1
|
Dera N, Kosińska-Kaczyńska K, Żeber-Lubecka N, Brawura-Biskupski-Samaha R, Massalska D, Szymusik I, Dera K, Ciebiera M. Impact of Early-Life Microbiota on Immune System Development and Allergic Disorders. Biomedicines 2025; 13:121. [PMID: 39857705 PMCID: PMC11762082 DOI: 10.3390/biomedicines13010121] [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: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: The shaping of the human intestinal microbiota starts during the intrauterine period and continues through the subsequent stages of extrauterine life. The microbiota plays a significant role in the predisposition and development of immune diseases, as well as various inflammatory processes. Importantly, the proper colonization of the fetal digestive system is influenced by maternal microbiota, the method of pregnancy completion and the further formation of the microbiota. In the subsequent stages of a child's life, breastfeeding, diet and the use of antibiotics influence the state of eubiosis, which determines proper growth and development from the neonatal period to adulthood. The literature data suggest that there is evidence to confirm that the intestinal microbiota of the infant plays an important role in regulating the immune response associated with the development of allergic diseases. However, the identification of specific bacterial species in relation to specific types of reactions in allergic diseases is the basic problem. Background: The main aim of the review was to demonstrate the influence of the microbiota of the mother, fetus and newborn on the functioning of the immune system in the context of allergies and asthma. Methods: We reviewed and thoroughly analyzed the content of over 1000 articles and abstracts between the beginning of June and the end of August 2024. Over 150 articles were selected for the detailed study. Results: The selection was based on the PubMed National Library of Medicine search engine, using selected keywords: "the impact of intestinal microbiota on the development of immune diseases and asthma", "intestinal microbiota and allergic diseases", "the impact of intrauterine microbiota on the development of asthma", "intrauterine microbiota and immune diseases", "intrauterine microbiota and atopic dermatitis", "intrauterine microbiota and food allergies", "maternal microbiota", "fetal microbiota" and "neonatal microbiota". The above relationships constituted the main criteria for including articles in the analysis. Conclusions: In the present review, we showed a relationship between the proper maternal microbiota and the normal functioning of the fetal and neonatal immune system. The state of eubiosis with an adequate amount and diversity of microbiota is essential in preventing the development of immune and allergic diseases. The way the microbiota is shaped, resulting from the health-promoting behavior of pregnant women, the rational conduct of the medical staff and the proper performance of the diagnostic and therapeutic process, is necessary to maintain the health of the mother and the child. Therefore, an appropriate lifestyle, rational antibiotic therapy as well as the way of completing the pregnancy are indispensable in the prevention of the above conditions. At the same time, considering the intestinal microbiota of the newborn in relation to the genera and phyla of bacteria that have a potentially protective effect, it is worth noting that the use of suitable probiotics and prebiotics seems to contribute to the protective effect.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, 02-781 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Diana Massalska
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Kacper Dera
- Pediatric Ward, Department of Pediatrics, Center of Postgraduate Medical Education, Bielański Hospital, 01-809 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| |
Collapse
|
2
|
Del Chierico F, Piazzesi A, Fiscarelli EV, Ristori MV, Pirona I, Russo A, Citerà N, Macari G, Santarsiero S, Bianco F, Antenucci V, Damiani V, Mercuri L, De Vincentis GC, Putignani L. Changes in the pharyngeal and nasal microbiota in pediatric patients with adenotonsillar hypertrophy. Microbiol Spectr 2024; 12:e0072824. [PMID: 39248478 PMCID: PMC11449029 DOI: 10.1128/spectrum.00728-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
The present study aimed to investigate the pharyngeal and nasal microbiota composition in children with adenotonsillar hypertrophy (AH) and assess longitudinal alterations in both microbiota after a probiotic oral spray treatment. A cohort of 57 AH patients were enrolled and randomly assigned to the probiotic and placebo groups for a 5-month treatment course. Pharyngeal and nasal swabs were collected before and after treatment and analyzed by 16S rRNA-based metataxonomics and axenic cultures for pathobiont identification. 16S rRNA sequences from pharyngeal and nasal swabs of 65 healthy children (HC) were used as microbiota reference profiles. We found that the pharyngeal and nasal microbiota of AH children were similar. When compared to HC, we observed an increase of the genera Rothia, Granulicatella, Streptococcus, Neisseria, and Haemophilus, as well as a reduction of Corynebacterium, Pseudomonas, Acinetobacter, and Moraxella in both microbiota of AH patients. After probiotic treatment, we confirmed the absence of adverse effects and a reduction of upper respiratory tract infections (URTI). Moreover, the composition of pharyngeal microbiota was positively influenced by the reduction of potential pathobionts, like Haemophilus spp., with an increase of beneficial microbial metabolic pathways. Finally, the probiotic reduced the abundance of the pathobionts Streptococcus mitis and Gemella haemolysans in relation to AH severity. In conclusion, our results highlight the alterations of the pharyngeal and nasal microbiota associated with AH. Moreover, probiotic administration conferred protection against URTI and reduced the presence of potential pathobionts in patients with AH. IMPORTANCE Adenotonsillar hypertrophy (AH) is considered the main cause of breathing disorders during sleep in children. AH patients, after significant morbidity and often multiple courses of antibiotics, often proceed to tonsillectomy and/or adenoidectomy. Given the potential risks associated with these procedures, there is a growing interest in the use of nonsurgical adjuvant therapies, such as probiotics, that could potentially reduce their need for surgical intervention. In this study, we investigated the pharyngeal and nasal microbiota in patients with AH compared with healthy children. Furthermore, we tested the effects of probiotic spray administration on both disease symptoms and microbiota profiles, to evaluate the possible use of this microbial therapy as an adjuvant for AH patients.
Collapse
Affiliation(s)
| | - Antonia Piazzesi
- Research Unit of
Microbiome, Bambino Gesù Children’s Hospital,
IRCCS, Rome,
Italy
| | - Ersilia Vita Fiscarelli
- Research Unit of
Diagnostical and Management Innovations, Bambino Gesù
Children’s Hospital, IRCCS,
Rome, Italy
| | | | | | - Alessandra Russo
- Unit of Microbiomics,
Bambino Gesù Children’s Hospital,
IRCCS, Rome,
Italy
| | - Nicoletta Citerà
- Research Unit of
Diagnostical and Management Innovations, Bambino Gesù
Children’s Hospital, IRCCS,
Rome, Italy
| | | | - Sara Santarsiero
- Unit of
Otorhinolaryngology, Bambino Gesù Children’s Hospital,
IRCCS, Rome,
Italy
| | - Fabrizio Bianco
- Quality Team Studi
Clinici, Bambino Gesù Children’s Hospital,
IRCCS, Rome,
Italy
| | - Valeria Antenucci
- Modelli Innovativi di
Regolamentazione in Pediatria, Bambino Gesù Children’s
Hospital, IRCCS, Rome,
Italy
| | | | | | | | - Lorenza Putignani
- Unit of Microbiomics
and Research Unit of Microbiome, IRCCS, Bambino Gesù
Children’s Hospital,
Rome, Italy
| |
Collapse
|
3
|
Tate DE, Tanprasertsuk J, Jones RB, Maughan H, Chakrabarti A, Khafipour E, Norton SA, Shmalberg J, Honaker RW. A Randomized Controlled Trial to Evaluate the Impact of a Novel Probiotic and Nutraceutical Supplement on Pruritic Dermatitis and the Gut Microbiota in Privately Owned Dogs. Animals (Basel) 2024; 14:453. [PMID: 38338095 PMCID: PMC10854619 DOI: 10.3390/ani14030453] [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: 12/13/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Pruritic dermatitis (PD) is a common presentation of canine allergic skin diseases, with diversity in severity and treatment response due to complex etiopathogenesis. Evidence suggests the gut microbiota (GM) may contribute to the development of canine allergies. A 10-week double-blind randomized controlled trial evaluated a novel probiotic and nutraceutical blend (PNB) on clinical signs of skin allergy, health measures, and the GM of privately owned self-reported pruritic dogs. A total of 105 dogs were enrolled, with 62 included in pruritus and health analysis and 50 in microbiome analysis. The PNB supported greater improvement of owner-assessed clinical signs of PD at week 2 than the placebo (PBO). More dogs that received the PNB shifted to normal pruritus (digital PVAS10-N: <2) by week 4, compared to week 7 for the PBO. While a placebo effect was identified, clinical differences were supported by changes in the GM. The PNB enriched three probiotic bacteria and reduced abundances of species associated with negative effects. The PBO group demonstrated increased abundances of pathogenic species and reduced abundances of several beneficial species. This trial supports the potential of the PNB as a supplemental intervention in the treatment of PD; however, further investigation is warranted, with stricter diagnostic criteria, disease biomarkers and direct veterinary examination.
