1
|
Peneva P, Pancheva R, Nikolova SP. Respiratory Microbiota and Health Risks in Children with Cerebral Palsy: A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2025; 12:358. [PMID: 40150639 PMCID: PMC11941506 DOI: 10.3390/children12030358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Children diagnosed with cerebral palsy (CP) frequently face a range of intricate health challenges that go beyond their main condition. Respiratory problems represent one of the most crucial factors contributing to morbidity and mortality. This review employed a systematic approach to identify and collate recent findings on the respiratory microbiota in children with CP. The review emphasizes notable microbial alterations in the respiratory systems of children with CP, marked by a decrease in beneficial bacteria (such as Corynebacterium spp. and Dolosigranulum spp.) and an increase in opportunistic pathogens like Staphylococcus aureus, Pseudomonas aeruginosa, and Klebsiella pneumonia. These changes probably increase the vulnerability of children with CP to frequent respiratory infections, ongoing inflammation, and infections that are resistant to antibiotics. Key factors influencing the composition of microbiota include living in urban areas, socioeconomic factors, seasonal variations, vaccination status, dietary habits, breastfeeding, etc. Although new research has shed significant light on this topic, there are still considerable gaps in our understanding of how these microbial communities develop and interact with the immune responses of the host. Future research should focus on longitudinal studies to track microbiota changes over time and identify interventions that optimize respiratory health in CP.
Collapse
Affiliation(s)
- Pavlina Peneva
- First Department of Internal Disease, Faculty of Medicine, Medical University, 9002 Varna, Bulgaria
| | - Rouzha Pancheva
- Department of Hygiene and Epidemiology, Faculty of Public Health, Medical University, 9002 Varna, Bulgaria;
| | - Silviya P. Nikolova
- Department of Social Medicine and Healthcare Organization, Faculty of Public Health, Medical University, 9002 Varna, Bulgaria;
| |
Collapse
|
2
|
Xian M, Maskey AR, Kopulos D, Li XM. The roles of bitter and sweet taste receptors in food allergy: Where are we now? Allergol Int 2025:S1323-8930(25)00010-3. [PMID: 40037957 DOI: 10.1016/j.alit.2025.01.005] [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: 11/04/2024] [Revised: 12/31/2024] [Accepted: 01/17/2025] [Indexed: 03/06/2025] Open
Abstract
Food allergy (FA) is a growing global concern, which contributes significantly to anaphylaxis and severe allergic reactions. Despite advancements in treatments like allergen immunotherapy and biologics, current approaches have notable limitations and there is a pressing need for new therapeutic strategies. Recent research into taste receptors has unveiled their potential role in FA, offering fresh perspectives for understanding and managing this condition. Taste receptors, particularly type 1 taste receptors (TAS1Rs/T1Rs, sweet taste receptors) and type 2 taste receptors (TAS2Rs/T2Rs, bitter taste receptors), are distributed not only in the oral cavity but also in various extra-oral tissues, and their interactions with immune responses are increasingly recognized. This review highlights the connections between taste receptors and FA, exploring how taste receptor mechanisms might contribute to FA pathogenesis and treatment. Taste receptors, especially TAS2Rs, which include multiple subtypes with varying ligand specificities, have been implicated in modulating allergic responses and could serve as targets for novel FA therapies. Additionally, compounds such as bitter agents and sweeteners that interact with taste receptors show promise in influencing FA outcomes. This review emphasizes the need for further research into the mechanisms of taste receptor involvement in FA and suggests that targeting these receptors could provide new avenues for therapeutic intervention in the future.
Collapse
Affiliation(s)
- Mo Xian
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Anish R Maskey
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Daniel Kopulos
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, USA; Department of Otolaryngology, New York Medical College, Valhalla, NY, USA; Department of Dermatology, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
3
|
Yang L, Wang P, Gao T, Huang X, Lin Z, Sweren E, Li Y, Chen L, Alphonse MP, Zhang J, Wang G. Melatonin treatment increases skin microbiota-derived propionic acid to alleviate atopic dermatitis. J Allergy Clin Immunol 2025; 155:880-891.e9. [PMID: 39579877 DOI: 10.1016/j.jaci.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Melatonin has been reported to relieve the inflammatory symptoms and improve sleep disturbance in patients with atopic dermatitis (AD). Recent studies showed that melatonin produced beneficial effects by remodeling intestinal microbiota composition; however, whether the beneficial effects of melatonin in AD were mediated by the modulation of skin microbiota remains unclear. OBJECTIVE We sought to investigate the mechanism by which melatonin treatment-induced changes in the skin microbiota composition further alleviated AD. METHODS The changes in skin bacterial composition after melatonin treatment were detected by 16S-rRNA sequencing. Further mechanisms were explored in calcipotriol (MC903)-induced AD mice and HaCaT cells through skin microbiota transplantation, quantification detection of short-chain fatty acids, transcriptome and single-cell sequencing analysis, quantitative RT-PCR, Western blotting, and Cell Counting Kit-8 assay. RESULTS We demonstrated that melatonin reshaped the skin microbiota in AD mice. The transplantation of skin microbiota from melatonin-treated mice alleviated AD symptoms in mice. Skin microbiota-derived short-chain fatty acids, especially propionic acid, were increased in the skin of melatonin-treated AD mice, which further inhibited FABP5 expression to alleviate AD. Propionic acid also inhibited FABP5 expression in HaCaT cells, which was reversed by the treatment of GPR43 inhibitor GLPG0974. GLPG0974 also blocked the therapeutic effects of melatonin on AD mice. CONCLUSIONS Our study demonstrated that melatonin alleviates AD through the skin microbiota/propionic acid/GPR43/FABP5 axis, highlighting a novel role of melatonin as a modulator of skin microbiota to alleviate AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Piao Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Ting Gao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinyu Huang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Evan Sweren
- University of Michigan Medical School, Ann Arbor, Mich
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
4
|
Liu Y, Dai J, Zhou G, Chen R, Bai C, Shi F. Innovative Therapeutic Strategies for Asthma: The Role of Gut Microbiome in Airway Immunity. J Asthma Allergy 2025; 18:257-267. [PMID: 39996012 PMCID: PMC11849427 DOI: 10.2147/jaa.s504571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
There is a growing acknowledgment of the gut microbiome's impact on widespread immune responses, which holds considerable importance for comprehending and addressing asthma. Recent research has clarified the complex interactions between gut microbiota and airway immune systems, demonstrating that microbial diversity and composition can affect both the initiation and advancement of asthma. Gut microbial species and metabolites primarily short-chain fatty acids (SCFAs) may either worsen or reduce airway inflammation by regulating the balance of helper T cell 1 (Th1) / helper T cell 2 (Th2) and other immune mediators. This interaction presents innovative therapeutic possibilities, including modulation of gut microbiome during early life through breastfeeding and control of antibiotic use, particularly with prebiotics, which could selectively stimulate the growth of beneficial bacteria, promote immune maturation, reducing susceptibility to asthma and allergic airway inflammation. Besides, investigating the gut-lung axis reveals new opportunities for personalized medicine in asthma treatment, emphasizing the necessity for integrated strategies that take individual microbiome profiles into account. This paper examines the latest developments in comprehending the mechanisms by which gut microbiota affect airway inflammation and hypersensitivity, especially focusing on treatment strategies.
Collapse
Affiliation(s)
- Yaqin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Junjie Dai
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Guibao Zhou
- Department of Pharmacy, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Rongchang Chen
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Chengwen Bai
- Emergency Department, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Fei Shi
- Department of Infectious Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| |
Collapse
|
5
|
Fan GZ, Duan BY, Xin FJ, Qu ZH. Assessment of the bidirectional causal association between Helicobacter pylori infection and allergic diseases by mendelian randomization analysis. Sci Rep 2025; 15:5746. [PMID: 39962134 PMCID: PMC11832746 DOI: 10.1038/s41598-025-89981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
This article employed a bidirectional Mendelian randomization (MR) analysis to deduce the causal relationship between H. pylori infection (Seven H. pylori antibodies: CagA, Catalase, GroEL, IgG, OMP, UREA, and VacA) and allergic diseases. This study primarily employed the Inverse-Variance Weighted (IVW)method, supplemented by MR-Egger regression and the Weighted median (WM) method approach, to comprehensively assess the causal relationship between exposure and outcome. Sensitivity analysis, including Cochran's Q test, MR-Egger regression intercept, MR-PRESSO test, and leave-one-out analysis, verified the reliability of the results. In the forward MR analysis, the IVW analysis outcomes showed the causal relationship existed between the allergic urticaria (AU) and Catalase antibody, allergic asthma (AA) and allergic rhinitis (AR) with OMP antibody, and allergic conjunctivitis (AC) and VacA antibody; in the reverse MR analysis, the results of the IVW analysis revealed that CagA antibody was positively associated with AU. Sensitivity analysis indicated that the causal relationship was robust. Higher levels of Catalase antibody may potentially increase the risk of AU development; increased OMP antibody levels might be associated with a higher risk for AA, yet could potentially be a protective factor against AR; greater VacA antibody levels might possibly decrease the incidence of AC; individuals with AU might have a higher likelihood of exhibiting elevated CagA antibody levels. It is suggested that H. pylori infection could potentially influence the onset and progression of allergic diseases via the "gut-skin", "gut-lung", "gut-nose", and "gut-eye" axis; moreover, skin diseases may potentially impact the gut microbiota imbalance through the "skin-gut" axis.
Collapse
Affiliation(s)
- Guo Zhen Fan
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bo Yang Duan
- Department of Pediatrics, Hefei First People's Hospital, Hefei, China
| | - Fang Jie Xin
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zheng Hai Qu
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
6
|
Xu Y, Wang Z, Li S, Su J, Gao L, Ou J, Lin Z, Luo OJ, Xiao C, Chen G. An in-depth understanding of the role and mechanisms of T cells in immune organ aging and age-related diseases. SCIENCE CHINA. LIFE SCIENCES 2025; 68:328-353. [PMID: 39231902 DOI: 10.1007/s11427-024-2695-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/28/2024] [Indexed: 09/06/2024]
Abstract
T cells play a critical and irreplaceable role in maintaining overall health. However, their functions undergo alterations as individuals age. It is of utmost importance to comprehend the specific characteristics of T-cell aging, as this knowledge is crucial for gaining deeper insights into the pathogenesis of aging-related diseases and developing effective therapeutic strategies. In this review, we have thoroughly examined the existing studies on the characteristics of immune organ aging. Furthermore, we elucidated the changes and potential mechanisms that occur in T cells during the aging process. Additionally, we have discussed the latest research advancements pertaining to T-cell aging-related diseases. These findings provide a fresh perspective for the study of T cells in the context of aging.
Collapse
Affiliation(s)
- Yudai Xu
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zijian Wang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junwen Ou
- Anti Aging Medical Center, Clifford Hospital, Guangzhou, 511495, China
| | - Zhanyi Lin
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| |
Collapse
|
7
|
Zeng X, Wu Z, Pan Y, Ma Y, Chen Y, Zhao Z. Effects of micronutrients and macronutrients on risk of allergic disease in the European population: a Mendelian randomization study. FOOD AGR IMMUNOL 2024; 35. [DOI: 10.1080/09540105.2024.2442369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/10/2024] [Indexed: 01/21/2025] Open
Affiliation(s)
- Xiangyue Zeng
- Department of Gastrointestinal Surgery, the Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Zhimin Wu
- Department of Otorhinolaryngology, Guiyang maternal and child health care hospital, the maternal and child health care hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
| | - Yipeng Pan
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yifei Ma
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
| | - Yi Chen
- Department of Breast and Thyroid Surgery, the Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Zeliang Zhao
- Department of Gastrointestinal Surgery, the Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, People’s Republic of China
| |
Collapse
|
8
|
Ma J, Palmer DJ, Geddes D, Lai CT, Rea A, Prescott SL, D'Vaz N, Stinson LF. Maternal Allergic Disease Phenotype and Infant Birth Season Influence the Human Milk Microbiome. Allergy 2024. [PMID: 39723602 DOI: 10.1111/all.16442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/11/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Early infancy is a critical period for immune development. In addition to being the primary food source during early infancy, human milk also provides multiple bioactive components that shape the infant gut microbiome and immune system and provides a constant source of exposure to maternal microbiota. Given the potential interplay between allergic diseases and the human microbiome, this study aimed to characterise the milk microbiome of allergic mothers. Full-length 16S rRNA gene sequencing was performed on milk samples collected at 3 and 6 months postpartum from 196 women with allergic disease. Multivariate linear mixed models were constructed to identify the maternal, infant, and environmental determinants of the milk microbiome. Human milk microbiome composition and beta diversity varied over time (PERMANOVA R2 = 0.011, p = 0.011). The season of infant birth emerged as the strongest determinant of the microbiome community structure (PERMANOVA R2 = 0.014, p = 0.011) with impacts on five of the most abundant taxa. The milk microbiome also varied according to the type of maternal allergic disease (allergic rhinitis, asthma, atopic dermatitis, and food allergy). Additionally, infant formula exposure reduced the relative abundance of several typical oral taxa in milk. In conclusion, the milk microbiome of allergic mothers was strongly shaped by the season of infant birth, maternal allergic disease phenotype, and infant feeding mode. Maternal allergic disease history and infant season of birth should therefore be considered in future studies of infant and maternal microbiota. Trial Registration: ClinicalTrials.gov identifier: ACTRN12606000281594.
