the inflammatory microenvironment. These data suggest that β-glucan improves experimental intestinal inflammation and delays the development of CAC. Therefore, β-glucan is feasible for treating chronic colitis and CAC in clinical practice.
Collapse
Affiliation(s)
- Yewen Xie
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Fang Shao
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xuehan Duan
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jun Ding
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Yongling Ning
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xiao Sun
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Lei Xia
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jie Pan
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Jie Chen
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Shuyan He
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu, China
| | - Dong Shen
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, Jiangsu, China,*Correspondence: Chunjian Qi, ; Dong Shen,
| | - Chunjian Qi
- Medical Research Center, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,Oncology Institute, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China,*Correspondence: Chunjian Qi, ; Dong Shen,
| |
Collapse
|
48
|
Yao H, Zhang D, Yu H, Yuan H, Shen H, Lan X, Liu H, Chen X, Meng F, Wu X, Zhang G, Wang X. Gut microbiota regulates chronic ethanol exposure-induced depressive-like behavior through hippocampal NLRP3-mediated neuroinflammation. Mol Psychiatry 2023; 28:919-930. [PMID: 36280756 PMCID: PMC9908543 DOI: 10.1038/s41380-022-01841-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Chronic ethanol exposure (CEE), which can lead to neuroinflammation, is an increasing risk factor for depression disorder, but the underlying mechanism is not clear. Recent observations have revealed the associations among psychiatric disorders, ethanol exposure and alterations of the gut microbiota. Here, we found that CEE induced depressive-like behavior, which could be alleviated by probiotics and transferred from donor to recipient mice by fecal microbiota transplantation (FMT). Neuroinflammation and the activation of the NLRP3 inflammasome were also observed in recipient mice. The downregulation of NLRP3 in the hippocampus mitigated CEE-induced depressive-like behavior and neuroinflammation but had no significant effect on FMT recipient mice. Moreover, elevated serum inflammatory factors in recipient mice showed a significant mediation effect between the gut microbiota and depressive-like behavior. Together, our study findings indicate that the gut microbiota contributes to both hippocampal NLRP3-mediated neuroinflammation and depressive-like behavior induced by CEE, which may open avenues for potential interventions against CEE-associated psychiatric disorders.
Collapse
Affiliation(s)
- Hui Yao
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Dalin Zhang
- grid.412636.40000 0004 1757 9485Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning PR China
| | - Hao Yu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Huiya Yuan
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884Department of Forensic Analytical Toxicology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China
| | - Hui Shen
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xinze Lan
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Hao Liu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xiaohuan Chen
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Fanyue Meng
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xu Wu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Guohua Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122, Liaoning, PR China. .,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, Liaoning, PR China. .,China Medical University Center of Forensic Investigation, Shenyang, 110122, Liaoning, PR China.
| | - Xiaolong Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122, Liaoning, PR China. .,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, Liaoning, PR China. .,China Medical University Center of Forensic Investigation, Shenyang, 110122, Liaoning, PR China.
| |
Collapse
|
49
|
Riaz F, Pan F, Wei P. Aryl hydrocarbon receptor: The master regulator of immune responses in allergic diseases. Front Immunol 2022; 13:1057555. [PMID: 36601108 PMCID: PMC9806217 DOI: 10.3389/fimmu.2022.1057555] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a widely studied ligand-activated cytosolic transcriptional factor that has been associated with the initiation and progression of various diseases, including autoimmune diseases, cancers, metabolic syndromes, and allergies. Generally, AhR responds and binds to environmental toxins/ligands, dietary ligands, and allergens to regulate toxicological, biological, cellular responses. In a canonical signaling manner, activation of AhR is responsible for the increase in cytochrome P450 enzymes which help individuals to degrade and metabolize these environmental toxins and ligands. However, canonical signaling cannot be applied to all the effects mediated by AhR. Recent findings indicate that activation of AhR signaling also interacts with some non-canonical factors like Kruppel-like-factor-6 (KLF6) or estrogen-receptor-alpha (Erα) to affect the expression of downstream genes. Meanwhile, enormous research has been conducted to evaluate the effect of AhR signaling on innate and adaptive immunity. It has been shown that AhR exerts numerous effects on mast cells, B cells, macrophages, antigen-presenting cells (APCs), Th1/Th2 cell balance, Th17, and regulatory T cells, thus, playing a significant role in allergens-induced diseases. This review discussed how AhR mediates immune responses in allergic diseases. Meanwhile, we believe that understanding the role of AhR in immune responses will enhance our knowledge of AhR-mediated immune regulation in allergic diseases. Also, it will help researchers to understand the role of AhR in regulating immune responses in autoimmune diseases, cancers, metabolic syndromes, and infectious diseases.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China,*Correspondence: Ping Wei, ; Fan Pan,
| | - Ping Wei
- Department of Otolaryngology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China,*Correspondence: Ping Wei, ; Fan Pan,
| |
Collapse
|
50
|
Cirillo G, Negrete-Diaz F, Yucuma D, Virtuoso A, Korai SA, De Luca C, Kaniusas E, Papa M, Panetsos F. Vagus Nerve Stimulation: A Personalized Therapeutic Approach for Crohn's and Other Inflammatory Bowel Diseases. Cells 2022; 11:cells11244103. [PMID: 36552867 PMCID: PMC9776705 DOI: 10.3390/cells11244103] [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: 08/25/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis, are incurable autoimmune diseases characterized by chronic inflammation of the gastrointestinal tract. There is increasing evidence that inappropriate interaction between the enteric nervous system and central nervous system and/or low activity of the vagus nerve, which connects the enteric and central nervous systems, could play a crucial role in their pathogenesis. Therefore, it has been suggested that appropriate neuroprosthetic stimulation of the vagus nerve could lead to the modulation of the inflammation of the gastrointestinal tract and consequent long-term control of these autoimmune diseases. In the present paper, we provide a comprehensive overview of (1) the cellular and molecular bases of the immune system, (2) the way central and enteric nervous systems interact and contribute to the immune responses, (3) the pathogenesis of the inflammatory bowel disease, and (4) the therapeutic use of vagus nerve stimulation, and in particular, the transcutaneous stimulation of the auricular branch of the vagus nerve. Then, we expose the working hypotheses for the modulation of the molecular processes that are responsible for intestinal inflammation in autoimmune diseases and the way we could develop personalized neuroprosthetic therapeutic devices and procedures in favor of the patients.
Collapse
Affiliation(s)
- Giovanni Cirillo
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Flor Negrete-Diaz
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias (IdISSC), Hospital Clinico San Carlos de Madrid, 28040 Madrid, Spain
| | - Daniela Yucuma
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Andalusian School of Public Health, University of Granada, 18011 Granada, Spain
| | - Assunta Virtuoso
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Sohaib Ali Korai
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | - Ciro De Luca
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
| | | | - Michele Papa
- Division of Human Anatomy, Neuronal Morphology Networks & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy
- SYSBIO Centre of Systems Biology ISBE-IT, University of Milano-Bicocca, 20126 Milan, Italy
- Correspondence: (M.P.); (F.P.)
| | - Fivos Panetsos
- Neurocomputing & Neurorobotics Research Group, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias (IdISSC), Hospital Clinico San Carlos de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, 28260 Madrid, Spain
- Correspondence: (M.P.); (F.P.)
| |
Collapse
|