Collapse
Affiliation(s)
- Devon E. Tate
- NomNomNow Inc., Nashville, TN 37207, USA; (D.E.T.); (J.T.); (R.B.J.)
| | | | - Roshonda B. Jones
- NomNomNow Inc., Nashville, TN 37207, USA; (D.E.T.); (J.T.); (R.B.J.)
| | | | | | - Ehsan Khafipour
- Cargill Inc., Wayzata, MN 55391, USA; (A.C.); (E.K.); (S.A.N.)
| | | | - Justin Shmalberg
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ryan W. Honaker
- NomNomNow Inc., Nashville, TN 37207, USA; (D.E.T.); (J.T.); (R.B.J.)
| |
Collapse
|
4
|
Li R, Li J, Zhou X. Lung microbiome: new insights into the pathogenesis of respiratory diseases. Signal Transduct Target Ther 2024; 9:19. [PMID: 38228603 PMCID: PMC10791971 DOI: 10.1038/s41392-023-01722-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
The lungs were long thought to be sterile until technical advances uncovered the presence of the lung microbial community. The microbiome of healthy lungs is mainly derived from the upper respiratory tract (URT) microbiome but also has its own characteristic flora. The selection mechanisms in the lung, including clearance by coughing, pulmonary macrophages, the oscillation of respiratory cilia, and bacterial inhibition by alveolar surfactant, keep the microbiome transient and mobile, which is different from the microbiome in other organs. The pulmonary bacteriome has been intensively studied recently, but relatively little research has focused on the mycobiome and virome. This up-to-date review retrospectively summarizes the lung microbiome's history, composition, and function. We focus on the interaction of the lung microbiome with the oropharynx and gut microbiome and emphasize the role it plays in the innate and adaptive immune responses. More importantly, we focus on multiple respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), fibrosis, bronchiectasis, and pneumonia. The impact of the lung microbiome on coronavirus disease 2019 (COVID-19) and lung cancer has also been comprehensively studied. Furthermore, by summarizing the therapeutic potential of the lung microbiome in lung diseases and examining the shortcomings of the field, we propose an outlook of the direction of lung microbiome research.
Collapse
Affiliation(s)
- Ruomeng Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Forouhandeh H, Soofiyani SR, Hosseini K, Beirami SM, Ahangari H, Moammer Y, Ebrahimzadeh S, Nejad MK, Farjami A, Khodaiefar F, Tarhriz V. Modulation of the Immune System Mechanisms using Probiotic Bacteria in Allergic Diseases: Focus on Allergic Retinitis and Food Allergies. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2024; 18:11-26. [PMID: 37842889 DOI: 10.2174/0127722708246899230928080651] [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: 01/31/2023] [Revised: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 10/17/2023]
Abstract
Allergic illnesses occur when an organism's immune system is excessively responsive to certain antigens, such as those that are presented in the environment. Some people suffer from a wide range of immune system-related illnesses including allergic rhinitis, asthma, food allergies, hay fever, and even anaphylaxis. Immunotherapy and medications are frequently used to treat allergic disorders. The use of probiotics in bacteriotherapy has lately gained interest. Probiotics are essential to human health by modulating the gut microbiota in some ways. Due to probiotics' immunomodulatory properties present in the gut microbiota of all animals, including humans, these bacterial strains can prevent a wide variety of allergic disorders. Probiotic treatment helps allergy patients by decreasing inflammatory cytokines and enhancing intestinal permeability, which is important in the battle against allergy. By altering the balance of Th1 and Th2 immune responses in the intestinal mucosa, probiotics can heal allergic disorders. Numerous studies have shown a correlation between probiotics and a reduced risk of allergy disorders. A wide range of allergic disorders, including atopic dermatitis, asthma, allergic retinitis and food allergies has been proven to benefit from probiotic bacteria. Therefore, the use of probiotics in the treatment of allergic diseases offers a promising perspective. Considering that probiotic intervention in the treatment of diseases is a relatively new field of study, more studies in this regard seem necessary.
Collapse
Affiliation(s)
- Haleh Forouhandeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saiedeh Razi Soofiyani
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sohrab Minaei Beirami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Ahangari
- Department of Food Science and Technology, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yusif Moammer
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Ebrahimzadeh
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoomeh Kashef Nejad
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Farjami
- Food and Drug Safety Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Khodaiefar
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
6
|
Yang J, Liu S, Zhao Q, Li X, Jiang K. Gut microbiota-related metabolite alpha-linolenic acid mitigates intestinal inflammation induced by oral infection with Toxoplasma gondii. MICROBIOME 2023; 11:273. [PMID: 38087373 PMCID: PMC10714487 DOI: 10.1186/s40168-023-01681-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/27/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Oral infection with cysts is the main transmission route of Toxoplasma gondii (T. gondii), which leads to lethal intestinal inflammation. It has been widely recognized that T. gondii infection alters the composition and metabolism of the gut microbiota, thereby affecting the progression of toxoplasmosis. However, the potential mechanisms remain unclear. In our previous study, there was a decrease in the severity of toxoplasmosis after T. gondii α-amylase (α-AMY) was knocked out. Here, we established mouse models of ME49 and Δα-amy cyst infection and then took advantage of 16S rRNA gene sequencing and metabolomics analysis to identify specific gut microbiota-related metabolites that mitigate T. gondii-induced intestinal inflammation and analyzed the underlying mechanism. RESULTS There were significant differences in the intestinal inflammation between ME49 cyst- and Δα-amy cyst-infected mice, and transferring feces from mice infected with Δα-amy cysts into antibiotic-treated mice mitigated colitis caused by T. gondii infection. 16S rRNA gene sequencing showed that the relative abundances of gut bacteria, such as Lactobacillus and Bacteroides, Bifidobacterium, [Prevotella], Paraprevotella and Macellibacteroides, were enriched in mice challenged with Δα-amy cysts. Spearman correlation analysis between gut microbiota and metabolites indicated that some fatty acids, including azelaic acid, suberic acid, alpha-linolenic acid (ALA), and citramalic acid, were highly positively correlated with the identified bacterial genera. Both oral administration of ALA and fecal microbiota transplantation (FMT) decreased the expression of pro-inflammatory cytokines and restrained the MyD88/NF-κB pathway, which mitigated colitis and ultimately improved host survival. Furthermore, transferring feces from mice treated with ALA reshaped the colonization of beneficial bacteria, such as Enterobacteriaceae, Proteobacteria, Shigella, Lactobacillus, and Enterococcus. CONCLUSIONS The present findings demonstrate that the host gut microbiota is closely associated with the severity of T. gondii infection. We provide the first evidence that ALA can alleviate T. gondii-induced colitis by improving the dysregulation of the host gut microbiota and suppressing the production of pro-inflammatory cytokines via the MyD88/NF-κB pathway. Our study provides new insight into the medical application of ALA for the treatment of lethal intestinal inflammation caused by Toxoplasma infection. Video Abstract.
Collapse
Affiliation(s)
- Jing Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Songhao Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Qian Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Xiaobing Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China.
| | - Kangfeng Jiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China.
| |
Collapse
|
7
|
Zhao Y, Sun H, Chen Y, Niu Q, Dong Y, Li M, Yuan Y, Yang X, Sun Q. Butyrate protects against MRSA pneumonia via regulating gut-lung microbiota and alveolar macrophage M2 polarization. mBio 2023; 14:e0198723. [PMID: 37754570 PMCID: PMC10653920 DOI: 10.1128/mbio.01987-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 09/28/2023] Open
Abstract
IMPORTANCE Pneumonia caused by methicillin-resistant Staphylococcus aureus (MRSA) continues to carry a high burden in terms of mortality. With the roles of gut microbiota in mediating lung diseases being gradually uncovered, the details of the molecular mechanism of the "gut-lung axis" mediated by beneficial microorganisms and small-molecule metabolites have gradually attracted the attention of researchers. However, further studies are still necessary to determine the efficacy of microbial-based interventions. Our findings indicate that sodium butyrate (NaB) alleviates MRSA-induced pulmonary inflammation by improving gut-lung microbiota and promoting M2 polarization of alveolar macrophages. Therefore, the preventive administration of NaB might be explored as an effective strategy to control MRSA pneumonia.
Collapse
Affiliation(s)
- Yan Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Haoming Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yiwei Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qiang Niu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yiting Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ye Yuan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qingzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
8
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 139] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
9
|
Wang M, Zhao L, Wang K, Qin Y, Jin J, Wang D, Yan H, You C. Changes of Gut Microbiome in Adolescent Patients with Chronic Spontaneous Urticaria After Omalizumab Treatment. CLINICAL, COSMETIC AND INVESTIGATIONAL DERMATOLOGY 2023; 16:345-357. [PMID: 36762258 PMCID: PMC9907007 DOI: 10.2147/ccid.s393406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
Purpose Omalizumab is a humanized anti-immunoglobulin (Ig) E monoclonal antibody that is effective in treating some patients with chronic spontaneous urticaria (CSU) who do not respond to antihistamines. Gut microbiome plays a role in the pathogenesis of allergies and autoimmune diseases. Here, we investigated differences in the gut microbiome of adolescent CSU patients before and after omalizumab treatment, which has not been previously reported. Patients and Methods Ten adolescent CSU patients were given 300 mg omalizumab subcutaneously in three treatments at 4-week intervals. Urticaria Activity Score (UAS7) was applied to evaluate the efficacy of each omalizumab treatment during follow-up. Fecal samples were collected before and 12 weeks after the first treatment. Total DNA of the gut microbiota in all fecal samples were extracted. The 16S rRNA gene-targeted sequencing technology was used for the analysis of the diversity and distribution of gut microbiome, followed by bioinformatics analysis. Results UAS7 scores decreased significantly after each treatment compared with the baseline (all P < 0.0001). There were five well-controlled responders and five non-responders after three treatment sessions of omalizumab. The dominant bacteria phyla in all fecal samples were Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. Alpha diversity analysis showed no significant difference before and after treatment (P > 0.05), whereas beta diversity analysis revealed a significant difference in the bacterial abundance before and after treatment (P < 0.01). The relative abundance of Alphaproteobacteria and Betaproteobacteria at the class level and Burkholderia, Rhodococcus, and Sphingomonas at the genus level decreased significantly after treatment (linear discriminant analysis > 4, P < 0.05). The functional prediction results showed that the dioxin and xylene degradation pathways were more abundant before treatment. Conclusion Omalizumab is effective in treating CSU and the abundance of Alphaproteobacteria and Betaproteobacteria was reduced after treatment, which may help improve the treatment outcomes in adolescent CSU patients.