Collapse
Affiliation(s)
- Jie Ma
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Debra J Palmer
- ABREAST Network, Perth, WA, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Donna Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| | - Alethea Rea
- Mathematics and Statistics, Harry Butler Institute, Murdoch University, Murdoch, WA, Australia
| | - Susan L Prescott
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medicine, The University of Western Australia, Perth, WA, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nina D'Vaz
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
- ABREAST Network, Perth, WA, Australia
- UWA Centre for Human Lactation Research and Translation, Perth, WA, Australia
| |
Collapse
|
9
|
Cantón R, De Lucas Ramos P, García-Botella A, García-Lledó A, Hernández-Sampelayo T, Gómez-Pavón J, González Del Castillo J, Martín-Delgado MC, Martín Sánchez FJ, Martínez-Sellés M, Molero García JM, Moreno Guillén S, Rodríguez-Artalejo FJ, Reigadas E, Del Campo R, Serrano S, Ruiz-Galiana J, Bouza E. Human intestinal microbiome: Role in health and disease. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2024; 37:438-453. [PMID: 38978509 PMCID: PMC11578434 DOI: 10.37201/req/056.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
The study of the microbiota and the microbiome, and specifically the intestinal one, has determined great interest due to the possible association of their alterations with numerous diseases. These include entities as diverse as Crohn's disease, autism, diabetes, cancer or situations as prevalent today as obesity. In view of this situation, different recommendations have been performed regarding the use of probiotics, prebiotics, and postbiotics as modulators of the microbiota and the microbiome, seeking both preventive and therapeutic effects, and faecal material transfer (FMT) is proposed as an alternative. The latter has emerged as the only proven beneficial intervention on the intestinal microbiome, specifically in the treatment of recurrent colitis associated with Clostridioides difficile (R-CDI). In the rest of the entities, the lowering of laboratory costs has favored the study of the microbiome, which is resolved by delivering reports with catalogs of microorganisms, metabolites or supposed biomarkers without consensus on their composition associated with healthy or diseased microbiota and the disease. There is still insufficient evidence in any disease for interventions on the microbiome beyond FMT and R-CDI. Multi- and multi-disciplinary work with extensive research and the application of artificial intelligence in this field may shed light on the questions raised currently. Ethical issues must also be resolved in light of possible interventions within the umbrella of personalized medicine.
Collapse
Affiliation(s)
- R Cantón
- Rafael Cantón. Servicio de Microbiología. Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria. CIBER de Enfermedades Infecciosas (CIBERINFEC). Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hou Y, Wang D, Zhou S, Huo C, Chen H, Li F, Ding M, Li H, Zhao H, He J, Da H, Ma Y, Qiang Z, Chen X, Bai C, Cui J, Gao N, Liu Y. Probiotics combined with prebiotics alleviated seasonal allergic rhinitis by altering the composition and metabolic function of intestinal microbiota: a prospective, randomized, double-blind, placebo-controlled clinical trial. Front Immunol 2024; 15:1439830. [PMID: 39555052 PMCID: PMC11563974 DOI: 10.3389/fimmu.2024.1439830] [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: 05/28/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024] Open
Abstract
Background Numerous studies have established that probiotics or prebiotics can relieve the symptoms of allergic rhinitis (AR), but their mechanism of action remain underexplored. This study aimed to observe the clinical efficacy of probiotics combined with prebiotics in seasonal AR patients and explore their underlying mechanisms. Methods We conducted a prospective, randomized, double-blind, placebo-controlled clinical trial. The test group was given probiotics combined with prebiotics, whereas the placebo group was administered simulated preparation for 90 days. Outcome measures included total nasal symptom score (TNSS), visual analog scale, rhinitis quality of life questionnaire, fractional exhaled nitric oxide, and the rate and intensity of Loratadine use. Serum TNF-α, INF-γ, IL-4, IL-17, and IgE levels were measured by enzyme-linked immunosorbent assay. Intestinal microbiota was detected by 16S rRNA gene sequencing and quantitative PCR. Short-chain fatty acids were analyzed by gas chromatography-mass spectrometry. Results 106 participants (N = 53 for both test group and placebo group) completed the study. From baseline to day 91, mean difference between groups (MDBG) in the reduction of TNSS was -1.1 (-2.2, -0.1) (P = 0.04); MDBG in the increment of TNF-α was 7.1 pg/ml (95% CI: 0.8, 13.4, P = 0.03); the INF-γ level was significantly increased (P = 0.01), whereas that of IL-17 (P = 0.005) was significantly decreased in the test group, whilst mean difference within groups was not statistically significant in the placebo group; MDBG in the increment of acetate was 12.4% (95% CI: 7.1%, 17.6%, P <0.001). After the administration of probiotics and prebiotics, the composition and metabolic function of the intestinal microbiota were significantly altered and positively related to the beneficial effect on seasonal AR patients. Conclusion Probiotics combined with prebiotics administered for 90 days significantly attenuated the symptoms of seasonal AR patients, which may related to fluctuations in the composition and metabolic function of the intestinal microbiota and further ameliorating host immunity.
Collapse
Affiliation(s)
- Yangfan Hou
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Shuru Zhou
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Caifang Huo
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Haijuan Chen
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Fangxia Li
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Minjuan Ding
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongxin Li
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongyan Zhao
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Jin He
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Hongju Da
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Yu Ma
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Zhihui Qiang
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Xiushan Chen
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
| | - Cairong Bai
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
- Department of Allergy, Yulin No.2 Hospital, Yulin, China
| | - Jing Cui
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
| | - Na Gao
- Department of Respiratory and Critical Care Medicine, Yulin No.2 Hospital, Yulin, China
- Department of Allergy, Yulin No.2 Hospital, Yulin, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| |
Collapse
|
11
|
Zhou Z, Yang J, Liu Q, Gao J, Ji W. Patho-immunological mechanisms of atopic dermatitis: The role of the three major human microbiomes. Scand J Immunol 2024; 100:e13403. [PMID: 39267301 DOI: 10.1111/sji.13403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/17/2024]
Abstract
Atopic dermatitis (AD) is a genetically predisposed allergic inflammatory dermatosis with chronic, pruritic, and recurrent features. Patients with AD have dry and itchy skin, often accompanied by chronic eczematous lesions, allergic rhinitis, or asthma, which has a considerable impact on their daily lives. With advances in genome sequencing technology, it has been demonstrated that microorganisms are involved in this disease, and the microorganisms associated with AD are attracting considerable research attention. An increasing number of studies conducted in recent years have demonstrated that an imbalanced microbiome in AD patients has substantial impact on disease prognosis, and the causes are closely tied to various immune mechanisms. However, the involvement of microorganisms in the pathogenesis of AD remains poorly understood. In this paper, we review the advances in research on the immunological mechanisms of the skin microbiome, intestinal microbiome, and lung microbiome that are related to AD prognosis and immunotherapy protocols. It is hoped that this approach will lay the foundation for exploring the pathogenesis of and emerging treatments for AD.
Collapse
Affiliation(s)
- Zhaosen Zhou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qin Liu
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jing Gao
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenting Ji
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Dong L, Tang Y, Wen S, He Y, Li F, Deng Y, Tao Z. Fecal Microbiota Transplantation Alleviates Allergic Rhinitis via CD4 + T Cell Modulation Through Gut Microbiota Restoration. Inflammation 2024; 47:1278-1297. [PMID: 38294580 DOI: 10.1007/s10753-024-01975-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: 11/26/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Allergic rhinitis (AR) is an allergic condition of the upper respiratory tract with a complex pathogenesis, including epithelial barrier disruption, immune regulation, and gut microbiota, which is not yet fully understood. Gut microbiota is closely linked to allergic diseases, including AR. Fecal microbiota transplantation (FMT) has recently been recognized as a potentially effective therapy for allergic diseases. However, the efficacy and mechanism of action of FMT in AR remain unknown. Herein, we aimed to observe the implications of gut microbiota on epithelial barrier function and T cell homeostasis, as well as the effect of FMT in AR, using the ovalbumin (OVA)-induced AR mice. The intestinal microbiota of recipient mice was cleared using an antibiotic cocktail; thereafter, FMT was performed. Subsequently, the nasal symptom scores and histopathological features of colon and nasal mucosa tissues of mice were monitored, and serum OVA-sIgE and cytokines of IL-4, IFNγ, IL-17A, and IL-10 cytokine concentrations were examined. Thereafter, tight junction protein and CD4+ T cell-related transcription factor and cytokine expressions were observed in the colon and nasal mucosa, and changes in the expression of PI3K/AKT/mTOR and NFκB signaling pathway were detected by WB assay in each group. Fecal DNA was extracted from the four mice groups for high-throughput 16S rRNA sequencing. FMT ameliorated nasal symptoms and reduced nasal mucosal inflammation in AR mice. Moreover, according to 16S rRNA sequencing, FMT restored the disordered gut microbiota in AR mice. Following FMT, ZO-1 and claudin-1 and Th1/Th2/Th17-related transcription factor and cytokine expressions were upregulated, whereas Treg cell-related Foxp3 and IL-10 expressions were downregulated. Mechanistic studies have revealed that FMT also inhibited PI3K/AKT/mTOR and NF-κB pathway protein phosphorylation in AR mouse tissues. FMT alleviates allergic inflammation in AR by repairing the epithelial barrier and modulating CD4+ T cell balance and exerts anti-inflammatory effects through the PI3K/AKT/mTOR and NF-κB signaling pathways. Moreover, gut microbiota disorders are involved in AR pathogenesis. Disturbed gut microbiota causes an altered immune-inflammatory state in mice and increases susceptibility to AR. This study suggested the regulatory role of the gut-nose axis in the pathogenesis of AR is an emerging field, which provides novel directions and ideas for the treatment of AR.
Collapse
Affiliation(s)
- Lin Dong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yulei Tang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Silu Wen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yan He
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Fen Li
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Yuqin Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
13
|
Benton LD, Lopez-Galvez N, Herman C, Caporaso JG, Cope EK, Rosales C, Gameros M, Lothrop N, Martínez FD, Wright AL, Carr TF, Beamer PI. Environmental and structural factors associated with bacterial diversity in household dust across the Arizona-Sonora border. Sci Rep 2024; 14:12803. [PMID: 38834753 PMCID: PMC11150412 DOI: 10.1038/s41598-024-63356-6] [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: 09/04/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
We previously reported that asthma prevalence was higher in the United States (US) compared to Mexico (MX) (25.8% vs. 8.4%). This investigation assessed differences in microbial dust composition in relation to demographic and housing characteristics on both sides of the US-MX Border. Forty homes were recruited in the US and MX. Home visits collected floor dust and documented occupants' demographics, asthma prevalence, housing structure, and use characteristics. US households were more likely to have inhabitants who reported asthma when compared with MX households (30% vs. 5%) and had significantly different flooring types. The percentage of households on paved roads, with flushing toilets, with piped water and with air conditioning was higher in the US, while dust load was higher in MX. Significant differences exist between countries in the microbial composition of the floor dust. Dust from Mexican homes was enriched with Alishewanella, Paracoccus, Rheinheimera genera and Intrasporangiaceae family. A predictive metagenomics analysis identified 68 significantly differentially abundant functional pathways between US and MX. This study documented multiple structural, environmental, and demographic differences between homes in the US and MX that may contribute to significantly different microbial composition of dust observed in these two countries.
