1
|
Zhou D, Gan G, Song S, Zi C, Bao Y, Hao W, Chen Q. The Impact of Immune Cells, Metabolites, Inflammatory Factors, and Circulating Proteins on Atopic Dermatitis: Insights from a Mendelian Randomization Study. Clin Cosmet Investig Dermatol 2024; 17:2999-3011. [PMID: 39723341 PMCID: PMC11669345 DOI: 10.2147/ccid.s495217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Background The onset of atopic dermatitis (AD) is complex, and its specific pathological mechanisms have not yet been fully elucidated. Methods Using circulating multi-omics as the exposure factors and AD as the outcome, we conducted univariable MR analysis. The circulating multi-omics data included immunomics (731 immune cell types), proteomics (4907 plasma proteins), metabolomics (1400 metabolites and 486 additional metabolites), and 91 inflammatory factors. MR analysis was conducted using IVW, WM, Simple Mode, Weighted Mode, and MR-Egger methods, with IVW as the primary analysis tool. To address horizontal pleiotropy, we utilized MR-Egger intercept tests and MR-PRESSO for correction, alongside the Cochrane Q statistic for heterogeneity assessment. Sensitivity analysis was performed using a leave-one-out strategy. To control for false positives due to multiple testing, we set a standard of a 5% false discovery rate. Additionally, we conducted F-statistics on the included SNPs to eliminate the impact of weak instrumental variables. Results IL-18R1 on AD (OR = 1.12, 95% CI: 1.08-1.17, P FDR < 0.01). Mannonate levels on AD (OR = 0.88, 95% CI: 0.83-0.94, P FDR = 0.03). Retinol (Vitamin A) to linoleoyl-arachidonoyl-glycerol (18:2 to 20:4) on AD (OR = 1.12, 95% CI: 1.06-1.18, PFDR = 0.03). HVEM on CM CD4+ cells on AD (OR = 0.81, 95% CI: 0.75-0.88, P FDR < 0.01). CR2 on AD (OR = 0.81, 95% CI: 0.72-0.90, P FDR = 0.04). MANSC1 on AD (OR = 0.87, 95% CI: 0.81-0.93, P FDR = 0.04). IL18R1 (4097 inflammatory markers) on AD (OR = 1.11, 95% CI: 1.06-1.17, P FDR = 0.01). HNRNPAB on AD (OR = 1.44, 95% CI: 1.23-1.70, P FDR < 0.01). Conclusion This study further explored the correlations between multi-omics data and AD. We identified seven previously unreported circulating substances with causal relationships to AD, filling a current theoretical gap.
Collapse
Affiliation(s)
- Dongqi Zhou
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Gaofeng Gan
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Shiwei Song
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Cangyan Zi
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Yichen Bao
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Wenfeng Hao
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, People’s Republic of China
| |
Collapse
|
2
|
Hanski E, Raulo A, Knowles SCL. Early-life gut microbiota assembly patterns are conserved between laboratory and wild mice. Commun Biol 2024; 7:1456. [PMID: 39511304 PMCID: PMC11543677 DOI: 10.1038/s42003-024-07039-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Assembly of the mammalian gut microbiota during early life is known to shape key aspects of organismal development, including immunity, metabolism and behaviour. While house mice (Mus musculus) are the major laboratory model organism for gut microbiota research, their artificial lab-based lifestyle could fundamentally alter ecological processes of microbiota assembly and dynamics, in ways that affect their usefulness as a model system. To examine this, here we directly compared patterns of gut microbiota assembly in house mice from the lab and from the wild, making use of a tractable, individually-marked wild population where we could examine patterns of gut microbiota assembly during early life. Despite lab and wild mice harbouring taxonomically distinct communities, we identify striking similarities in multiple patterns of their gut microbiota assembly. Specifically, age-related changes in both alpha and beta diversity, as well as the abundance of predominant phyla and aerotolerance of the microbiota followed parallel trajectories in both settings. These results suggest some degree of intrinsic programme in gut microbiota assembly that transcends variation in taxonomic profiles, and the genetic and environmental background of the host. They further support the notion that despite their artificial environment, lab mice can provide meaningful insights into natural microbiota ecological dynamics in early life and their interplay with host development.