Collapse
Affiliation(s)
- Mei Wang
- Department of Dermatology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Leran Zhao
- Department of Dermatology and Venereology, The General Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| | - Kun Wang
- Department of Dermatology and Venereology, Candidate Branch of National Clinical Research Centre for Skin and Immune Diseases, First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Yongzhang Qin
- Department of Endocrinology, First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Jingji Jin
- Department of Dermatology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Dong Wang
- Department of Dermatology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Huimin Yan
- Department of Dermatology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Cong You
- Department of Dermatology and Venereology, Candidate Branch of National Clinical Research Centre for Skin and Immune Diseases, First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China,Correspondence: Cong You, Department of Dermatology and Venereology, Candidate Branch of National Clinical Research Centre for Skin and Immune Diseases, First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China, Tel +8615979766532, Email
| |
Collapse
|
10
|
Sadeghi M, Asadirad A, Koushki K, Keshavarz Shahbaz S, Dehnavi S. Recent advances in improving intranasal allergen-specific immunotherapy; focus on delivery systems and adjuvants. Int Immunopharmacol 2022; 113:109327. [PMID: 36257257 DOI: 10.1016/j.intimp.2022.109327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
11
|
Zeinali T, Faraji N, Joukar F, Khan Mirzaei M, Kafshdar Jalali H, Shenagari M, Mansour-Ghanaei F. Gut bacteria, bacteriophages, and probiotics: Tripartite mutualism to quench the SARS-CoV2 storm. Microb Pathog 2022; 170:105704. [PMID: 35948266 PMCID: PMC9357283 DOI: 10.1016/j.micpath.2022.105704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
Patients with SARS-CoV-2 infection, exhibit various clinical manifestations and severity including respiratory and enteric involvements. One of the main reasons for death among covid-19 patients is excessive immune responses directed toward cytokine storm with a low chance of recovery. Since the balanced gut microbiota could prepare health benefits by protecting against pathogens and regulating immune homeostasis, dysbiosis or disruption of gut microbiota could promote severe complications including autoimmune disorders; we surveyed the association between the imbalanced gut bacteria and the development of cytokine storm among COVID-19 patients, also the impact of probiotics and bacteriophages on the gut bacteria community to alleviate cytokine storm in COVID-19 patients. In present review, we will scrutinize the mechanism of immunological signaling pathways which may trigger a cytokine storm in SARS-CoV2 infections. Moreover, we are explaining in detail the possible immunological signaling pathway-directing by the gut bacterial community. Consequently, the specific manipulation of gut bacteria by using probiotics and bacteriophages for alleviation of the cytokine storm will be investigated. The tripartite mutualistic cooperation of gut bacteria, probiotics, and phages as a candidate prophylactic or therapeutic approach in SARS-CoV-2 cytokine storm episodes will be discussed at last.
Collapse
Affiliation(s)
- Tahereh Zeinali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764, Neuherberg, Germany
| | - Hossnieh Kafshdar Jalali
- Department of Microbiology, Faculty of Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Caspian Digestive Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
12
|
Debnath N, Kumar A, Yadav AK. Probiotics as a biotherapeutics for the management and prevention of respiratory tract diseases. Microbiol Immunol 2022; 66:277-291. [DOI: 10.1111/1348-0421.12980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/20/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology Central University of Jammu Samba 181143 Jammu and Kashmir (UT) India
| | - Ashwani Kumar
- Department of Nutrition Biology Central University of Haryana, Mahendergarh Jant‐Pali 123031 Haryana India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology Central University of Jammu Samba 181143 Jammu and Kashmir (UT) India
| |
Collapse
|
13
|
Coppola S, Avagliano C, Sacchi A, Laneri S, Calignano A, Voto L, Luzzetti A, Berni Canani R. Potential Clinical Applications of the Postbiotic Butyrate in Human Skin Diseases. Molecules 2022; 27:1849. [PMID: 35335213 PMCID: PMC8949901 DOI: 10.3390/molecules27061849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/01/2023] Open
Abstract
Human skin is the largest organ and the most external interface between the environment and the body. Vast communities of viruses, bacteria, archaea, fungi, and mites, collectively named the skin microbiome (SM), cover the skin surface and connected structures. Skin-resident microorganisms contribute to the establishment of cutaneous homeostasis and can modulate host inflammatory responses. Imbalances in the SM structure and function (dysbiosis) are associated with several skin conditions. Therefore, novel target for the skincare field could be represented by strategies, which restore or preserve the SM natural/individual balance. Several of the beneficial effects exerted by the SM are aroused by the microbial metabolite butyrate. Since butyrate exerts a pivotal role in preserving skin health, it could be used as a postbiotic strategy for preventing or treating skin diseases. Herein, we describe and share perspectives of the potential clinical applications of therapeutic strategies using the postbiotic butyrate against human skin diseases.
Collapse
Affiliation(s)
- Serena Coppola
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (L.V.); (A.L.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.A.); (A.S.); (S.L.); (A.C.)
| | - Antonia Sacchi
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.A.); (A.S.); (S.L.); (A.C.)
| | - Sonia Laneri
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.A.); (A.S.); (S.L.); (A.C.)
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.A.); (A.S.); (S.L.); (A.C.)
| | - Luana Voto
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (L.V.); (A.L.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Luzzetti
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (L.V.); (A.L.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (L.V.); (A.L.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, 80131 Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
14
|
Diallo D, Somboro AM, Diabate S, Baya B, Kone A, Sarro YS, Kone B, Diarra B, Diallo S, Diakite M, Doumbia S, Toloba Y, Murphy RL, Maiga M. Antituberculosis Therapy and Gut Microbiota: Review of Potential Host Microbiota Directed-Therapies. Front Cell Infect Microbiol 2021; 11:673100. [PMID: 34950603 PMCID: PMC8688706 DOI: 10.3389/fcimb.2021.673100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022] Open
Abstract
Tuberculosis (TB) remains a major public health concern with millions of deaths every year. The overlap with HIV infections, long treatment duration, and the emergence of drug resistance are significant obstacles to the control of the disease. Indeed, the standard first-line regimen TB treatment takes at least six months and even longer for the second-line therapy, resulting in relapses, drug resistance and re-infections. Many recent reports have also shown prolonged and significant damage of the gut microbial community (dysbiosis) from anti-TB drugs that can detrimentally persist several months after the cessation of treatment and could lead to the impairment of the immune response, and thus re-infections and drug resistance. A proposed strategy for shortening the treatment duration is thus to apply corrective measures to the dysbiosis for a faster bacterial clearance and a better treatment outcome. In this review, we will study the role of the gut microbiota in both TB infection and treatment, and its potential link with treatment duration. We will also discuss, the new concept of "Host Microbiota Directed-Therapies (HMDT)" as a potential adjunctive strategy to improve the treatment effectiveness, reduce its duration and or prevent relapses. These strategies include the use of probiotics, prebiotics, gut microbiota transfer, and other strategies. Application of this innovative solution could lead to HMDT as an adjunctive tool to shorten TB treatment, which will have enormous public health impacts for the End TB Strategy worldwide.
Collapse
Affiliation(s)
- Dramane Diallo
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Anou M Somboro
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Seydou Diabate
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bacar Baya
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Amadou Kone
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Yeya S Sarro
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bourahima Kone
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bassirou Diarra
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Souleymane Diallo
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Mahamadou Diakite
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Seydou Doumbia
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Yacouba Toloba
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Robert L Murphy
- Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Mamoudou Maiga
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali.,Institute for Global Health, Northwestern University, Chicago, IL, United States
| |
Collapse
|
15
|
Hu T, Dong Y, Yang C, Zhao M, He Q. Pathogenesis of Children's Allergic Diseases: Refocusing the Role of the Gut Microbiota. Front Physiol 2021; 12:749544. [PMID: 34721073 PMCID: PMC8551706 DOI: 10.3389/fphys.2021.749544] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Allergic diseases comprise a genetically heterogeneous cluster of immunologically mediated diseases, including asthma, food allergy (FA), allergic rhinitis (AR) and eczema, that have become major worldwide health problems. Over the past few decades, the spread of allergic diseases has displayed an increasing trend, and it has been reported that 22% of 1.39 billion people in 30 countries have a type of allergic disease. Undoubtedly, allergic diseases, which can be chronic, with significant morbidity, mortality and dynamic progression, impose major economic burdens on society and families; thus, exploring the cause of allergic diseases and reducing their prevalence is a top priority. Recently, it has been reported that the gastrointestinal (GI) microbiota can provide vital signals for the development, function, and regulation of the immune system, and the above-mentioned contributions make the GI microbiota a key player in allergic diseases. Notably, the GI microbiota is highly influenced by the mode of delivery, infant diet, environment, antibiotic use and so on. Specifically, changes in the environment can result in the dysbiosis of the GI microbiota. The proper function of the GI microbiota depends on a stable cellular composition which in the case of the human microbiota consists mainly of bacteria. Large shifts in the ratio between these phyla or the expansion of new bacterial groups lead to a disease-promoting imbalance, which is often referred to as dysbiosis. And the dysbiosis can lead to alterations of the composition of the microbiota and subsequent changes in metabolism. Further, the GI microbiota can affect the physiological characteristics of the human host and modulate the immune response of the host. The objectives of this review are to evaluate the development of the GI microbiota, the main drivers of the colonization of the GI tract, and the potential role of the GI microbiota in allergic diseases and provide a theoretical basis as well as molecular strategies for clinical practice.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yinmiao Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chenghao Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Jin BY, Li Z, Xia YN, Li LX, Zhao ZX, Li XY, Li Y, Li B, Zhou RC, Fu SC, Li SY, Li YQ. Probiotic Interventions Alleviate Food Allergy Symptoms Correlated With Cesarean Section: A Murine Model. Front Immunol 2021; 12:741371. [PMID: 34650564 PMCID: PMC8505808 DOI: 10.3389/fimmu.2021.741371] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Delivery by cesarean section (CS) is linked to an increased incidence of food allergies in children and affects early gut microbiota colonization. Furthermore, emerging evidence has connected disordered intestinal microbiota to food allergies. Here, we investigated the impact of CS on a rat model for food allergy to ovalbumin (OVA). Rats delivered by CS were found to be more responsive to OVA sensitization than vaginally born ones, displaying a greater reduction in rectal temperature upon challenge, worse diarrhea, and higher levels of OVA-specific antibodies and histamine. 16S rRNA sequencing of feces revealed reduced levels of Lactobacillus and Bifidobacterium in the CS rats. Preventative supplementation with a probiotic combination containing Lactobacillus and Bifidobacterium could protect CS rats against an allergic response to OVA, indicating that the microbiota dysbiosis contributes to CS-related response. Additionally, probiotic intervention early in life might help to rebuild aberrant Th2 responses and tight junction proteins, both of which have been linked to CS-related high allergic reactions. Taken together, this study shows that disordered intestinal microbiota plays an essential role in the pathogenesis of food allergy mediated by CS. More importantly, interventions that modulate the microbiota composition in early life are therapeutically relevant for CS-related food allergies.