Collapse
Affiliation(s)
- Lauren D Benton
- Department of Pediatrics, Steele Children's Research Center, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA.
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA.
| | - Nicolas Lopez-Galvez
- Mel and Enid Zuckerman College of Public Health, University of Arizona, 1295 N. Martin Ave, PO 245210, Tucson, AZ, 85724, USA
- San Diego State University Research Foundation, San Diego State University, 5250 Campanile Dr, San Diego, CA, 92182, USA
| | - Chloe Herman
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, 1350 S Knoles Dr, Flagstaff, AZ, 86011, USA
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - J Gregory Caporaso
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, 1350 S Knoles Dr, Flagstaff, AZ, 86011, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Emily K Cope
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, 1350 S Knoles Dr, Flagstaff, AZ, 86011, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Cecilia Rosales
- Mel and Enid Zuckerman College of Public Health, University of Arizona, 1295 N. Martin Ave, PO 245210, Tucson, AZ, 85724, USA
| | - Mercedes Gameros
- Mel and Enid Zuckerman College of Public Health, University of Arizona, 1295 N. Martin Ave, PO 245210, Tucson, AZ, 85724, USA
| | - Nathan Lothrop
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA
- Mel and Enid Zuckerman College of Public Health, University of Arizona, 1295 N. Martin Ave, PO 245210, Tucson, AZ, 85724, USA
| | - Fernando D Martínez
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA
| | - Anne L Wright
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA
| | - Tara F Carr
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA
| | - Paloma I Beamer
- Asthma and Airway Disease Research Center, University of Arizona, College of Medicine, University of Arizona Health Sciences, 1501 N. Campbell Avenue, Tucson, AZ, 85724, USA
- Mel and Enid Zuckerman College of Public Health, University of Arizona, 1295 N. Martin Ave, PO 245210, Tucson, AZ, 85724, USA
| |
Collapse
|
14
|
Song X, Ding X, Niu P, Chen T, Yan T. The Associations between Exposure to Multiple Heavy Metals and Total Immunoglobulin E in U.S. Adults. TOXICS 2024; 12:116. [PMID: 38393211 PMCID: PMC10891582 DOI: 10.3390/toxics12020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024]
Abstract
Immunoglobulin E (IgE) is a type of immunoglobulin, and elevated serum total IgE is often present in allergic diseases. Exposure to environmental heavy metals has been markedly linked to allergic diseases, leading to elevated total IgE levels. However, studies concerning the effects of multiple metal exposures on total IgE levels are limited. Therefore, the current study seeks to explore the correlation between heavy-metal co-exposure and total IgE levels based on the National Health and Nutrition Examination Survey (NHANES, 2005-2006). Participants possessed complete data on total IgE levels, 11 urinary metal concentrations and other covariates. The correlations between 11 metals and total IgE levels were analyzed using multiple linear regression, and total IgE levels were a continuous variable. Total IgE levels exceeding 150 kU/L were considered sensitized. Binary logistic regression analyses were employed to assess the correlation between metal exposure and the occurrence of an allergic state. Then, the association between co-exposure to the 11 metals and total IgE levels or the occurrence of sensitization status was further analyzed by Bayesian kernel machine regression (BKMR), a multi-contaminant model. There were 1429 adults with complete data included. Based on the median concentration, molybdenum (Mo) had the highest concentration (46.60 μg/L), followed by cesium (Cs), barium (Ba), lead (Pb), and mercury (Hg). And the median (interquartile range) for total IgE levels was 43.7 (17.3, 126.0) kU/L. Multiple linear regression results showed that Pb was significantly and positively associated with total IgE levels (β = 0.165; 95% CI: 0.046, 0.284). Binary logistic regression showed a significant positive correlation between urinary Pb (OR: 1.258; 95% CI: 1.052, 1.510) and tungsten (W) (OR: 1.251; 95% CI: 1.082, 1.447). Importantly, the BKMR model found a positive correlation between combined-metal exposure and total IgE levels and the occurrence of sensitization status. The mixed heavy-metal exposure was associated with increased total IgE levels, and this association may be driven primarily by the exposure of Pb and W. This study provides new insights into the relationship between heavy-metal exposure and allergic diseases. More research is needed to confirm these findings.
Collapse
Affiliation(s)
- Xin Song
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; (X.S.)
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaowen Ding
- Beijing Institute of Occupational Disease Prevention and Treatment, Beijing 100093, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; (X.S.)
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Tian Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; (X.S.)
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Tenglong Yan
- Beijing Institute of Occupational Disease Prevention and Treatment, Beijing 100093, China
| |
Collapse
|
15
|
Kim K, Jang H, Kim E, Kim H, Sung GY. Recent advances in understanding the role of the skin microbiome in the treatment of atopic dermatitis. Exp Dermatol 2023; 32:2048-2061. [PMID: 37767872 DOI: 10.1111/exd.14940] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023]
Abstract
The skin is the largest organ in the human body, and histologically consists of the epidermis, dermis and subcutaneous tissue. Humans maintain a cooperative symbiotic relationship with their skin microbiota, a complex community of bacteria, fungi and viruses that live on the surface of the skin, and which act as a barrier to protect the body from the inside and outside. The skin is a 'habitat' and vast 'ecosystem' inhabited by countless microbes; as such, relationships have been forged through millions of years of coevolution. It is not surprising then that microbes are key participants in shaping and maintaining essential physiological processes. In addition to maintaining barrier function, the unique symbiotic microbiota that colonizes the skin increases the immune response and provides protection against pathogenic microbes. This review examines our current understanding of skin microbes in shaping and enhancing the skin barrier, as well as skin microbiome-host interactions and their roles in skin diseases, such as atopic dermatitis (AD). We also report on the current status of AD therapeutic drugs that target the skin microbiome, related research on current therapeutic strategies, and the limitations and future considerations of skin microbiome research. In particular, as a future strategy, we discuss the need for a skin-on-a-chip-based microphysiological system research model amenable to biomimetic in vitro studies and human skin equivalent models, including skin appendages.
Collapse
Affiliation(s)
- Kyunghee Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeji Jang
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Eunyul Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeju Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
- Major in Materials Science and Engineering, Hallym University, Chuncheon, Korea
| |
Collapse
|
16
|
Wallen-Russell C, Pearlman N, Wallen-Russell S, Cretoiu D, Thompson DC, Voinea SC. A Catastrophic Biodiversity Loss in the Environment Is Being Replicated on the Skin Microbiome: Is This a Major Contributor to the Chronic Disease Epidemic? Microorganisms 2023; 11:2784. [PMID: 38004795 PMCID: PMC10672968 DOI: 10.3390/microorganisms11112784] [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: 08/30/2023] [Revised: 10/16/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
There has been a catastrophic loss of biodiversity in ecosystems across the world. A similar crisis has been observed in the human gut microbiome, which has been linked to "all human diseases affecting westernized countries". This is of great importance because chronic diseases are the leading cause of death worldwide and make up 90% of America's healthcare costs. Disease development is complex and multifactorial, but there is one part of the body's interlinked ecosystem that is often overlooked in discussions about whole-body health, and that is the skin microbiome. This is despite it being a crucial part of the immune, endocrine, and nervous systems and being continuously exposed to environmental stressors. Here we show that a parallel biodiversity loss of 30-84% has occurred on the skin of people in the developed world compared to our ancestors. Research has shown that dysbiosis of the skin microbiome has been linked to many common skin diseases and, more recently, that it could even play an active role in the development of a growing number of whole-body health problems, such as food allergies, asthma, cardiovascular diseases, and Parkinson's, traditionally thought unrelated to the skin. Damaged skin is now known to induce systemic inflammation, which is involved in many chronic diseases. We highlight that biodiversity loss is not only a common finding in dysbiotic ecosystems but also a type of dysbiosis. As a result, we make the case that biodiversity loss in the skin microbiome is a major contributor to the chronic disease epidemic. The link between biodiversity loss and dysbiosis forms the basis of this paper's focus on the subject. The key to understanding why biodiversity loss creates an unhealthy system could be highlighted by complex physics. We introduce entropy to help understand why biodiversity has been linked with ecosystem health and stability. Meanwhile, we also introduce ecosystems as being governed by "non-linear physics" principles-including chaos theory-which suggests that every individual part of any system is intrinsically linked and implies any disruption to a small part of the system (skin) could have a significant and unknown effect on overall system health (whole-body health). Recognizing the link between ecosystem health and human health allows us to understand how crucial it could be to maintain biodiversity across systems everywhere, from the macro-environment we inhabit right down to our body's microbiome. Further, in-depth research is needed so we can aid in the treatment of chronic diseases and potentially change how we think about our health. With millions of people currently suffering, research to help mitigate the crisis is of vital importance.
Collapse
Affiliation(s)
| | - Nancy Pearlman
- Ecology Center of Southern California, Los Angeles, CA 90035, USA;
| | | | - Dragos Cretoiu
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 011062 Bucharest, Romania
| | - Dana Claudia Thompson
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 011062 Bucharest, Romania
| | - Silviu Cristian Voinea
- Department of Surgical Oncology, Prof. Dr. Al. Trestioreanu Oncology Institute, Carol Davila University of Medicine and Pharmacy, 022328 Bucharest, Romania
| |
Collapse
|
17
|
Liu W, Peng L, Chen L, Wan J, Lou S, Yang T, Shen Z. Skin microbial dysbiosis is a characteristic of systemic drug-related intertriginous and flexural exanthema-like lesions induced by EGFR inhibitor. Heliyon 2023; 9:e21690. [PMID: 38028014 PMCID: PMC10661433 DOI: 10.1016/j.heliyon.2023.e21690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives To investigate the characteristics of the skin microbiome in severe afatinib-induced skin toxicity. Methods Body site-matched skin surface samples were collected from the lesions on seven flexural sites of one lung cancer (Patient 1) with serious systemic drug-related intertriginous and flexural exanthema (SDRIFE)-like toxicity induced by EGFR-TKI and three healthy age/sex matched controls for whole metagenomics sequencing analysis. Lung cancer Patient 1 and Patient 2 were prescribed minocycline and followed up. Results In SDRIFE-like toxicities induced by afatinib, lesion microbiota richness (ACE and Chao1 index: p < 0.001) and diversity (Shannon's and Simpson's diversity indices: p < 0.01) were reduced. Similarly, the beta diversity analysis (R = 1, p = 0.002 for ANOSIM) showed that the apparent difference in the microbiota composition was statistically significant. The microbial taxa composition in the patient showed an increased abundance of pathogenic bacteria and a decreased abundance of commensal bacteria. LEfSe analysis identified strong bacterial pathogenicity in the patient, while healthy controls exhibited enrichment in several pathways that are beneficial for skin commensal bacteria and skin physiology, including key amino acid metabolism, energy/lipid/glycan biosynthesis/metabolism, and cofactors/vitamins biosynthesis. Ultimately, the patients experienced significant improvement with minocycline. Conclusion Microbial dysbiosis is a characteristic of severe SDRIFE-like toxicity induced by afatinib.
Collapse
Affiliation(s)
- Wenqi Liu
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Lu Peng
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ling Chen
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jianji Wan
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuang Lou
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Tingting Yang
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
18
|
Benton L, Lopez-Galvez N, Herman C, Caporaso G, Cope E, Rosales C, Gameros M, Lothrop N, Martínez F, Wright A, Carr T, Beamer P. Environmental and Structural Factors Associated with Bacterial Diversity in Household Dust Across the Arizona-Sonora Border. RESEARCH SQUARE 2023:rs.3.rs-3325336. [PMID: 37841844 PMCID: PMC10571632 DOI: 10.21203/rs.3.rs-3325336/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
We previously reported that asthma prevalence was higher in the United States (US) compared to Mexico (MX) (25.8% vs 8.4%). This investigation assessed differences in microbial dust composition in relation to demographic and housing characteristics on both sides of the US-MX Border. Forty homes were recruited in the US and MX. Home visits collected floor dust and documented occupants' demographics, asthma prevalence, and housing structure and use characteristics. US households were more likely to have inhabitants who reported asthma when compared with MX households (30% vs 5%) and had significantly different flooring types. The percentage of households on paved roads, with flushing toilets, with piped water and with air conditioning was higher in the US, while dust load was higher in MX. Significant differences exist between countries in the microbial composition of the floor dust. Dust from US homes was enriched with Geodermatophilus, whereas dust from Mexican homes was enriched with Alishewanella and Chryseomicrobium. A predictive metagenomics analysis identified 68 significantly differentially abundant functional pathways between US and MX. This study documented multiple structural, environmental, and demographic differences between homes in the US and MX that may contribute to significantly different microbial composition of dust observed in these two countries.