Collapse
Affiliation(s)
- Eveliina Hanski
- Department of Biology, University of Oxford, Oxford, Oxfordshire, UK.
- Faculty of Medicine, University of Helsinki, Helsinki, Southern Finland, Finland.
| | - Aura Raulo
- Department of Biology, University of Oxford, Oxford, Oxfordshire, UK
- Department of Computing, University of Turku, Turku, Western Finland, Finland
| | - Sarah C L Knowles
- Department of Biology, University of Oxford, Oxford, Oxfordshire, UK.
| |
Collapse
|
3
|
Vassilopoulou E, Venter C, Roth-Walter F. Malnutrition and Allergies: Tipping the Immune Balance towards Health. J Clin Med 2024; 13:4713. [PMID: 39200855 PMCID: PMC11355500 DOI: 10.3390/jcm13164713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Malnutrition, which includes macro- and micronutrient deficiencies, is common in individuals with allergic dermatitis, food allergies, rhinitis, and asthma. Prolonged deficiencies of proteins, minerals, and vitamins promote Th2 inflammation, setting the stage for allergic sensitization. Consequently, malnutrition, which includes micronutrient deficiencies, fosters the development of allergies, while an adequate supply of micronutrients promotes immune cells with regulatory and tolerogenic phenotypes. As protein and micronutrient deficiencies mimic an infection, the body's innate response limits access to these nutrients by reducing their dietary absorption. This review highlights our current understanding of the physiological functions of allergenic proteins, iron, and vitamin A, particularly regarding their reduced bioavailability under inflamed conditions, necessitating different dietary approaches to improve their absorption. Additionally, the role of most allergens as nutrient binders and their involvement in nutritional immunity will be briefly summarized. Their ability to bind nutrients and their close association with immune cells can trigger exaggerated immune responses and allergies in individuals with deficiencies. However, in nutrient-rich conditions, these allergens can also provide nutrients to immune cells and promote health.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
- Department of Clinical Sciences and Community Health, Univertià degli Studi die Milano, 20122 Milan, Italy
| | - Carina Venter
- Pediatrics, Section of Allergy & Immunology, University of Colorado Denver School of Medicine, Children’s Hospital Colorado, Box B518, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Franziska Roth-Walter
- Messerli Research Institute, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, 1210 Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
4
|
Wang L, Ren B, Wu S, Song H, Xiong L, Wang F, Shen X. Current research progress, opportunities, and challenges of Limosillactobacillus reuteri-based probiotic dietary strategies. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38920093 DOI: 10.1080/10408398.2024.2369946] [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: 06/27/2024]
Abstract
Limosillactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., stands out as the most extensively researched probiotic. Its remarkable intestinal adhesion has led to widespread applications in both the food and medical sectors. Notably, recent research highlights the probiotic efficacy of L. reuteri sourced from breast milk, particularly in influencing social behavior and mitigating atopic dermatitis. In this review, our emphasis is on surveying recent literature regarding the promotion of host's health by L. reuteri. We aim to provide a concise summary of the latest regulatory effects and potential mechanisms attributed to L. reuteri in the realms of metabolism, brain- and immune-related functions. The mechanism through which L. reuteri promotes host health by modulating the intestinal microenvironment primarily involves promoting intestinal epithelial renewal, bolstering intestinal barrier function, regulating gut microbiota and its metabolites, and suppressing inflammation and immune responses. Additionally, this review delves into new technologies, identifies shortcomings, and addresses challenges in current L. reuteri research. Finally, the application prospects of L. reuteri are provided. Therefore, a better understanding of the role and mechanisms of L. reuteri will contribute significantly to the development of new probiotic functional foods and enable precise, targeted interventions for various diseases.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Shufeng Wu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
5
|
C. Bergeron C, Costa MC, Segura M, de Souza LB, Bleuzé M, Sauvé F. Bacterial microbiota and proinflammatory cytokines in the anal sacs of treated and untreated atopic dogs: Comparison with a healthy control group. PLoS One 2024; 19:e0298361. [PMID: 38814946 PMCID: PMC11139270 DOI: 10.1371/journal.pone.0298361] [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: 09/27/2023] [Accepted: 01/24/2024] [Indexed: 06/01/2024] Open
Abstract