Collapse
Affiliation(s)
- Bi-Ying Jin
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Ya-Nan Xia
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Li-Xiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Zi-Xiao Zhao
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Yu Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Yan Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Bing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Ru-Chen Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Shi-Chen Fu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| | - Shi-Yang Li
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China.,Robot Engineering Laboratory for Precise Diagnosis and Therapy of Gastrointestinal Tumor, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Chan Y, Raju Allam VSR, Paudel KR, Singh SK, Gulati M, Dhanasekaran M, Gupta PK, Jha NK, Devkota HP, Gupta G, Hansbro PM, Oliver BGG, Chellappan DK, Dua K. Nutraceuticals: unlocking newer paradigms in the mitigation of inflammatory lung diseases. Crit Rev Food Sci Nutr 2021:1-31. [PMID: 34613853 DOI: 10.1080/10408398.2021.1986467] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Persistent respiratory tract inflammation contributes to the pathogenesis of various chronic respiratory diseases, such as asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis. These inflammatory respiratory diseases have been a major public health concern as they are the leading causes of worldwide mortality and morbidity, resulting in heavy burden on socioeconomic growth throughout these years. Although various therapeutic agents are currently available, the clinical applications of these agents are found to be futile due to their adverse effects, and most patients remained poorly controlled with a low quality of life. These drawbacks have necessitated the development of novel, alternative therapeutic agents that can effectively improve therapeutic outcomes. Recently, nutraceuticals such as probiotics, vitamins, and phytochemicals have gained increasing attention due to their nutritional properties and therapeutic potential in modulating the pathological mechanisms underlying inflammatory respiratory diseases, which could ultimately result in improved disease control and overall health outcomes. As such, nutraceuticals have been held in high regard as the possible alternatives to address the limitations of conventional therapeutics, where intensive research are being performed to identify novel nutraceuticals that can positively impact various inflammatory respiratory diseases. This review provides an insight into the utilization of nutraceuticals with respect to their molecular mechanisms targeting multiple signaling pathways involved in the pathogenesis of inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Yinghan Chan
- School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama, USA
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Brian Gregory George Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
18
|
Elgamal Z, Singh P, Geraghty P. The Upper Airway Microbiota, Environmental Exposures, Inflammation, and Disease. ACTA ACUST UNITED AC 2021; 57:medicina57080823. [PMID: 34441029 PMCID: PMC8402057 DOI: 10.3390/medicina57080823] [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: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
Along with playing vital roles in pathogen exclusion and immune system priming, the upper airways (UAs) and their microbiota are essential for myriad physiological functions such as conditioning and transferring inhaled air. Dysbiosis, a microbial imbalance, is linked with various diseases and significantly impedes the quality of one’s life. Daily inhaled exposures and/or underlying conditions contribute to adverse changes to the UA microbiota. Such variations in the microbial community exacerbate UA and pulmonary disorders via modulating inflammatory and immune pathways. Hence, exploring the UA microbiota’s role in maintaining homeostasis is imperative. The microbial composition and subsequent relationship with airborne exposures, inflammation, and disease are crucial for strategizing innovating UA diagnostics and therapeutics. The development of a healthy UA microbiota early in life contributes to normal respiratory development and function in the succeeding years. Although different UA cavities present a unique microbial profile, geriatrics have similar microbes across their UAs. This lost community segregation may contribute to inflammation and disease, as it stimulates disadvantageous microbial–microbial and microbial–host interactions. Varying inflammatory profiles are associated with specific microbial compositions, while the same is true for many disease conditions and environmental exposures. A shift in the microbial composition is also detected upon the administration of numerous therapeutics, highlighting other beneficial and adverse side effects. This review examines the role of the UA microbiota in achieving homeostasis, and the impact on the UAs of environmental airborne pollutants, inflammation, and disease.
Collapse
Affiliation(s)
- Ziyad Elgamal
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada;
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA
| | - Pratyush Singh
- Department of Biology, University of Western Ontario, London, ON N6A 5B7, Canada;
| | - Patrick Geraghty
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA
- Correspondence: ; Tel.: +1-718-270-3141
| |
Collapse
|
19
|
Zhang J, Ma J, Li Q, Su H, Sun X. Exploration of the effect of mixed probiotics on microbiota of allergic asthma mice. Cell Immunol 2021; 367:104399. [PMID: 34192623 DOI: 10.1016/j.cellimm.2021.104399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 12/13/2022]
Abstract
Co-relation between allergic asthma and microbiota varying with diet has been extensively investigated, implicating that oral supplement of alternative pharmaceuticals are potential for asthma control. Probiotics are of great concern due to its beneficial effects on the host, whereas the potential mechanisms and the optimal dose need to be further explored. In the present study, three different doses of mixed strains were given orally to mouse model of allergic asthma induced by ovalbumin (OVA). Continuous administration of mixed probiotics could alleviate OVA-induced allergic inflammation through reducing costimulatory molecules on the surface of dendritic cells (DCs) and increasing mucosal gut-primed Tregs induced by mesenteric CD103+DCs. Mixed probiotics-induced protective effect relates to gut microbiota instead of lung microbes. Microbial diversity and Bacteroidetes/Firmicutes (B/F) ratio are augmented upon probiotic strains. Interestingly, treatment with mixed strain lead to an increased levels of genus Lactobacillus. Lactobacillus genus-Operational Taxonomic Unit (OUT) analysis indicated that OTU9 may be the dominant strain from mixed probiotics providing protective effect. The low dose seems to be the best in the whole study. Our results provide new evidence for probiotic application in allergic diseases and support the idea that targeted gut microbiota will be an effective approach for allergic airway diseases.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Jingyi Ma
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Qiuhong Li
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
20
|
El-Sayed A, Aleya L, Kamel M. The link among microbiota, epigenetics, and disease development. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:28926-28964. [PMID: 33860421 DOI: 10.1007/s11356-021-13862-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
The microbiome is a community of various microorganisms that inhabit or live on the skin of humans/animals, sharing the body space with their hosts. It is a sort of complex ecosystem of trillions of commensals, symbiotic, and pathogenic microorganisms, including trillions of bacteria, archaea, protozoa, fungi, and viruses. The microbiota plays a role in the health and disease status of the host. Their number, species dominance, and viability are dynamic. Their long-term disturbance is usually accompanied by serious diseases such as metabolic disorders, cardiovascular diseases, or even cancer. While epigenetics is a term that refers to different stimuli that induce modifications in gene expression patterns without structural changes in the inherited DNA sequence, these changes can be reversible or even persist for several generations. Epigenetics can be described as cell memory that stores experience against internal and external factors. Results from multiple institutions have contributed to the role and close interaction of both microbiota and epigenetics in disease induction. Understanding the mechanisms of both players enables a better understanding of disease induction and development and also opens the horizon to revolutionary therapeutic approaches. The present review illustrates the roles of diet, microbiome, and epigenetics in the induction of several chronic diseases. In addition, it discusses the application of epigenetic data to develop diagnostic biomarkers and therapeutics and evaluate their safety for patients. Understanding the interaction among all these elements enables the development of innovative preventive/therapeutic approaches for disease control.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| |
Collapse
|
21
|
|
22
|
Lin CH, Tseng CY, Chao MW. Administration of Lactobacillus paracasei HB89 mitigates PM2.5-induced enhancement of inflammation and allergic airway response in murine asthma model. PLoS One 2020; 15:e0243062. [PMID: 33284823 PMCID: PMC7721166 DOI: 10.1371/journal.pone.0243062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 11/16/2020] [Indexed: 01/05/2023] Open
Abstract
PM2.5 causes abnormal immune response and asthma in animals. In this study, a Balb/c mouse animal model was exposed to PM2.5 to induce asthma. Lactobacillus paracasei HB89 was fed at the same time, in order to observe whether L. paracasei HB89 mitigates respiratory tract allergies stimulated by PM2.5. The results showed that PM2.5 stimulated a significant increase in white blood cells and immunoglobulin (IgE) in OVA-induced allergic Balb/c mice, and IgE in the blood further triggered the release of histamine in the lung immune cells. This not only increased overall immune cell counts, but the lymphocyte counts also increased significantly, resulting in significant inhibitions of cytokines INF-r and TGF-β, and induction of IL-4, IL-5, IL-13 and IL-17a. After feeding with HB89, apart from the absence of observable changes in body weight, the total white blood cell count in the animal blood and IgE response were also be reduced; the proliferation of immune cells in the lungs caused by PM2.5 was slowed down; and histamine and cytokines INF-r and TGF-β were secreted in large quantities, but IL- 4, IL-5, IL-13, IL-17a were inhibited, which effectively reduced the possibility of asthma induction.