Collapse
|
19
|
Hong M, Tong L, Mehta JS, Ong HS. Impact of Exposomes on Ocular Surface Diseases. Int J Mol Sci 2023; 24:11273. [PMID: 37511032 PMCID: PMC10379833 DOI: 10.3390/ijms241411273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Ocular surface diseases (OSDs) are significant causes of ocular morbidity, and are often associated with chronic inflammation, redness, irritation, discomfort, and pain. In severe OSDs, loss of vision can result from ocular surface failure, characterised by limbal stem cell deficiencies, corneal vascularisation, corneal opacification, and surface keratinisation. External and internal exposomes are measures of environmental factors that individuals are exposed to, and have been increasingly studied for their impact on ocular surface diseases. External exposomes consist of external environmental factors such as dust, pollution, and stress; internal exposomes consist of the surface microbiome, gut microflora, and oxidative stress. Concerning internal exposomes, alterations in the commensal ocular surface microbiome of patients with OSDs are increasingly reported due to advancements in metagenomics using next-generation sequencing. Changes in the microbiome may be a consequence of the underlying disease processes or may have a role in the pathogenesis of OSDs. Understanding the changes in the ocular surface microbiome and the impact of various other exposomes may also help to establish the causative factors underlying ocular surface inflammation and scarring, the hallmarks of OSDs. This review provides a summary of the current evidence on exposomes in various OSDs.
Collapse
Affiliation(s)
- Merrelynn Hong
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
| | - Louis Tong
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
- Ocular Surface Group, Singapore Eye Research Institute, Singapore 169856, Singapore
- Department of Ophthalmology and Visual Science, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jodhbir S Mehta
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
- Department of Ophthalmology and Visual Science, Duke-NUS Medical School, Singapore 169857, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Hon Shing Ong
- Corneal and External Diseases Department, Singapore National Eye Centre, Singapore 168751, Singapore
- Department of Ophthalmology and Visual Science, Duke-NUS Medical School, Singapore 169857, Singapore
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| |
Collapse
|
20
|
Zhang X, Lu C, Li Y, Norbäck D, Murthy P, Sram RJ, Deng Q. Early-life exposure to air pollution associated with food allergy in children: Implications for 'one allergy' concept. ENVIRONMENTAL RESEARCH 2023; 216:114713. [PMID: 36347392 DOI: 10.1016/j.envres.2022.114713] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/23/2022] [Accepted: 10/30/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The rapid increase of food allergy (FA) has become the "second wave" of allergy epidemic and is now a major global public health concern. Mounting evidence indicates that early life exposure to air pollution is associated with the "first wave" of allergy epidemic (including asthma, allergic rhinitis and eczema) in children, but little is known about its association with FA. OBJECTIVES We hypothesize FA has triple exposure pathways, gut-skin-airway, and investigate the effects of airway exposure to outdoor and indoor air pollution on childhood FA. METHODS A cohort study of 2598 preschool children aged 3-6 years old was conducted in Changsha, China. The prevalence of FA was surveyed using a standard questionnaire by International Study of Asthma and Allergies in Childhood (ISAAC). Exposure to indoor air pollution was assessed by four indicators: new furniture, redecoration, mold or dampness, and window condensation. Exposure to outdoor air pollution was evaluated by the concentrations of PM10, SO2 and NO2, which were obtained from the monitored stations. Both prenatal and postnatal exposure windows were considered. The association between exposure to outdoor/indoor air pollution and childhood FA was estimated by multiple logistic regression models using odds ratio (OR) and a 95% confidence interval (CI). RESULTS A total of 14.9% children reported FA. The prevalence was significantly associated with exposure to indoor air pollution, OR (95% CI) = 1.93 (1.35-2.75) for prenatal exposure to mold/dampness and 1.49 (1.07-2.10) and 1.41 (1.04-1.89) respectively for postnatal exposure to new furniture and window condensation. The prevalence of FA was also associated with prenatal and postnatal exposure to outdoor air pollution, particularly the traffic-related air pollutant NO2, with adjusted ORs (95% Cls) respectively 1.24 (1.00-1.54) and 1.38 (1.03-1.85) per interquartile range (IQR) increase. Sensitivity analysis showed that the association between outdoor/indoor air pollution and childhood FA was significant only in young children aged 3-4 years. CONCLUSION Early-life exposure to high levels of outdoor and indoor air pollution in China due to the rapid economic growth and fast urbanization in the past decades may contribute to the rapid increase of food allergy (FA) in children. Our study indicates that, in addition to gut and skin, airway may be a new route of food sensitization. Air pollution leads to the first and second waves of allergy epidemics, suggesting a concept of 'one allergy' disease.
Collapse
Affiliation(s)
- Xin Zhang
- School of Energy Science and Engineering, Central South University, Changsha 410083, China
| | - Chan Lu
- XiangYa School of Public Health, Central South University, Changsha 410078, China
| | - Yuguo Li
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China
| | - Dan Norbäck
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Padmini Murthy
- Department of Public Health, School of Health Sciences and Practice, New York Medical College, Hawthorne NY 10532, USA
| | - Radim J Sram
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Qihong Deng
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
21
|
Lorentz A, Bilotta S, Civelek M. Molecular links between allergy and cancer. Trends Mol Med 2022; 28:1070-1081. [PMID: 35794030 DOI: 10.1016/j.molmed.2022.06.003] [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: 04/21/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 01/21/2023]
Abstract
Epidemiologic studies show both positive and negative associations between allergies and cancer. Allergic diseases may protect against tumorigenesis by promoting the immune surveillance, while carcinogenesis may be promoted through inflammatory responses from allergies. Histamine receptor antagonists are the focus of recent cancer studies because of their promising beneficial effect on tumor development. Also, cytokines, particularly IL-4 or IL-33, IgE as well as allergy-related immune cells such as eosinophils can contribute to tumor growth suppression. Depending on cancer types, cancer therapy may be more beneficial when considering combinatorial immunotherapy. In this review, we give an overview on molecular links between allergies and cancer.
Collapse
Affiliation(s)
- Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany.
| | - Sabrina Bilotta
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany
| | - Mehtap Civelek
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
22
|
Salava A, Salo V, Leppänen J, Lauerma A, Remitz A. Factors associated with severity of atopic dermatitis - a Finnish cross-sectional study. J Eur Acad Dermatol Venereol 2022; 36:2130-2139. [PMID: 35766133 PMCID: PMC9796899 DOI: 10.1111/jdv.18378] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/02/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Severity-associated factors in atopic dermatitis (AD) have focussed on early onset, concomitant atopic diseases, markers of Th2-shifted inflammation and filaggrin mutations. OBJECTIVES To investigate factors associated with severe AD in Finnish patients. METHODS We conducted a single-centre, cross-sectional observational study with 502 AD patients aged 4.79 to 79.90 years (mean 32.08 years). Disease severity was assessed with the Rajka-Langeland severity score and EASI and associated clinical signs were evaluated. Data regarding onset, relatives, atopic and other comorbidities was gathered retrospectively. We investigated total serum IgE-levels, a panel of filaggrin null mutations and functional variants of genes associated with skin barrier defects. RESULTS Factors more frequent in severe AD included early onset (P = 0.004, 95%CI 0.000-0.024), male sex (P = 0.002, 95%CI 0.000-0.11), history of smoking (P = 0.012, 95%CI 0.000-0.024), concomitant asthma (P = 0.001, 95%CI 0.000-0.011), palmar hyperlinearity (P = 0.013, 95%CI 0.014-0.059), hand dermatitis (P = 0.020, 95%CI 0.000-0.029) and history of contact allergy (P = 0.042, 95%CI 0.037-0.096). Body mass indices (P < 0.000, 95%CI 0.000-0.011) and total serum IgE-levels (P < 0.000, 95%CI 0.000-0.011) were higher in severe AD. No differences were observed for allergic rhinitis, allergic conjunctivitis, food allergy, peanut allergy, prick positivity, keratosis pilaris, history of herpes simplex infections, filaggrin null mutations and other gene variants. CONCLUSIONS Severity determinants in Finnish patients seem to be early-onset, male sex, smoking, overweight, concomitant asthma, palmar hyperlinearity, hand dermatitis and high IgE-levels. A sub-typing of patients in relation to confirmed severity determinants may be useful for course prediction, prognosis and targeted AD management.
Collapse
Affiliation(s)
- A. Salava
- Skin and Allergy HospitalHelsinki University HospitalHelsinkiFinland
| | - V. Salo
- Skin and Allergy HospitalHelsinki University HospitalHelsinkiFinland
| | - J. Leppänen
- Skin and Allergy HospitalHelsinki University HospitalHelsinkiFinland
| | - A. Lauerma
- Skin and Allergy HospitalHelsinki University HospitalHelsinkiFinland
| | - A. Remitz
- Skin and Allergy HospitalHelsinki University HospitalHelsinkiFinland
| |
Collapse
|
23
|
Wang Y, Wang H. The emerging role of histone deacetylase 1 in allergic diseases. Front Immunol 2022; 13:1027403. [PMID: 36311721 PMCID: PMC9597694 DOI: 10.3389/fimmu.2022.1027403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Histone deacetylase 1 (HDAC1) is a unique member of the classes I HDACs and helps to regulate acute and chronic adaptation to environmental stimuli such as allergen, stress. Allergic diseases are complex diseases resulting from the effect of multiple genetic and interacting foreign substances. Epigenetics play an important role in both pathological and immunomodulatory conditions of allergic diseases. To be consistent with this role, recent evidence strongly suggests that histone deacetylase 1 (HDAC1) plays a critical role in allergic response. HDAC1 expression is stimulated by allergen and attributes to increase T helper 2 (Th2) cytokine levels, decrease Th1/Th17 cells and anti-inflammatory cytokine Interleukin-10 (IL-10), and TWIK-related potassium channel-1 (Trek-1) expression. This review focuses on the contribution of HDAC1 and the regulatory role in characterizing allergic endotypes with common molecular pathways and understanding allergic multimorbidity relationships, as well as addressing their potential as therapeutic targets for these conditions.
Collapse
Affiliation(s)
| | - Huiying Wang
- Department of Allergy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Radzikowska U, Baerenfaller K, Cornejo‐Garcia JA, Karaaslan C, Barletta E, Sarac BE, Zhakparov D, Villaseñor A, Eguiluz‐Gracia I, Mayorga C, Sokolowska M, Barbas C, Barber D, Ollert M, Chivato T, Agache I, Escribese MM. Omics technologies in allergy and asthma research: An EAACI position paper. Allergy 2022; 77:2888-2908. [PMID: 35713644 PMCID: PMC9796060 DOI: 10.1111/all.15412] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023]
Abstract
Allergic diseases and asthma are heterogenous chronic inflammatory conditions with several distinct complex endotypes. Both environmental and genetic factors can influence the development and progression of allergy. Complex pathogenetic pathways observed in allergic disorders present a challenge in patient management and successful targeted treatment strategies. The increasing availability of high-throughput omics technologies, such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics allows studying biochemical systems and pathophysiological processes underlying allergic responses. Additionally, omics techniques present clinical applicability by functional identification and validation of biomarkers. Therefore, finding molecules or patterns characteristic for distinct immune-inflammatory endotypes, can subsequently influence its development, progression, and treatment. There is a great potential to further increase the effectiveness of single omics approaches by integrating them with other omics, and nonomics data. Systems biology aims to simultaneously and longitudinally understand multiple layers of a complex and multifactorial disease, such as allergy, or asthma by integrating several, separated data sets and generating a complete molecular profile of the condition. With the use of sophisticated biostatistics and machine learning techniques, these approaches provide in-depth insight into individual biological systems and will allow efficient and customized healthcare approaches, called precision medicine. In this EAACI Position Paper, the Task Force "Omics technologies in allergic research" broadly reviewed current advances and applicability of omics techniques in allergic diseases and asthma research, with a focus on methodology and data analysis, aiming to provide researchers (basic and clinical) with a desk reference in the field. The potential of omics strategies in understanding disease pathophysiology and key tools to reach unmet needs in allergy precision medicine, such as successful patients' stratification, accurate disease prognosis, and prediction of treatment efficacy and successful prevention measures are highlighted.