The pathogenesis of anal sacculitis has not been extensively investigated, although atopic dogs seem to be predisposed to the disease. The aim of this study was therefore to characterize and compare the bacterial microbiota and pro-inflammatory cytokines in the anal sacs of dogs from three groups (healthy dogs, untreated atopic dogs and atopic dogs receiving antipruritic treatment or allergen-specific immunotherapy) in order to determine whether changes could be at the origin of anal sacculitis in atopic dogs. Bacterial populations of anal sac secretions from fifteen healthy dogs, fourteen untreated and six treated atopic dogs were characterized by sequencing the V4 region of the 16S rRNA gene using Illumina technology. Proinflammatory cytokines were analyzed with the Luminex multiplex test. Community membership and structure were significantly different between the anal sacs of healthy and untreated atopic dogs (P = 0.002 and P = 0.003, respectively) and between those of untreated and treated atopic dogs (P = 0.012 and P = 0.017, respectively). However, the community structure was similar in healthy and treated atopic dogs (P = 0.332). Among the proinflammatory cytokines assessed, there was no significant difference between groups, except for interleukin 8 which was higher in the anal sacs of untreated atopic dogs compared to treated atopic dogs (P = 0.02), and tumor necrosis factor-alpha which was lower in the anal sacs of healthy dogs compared to treated atopic dogs (P = 0.04). These results reveal a dysbiosis in the anal sacs of atopic dogs, which may partially explain the predisposition of atopic dogs to develop bacterial anal sacculitis. Treatments received by atopic dogs (oclacitinib, desloratadine and allergen-specific immunotherapy) shift the microbiota of the anal sacs towards that of healthy dogs. Further studies are required to identify significant cytokines contributing to anal sacculitis in atopic dogs.
Collapse
Affiliation(s)
- Camylle C. Bergeron
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marcio Carvalho Costa
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Mariela Segura
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Lucilene Bernardi de Souza
- Centre Hospitalier Universitaire Vétérinaire, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marêva Bleuzé
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Frédéric Sauvé
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
6
|
Roth-Walter F, Berni Canani R, O'Mahony L, Peroni D, Sokolowska M, Vassilopoulou E, Venter C. Nutrition in chronic inflammatory conditions: Bypassing the mucosal block for micronutrients. Allergy 2024; 79:353-383. [PMID: 38084827 DOI: 10.1111/all.15972] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Nutritional Immunity is one of the most ancient innate immune responses, during which the body can restrict nutrients availability to pathogens and restricts their uptake by the gut mucosa (mucosal block). Though this can be a beneficial strategy during infection, it also is associated with non-communicable diseases-where the pathogen is missing; leading to increased morbidity and mortality as micronutritional uptake and distribution in the body is hindered. Here, we discuss the acute immune response in respect to nutrients, the opposing nutritional demands of regulatory and inflammatory cells and particularly focus on some nutrients linked with inflammation such as iron, vitamins A, Bs, C, and other antioxidants. We propose that while the absorption of certain micronutrients is hindered during inflammation, the dietary lymph path remains available. As such, several clinical trials investigated the role of the lymphatic system during protein absorption, following a ketogenic diet and an increased intake of antioxidants, vitamins, and minerals, in reducing inflammation and ameliorating disease.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Liam O'Mahony
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Diego Peroni
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Vassilopoulou
- Pediatric Area, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carina Venter
- Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
7
|
Zachariassen LF, Ebert MBB, Mentzel CMJ, Deng L, Krych L, Nielsen DS, Stokholm J, Hansen CHF. Cesarean section induced dysbiosis promotes type 2 immunity but not oxazolone-induced dermatitis in mice. Gut Microbes 2023; 15:2271151. [PMID: 37889696 PMCID: PMC10730161 DOI: 10.1080/19490976.2023.2271151] [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: 03/27/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Delivery by cesarean section (CS) is associated with an altered gut microbiota (GM) colonization and a higher risk of later chronic inflammatory diseases. Studies investigating the association between CS and atopic dermatitis (AD) are contradictive and often biased by confounding factors. The aim of this study was therefore to provide experimental evidence for the association between CS and AD in a mouse model and clarify the role of the GM changes associated with CS. It was hypothesized that CS-delivered mice, and human CS-GM transplanted mice develop severe dermatitis due to early dysbiosis. BALB/c mice delivered by CS or vaginally (VD) as well as BALB/c mice transplanted with GM from CS or VD human donors were challenged with oxazolone on the ear. The severity of dermatitis was evaluated by ear thickness and clinical and histopathological assessment which were similar between all groups. The immune response was assessed by serum IgE concentration, local cytokine response, and presence of immune cells in the draining lymph node. Both CS-delivered mice and mice inoculated with human CS-GM had a higher IgE concentration. A higher proportion of Th2 cells were also found in the CS-GM inoculated mice, but no differences were seen in the cytokine levels in the affected ears. In support of the experimental findings, a human cohort analysis from where the GM samples were obtained found that delivery mode did not affect the children's risk of developing AD. In conclusion, CS-GM enhanced a Th2 biased immune response, but had no effect on oxazolone-induced dermatitis in mice.