Collapse
Affiliation(s)
- Ching-Hung Lin
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan, Taiwan
- Center for Nanotechnology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
- Center for Nanotechnology, Chung Yuan Christian University, Taoyuan, Taiwan
| |
Collapse
|
23
|
Probiotics function and modulation of the immune system in allergic diseases. Allergol Immunopathol (Madr) 2020; 48:771-788. [PMID: 32763025 DOI: 10.1016/j.aller.2020.04.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
Allergic diseases have been a global problem over the past few decades. The effect of allergic diseases on healthcare systems and society is generally remarkable and is considered as one of the most common causes of chronic and hospitalized disease. The functional ability of probiotics to modulate the innate/acquired immune system leads to the initiation of mucosal/systemic immune responses. Gut microbiota plays a beneficial role in food digestion, development of the immune system, control/growth of the intestinal epithelial cells and their differentiation. Prescribing probiotics causes a significant change in the intestinal microflora and modulates cytokine secretion, including networks of genes, TLRs, signaling molecules and increased intestinal IgA responses. The modulation of the Th1/Th2 balance is done by probiotics, which suppress Th2 responses with shifts to Th1 and thereby prevent allergies. In general, probiotics are associated with a decrease in inflammation by increasing butyrate production and induction of tolerance with an increase in the ratio of cytokines such as IL-4, IL-10/IFN-γ, Treg/TGF-β, reducing serum eosinophil levels and the expression of metalloproteinase-9 which contribute to the improvement of the allergic disease's symptoms. Finally, it can be said that the therapeutic approach to immunotherapy and the reduction of the risk of side effects in the treatment of allergic diseases is the first priority of treatment and the final approach that completes the first priority in maintaining the condition and sustainability of the tolerance along with the recovery of the individual.
Collapse
|
24
|
Navarro-Tapia E, Sebastiani G, Sailer S, Toledano LA, Serra-Delgado M, García-Algar Ó, Andreu-Fernández V. Probiotic Supplementation During the Perinatal and Infant Period: Effects on Gut Dysbiosis and Disease. Nutrients 2020; 12:E2243. [PMID: 32727119 PMCID: PMC7468726 DOI: 10.3390/nu12082243] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The perinatal period is crucial to the establishment of lifelong gut microbiota. The abundance and composition of microbiota can be altered by several factors such as preterm delivery, formula feeding, infections, antibiotic treatment, and lifestyle during pregnancy. Gut dysbiosis affects the development of innate and adaptive immune responses and resistance to pathogens, promoting atopic diseases, food sensitization, and infections such as necrotizing enterocolitis (NEC). Recent studies have indicated that the gut microbiota imbalance can be restored after a single or multi-strain probiotic supplementation, especially mixtures of Lactobacillus and Bifidobacterium strains. Following the systematic search methodology, the current review addresses the importance of probiotics as a preventive or therapeutic tool for dysbiosis produced during the perinatal and infant period. We also discuss the safety of the use of probiotics in pregnant women, preterm neonates, or infants for the treatment of atopic diseases and infections.
Collapse
Affiliation(s)
- Elisabet Navarro-Tapia
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
| | - Giorgia Sebastiani
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Sebastian Sailer
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Laura Almeida Toledano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| |
Collapse
|
25
|
Wu R, Li X, Ma N, Jin X, Yuan X, Qu C, Tang H, Liu Z, Zhang Z. Bacterial Quorum Sensing Molecules Promote Allergic Airway Inflammation by Activating the Retinoic Acid Response. iScience 2020; 23:101288. [PMID: 32622265 PMCID: PMC7334417 DOI: 10.1016/j.isci.2020.101288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/24/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
IgE and IgG1 production in the type 2 immune response is the characteristic feature of an allergic reaction. However, whether bacterial molecules modulate IgE and IgG1 production remains obscure. Here, we demonstrate that the bacterial quorum sensing molecules acyl homoserine lactones (AHLs) induce IgE and IgG1 production by activating the RARE (retinoic acid response element) response in dendritic cells (DCs) in vivo. DC-specific knockout of the retinoic acid transcriptional factor Rara diminished the AHL-stimulated type 2 immune response in vitro. AHLs altered DC phenotype, upregulated OX40L and IFN-I signature, and promoted T helper 2 cell differentiation in vitro. Finally, AHLs activated the RARE response by inhibiting AKT phosphorylation in vitro, as the AKT agonists IGF-1 and PDGF abolished the effect of AHLs on the RARE response. This study demonstrates a mechanism by which AHLs drive allergic airway inflammation through activating retinoic acid signaling in DCs. Acyl homoserine lactones (AHLs) exacerbate allergic airway inflammation AHLs induce IgE and IgG1 production AHLs activate the RARE response in dendritic cells AHLs inhibit AKT phosphorylation
Collapse
Affiliation(s)
- Renlan Wu
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Xingjie Li
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ning Ma
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiufeng Jin
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiefang Yuan
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chen Qu
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Hongmei Tang
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhigang Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Zongde Zhang
- Inflammation & Allergic Diseases Research Unit, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China; Model Animal Research Center, Nanjing University, Nanjing 210061, China.
| |
Collapse
|
26
|
Pratap K, Taki AC, Johnston EB, Lopata AL, Kamath SD. A Comprehensive Review on Natural Bioactive Compounds and Probiotics as Potential Therapeutics in Food Allergy Treatment. Front Immunol 2020; 11:996. [PMID: 32670266 PMCID: PMC7326084 DOI: 10.3389/fimmu.2020.00996] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Food allergy is rising at an alarming rate and is a major public health concern. Globally, food allergy affects over 500 million people, often starting in early childhood and increasingly reported in adults. Commercially, only one approved oral immunotherapy-based treatment is currently available and other allergen-based immunotherapeutic are being investigated in clinical studies. As an alternative approach, a substantial amount of research has been conducted on natural compounds and probiotics, focusing on the immune modes of action, and therapeutic uses of such sources to tackle various immune-related diseases. Food allergy is primarily mediated by IgE antibodies and the suppression of allergic symptoms seems to be mostly modulated through a reduction of allergen-specific IgE antibodies, upregulation of blocking IgG, and downregulation of effector cell activation (e.g., mast cells) or expression of T-helper 2 (Th-2) cytokines. A wide variety of investigations conducted in small animal models or cell-based systems have reported on the efficacy of natural bioactive compounds and probiotics as potential anti-allergic therapeutics. However, very few lead compounds, unlike anti-cancer and anti-microbial applications, have been selected for clinical trials in the treatment of food allergies. Natural products or probiotic-based approaches appear to reduce the symptoms and/or target specific pathways independent of the implicated food allergen. This broad range therapeutic approach essentially provides a major advantage as several different types of food allergens can be targeted with one approach and potentially associated with a lower cost of development. This review provides a brief overview of the immune mechanisms underlying food allergy and allergen-specific immunotherapy, followed by a comprehensive collection of current studies conducted to investigate the therapeutic applications of natural compounds and probiotics, including discussions of their mode of action and immunological aspects of their disease-modifying capabilities.