Collapse
Affiliation(s)
- Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland,Christine‐Kühne Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Katja Baerenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland,Swiss Institute of Bioinformatics (SIB)DavosSwitzerland
| | - José Antonio Cornejo‐Garcia
- Research LaboratoryIBIMA, ARADyAL Instituto de Salud Carlos III, Regional University Hospital of Málaga, UMAMálagaSpain
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology SectionFaculty of ScienceHacettepe UniversityAnkaraTurkey
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland,Swiss Institute of Bioinformatics (SIB)DavosSwitzerland
| | - Basak Ezgi Sarac
- Department of Biology, Molecular Biology SectionFaculty of ScienceHacettepe UniversityAnkaraTurkey
| | - Damir Zhakparov
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland,Swiss Institute of Bioinformatics (SIB)DavosSwitzerland
| | - Alma Villaseñor
- Centre for Metabolomics and Bioanalysis (CEMBIO)Department of Chemistry and BiochemistryFacultad de FarmaciaUniversidad San Pablo‐CEU, CEU UniversitiesMadridSpain,Institute of Applied Molecular Medicine Nemesio Diaz (IMMAND)Department of Basic Medical SciencesFacultad de MedicinaUniversidad San Pablo CEU, CEU UniversitiesMadridSpain
| | - Ibon Eguiluz‐Gracia
- Allergy UnitHospital Regional Universitario de MálagaMálagaSpain,Allergy Research GroupInstituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain
| | - Cristobalina Mayorga
- Allergy UnitHospital Regional Universitario de MálagaMálagaSpain,Allergy Research GroupInstituto de Investigación Biomédica de Málaga‐IBIMAMálagaSpain,Andalusian Centre for Nanomedicine and Biotechnology – BIONANDMálagaSpain
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland,Christine‐Kühne Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO)Department of Chemistry and BiochemistryFacultad de FarmaciaUniversidad San Pablo‐CEU, CEU UniversitiesMadridSpain
| | - Domingo Barber
- Institute of Applied Molecular Medicine Nemesio Diaz (IMMAND)Department of Basic Medical SciencesFacultad de MedicinaUniversidad San Pablo CEU, CEU UniversitiesMadridSpain
| | - Markus Ollert
- Department of Infection and ImmunityLuxembourg Institute of HealthyEsch‐sur‐AlzetteLuxembourg,Department of Dermatology and Allergy CenterOdense Research Center for AnaphylaxisOdense University Hospital, University of Southern DenmarkOdenseDenmark
| | - Tomas Chivato
- Institute of Applied Molecular Medicine Nemesio Diaz (IMMAND)Department of Basic Medical SciencesFacultad de MedicinaUniversidad San Pablo CEU, CEU UniversitiesMadridSpain,Department of Clinic Medical SciencesFacultad de MedicinaUniversidad San Pablo CEU, CEU UniversitiesMadridSpain
| | | | - Maria M. Escribese
- Institute of Applied Molecular Medicine Nemesio Diaz (IMMAND)Department of Basic Medical SciencesFacultad de MedicinaUniversidad San Pablo CEU, CEU UniversitiesMadridSpain
| |
Collapse
|
25
|
Yang Y, Qu L, Mijakovic I, Wei Y. Advances in the human skin microbiota and its roles in cutaneous diseases. Microb Cell Fact 2022; 21:176. [PMID: 36038876 PMCID: PMC9422115 DOI: 10.1186/s12934-022-01901-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
Skin is the largest organ in the human body, and the interplay between the environment factors and human skin leads to some skin diseases, such as acne, psoriasis, and atopic dermatitis. As the first line of human immune defense, skin plays significant roles in human health via preventing the invasion of pathogens that is heavily influenced by the skin microbiota. Despite being a challenging niche for microbes, human skin is colonized by diverse commensal microorganisms that shape the skin environment. The skin microbiota can affect human health, and its imbalance and dysbiosis contribute to the skin diseases. This review focuses on the advances in our understanding of skin microbiota and its interaction with human skin. Moreover, the potential roles of microbiota in skin health and diseases are described, and some key species are highlighted. The prevention, diagnosis and treatment strategies for microbe-related skin diseases, such as healthy diets, lifestyles, probiotics and prebiotics, are discussed. Strategies for modulation of skin microbiota using synthetic biology are discussed as an interesting venue for optimization of the skin-microbiota interactions. In summary, this review provides insights into human skin microbiota recovery, the interactions between human skin microbiota and diseases, and the strategies for engineering/rebuilding human skin microbiota.
Collapse
Affiliation(s)
- Yudie Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450051, China
- Laboratory of Synthetic Biology, Zhengzhou University, Zhengzhou, 450051, China
| | - Lingbo Qu
- Laboratory of Synthetic Biology, Zhengzhou University, Zhengzhou, 450051, China
- College of Chemistry, Zhengzhou University, Zhengzhou, China
| | - Ivan Mijakovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Yongjun Wei
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450051, China.
- Laboratory of Synthetic Biology, Zhengzhou University, Zhengzhou, 450051, China.
| |
Collapse
|
26
|
Huang L, Zhang H, Liu Y, Long Y. The Role of Gut and Airway Microbiota in Pulmonary Arterial Hypertension. Front Microbiol 2022; 13:929752. [PMID: 35910623 PMCID: PMC9326471 DOI: 10.3389/fmicb.2022.929752] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe clinical condition that is characterized pathologically by perivascular inflammation and pulmonary vascular remodeling that ultimately leads to right heart failure. However, current treatments focus on controlling vasoconstriction and have little effect on pulmonary vascular remodeling. Better therapies of PAH require a better understanding of its pathogenesis. With advances in sequencing technology, researchers have begun to focus on the role of the human microbiota in disease. Recent studies have shown that the gut and airway microbiota and their metabolites play an important role in the pathogenesis of PAH. In this review, we summarize the current literature on the relationship between the gut and airway microbiota and PAH. We further discuss the key crosstalk between the gut microbiota and the lung associated with PAH, and the potential link between the gut and airway microbiota in the pathogenesis of PAH. In addition, we discuss the potential of using the microbiota as a new target for PAH therapy.
Collapse
Affiliation(s)
- Linlin Huang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Hongdie Zhang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yijun Liu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yang Long
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Yang Long
| |
Collapse
|
27
|
Bellinghausen I, Khatri R, Saloga J. Current Strategies to Modulate Regulatory T Cell Activity in Allergic Inflammation. Front Immunol 2022; 13:912529. [PMID: 35720406 PMCID: PMC9205643 DOI: 10.3389/fimmu.2022.912529] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022] Open
Abstract
Over the past decades, atopic diseases, including allergic rhinitis, asthma, atopic dermatitis, and food allergy, increased strongly worldwide, reaching up to 50% in industrialized countries. These diseases are characterized by a dominating type 2 immune response and reduced numbers of allergen-specific regulatory T (Treg) cells. Conventional allergen-specific immunotherapy is able to tip the balance towards immunoregulation. However, in mouse models of allergy adaptive transfer of Treg cells did not always lead to convincing beneficial results, partially because of limited stability of their regulatory phenotype activity. Besides genetic predisposition, it has become evident that environmental factors like a westernized lifestyle linked to modern sanitized living, the early use of antibiotics, and the consumption of unhealthy foods leads to epithelial barrier defects and dysbiotic microbiota, thereby preventing immune tolerance and favoring the development of allergic diseases. Epigenetic modification of Treg cells has been described as one important mechanism in this context. In this review, we summarize how environmental factors affect the number and function of Treg cells in allergic inflammation and how this knowledge can be exploited in future allergy prevention strategies as well as novel therapeutic approaches.
Collapse
Affiliation(s)
- Iris Bellinghausen
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Rahul Khatri
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Joachim Saloga
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
28
|
Zhang L, Ai T, Xie C, Xia W, Zhang Y, Liao H, Jia L, Fan Y, Xu J. Lower airway microbiome of children with recurrent wheezing: a clinical cohort study. Transl Pediatr 2022; 11:696-705. [PMID: 35685081 PMCID: PMC9173878 DOI: 10.21037/tp-22-165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/07/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Wheezing is one of the most common respiratory symptoms in childhood especially in infants. In recent years, the incidence of recurrent wheezing is on the rise worldwide. To investigate the lower airway microbiota in patients with recurrent wheezing and provide insights into clinical diagnosis and treatment. METHODS This study initially enrolled 45 hospitalised children with recurrent wheezing symptoms awaiting complete fiberoptic bronchoscopy. Of these, 13 children with tracheobronchomalacia were excluded. The final population included 32 participants (group A). The control group comprised 23 children who inhaled a foreign body and were admitted to the hospital for fiberoptic bronchoscopy within 24 hours (group B). Deoxyribonucleic acid (DNA) was extracted from the bronchoalveolar lavage fluid (BALF) and amplified for the 16S ribosomal Ribonucleic Acid (rRNA) gene, and sequencing of the microbiome was performed using the Illumina Nova Seq 6000 system. RESULTS There were significant differences in the gestational duration (P=0.0458), mode of delivery (P=0.0261), and allergy status (P=0.0000) between groups A and B, but they had similar richness (P=0.8574). There was also a marked difference in the diversity of flora composition between the two groups (P=0.0095). The three most common phyla of microbiota in the two groups were Proteobacteria, Firmicutes, and Bacteroidetes. Species with notably different phyla included Proteobacteria, Bacteroidota, Fusobacteriota, and Acidobacteriota. There was a significant enrichment in the of Proteobacteria and lower levels of Bacteroidota, Fusobacteriota, and Acidobacteriota in group A compared to that in group B. CONCLUSIONS Significant changes occur in the lower airway microbiota during recurrent wheezing in children. The discovery of beneficial airway bacteria may facilitate the prevention and treatment of recurrent wheezing or asthma in children.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Ai
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Cheng Xie
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wanmin Xia
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Zhang
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huling Liao
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liangqin Jia
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yinghong Fan
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jia Xu
- Department of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
29
|
Song Y, Hou J, Kwok JSL, Weng H, Tang MF, Wang MH, Leung ASY, Tao KP, Wong GWK, Chan RWY, Tsui SKW, Leung TF. Whole-Genome Shotgun Sequencing for Nasopharyngeal Microbiome in Pre-school Children With Recurrent Wheezing. Front Microbiol 2022; 12:792556. [PMID: 35250904 PMCID: PMC8889122 DOI: 10.3389/fmicb.2021.792556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
Microbiome mediates early life immune deviation in asthma development. Recurrent wheeze (RW) in pre-school years is a risk factor for asthma diagnosis in school-age children. Dysbiosis exists in asthmatic airways, while its origin in pre-school years and relationship to RW is not clearly defined. This study investigated metagenomics of nasopharyngeal microbiome in pre-school children with RW. We applied whole-genome shotgun sequencing and human rhinovirus (HRV) detection on nasopharyngeal samples collected from three groups of pre-school children: (i) RW group: 16 children at-risk for asthma who were hospitalized for RW, (ii) inpatient control (IC): 18 subjects admitted for upper respiratory infection, and (iii) community control (CC): 36 children without respiratory syndromes. Sequence reads were analyzed by MetaPhlAn2 and HUMAnN2 algorithm for taxonomic and functional identification. Linear discriminant analysis effect size (LEfSe) analysis was used to identify discriminative features. We identified that Moraxella catarrhalis and Dolosigranulum pigrum were predominant species in nasopharynx. RW had lower alpha diversity (Shannon diversity index) than CC (0.48 vs. 1.07; Padj = 0.039), characterized by predominant Proteobacteria. LEfSe analysis revealed D. pigrum was the only discriminative species across groups (LDA = 5.57, P = 0.002), with its relative abundance in RW, IC, and CC being 9.6, 14.2, and 37.3%, respectively (P < 0.05). LEfSe identified five (ribo)nucleotides biosynthesis pathways to be group discriminating. Adjusting for HRV status, pre-school children with RW have lower nasopharyngeal biodiversity, which is associated with Proteobacteria predominance and lower abundance of D. pigrum. Along with discriminative pathways found in RW and CC, these microbial biomarkers help to understand RW pathogenesis.