Collapse
Affiliation(s)
- Line Fisker Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maria Bernadette Bergh Ebert
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Caroline Märta Junker Mentzel
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Deng
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Dennis Sandris Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Jakob Stokholm
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, Gentofte, Denmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
8
|
Yu R, Ma Y, Luo Z, Qi C, Xie A, Jiang Y, Zhu B, Sun J. Maternal supplementation with Limosilactobacillus reuteri FN041 for preventing infants with atopic dermatitis: study protocol for a randomized controlled trial. Front Microbiol 2023; 14:1267448. [PMID: 37869669 PMCID: PMC10588667 DOI: 10.3389/fmicb.2023.1267448] [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: 07/26/2023] [Accepted: 08/28/2023] [Indexed: 10/24/2023] Open
Abstract
Background Atopic dermatitis (AD) has increased rapidly with rapid urbanization; however, the treatment options for AD are lacking because the commonly used therapies can only alleviate symptoms. Limosilactobacillus reuteri (L. reuteri), FN041 is a specific strain isolated from human breast milk, and its protective potential against AD has been confirmed. This study aims to assess the efficacy of maternal consumption of L. reuteri FN041 during late pregnancy and lactation in preventing infantile AD. Methods First, a randomized, double-blind, placebo-controlled intervention study will be conducted on 340 pregnant females with babies at high risk for AD. These subjects will be randomly divided into four groups of different doses of L. reuteri FN041 (1 × 109, 5 × 109, and 1 × 1010 CFU/d) along with a placebo. The safety and efficacy of maternal use of L. reuteri FN041 for preventing infantile AD will be analyzed, and the most efficient dosage of L. reuteri FN041 will be determined. Subsequently, a multicenter cohort study of 500 pregnant females with babies at high risk for AD will be conducted to promote the maternal application of L. reuteri FN041. These subjects will be administered L. reuteri FN041 at the optimal dose determined during the first stage of late pregnancy and lactation, and their babies will be analyzed for AD development. Recruitment was initiated in October 2022. Discussion The primary outcome is the cumulative incidence of AD at 24 months after maternal consumption of L. reuteri FN041 during late pregnancy and lactation, whereas the secondary outcome is the efficiency of L. reuteri FN041 transfer from the mother's gut to breast milk and then the infant's gut after oral supplementation. This study will demonstrate the efficacy of edible probiotics isolated from breast milk in preventing or treating AD in infants. Accordingly, we provide population-based advice for administering specific probiotics for the primary prevention of AD in pregnant females. Understanding the underlying mechanisms of probiotic strains derived from breast milk can promote their application in preventing infant diseases associated with intestinal microbiota imbalance and immune disorders. Clinical trial registration https://www.chictr.org.cn/, identifier [ChiCTR2300075611].