Collapse
Affiliation(s)
- Kunal Pratap
- Molecular Allergy Research Laboratory, Discipline of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.,Center for Molecular Therapeutics, James Cook University, Townsville, QLD, Australia
| | - Aya C Taki
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Elecia B Johnston
- Molecular Allergy Research Laboratory, Discipline of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.,Center for Molecular Therapeutics, James Cook University, Townsville, QLD, Australia
| | - Andreas L Lopata
- Molecular Allergy Research Laboratory, Discipline of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.,Center for Molecular Therapeutics, James Cook University, Townsville, QLD, Australia
| | - Sandip D Kamath
- Molecular Allergy Research Laboratory, Discipline of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.,Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.,Center for Molecular Therapeutics, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
27
|
Ren C, Faas MM, de Vos P. Disease managing capacities and mechanisms of host effects of lactic acid bacteria. Crit Rev Food Sci Nutr 2020; 61:1365-1393. [PMID: 32366110 DOI: 10.1080/10408398.2020.1758625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Consumption of lactic acid bacteria (LAB) has been suggested to confer health-promoting effects on the host. However, effects of LABs have been reported to be species- and strain-specific and the mechanisms involved are subjects of discussion. Here, the possible mechanisms by which LABs induce antipathogenic, gut barrier enhancing and immune modulating effects in consumers are reviewed. Specific strains for which it has been proven that health is improved by these mechanisms are discussed. However, most strains probably act via several or combinations of mechanisms depending on which effector molecules they express. Current insight is that these effector molecules are either present on the cell wall of LAB or are excreted. These molecules are reviewed as well as the ligand binding receptors in the host. Also postbiotics are discussed. Finally, we provide an overview of the efficacy of LABs in combating infections caused by Helicobacter pylori, Salmonella, Escherichia coli, Streptococcus pneumoniae, and influenza virus, in controlling gut inflammatory diseases, in managing allergic disorders, and in alleviating cancer.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Alfano A, Perillo F, Fusco A, Savio V, Corsaro MM, Donnarumma G, Schiraldi C, Cimini D. Lactobacillus brevis CD2: Fermentation Strategies and Extracellular Metabolites Characterization. Probiotics Antimicrob Proteins 2020; 12:1542-1554. [PMID: 32279232 DOI: 10.1007/s12602-020-09651-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Functional foods and nutraceuticals frequently contain viable probiotic strains that, at certain titers, are considered to be responsible of beneficial effects on health. Recently, it was observed that secreted metabolites might play a key role in this respect, especially in immunomodulation. Exopolysaccharides produced by probiotics, for example, are used in the food, pharmaceutical, and biomedical fields, due to their unique properties. Lactobacillus brevis CD2 demonstrated the ability to inhibit oral pathogens causing mucositis and periodontal inflammation and to reduce Helycobacter pylori infections. Due to the lack of literature, for this strain, on the development of fermentation processes that can increase the titer of viable cells and associated metabolites to industrially attractive levels, different batch and fed-batch strategies were investigated in the present study. In particular, aeration was shown to improve the growth rate and the yields of lactic acid and biomass in batch cultures. The use of an exponential feeding profile in fed-batch experiments allowed to produce 9.3 ± 0.45 × 109 CFU/mL in 42 h of growth, corresponding to a 20-fold increase of viable cells compared with that obtained in aerated batch processes; moreover, also increased titers of exopolysaccharides and lactic acid (260 and 150%, respectively) were observed. A purification process based on ultrafiltration, charcoal treatment, and solvent precipitation was applied to partially purify secreted metabolites and separate them into two molecular weight fractions (above and below 10 kDa). Both fractions inhibited growth of the known gut pathogen, Salmonella typhimurium, demonstrating that lactic acid plays a major role in pathogen growth inhibition, which is however further enhanced by the presence of Lact. brevis CD2 exopolysaccharides. Finally, the EPS produced from Lact. brevis CD2 was characterized by NMR for the first time up to date.
Collapse
Affiliation(s)
- Alberto Alfano
- Department of Experimental Medicine, Section of Biotechnology Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy
| | - Filomena Perillo
- Department of Experimental Medicine, Section of Biotechnology Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy
| | - Alessandra Fusco
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, Università della Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy
| | - Vittoria Savio
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, Università della Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy
| | - Maria Michela Corsaro
- Department of Chemic1al Science, University Federico II, Complesso Universitario Monte S.Angelo, via Cintia 4, 80126, Naples, Italy
| | - Giovanna Donnarumma
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, Università della Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy
| | - Chiara Schiraldi
- Department of Experimental Medicine, Section of Biotechnology Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy.
| | - Donatella Cimini
- Department of Experimental Medicine, Section of Biotechnology Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", via L. De Crecchio n 7, 80138, Naples, Italy.
| |
Collapse
|
29
|
Qosimah D, Murwani S, Amri IA, Prasetyo D, Rifai M, Kusuma ID, Purba O, Pranoto YN, Rahmadiani F, Sanjoyo AM. Preventive effect of kefir in BALB-c (Mus musculus) induced by ovalbumin towards relative amount of B cell-Ig-E. ACTA ACUST UNITED AC 2020. [DOI: 10.1088/1742-6596/1430/1/012015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
In Vitro Evaluation of Probiotic Potential and Safety Assessment of Lactobacillus mucosae Strains Isolated from Donkey’s Lactation. Probiotics Antimicrob Proteins 2019; 12:1045-1056. [DOI: 10.1007/s12602-019-09610-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
31
|
Kumpitsch C, Koskinen K, Schöpf V, Moissl-Eichinger C. The microbiome of the upper respiratory tract in health and disease. BMC Biol 2019; 17:87. [PMID: 31699101 PMCID: PMC6836414 DOI: 10.1186/s12915-019-0703-z] [Citation(s) in RCA: 241] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 02/08/2023] Open
Abstract
The human upper respiratory tract (URT) offers a variety of niches for microbial colonization. Local microbial communities are shaped by the different characteristics of the specific location within the URT, but also by the interaction with both external and intrinsic factors, such as ageing, diseases, immune responses, olfactory function, and lifestyle habits such as smoking. We summarize here the current knowledge about the URT microbiome in health and disease, discuss methodological issues, and consider the potential of the nasal microbiome to be used for medical diagnostics and as a target for therapy.
Collapse
Affiliation(s)
- Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Kaisa Koskinen
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Veronika Schöpf
- Institute of Psychology, University of Graz, Universitaetsplatz 2, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
- Present address: Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
32
|
Fabbrizzi A, Amedei A, Lavorini F, Renda T, Fontana G. The lung microbiome: clinical and therapeutic implications. Intern Emerg Med 2019; 14:1241-1250. [PMID: 31667699 DOI: 10.1007/s11739-019-02208-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022]
Abstract
The human respiratory tract, usually considered sterile, is currently being investigated for human-associated microbial communities. According to Dickson's conceptual model, the lung microbiota (LMt) is a dynamic ecosystem, whose composition, in healthy lungs, is likely to reflect microbial migration, reproduction, and elimination. However, which microbial genera constitutes a "healthy microbiome" per se remains hotly debated. It is now widely accepted that a bi-directional gut-lung axis connects the intestinal with the pulmonary microbiota and that the diet could have a role in modulating both microbiotas as in health as in pathological status. The LMt is altered in numerous respiratory disorders such as obstructive airway diseases, interstitial lung diseases, infections, and lung cancer. Some authors hypothesize that the use of specific bacterial strains, termed "probiotics," with positive effects on the host immunity and/or against pathogens, could have beneficial effects in the treatment of intestinal disorders and pulmonary diseases. In this manuscript, we have reviewed the literature available on the LMt to delineate and discuss the potential relationship between composition alterations of LMt and lung diseases. Finally, we have reported some meaningful clinical studies that used integrated probiotics' treatments to contrast some lung-correlated disorders.
Collapse
Affiliation(s)
- Alessio Fabbrizzi
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy.
- Sod of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), University of Florence, 50134, Florence, Italy.
| | - Federico Lavorini
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Teresa Renda
- Respiratory Unit, Careggi University Hospital, Florence, Italy
| | - Giovanni Fontana
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
33
|
van den Broek MFL, De Boeck I, Kiekens F, Boudewyns A, Vanderveken OM, Lebeer S. Translating Recent Microbiome Insights in Otitis Media into Probiotic Strategies. Clin Microbiol Rev 2019; 32:e00010-18. [PMID: 31270125 PMCID: PMC6750133 DOI: 10.1128/cmr.00010-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The microbiota of the upper respiratory tract (URT) protects the host from bacterial pathogenic colonization by competing for adherence to epithelial cells and by immune response regulation that includes the activation of antimicrobial and (anti-)inflammatory components. However, environmental or host factors can modify the microbiota to an unstable community that predisposes the host to infection or inflammation. One of the URT diseases most often encountered in children is otitis media (OM). The role of pathogenic bacteria like Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis in the pathogenesis of OM is well documented. Results from next-generation-sequencing (NGS) studies reveal other bacterial taxa involved in OM, such as Turicella and Alloiococcus Such studies can also identify bacterial taxa that are potentially protective against URT infections, whose beneficial action needs to be substantiated in relevant experimental models and clinical trials. Of note, lactic acid bacteria (LAB) are members of the URT microbiota and associated with a URT ecosystem that is deemed healthy, based on NGS and some experimental and clinical studies. These observations have formed the basis of this review, in which we describe the current knowledge of the molecular and clinical potential of LAB in the URT, which is currently underexplored in microbiome and probiotic research.
Collapse
Affiliation(s)
- Marianne F L van den Broek
- Environmental Ecology and Applied Microbiology Research Group, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Ilke De Boeck
- Environmental Ecology and Applied Microbiology Research Group, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Filip Kiekens
- Laboratory of Pharmaceutical Technology and Biopharmacy, Department of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - An Boudewyns
- Department of Otorhinolaryngology, Head and Neck Surgery, Antwerp University Hospital, Edegem, Belgium
| | - Olivier M Vanderveken
- Department of Otorhinolaryngology, Head and Neck Surgery, Antwerp University Hospital, Edegem, Belgium
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Environmental Ecology and Applied Microbiology Research Group, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
34
|
Yan DC, Hung CH, Sy LB, Lue KH, Shih IH, Yang CY, Chen LC, Sun HL, Lee MS, Chambard J, Tanguy J, Hughes-Formella B, Nutten S, Blanchard C. A Randomized, Double-Blind, Placebo-Controlled Trial Assessing the Oral Administration of a Heat-Treated Lactobacillus paracasei Supplement in Infants with Atopic Dermatitis Receiving Topical Corticosteroid Therapy. Skin Pharmacol Physiol 2019; 32:201-211. [PMID: 31117100 DOI: 10.1159/000499436] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/05/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Atopic dermatitis (AD) is a common disease in infancy, for which topical steroids are the first-line therapy but have side effects. Innovative approaches are needed to reduce the burden of AD and corticosteroid usage in infants. METHODS The once-daily consumption of heat-treated probiotic Lactobacillus paracasei GM-080 or placebo for 16 weeks as supplementary approach to topical treatment with fluticasone propionate cream was compared in AD infants aged 4-30 months. Outcomes were SCORAD and its subscores, TEWL, Infants' Dermatitis Quality of Life Index (IDQOL), corticoid "sparing effect," CCL17/TARC, and IgE status. RESULTS SCORAD, objective SCORAD, itching, and IDQOL decreased significantly (p < 0.001) over the treatment period in both treatment groups. Slight decreases (ns) were noted in TEWL in lesional and unaffected skin and CCL17 levels. There were no differences between the treatment groups. Total IgE increased over the treatment period in both groups, with significantly higher increase in the heat-treated probiotic group (p = 0.038). There was no evidence of a corticoid "sparing effect" by the probiotic. CONCLUSIONS In this design, the probiotic L. paracasei was not beneficial as a complementary approach to topical corticosteroids in infants with AD. However, slight beneficial effects may have been masked by the moderate potency corticoid.