Collapse
Affiliation(s)
- Yuping Song
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Jinpao Hou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Jamie Sui Lam Kwok
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Haoyi Weng
- Jockey Club School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Man Fung Tang
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Maggie Haitian Wang
- Jockey Club School of Public Health and Primary Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Agnes Sze Yin Leung
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Kin Pong Tao
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Gary Wing Kin Wong
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Renee Wan Yi Chan
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Stephen Kwok Wing Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Ting Fan Leung
- Department of Pediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.,The Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| |
Collapse
|
30
|
Selway CA, Sudarpa J, Weyrich LS. Moving beyond the gut microbiome: combining systems biology and multi-site microbiome analyses to combat non-communicable diseases. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
31
|
Zhou Y, Zhao H, Wang T, Zhao X, Wang J, Wang Q. Anti-Inflammatory and Anti-asthmatic Effects of TMDCT Decoction in Eosinophilic Asthma Through Treg/Th17 Balance. Front Pharmacol 2022; 13:819728. [PMID: 35211018 PMCID: PMC8861319 DOI: 10.3389/fphar.2022.819728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/24/2022] [Indexed: 12/28/2022] Open
Abstract
Tuo-Min-Ding-Chuan decoction (TMDCT) is a Traditional Chinese Medicine (TCM) formula consisting of twelve herbs that can relieve the symptoms and treat allergic asthma. Yet, the underlying mechanism of action is still unclear. In this study, we investigated the effect of TMDCT in regulating Treg/Th17 cells immune balance and explored potential metabolic and gut biomarkers associated with Treg and Th17 cells in eosinophilic asthma mice treated by TMDCT. We found that TMDCT increases Treg cells percentage and decreases Th17 cells percentage in the ovalbumin (OVA) -induced eosinophilic asthma mice model. Furthermore, Imidazoleacetic acid, dL-glutamine, L-pyroglutamic acid, 2-deoxy-d-glucose were preliminary identified as biomarkers in plasma metabolites treated by TMDCT, meanwhile genus Desulfovibrio, genus Butyricimonas and genus Prevotella 9 were preliminary identified as gut microbiota biomarkers after TMDCT treatment. These results provide an experimental foundation for the treatment of allergic asthma with Chinese herbal compounds.
Collapse
Affiliation(s)
- Yumei Zhou
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Haihong Zhao
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhao
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ji Wang
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Characterization of Conjunctival Sac Microbiome from Patients with Allergic Conjunctivitis. J Clin Med 2022; 11:jcm11041130. [PMID: 35207407 PMCID: PMC8875969 DOI: 10.3390/jcm11041130] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Conjunctival sac microbiome alterations have been reported to be closely associated with many ocular diseases. However, the characteristic of conjunctival sac microbiome in allergic conjunctivitis (AC) was scarcely described. In this study, we aimed to identify the differences of the conjunctival sac microbiome composition in AC patients compared with normal controls (NCs) using high-throughput 16S rDNA sequencing metagenomic analysis. The conjunctival sac microbiome samples from 28 AC patients and 39 NC patients were collected. The V3-V4 region of 16S rRNA gene high-throughput sequencing was performed on the illumina MiSeq platform. Alpha diversity, beta diversity and the relative abundance at the phylum and genus levels were analyzed using QIIME. Alpha diversity demonstrated by Chao1, Observed_species and PD_whole_tree indexes did not show significant difference between the AC and NC groups, while the Shannon index was higher in the AC group. Beta diversity showed divergent microbiome composition in different groups (p < 0.005). The top five abundant phyla were Firmicutes, Proteobacteria, Actinobacteriota, Bacteroidota and Cyanobacteria in both groups. The top five abundant genera were Bacillus, Staphylococcus, Corynebacterium, Acinetobacter and Ralstonia in the AC group and Acinetobacter, Staphylococcus, Bacillus, Clostridium_sensu_stricto_1, Corynebacterium and Geobacillus in the NC group. The Firmicutes/Bacteroidetes (F/B) ratio at the phylum level was similar between groups (p = 0.144). The Bacillus/Acinetobacter (B/A) ratio at the genus level was higher in the AC group (p = 0.021). The dysbiosis detected in this study might provide further evidence to investigate the mechanism and treatment methods for allergic conjunctivitis.
Collapse
|
33
|
Vitte J, Vibhushan S, Bratti M, Montero-Hernandez JE, Blank U. Allergy, Anaphylaxis, and Nonallergic Hypersensitivity: IgE, Mast Cells, and Beyond. Med Princ Pract 2022; 31:501-515. [PMID: 36219943 PMCID: PMC9841766 DOI: 10.1159/000527481] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/06/2022] [Indexed: 01/20/2023] Open
Abstract
IgE-mediated type I hypersensitivity reactions have many reported beneficial functions in immune defense against parasites, venoms, toxins, etc. However, they are best known for their role in allergies, currently affecting almost one third of the population worldwide. IgE-mediated allergic diseases result from a maladaptive type 2 immune response that promotes the synthesis of IgE antibodies directed at a special class of antigens called allergens. IgE antibodies bind to type I high-affinity IgE receptors (FcεRI) on mast cells and basophils, sensitizing them to get triggered in a subsequent encounter with the cognate allergen. This promotes the release of a large variety of inflammatory mediators including histamine responsible for the symptoms of immediate hypersensitivity. The development of type 2-driven allergies is dependent on a complex interplay of genetic and environmental factors at barrier surfaces including the host microbiome that builds up during early life. While IgE-mediated immediate hypersensitivity reactions are undoubtedly at the origin of the majority of allergies, it has become clear that similar responses and symptoms can be triggered by other types of adaptive immune responses mediated via IgG or complement involving other immune cells and mediators. Likewise, various nonadaptive innate triggers via receptors expressed on mast cells have been found to either directly launch a hypersensitivity reaction and/or to amplify existing IgE-mediated responses. This review summarizes recent findings on both IgE-dependent and IgE-independent mechanisms in the development of allergic hypersensitivities and provides an update on the diagnosis of allergy.
Collapse
Affiliation(s)
- Joana Vitte
- Aix-Marseille Université, IRD, APHM, MEPHI, Marseille, France
- IHU Méditerranée Infection, Marseille, France
- IDESP, INSERM UMR UA 11, Montpellier, France
| | - Shamila Vibhushan
- Université Paris Cité - Centre de Recherche sur l'Inflammation, INSERM UMRS 1149, CNRS EMR8252, Laboratoire d'Excellence Inflamex, Paris, France
| | - Manuela Bratti
- Université Paris Cité - Centre de Recherche sur l'Inflammation, INSERM UMRS 1149, CNRS EMR8252, Laboratoire d'Excellence Inflamex, Paris, France
| | - Juan Eduardo Montero-Hernandez
- Université Paris Cité - Centre de Recherche sur l'Inflammation, INSERM UMRS 1149, CNRS EMR8252, Laboratoire d'Excellence Inflamex, Paris, France
| | - Ulrich Blank
- Université Paris Cité - Centre de Recherche sur l'Inflammation, INSERM UMRS 1149, CNRS EMR8252, Laboratoire d'Excellence Inflamex, Paris, France
- *Ulrich Blank,
| |
Collapse
|
34
|
Parrish A, Boudaud M, Kuehn A, Ollert M, Desai MS. Intestinal mucus barrier: a missing piece of the puzzle in food allergy. Trends Mol Med 2021; 28:36-50. [PMID: 34810087 DOI: 10.1016/j.molmed.2021.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of food allergies has reached epidemic levels but the cause remains largely unknown. We discuss the clinical relevance of the gut mucosal barrier as a site for allergic sensitization to food. In this context, we focus on an important but overlooked part of the mucosal barrier in pathogenesis, the glycoprotein-rich mucus layer, and call attention to both beneficial and detrimental aspects of mucus-gut microbiome interactions. Studying the intricate links between the mucus barrier, the associated bacteria, and the mucosal immune system may advance our understanding of the mechanisms and inform prevention and treatment strategies in food allergy.
Collapse
Affiliation(s)
- Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark.
| |
Collapse
|
35
|
Suaini NHA, Siah KTH, Tham EH. Role of the gut-skin axis in IgE-mediated food allergy and atopic diseases. Curr Opin Gastroenterol 2021; 37:557-564. [PMID: 34411036 DOI: 10.1097/mog.0000000000000780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW In recent years, landmark clinical trials investigating the role of early oral exposure to food antigens for food allergy (FA) prevention have highlighted the importance of immunoregulatory pathways in the 'gut-skin axis'. This review highlights recent literature on the mechanisms of the immune system and microbiome involved in the gut-skin axis, contributing to the development of atopic dermatitis (AD), FA, allergic rhinitis (AR) and asthma. Therapeutic interventions harnessing the gut-skin axis are also discussed. RECENT FINDINGS Epicutaneous sensitization in the presence of AD is capable of inducing Th2 allergic inflammation in the intestinal tract and lower respiratory airways, predisposing one to the development of AR and asthma. Probiotics have demonstrated positive effects in preventing and treating AD, though there is no evident relationship of its beneficial effects on other allergic diseases. Prophylactic skin emollients use has not shown consistent protection against AD, whereas there is some evidence for the role of dietary changes in alleviating AD and airway inflammation. More randomized controlled trials are needed to clarify the potential of epicutaneous immunotherapy as a therapeutic strategy for patients with FA. SUMMARY The growing understanding of the gut-skin interactions on allergic disease pathogenesis presents novel avenues for therapeutic interventions which target modulation of the gut and/or skin.
Collapse
Affiliation(s)
| | - Kewin Tien Ho Siah
- Division of Gastroenterology & Hepatology, University Medicine Cluster, National University Hospital
- Department of Medicine, Yong Loo Lin School of Medicine
| | - Elizabeth Huiwen Tham
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A STAR)
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS)
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System (NUHS)
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
36
|
Gallegos-Alcalá P, Jiménez M, Cervantes-García D, Salinas E. The Keratinocyte as a Crucial Cell in the Predisposition, Onset, Progression, Therapy and Study of the Atopic Dermatitis. Int J Mol Sci 2021; 22:ijms221910661. [PMID: 34639001 PMCID: PMC8509070 DOI: 10.3390/ijms221910661] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
The keratinocyte (KC) is the main functional and structural component of the epidermis, the most external layer of the skin that is highly specialized in defense against external agents, prevention of leakage of body fluids and retention of internal water within the cells. Altered epidermal barrier and aberrant KC differentiation are involved in the pathophysiology of several skin diseases, such as atopic dermatitis (AD). AD is a chronic inflammatory disease characterized by cutaneous and systemic immune dysregulation and skin microbiota dysbiosis. Nevertheless, the pathological mechanisms of this complex disease remain largely unknown. In this review, we summarize current knowledge about the participation of the KC in different aspects of the AD. We provide an overview of the genetic predisposing and environmental factors, inflammatory molecules and signaling pathways of the KC that participate in the physiopathology of the AD. We also analyze the link among the KC, the microbiota and the inflammatory response underlying acute and chronic skin AD lesions.
Collapse
Affiliation(s)
- Pamela Gallegos-Alcalá
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Mariela Jiménez
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Daniel Cervantes-García
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- National Council of Science and Technology, Ciudad de México 03940, Mexico
| | - Eva Salinas
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- Correspondence: ; Tel.: +52-449-9108424
| |
Collapse
|
37
|
Pariser DN, Hilt ZT, Ture SK, Blick-Nitko SK, Looney MR, Cleary SJ, Roman-Pagan E, Saunders J, Georas SN, Veazey J, Madere F, Santos LT, Arne A, Huynh NP, Livada AC, Guerrero-Martin SM, Lyons C, Metcalf-Pate KA, McGrath KE, Palis J, Morrell CN. Lung megakaryocytes are immune modulatory cells. J Clin Invest 2021; 131:137377. [PMID: 33079726 DOI: 10.1172/jci137377] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023] Open
Abstract
Although platelets are the cellular mediators of thrombosis, they are also immune cells. Platelets interact both directly and indirectly with immune cells, impacting their activation and differentiation, as well as all phases of the immune response. Megakaryocytes (Mks) are the cell source of circulating platelets, and until recently Mks were typically only considered bone marrow-resident (BM-resident) cells. However, platelet-producing Mks also reside in the lung, and lung Mks express greater levels of immune molecules compared with BM Mks. We therefore sought to define the immune functions of lung Mks. Using single-cell RNA sequencing of BM and lung myeloid-enriched cells, we found that lung Mks, which we term MkL, had gene expression patterns that are similar to antigen-presenting cells. This was confirmed using imaging and conventional flow cytometry. The immune phenotype of Mks was plastic and driven by the tissue immune environment, as evidenced by BM Mks having an MkL-like phenotype under the influence of pathogen receptor challenge and lung-associated immune molecules, such as IL-33. Our in vitro and in vivo assays demonstrated that MkL internalized and processed both antigenic proteins and bacterial pathogens. Furthermore, MkL induced CD4+ T cell activation in an MHC II-dependent manner both in vitro and in vivo. These data indicated that MkL had key immune regulatory roles dictated in part by the tissue environment.