Collapse
Affiliation(s)
- Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yizhe Ma
- Department of Pediatric, Jiangyin People's Hospital of Nantong University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Anni Xie
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yifan Jiang
- School of Medicine, Nantong University, Nantong, China
| | - Baoli Zhu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Xie A, Chen A, Chen Y, Luo Z, Jiang S, Chen D, Yu R. Lactobacillus for the treatment and prevention of atopic dermatitis: Clinical and experimental evidence. Front Cell Infect Microbiol 2023; 13:1137275. [PMID: 36875529 PMCID: PMC9978199 DOI: 10.3389/fcimb.2023.1137275] [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: 01/04/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease, accompanied by itching and swelling. The main pathological mechanism of AD is related to the imbalance between Type 2 helper cells (Th2 cells) and Type 1 helper cells (Th1 cells). Currently, no safe and effective means to treat and prevent AD are available; moreover, some treatments have side effects. Probiotics, such as some strains of Lactobacillus, can address these concerns via various pathways: i) facilitating high patient compliance; ii) regulating Th1/Th2 balance, increasing IL-10 secretion, and reducing inflammatory cytokines; iii) accelerating the maturation of the immune system, maintaining intestinal homeostasis, and improving gut microbiota; and iv) improving the symptoms of AD. This review describes the treatment and prevention of AD using 13 species of Lactobacillus. AD is commonly observed in children. Therefore, the review includes a higher proportion of studies on AD in children and fewer in adolescents and adults. However, there are also some strains that do not improve the symptoms of AD and even worsen allergies in children. In addition, a subset of the genus Lactobacillus that can prevent and relieve AD has been identified in vitro. Therefore, future studies should include more in vivo studies and randomized controlled clinical trials. Given the advantages and disadvantages mentioned above, further research in this area is urgently required.
Collapse
Affiliation(s)
- Anni Xie
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuqing Chen
- Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Daozhen Chen, ; Renqiang Yu,
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Daozhen Chen, ; Renqiang Yu,
| |
Collapse
|
10
|
Xie R, Zhang H, Zhang H, Li C, Cui D, Li S, Li Z, Liu H, Huang J. Hemagglutinin expressed by yeast reshapes immune microenvironment and gut microbiota to trigger diverse anti-infection response in infected birds. Front Immunol 2023; 14:1125190. [PMID: 37143654 PMCID: PMC10151582 DOI: 10.3389/fimmu.2023.1125190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The H5N8 influenza virus is a highly pathogenic pathogen for poultry and human. Vaccination is the most effective method to control the spread of the virus right now. The traditional inactivated vaccine, though well developed and used widely, is laborious during application and more interests are stimulated in developing alternative approaches. Methods In this study, we developed three hemagglutinin (HA) gene-based yeast vaccine. In order to explore the protective efficacy of the vaccines, the gene expression level in the bursa of Fabricius and the structure of intestinal microflora in immunized animals were analyzed by RNA seq and 16SrRNA sequencing, and the regulatory mechanism of yeast vaccine was evaluated. Results All of these vaccines elicited the humoral immunity, inhibited viral load in the chicken tissues, and provided partial protective efficacy due to the high dose of the H5N8 virus. Molecular mechanism studies suggested that, compared to the traditional inactivated vaccine, our engineered yeast vaccine reshaped the immune cell microenvironment in bursa of Fabricius to promote the defense and immune responses. Analysis of gut microbiota further suggested that oral administration of engineered ST1814G/H5HA yeast vaccine increased the diversity of gut microbiota and the increasement of Reuteri and Muciniphila might benefit the recovery from influenza virus infection. These results provide strong evidence for further clinical use of these engineered yeast vaccine in poultry.
Collapse
Affiliation(s)
- Ruyu Xie
- School of Life Science, Tianjin University, Tianjin, China
| | - Huixia Zhang
- School of Life Science, Tianjin University, Tianjin, China
| | - Han Zhang
- School of Life Science, Tianjin University, Tianjin, China
| | - Changyan Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Daqing Cui
- School of Life Science, Tianjin University, Tianjin, China
| | - Shujun Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Zexing Li
- School of Life Science, Tianjin University, Tianjin, China
| | - Hualei Liu
- China Animal Health and Epidemiology Center, Qingdao, Shandong, China
- *Correspondence: Hualei Liu, ; Jinhai Huang,
| | - Jinhai Huang
- School of Life Science, Tianjin University, Tianjin, China
- *Correspondence: Hualei Liu, ; Jinhai Huang,
| |
Collapse
|