Collapse
Affiliation(s)
- Dah-Chin Yan
- Taipei Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Hsing Hung
- Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung City, Taiwan
| | | | - Ko-Huang Lue
- Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - I-Hsin Shih
- Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chin-Yi Yang
- Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Li-Chen Chen
- Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Hai-Lun Sun
- Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Min-Sheng Lee
- Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City, Taiwan
| | | | | | | | | | | |
Collapse
|
35
|
Cutting Edge: Probiotics and Fecal Microbiota Transplantation in Immunomodulation. J Immunol Res 2019; 2019:1603758. [PMID: 31143780 PMCID: PMC6501133 DOI: 10.1155/2019/1603758] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
Probiotics are commensal or nonpathogenic microbes that confer beneficial effects on the host through several mechanisms such as competitive exclusion, antibacterial effects, and modulation of immune responses. Some probiotics have been found to regulate immune responses via immune regulatory mechanisms. T regulatory (Treg) cells, T helper cell balances, dendritic cells, macrophages, B cells, and natural killer (NK) cells can be considered as the most determinant dysregulated mediators in immunomodulatory status. Recently, fecal microbiota transplantation (FMT) has been defined as the transfer of distal gut microbial communities from a healthy individual to a patient's intestinal tract to cure some immune disorders (mainly inflammatory bowel diseases). The aim of this review was followed through the recent literature survey on immunomodulatory effects and mechanisms of probiotics and FMT and also efficacy and safety of probiotics and FMT in clinical trials and applications.
Collapse
|
36
|
Wu CT, Lin FH, Lee YT, Ku MS, Lue KH. Effect of Lactobacillus rhamnosus GG immunopathologic changes in chronic mouse asthma model. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:911-919. [PMID: 30952512 DOI: 10.1016/j.jmii.2019.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/28/2019] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma is a heterogeneous inflammatory disorder of the airway. A Th2 response usually contributes to high levels of allergen-specific IgE and eosinophilic airway inflammation. Several findings have demonstrated that neutrophils, not eosinophils, are the major inflammatory cells in chronic asthma patients with steroid-resistance. Lactobacillus rhammosus GG (LGG) exhibits anti-inflammatory properties on OVA-induced acute airway inflammation. OBJECTIVE We hypothesized that orally administrated LGG should reduce airway remodeling in chronic experimental models. METHODS Female Balb/c mice were sensitized with OVA. LGG was used to investigate whether oral administrations of LGG inhibited OVA-induced airway inflammation in a chronic asthma model and the different intervention times between LGG pre-treatment and post-treatment groups. BALF was analyzed with Liu's stain and ELISA assay. Lung histopathology was assayed with HE, IHC and Masson's trichrome staining. Lung tissues were assayed with PCR (T-bet, GATA3, RORrt and Foxp3). Many cytokines were detected in the serum and BALF. RESULTS LGG significantly decreased the number of infiltrating inflammatory cells. We also found that the oral LGG group suppressed not only Th2 cytokine, but also IL-17, TNF-α and HMGB1 in the BALF levels. However, GATA3 and RORrt decreased significantly in the RNA level in the LGG groups, but the T-bet and Foxp3 increased in the RNA level. CONCLUSIONS LGG not only had anti-inflammatory effects on OVA-induced airway inflammation, but also improved airway remodeling and collagen expression in the chronic asthma mouse model. Moreover, LGG might be an additional or supplementary therapy for allergic airway diseases.
Collapse
Affiliation(s)
- Chia-Ta Wu
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, Taiwan 402; Department of Emergency Medicine, Changhua Christian Hospital, Changhua, Taiwan.
| | - Fei-Hung Lin
- Department of Emergency Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Yu-Tzu Lee
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, Taiwan 402
| | - Min-Sho Ku
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Ko-Haung Lue
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, Taiwan 402; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
37
|
Reddel S, Del Chierico F, Quagliariello A, Giancristoforo S, Vernocchi P, Russo A, Fiocchi A, Rossi P, Putignani L, El Hachem M. Gut microbiota profile in children affected by atopic dermatitis and evaluation of intestinal persistence of a probiotic mixture. Sci Rep 2019; 9:4996. [PMID: 30899033 PMCID: PMC6428866 DOI: 10.1038/s41598-019-41149-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Atopic dermatitis (AD) has been hypothesised to be associated with gut microbiota (GM) composition. We performed a comparative study of the GM profile of 19 AD children and 18 healthy individuals aimed at identifying bacterial biomarkers associated with the disease. The effect of probiotic intake (Bifidobacterium breve plus Lactobacillus salivarius) on the modulation of GM and the probiotic persistence in the GM were also evaluated. Faecal samples were analysed by real-time PCR and 16S rRNA targeted metagenomics. Although the probiotics, chosen for this study, did not shape the entire GM profile, we observed the ability of these species to pass through the gastrointestinal tract and to persist (only B. breve) in the GM. Moreover, the GM of patients compared to CTRLs showed a dysbiotic status characterised by an increase of Faecalibacterium, Oscillospira, Bacteroides, Parabacteroides and Sutterella and a reduction of short-chain fatty acid (SCFA)-producing bacteria (i.e., Bifidobacterium, Blautia, Coprococcus, Eubacterium and Propionibacterium). Taken togheter these results show an alteration in AD microbiota composition with the depletion or absence of some species, opening the way to future probiotic intervention studies.
Collapse
Affiliation(s)
- Sofia Reddel
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | | | | | - Pamela Vernocchi
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alessandra Russo
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alessandro Fiocchi
- Unit of Allergology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paolo Rossi
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit and Parasitology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy.
| | - May El Hachem
- Dermatology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
38
|
E. coli Nissle 1917 is a safe mucosal delivery vector for a birch-grass pollen chimera to prevent allergic poly-sensitization. Mucosal Immunol 2019; 12:132-144. [PMID: 30242254 DOI: 10.1038/s41385-018-0084-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/27/2018] [Accepted: 08/25/2018] [Indexed: 02/04/2023]
Abstract
Allergic poly-sensitization affects a large number of allergic patients and poses a great challenge for their treatment. In this study we evaluated the effects of the probiotic Escherichia coli Nissle 1917 (EcN) expressing a birch and grass pollen allergen chimera 'Bet v 1, Phl p 1 and Phl p 5' (EcN-Chim) on allergy prevention after oral or intranasal application in poly-sensitized mice. In contrast to oral application, intranasal pretreatment with EcN-Chim prior to poly-sensitization led to a significant reduction of lung inflammation (eosinophils, IL-5, and IL-13 in bronchoalveolar lavage) along with suppressed levels of allergen-specific serum IgE. The suppression was associated with increased levels of allergen-specific IgA in lungs and serum IgG2a along with increased Foxp3, TGF-β, and IL-10 mRNA in bronchial lymph nodes. In vitro EcN induced high levels of IL-10 and IL-6 in both lung and intestinal epithelial cells. Importantly, using in vivo imaging techniques we demonstrated that intranasally applied EcN do not permanently colonize nose, lung, and gut and this strain might therefore be a safe delivery vector against allergy in humans. In conclusion, our data show that intranasal application of recombinant EcN expressing a multiallergen chimera presents a novel and promising treatment strategy for prevention of allergic poly-sensitization.
Collapse
|
39
|
|
40
|
Kim HW, Hong R, Choi EY, Yu K, Kim N, Hyeon JY, Cho KK, Choi IS, Yun CH. A Probiotic Mixture Regulates T Cell Balance and Reduces Atopic Dermatitis Symptoms in Mice. Front Microbiol 2018; 9:2414. [PMID: 30374337 PMCID: PMC6196311 DOI: 10.3389/fmicb.2018.02414] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 09/20/2018] [Indexed: 01/20/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder with a complex etiology involving the immune response. Recent studies have demonstrated the role of certain probiotics in the treatment and prevention of AD. However, the mechanism by which these probiotics regulate the immune system remains unclear. In this study, we examined the immunomodulatory capacity of Duolac ATP, a mixed formulation of probiotics, both in vitro and in vivo. Results showed that the expression of programmed death-ligand 1(PD-L1) was significantly upregulated on bone marrow-derived dendritic cells (BMDCs) treated with Duolac ATP. Furthermore, the anti-inflammatory cytokines IL-10 and TGF-beta were both upregulated when BMDCs were treated with Duolac ATP. The percentage of proliferated regulatory T cells (Tregs) was enhanced when CD4+ T cells were co-cultured with Duolac ATP-treated BMDCs on plates coated with anti-CD3/CD28 antibodies. Intriguingly, IL-10 secretion from CD4+ T cells was also observed. The AD symptoms, histologic scores, and serum IgE levels in AD mice were significantly decreased after oral treatment with Duolac ATP. Moreover, the Th1-mediated response in AD-induced mice treated with oral Duolac ATP showed upregulation of IL-2 and IFN-gamma as well as of downstream signaling molecules T-bet, STAT-1, and STAT-4. Conversely, Duolac ATP suppressed Th2 and Th17 responses in AD-like mice, as evidenced by the downregulation of GATA-3, C-maf, IL-4, IL-5, and IL-17. Additionally, Duolac ATP increased the number of Tregs found at Peyer’s patches (PP) in treated AD mice. These results suggest that Duolac ATP modulates DCs to initiate both Th1 and Treg responses in AD mice. Thus, Duolac ATP represents a potential preventative agent against AD and could serve as an effective immunomodulator in AD patients.