Collapse
Affiliation(s)
- Daphne N Pariser
- Aab Cardiovascular Research Institute and.,Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | | - Mark R Looney
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Simon J Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Jerry Saunders
- Center for Pediatric Biomedical Research, Department of Pediatrics, and
| | - Steve N Georas
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Janelle Veazey
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Ferralita Madere
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Laura Tesoro Santos
- Cardiovascular Research Department, University Hospital Ramón y Cajal Biotechnology, Medicine and Health Sciences PhD Program, University Francisco de Vitoria, Madrid, Spain
| | | | - Nguyen Pt Huynh
- Genomics Research Center, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Alison C Livada
- Aab Cardiovascular Research Institute and.,Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Selena M Guerrero-Martin
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Claire Lyons
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelly A Metcalf-Pate
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - James Palis
- Center for Pediatric Biomedical Research, Department of Pediatrics, and
| | - Craig N Morrell
- Aab Cardiovascular Research Institute and.,Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.,Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
38
|
Hsieh CS, Rengarajan S, Kau A, Tarazona-Meza C, Nicholson A, Checkley W, Romero K, Hansel NN. Altered IgA Response to Gut Bacteria Is Associated with Childhood Asthma in Peru. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:398-407. [PMID: 34193598 PMCID: PMC8516662 DOI: 10.4049/jimmunol.2001296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/28/2021] [Indexed: 12/24/2022]
Abstract
Alterations in gut microbiota in early life have been associated with the development of asthma; however, the role of gut bacteria or the IgA response to gut bacteria in school-aged children with asthma is unclear. To address this question, we profiled the microbial populations in fecal and nasal swab samples by 16S rRNA sequencing from 40 asthma and 40 control children aged 9-17 y from Peru. Clinical history and laboratory evaluation of asthma and allergy were obtained. Fecal samples were analyzed by flow cytometry and sorted into IgA+ and IgA- subsets for 16S rRNA sequencing. We found that the fecal or nasal microbial 16S rRNA diversity and frequency of IgA+ fecal bacteria did not differ between children with or without asthma. However, the α diversity of fecal IgA+ bacteria was decreased in asthma compared with control. Machine learning analysis of fecal bacterial IgA-enrichment data revealed loss of IgA binding to the Blautia, Ruminococcus, and Lachnospiraceae taxa in children with asthma compared with controls. In addition, this loss of IgA binding was associated with worse asthma control (Asthma Control Test) and increased odds of severe as opposed to mild to moderate asthma. Thus, despite little to no change in the microbiota, children with asthma exhibit an altered host IgA response to gut bacteria compared with control participants. Notably, the signature of altered IgA responses is loss of IgA binding, in particular to members of Clostridia spp., which is associated with greater severity of asthma.
Collapse
Affiliation(s)
- Chyi-Song Hsieh
- Division of Rheumatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO;
| | - Sunaina Rengarajan
- Division of Rheumatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Andrew Kau
- Division of Allergy and Immunology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Carla Tarazona-Meza
- Asociacion Benefica Prisma, PRISMA, Lima, Peru
- Center for Global Non-Communicable Disease Research and Training, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew Nicholson
- Program in Global Disease Epidemiology and Control, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and
| | - William Checkley
- Center for Global Non-Communicable Disease Research and Training, Johns Hopkins University School of Medicine, Baltimore, MD
- Program in Global Disease Epidemiology and Control, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Karina Romero
- Asociacion Benefica Prisma, PRISMA, Lima, Peru
- Center for Global Non-Communicable Disease Research and Training, Johns Hopkins University School of Medicine, Baltimore, MD
- Program in Global Disease Epidemiology and Control, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
39
|
Mahdavinia M, Greenfield LR, Moore D, Botha M, Engen P, Gray C, Lunjani N, Hlela C, Basera W, Hobane L, Watkins A, Mankahla A, Gaunt B, Facey-Thomas H, Landay A, Keshavarzian A, Levin ME. House dust microbiota and atopic dermatitis; effect of urbanization. Pediatr Allergy Immunol 2021; 32:1006-1012. [PMID: 33570236 DOI: 10.1111/pai.13471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Previous studies have shown that a child's risk of developing atopic disease is impacted by both genetic and environmental factors. Because small children spend the majority of their time in their homes, exposure to microbial factors in their home environment may be protective or risk factors for development of atopic diseases, such as atopic dermatitis. METHODS Dust samples from the homes of 86 Black South African children 12 to 36 months old were collected for analysis of the bacterial microbiome. This case-control study design included children with and without atopic dermatitis from rural and urban environments. RESULTS Significant differences in bacterial composition and diversity were found when comparing children with and without atopic dermatitis. Furthermore, house dust microbiota was significantly different in rural and urban areas. Differences were best accounted for by higher relative abundance of Ruminococcaceae, Lachnospiraceae, and Bacteroidaceae families in rural compared with urban houses. Levels of Ruminococcaceae were also found to be significantly depleted in the house dust of rural children with atopic dermatitis as compared to control children. CONCLUSIONS House dust composition may be an important risk factor for the development of atopic disease, and this association may be driven in part by the gut microbiome. Low levels of the Ruminococcaceae family from Clostridia class in particular may explain the association between urban living and atopy. However, further research is needed to elucidate these links.
Collapse
Affiliation(s)
- Mahboobeh Mahdavinia
- Department of Internal Medicine, Allergy and Immunology Division, Rush University Medical Center, Chicago, IL, USA.,InVivo Planetary Health Network, Chicago, IL, USA
| | - Leah R Greenfield
- Department of Internal Medicine, Allergy and Immunology Division, Rush University Medical Center, Chicago, IL, USA.,Rush Medical College, Chicago, IL, USA
| | - Donyea Moore
- Department of Internal Medicine, Allergy and Immunology Division, Rush University Medical Center, Chicago, IL, USA
| | - Maresa Botha
- InVivo Planetary Health Network, Chicago, IL, USA.,Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | | | - Claudia Gray
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa.,Department of Dermatology, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla Lunjani
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Carol Hlela
- Department of Dermatology, University of Cape Town, Cape Town, South Africa
| | - Wisdom Basera
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Lelani Hobane
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Alexandra Watkins
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Avumile Mankahla
- Eastern Cape Department of Health, Zithulele Hospital, Mqanduli, South Africa
| | - Ben Gaunt
- Division of Dermatology, Department of Medicine and Pharmacology, Walter Sisulu University, Mthatha, South Africa
| | - Heidi Facey-Thomas
- Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Alan Landay
- Geriatrics Division, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, USA
| | - Michael E Levin
- InVivo Planetary Health Network, Chicago, IL, USA.,Division of Allergy, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
40
|
Dental Biofilm and Saliva Microbiome and Its Interplay with Pediatric Allergies. Microorganisms 2021; 9:microorganisms9061330. [PMID: 34207425 PMCID: PMC8235788 DOI: 10.3390/microorganisms9061330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
Little is known about the interplay and contribution of oral microorganisms to allergic diseases, especially in children. The aim of the clinical study was to associate saliva and dental biofilm microbiome with allergic disease, in particular with allergic asthma. In a single-center study, allergic/asthmatic children (n = 15; AA-Chd; age 10.7 ± 2.9), atopic/allergic children (n = 16; AT/AL-Chd; 11.3 ± 2.9), and healthy controls (n = 15; CON-Chd; age 9.9 ± 2.2) were recruited. After removing adhering biofilms from teeth and collecting saliva, microbiome was analyzed by using a 16s-rRNA gene-based next-generation sequencing in these two mediums. Microbiome structure differed significantly between saliva and dental biofilms (β-diversity). Within the groups, the dental biofilm microbiome of AA-Chd and AT/AL-Chd showed a similar microbial fingerprint characterized by only a small number of taxa that were enriched or depleted (4) compared to the CON-Chd, while both diseased groups showed a stronger microbial shift compared to CON-Chd, revealing 14 taxa in AA-Chd and 15 taxa in AT/AL-Chd that were different. This could be the first note to the contribution of dental biofilm and its metabolic activity to allergic health or disease.
Collapse
|
41
|
Fang Z, Li L, Lu W, Zhao J, Zhang H, Lee YK, Chen W. Bifidobacterium affected the correlation between gut microbial composition, SCFA metabolism, and immunity in mice with DNFB-induced atopic dermatitis. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
42
|
The microbiome in atopic patients and potential modifications in the context of the severe acute respiratory syndrome coronavirus 2 pandemic. Curr Opin Allergy Clin Immunol 2021; 21:245-251. [PMID: 33769313 DOI: 10.1097/aci.0000000000000738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Data regarding the effects of coronavirus disease 2019 (COVID-19) on host-microbiome alteration and subsequent effects on susceptibility and clinical course of COVID-19, especially in atopic patients, are currently limited. Here, we review the studies regarding the microbiome of atopic patients with other respiratory infections and discuss the potential role of probiotics as therapeutic targets for COVID-19 to decrease its susceptibility and severity of COVID-19. RECENT FINDINGS Respiratory tract virus infection affects the gut and airway microbiome structures and host's immune function. Diverse factors in atopic diseases affect the airway and gut microbiome structures, which are expected to negatively influence host health. However, response to respiratory virus infection in atopic hosts depends on the preexisting microbiome and immune responses. This may explain the inconclusiveness of the effects of COVID-19 on the susceptibility, morbidity, and mortality of patients with atopic diseases. Beneficial probiotics may be a therapeutic adjuvant in COVID-19 infection as the beneficial microbiome can decrease the viral load in the early phase of respiratory virus infection and improve the morbidity and mortality. SUMMARY Application of probiotics can be a potential adjuvant treatment in respiratory virus infection to improve host immune responses and disturbed microbiome structures in atopic patients. Further related studies involving COVID-19 are warranted in near future.
Collapse
|
43
|
Paparo L, Nocerino R, Ciaglia E, Di Scala C, De Caro C, Russo R, Trinchese G, Aitoro R, Amoroso A, Bruno C, Di Costanzo M, Passariello A, Messina F, Agangi A, Napolitano M, Voto L, Gatta GD, Pisapia L, Montella F, Mollica MP, Calignano A, Puca A, Berni Canani R. Butyrate as a bioactive human milk protective component against food allergy. Allergy 2021; 76:1398-1415. [PMID: 33043467 PMCID: PMC8247419 DOI: 10.1111/all.14625] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/31/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Food allergy (FA) is a growing health problem worldwide. Effective strategies are advocated to limit the disease burden. Human milk (HM) could be considered as a protective factor against FA, but its mechanisms remain unclear. Butyrate is a gut microbiota-derived metabolite able to exert several immunomodulatory functions. We aimed to define the butyrate concentration in HM, and to see whether the butyrate concentration detected in HM is able to modulate the mechanisms of immune tolerance. METHODS HM butyrate concentration from 109 healthy women was assessed by GS-MS. The effect of HM butyrate on tolerogenic mechanisms was assessed in in vivo and in vitro models. RESULTS The median butyrate concentration in mature HM was 0.75 mM. This butyrate concentration was responsible for the maximum modulatory effects observed in all experimental models evaluated in this study. Data from mouse model show that in basal condition, butyrate up-regulated the expression of several biomarkers of gut barrier integrity, and of tolerogenic cytokines. Pretreatment with butyrate significantly reduced allergic response in three animal models of FA, with a stimulation of tolerogenic cytokines, inhibition of Th2 cytokines production and a modulation of oxidative stress. Data from human cell models show that butyrate stimulated human beta defensin-3, mucus components and tight junctions expression in human enterocytes, and IL-10, IFN-γ and FoxP3 expression through epigenetic mechanisms in PBMCs from FA children. Furthermore, it promoted the precursors of M2 macrophages, DCs and regulatory T cells. CONCLUSION The study's findings suggest the importance of butyrate as a pivotal HM compound able to protect against FA.
Collapse
Affiliation(s)
- Lorella Paparo
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
- European Laboratory for the Investigation of Food‐Induced Diseases University of Naples Federico II Naples Italy
| | - Rita Nocerino
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno Fisciano Italy
| | - Carmen Di Scala
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Carmen De Caro
- Department of Pharmacy University of Naples Federico II Naples Italy
| | - Roberto Russo
- Department of Pharmacy University of Naples Federico II Naples Italy
| | | | - Rosita Aitoro
- Department of Translational Medical Science University of Naples Federico II Naples Italy
| | - Antonio Amoroso
- Department of Translational Medical Science University of Naples Federico II Naples Italy
| | - Cristina Bruno
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Margherita Di Costanzo
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Annalisa Passariello
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- Department of Pediatric Cardiology Monaldi Hospital Naples Italy
| | - Francesco Messina
- Neonatal Intensive Care Unit "Betania" Evangelical Hospital Naples Italy
| | - Annalisa Agangi
- Neonatal Intensive Care Unit "Betania" Evangelical Hospital Naples Italy
| | | | - Luana Voto
- Department of Translational Medical Science University of Naples Federico II Naples Italy
| | - Giusy Della Gatta
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Laura Pisapia
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
| | - Francesco Montella
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana" University of Salerno Fisciano Italy
| | | | - Antonio Calignano
- Department of Pharmacy University of Naples Federico II Naples Italy
| | - Annibale Puca
- European Laboratory for the Investigation of Food‐Induced Diseases University of Naples Federico II Naples Italy
- Cardiovascular Research Unit IRCCS MultiMedica Milan Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science University of Naples Federico II Naples Italy
- ImmunoNutritionLab at the CEINGE‐Biotecnologie Avanzate s.c.ar.l Research Center University of Naples Federico II Naples Italy
- European Laboratory for the Investigation of Food‐Induced Diseases University of Naples Federico II Naples Italy
- Task Force for Microbiome Studies University of Naples Federico II Naples Italy
| |
Collapse
|
44
|
Hu C, van Meel ER, Medina-Gomez C, Kraaij R, Barroso M, Kiefte-de Jong J, Radjabzadeh D, Pasmans SGMA, de Jong NW, de Jongste JC, Moll HA, Nijsten T, Rivadeneira F, Pardo LM, Duijts L. A population-based study on associations of stool microbiota with atopic diseases in school-age children. J Allergy Clin Immunol 2021; 148:612-620. [PMID: 33862008 DOI: 10.1016/j.jaci.2021.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/27/2021] [Accepted: 04/01/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Infants with less diverse gut microbiota seem to have higher risks of atopic diseases in early life, but any associations at school age are unclear. OBJECTIVES This study sought to examine the associations of diversity, relative abundance, and functional pathways of stool microbiota with atopic diseases in school-age children. METHODS We performed a cross-sectional study within an existing population-based prospective cohort among 1440 children 10 years of age. On stool samples, 16S ribosomal RNA gene sequencing was performed, and taxonomic and functional tables were produced. Physician-diagnosed eczema, allergy, and asthma were measured by questionnaires, allergic sensitization by skin prick tests, and lung function by spirometry. RESULTS The α-diversity of stool microbiota was associated with a decreased risk of eczema (odds ratio [OR], 0.98; 95% CI, 0.97, 1.00), and β-diversity was associated with physician-diagnosed inhalant allergy (R2 = 0.001; P = .047). Lachnospiraceae, Ruminococcaceae_UCG-005, and Christensenellaceae_R-7_group species were associated with decreased risks of eczema, inhalant allergic sensitization, and physician-diagnosed inhalant allergy (OR range, 0.88-0.94; 95% CI range, 0.79-0.96 to 0.88-0.98), while Agathobacter species were associated with an increased risk of physician-diagnosed inhalant allergy (OR, 1.23; 95% CI, 1.08-1.42). Functional pathways related to heme and terpenoid biosynthesis were associated with decreased risks of physician-diagnosed inhalant allergy and asthma (OR range, 0.89-0.86; 95% CI range, 0.80-0.99 to 0.73-1.02). No associations of stool microbiota with lung function were observed. CONCLUSIONS The diversity, relative abundance and functional pathways of stool microbiota were most consistently associated with physician-diagnosed inhalant allergy in school-age children and less consistently with other atopic diseases.
Collapse
Affiliation(s)
- Chen Hu
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Evelien R van Meel
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Division of Respiratory Medicine and Allergolog, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Monica Barroso
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jessica Kiefte-de Jong
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Public Health and Primary Care/Leiden University Medical Center Campus The Hague, Leiden University Medical Center, Leiden, The Netherlands
| | - Djawad Radjabzadeh
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Suzanne G M A Pasmans
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicolette W de Jong
- Divison of Allergy and Clinical Immunology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Division of Respiratory Medicine and Allergolog, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henriette A Moll
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Luba M Pardo
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Division of Respiratory Medicine and Allergolog, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
45
|
Li Y, Zhang Y, Zeng X. γδ T Cells Participating in Nervous Systems: A Story of Jekyll and Hyde. Front Immunol 2021; 12:656097. [PMID: 33868300 PMCID: PMC8044362 DOI: 10.3389/fimmu.2021.656097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/15/2021] [Indexed: 11/18/2022] Open
Abstract
γδ T cells are distributed in various lymphoid and nonlymphoid tissues, and act as early responders in many conditions. Previous studies have proven their significant roles in infection, cancer, autoimmune diseases and tissue maintenance. Recently, accumulating researches have highlighted the crosstalk between γδ T cells and nervous systems. In these reports, γδ T cells maintain some physiological functions of central nervous system by secreting interleukin (IL) 17, and neurons like nociceptors can in turn regulate the activity of γδ T cells. Moreover, γδ T cells are involved in neuroinflammation such as stroke and multiple sclerosis. This review illustrates the relationship between γδ T cells and nervous systems in physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | - Xun Zeng
- 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, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
46
|
Shaikh SB, Bhandary YP. Therapeutic properties of Punica granatum L (pomegranate) and its applications in lung-based diseases: A detailed review. J Food Biochem 2021; 45:e13684. [PMID: 33709449 DOI: 10.1111/jfbc.13684] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/26/2022]
Abstract
Respiratory diseases are the prime cause of death and disability worldwide. The majority of lung-based diseases are resistant to treatment. Hence, research on unique drugs/compounds with a more efficient and minimum side effect for treating lung diseases is urgent. Punica granatum L (pomegranate) fruit has been used in the prevention and treatment of various respiratory disorders in recent times. In vivo and in vitro studies have demonstrated that pomegranate fruit, as well as its juice, extract, peel powder, and oil, exert anti-proliferative, anti-oxidant, anti-microbial, anti-inflammatory, anti-cancer, and anti-tumorigenic properties by attenuating various respiratory conditions such as asthma, lung fibrosis, lung cancer, chronic obstructive pulmonary disease (COPD), and alveolar inflammation via modulating various signaling pathways. The current review summarizes the potential properties and medical benefits of pomegranate against different lung-based diseases, also highlighting its possible role in the lung fibrinolytic system. The available data suggest that pomegranate is effective in controlling the disease progressions and could be a potential therapeutic target benefiting human health status. Furthermore, this review also outlines the preclinical and clinical studies highlighting the role of pomegranate in lung diseases further evoking future studies to investigate the effect of intake of this anti-oxidant fruit in larger and well-defined human clinical trials. PRACTICAL APPLICATIONS: This review outlines the putative pharmacologic benefits of P. granatum L (pomegranate) in treating various chronic lung-based diseases such as lung cancer, COPD, ARDS, asthma, lung fibrosis, and cystic fibrosis. This review also highlights the possible inhibitory role of P. granatum L (pomegranate) in the lung fibrinolytic system triggering the fibrinolytic markers. This review summarizes the preclinical and clinical studies using in vitro, in vivo, and human models highlighting the potential role of P. granatum L (pomegranate) in lung diseases. This review evokes future research to investigate the effect of intake of pomegranate fruit in well-defined human clinical trials.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | | |
Collapse
|
47
|
Mowat AM. Historical Perspective: Metchnikoff and the intestinal microbiome. J Leukoc Biol 2021; 109:513-517. [PMID: 33630385 DOI: 10.1002/jlb.4ri0920-599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Metchnikoff's essay, Intestinal Bacteriotherapy, was written when the study of microbiology was still in its infancy and few intestinal diseases had been ascribed to a specific bacterial infection. Metchnikoff offered perceptive ideas that have become standard in today's science. This Historical Perspectives commentary examines how Metchnikoff's article influenced our field. An accompanying editorial by Siamon Gordon explores this topic further and describes the relevance of Metchnikoff's work to the current Covid-19 infection. We also include a translation of this fundamental article by Metchnikoff, as presented by Claudine Neyen.
Collapse
Affiliation(s)
- Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunology and Inflammation, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| |
Collapse
|
48
|
Liang Q, Li J, Zhang S, Liao Y, Guo S, Liang J, Deng X, Liu Y, Zou B, Wen X, Liang L, Wei L. Characterization of conjunctival microbiome dysbiosis associated with allergic conjunctivitis. Allergy 2021; 76:596-600. [PMID: 33080059 DOI: 10.1111/all.14635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Qiaoxing Liang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Jing Li
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Shiyao Zhang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yinglin Liao
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Shixin Guo
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Juanran Liang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xiuli Deng
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Yu Liu
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Bin Zou
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Xiaofeng Wen
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Lingyi Liang
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| | - Lai Wei
- State Key Laboratory of Ophthalmology Zhongshan Ophthalmic Center Sun Yat‐sen University Guangzhou China
| |
Collapse
|
49
|
Simultaneous allergic traits in dogs and their owners are associated with living environment, lifestyle and microbial exposures. Sci Rep 2020; 10:21954. [PMID: 33319851 PMCID: PMC7738549 DOI: 10.1038/s41598-020-79055-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
Both humans and pet dogs are more prone to develop allergies in urban than in rural environments, which has been associated with the differing microbial exposures between areas. However, potential similarities in the microbiota, that associate with environmental exposures, in allergic dogs and owners has not been investigated. We evaluated skin and gut microbiota, living environment, and lifestyle in 168 dog-owner pairs. Due to partly different manifestations of allergies between species, we focused on aeroallergen sensitized humans and dogs with owner-reported allergic symptoms. Our results agree with previous studies: dog-owner pairs suffered simultaneously from these allergic traits, higher risk associated with an urban environment, and the skin, but not gut, microbiota was partly shared by dog-owner pairs. We further discovered that urban environment homogenized both dog and human skin microbiota. Notably, certain bacterial taxa, which were associated with living environment and lifestyle, were also related with allergic traits, but these taxa differed between dogs and humans. Thus, we conclude that dogs and humans can be predisposed to allergy in response to same risk factors. However, as shared predisposing or protective bacterial taxa were not discovered, other factors than environmental microbial exposures can mediate the effect or furry dog and furless human skin select different taxa.
Collapse
|
50
|
Maturation of the gut microbiome during the first year of life contributes to the protective farm effect on childhood asthma. Nat Med 2020; 26:1766-1775. [PMID: 33139948 DOI: 10.1038/s41591-020-1095-x] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 09/08/2020] [Indexed: 12/19/2022]
Abstract
Growing up on a farm is associated with an asthma-protective effect, but the mechanisms underlying this effect are largely unknown. In the Protection against Allergy: Study in Rural Environments (PASTURE) birth cohort, we modeled maturation using 16S rRNA sequence data of the human gut microbiome in infants from 2 to 12 months of age. The estimated microbiome age (EMA) in 12-month-old infants was associated with previous farm exposure (β = 0.27 (0.12-0.43), P = 0.001, n = 618) and reduced risk of asthma at school age (odds ratio (OR) = 0.72 (0.56-0.93), P = 0.011). EMA mediated the protective farm effect by 19%. In a nested case-control sample (n = 138), we found inverse associations of asthma with the measured level of fecal butyrate (OR = 0.28 (0.09-0.91), P = 0.034), bacterial taxa that predict butyrate production (OR = 0.38 (0.17-0.84), P = 0.017) and the relative abundance of the gene encoding butyryl-coenzyme A (CoA):acetate-CoA-transferase, a major enzyme in butyrate metabolism (OR = 0.43 (0.19-0.97), P = 0.042). The gut microbiome may contribute to asthma protection through metabolites, supporting the concept of a gut-lung axis in humans.
Collapse
|