Collapse
Affiliation(s)
- Han Wool Kim
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Rira Hong
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Eun Young Choi
- Department of Biological Science, College of Medical and Life Sciences, Silla University, Busan, South Korea
| | - KeeSun Yu
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Narae Kim
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Jin Yi Hyeon
- Department of Biological Science, College of Medical and Life Sciences, Silla University, Busan, South Korea
| | - Kwang Keun Cho
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju, South Korea
| | - In Soon Choi
- Department of Biological Science, College of Medical and Life Sciences, Silla University, Busan, South Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, South Korea
| |
Collapse
|
41
|
Martens K, Pugin B, De Boeck I, Spacova I, Steelant B, Seys SF, Lebeer S, Hellings PW. Probiotics for the airways: Potential to improve epithelial and immune homeostasis. Allergy 2018; 73:1954-1963. [PMID: 29869783 DOI: 10.1111/all.13495] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2018] [Indexed: 12/30/2022]
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefit on the host. The therapeutic effects of probiotics have been mostly studied in the gastrointestinal tract, but recent evidence points toward the potential of these bacteria to prevent and/or treat chronic airway diseases. In this review, possible mechanisms of action of probiotics in the airways are described, with a particular focus on their capacity to modulate the epithelial barrier function and their mode of interaction with the immune system. Indeed, probiotic bacteria, mostly lactobacilli, can promote the expression and regulation of tight junctions and adherence junctions, resulting in the restoration of a defective epithelial barrier. These bacteria interact with the epithelial barrier and immune cells through pattern recognition receptors, such as Toll-like receptors, which upon activation can stimulate or suppress various immune responses. Finally, the clinical potential of probiotics to treat inflammatory diseases of the upper and lower respiratory tract, and the difference between their mode of application (eg, oral or nasal) are discussed here.
Collapse
Affiliation(s)
- K. Martens
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - B. Pugin
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - I. De Boeck
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - I. Spacova
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - B. Steelant
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. F. Seys
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. Lebeer
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - P. W. Hellings
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
- Clinical Division of Otorhinolaryngology, Head and Neck Surgery University Hospitals Leuven Leuven Belgium
- Department of Otorhinolaryngology University Hospitals Ghent Ghent Belgium
- Department of Otorhinolaryngology Academic Medical Center University of Amsterdam Amsterdam the Netherlands
| |
Collapse
|
42
|
Anand S, Mande SS. Diet, Microbiota and Gut-Lung Connection. Front Microbiol 2018; 9:2147. [PMID: 30283410 PMCID: PMC6156521 DOI: 10.3389/fmicb.2018.02147] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022] Open
Abstract
The gut microbial community (Gut microbiota) is known to impact metabolic functions as well as immune responses in our body. Diet plays an important role in determining the composition of the gut microbiota. Gut microbes help in assimilating dietary nutrients which are indigestible by humans. The metabolites produced by them not only modulate gastro-intestinal immunity, but also impact distal organs like lung and brain. Micro-aspiration of gut bacteria or movement of sensitized immune cells through lymph or bloodstream can also influence immune response of other organs. Dysbiosis in gut microbiota has been implicated in several lung diseases, including allergy, asthma and cystic fibrosis. The bi-directional cross-talk between gut and lung (termed as Gut-Lung axis) is best exemplified by intestinal disturbances observed in lung diseases. Some of the existing probiotics show beneficial effects on lung health. A deeper understanding of the gut microbiome which comprises of all the genetic material within the gut microbiota and its role in respiratory disorders is likely to help in designing appropriate probiotic cocktails for therapeutic applications.
Collapse
Affiliation(s)
- Swadha Anand
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| | - Sharmila S Mande
- Bio-Sciences R&D Division, TCS Research, Tata Consultancy Services Ltd., Pune, India
| |
Collapse
|
43
|
Rezazadeh A, Shahabi S, Bagheri M, Nabizadeh E, Jazani NH. The protective effect of Lactobacillus and Bifidobacterium as the gut microbiota members against chronic urticaria. Int Immunopharmacol 2018; 59:168-173. [PMID: 29655058 DOI: 10.1016/j.intimp.2018.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/29/2018] [Accepted: 04/05/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic Urticaria is a common disorder which is defined by recurrent occurrence of wheals and sometimes angioedema. It has a notable influence on the patients' quality of life. Regulation of the immune system is one of the important roles of the gut microbiota. The effect of dysbiosis considering some members of gut microbiota in patients with chronic urticaria has been demonstrated in our previous study. OBJECTIVE Comparing the frequency and bacterial load of Lactobacillus, Bifidobacterium, and Bacteroides between patients with chronic urticaria and healthy controls. METHODS 20 patients with chronic urticaria and 20 age and sex matched healthy individuals were included in the present study. Stool samples were analyzed for determining the frequency and bacterial load of Lactobacillus, Bifidobacterium, and Bacteroides genera. RESULTS There were no significant differences among the frequencies of detectable Lactobacillus, Bifidobacterium, or Bacteroides in stool samples of patients with chronic urticaria and healthy controls. The relative amounts of Lactobacillus and Bifidobacterium were significantly higher in fecal samples from controls compared to patients with chronic urticaria (P = 0.038 and 0.039, respectively). CONCLUSION It is the first study on the implication of Lactobacillus, Bifidobacterium, and Bacteroides genera as gut microbiota members in patients with chronic urticaria.
Collapse
Affiliation(s)
- Akram Rezazadeh
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahram Shahabi
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Morteza Bagheri
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Edris Nabizadeh
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nima Hosseini Jazani
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
44
|
Partida-Rodríguez O, Serrano-Vázquez A, Nieves-Ramírez ME, Moran P, Rojas L, Portillo T, González E, Hernández E, Finlay BB, Ximenez C. Human Intestinal Microbiota: Interaction Between Parasites and the Host Immune Response. Arch Med Res 2017; 48:690-700. [PMID: 29290328 DOI: 10.1016/j.arcmed.2017.11.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
The human gut is a highly complex ecosystem with an extensive microbial community, and the influence of the intestinal microbiota reaches the entire host organism. For example, the microbiome regulates fat storage, stimulates or renews epithelial cells, and influences the development and maturation of the brain and the immune system. Intestinal microbes can protect against infection by pathogenic bacteria, viruses, fungi and parasites. Hence, the maintenance of homeostasis between the gut microbiota and the rest of the body is crucial for health, with dysbiosis affecting disease. This review focuses on intestinal protozoa, especially those still representing a public health problem in Mexico, and their interactions with the microbiome and the host. The decrease in prevalence of intestinal helminthes in humans left a vacant ecological niche that was quickly occupied by protozoa. Although the mechanisms governing the interaction between intestinal microbiota and protozoa are poorly understood, it is known that the composition of the intestinal bacterial populations modulates the progression of protozoan infection and the outcome of parasitic disease. Most reports on the complex interactions between intestinal bacteria, protozoa and the immune system emphasize the protective role of the microbiota against protozoan infection. Insights into such protection may facilitate the manipulation of microbiota components to prevent and treat intestinal protozoan infections. Here we discuss recent findings about the immunoregulatory effect of intestinal microbiota with regards to intestinal colonization by protozoa, focusing on infections by Entamoeba histolytica, Blastocystis spp, Giardia duodenalis, Toxoplasma gondii and Cryptosporidium parvum. The possible consequences of the microbiota on parasitic, allergic and autoimmune disorders are also considered.
Collapse
Affiliation(s)
- Oswaldo Partida-Rodríguez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México; Michael Smith Laboratories, University of Brithish Columbia, Vancouver, Canada
| | - Angélica Serrano-Vázquez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Miriam E Nieves-Ramírez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Patricia Moran
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Liliana Rojas
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Tobias Portillo
- Unidad de Bioinformática, Bioestadística y Biología Computacional. Red de Apoyo a la Investigación Científica, Universidad Nacional Autónoma de México, Instituto Nacional De Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Enrique González
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Eric Hernández
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - B Brett Finlay
- Michael Smith Laboratories, University of Brithish Columbia, Vancouver, Canada
| | - Cecilia Ximenez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
45
|
Cerdó T, Ruíz A, Suárez A, Campoy C. Probiotic, Prebiotic, and Brain Development. Nutrients 2017; 9:E1247. [PMID: 29135961 PMCID: PMC5707719 DOI: 10.3390/nu9111247] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/02/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023] Open
Abstract
Recently, a number of studies have demonstrated the existence of a link between the emotional and cognitive centres of the brain and peripheral functions through the bi-directional interaction between the central nervous system and the enteric nervous system. Therefore, the use of bacteria as therapeutics has attracted much interest. Recent research has found that there are a variety of mechanisms by which bacteria can signal to the brain and influence several processes in relation to neurotransmission, neurogenesis, and behaviour. Data derived from both in vitro experiments and in vivo clinical trials have supported some of these new health implications. While recent molecular advancement has provided strong indications to support and justify the role of the gut microbiota on the gut-brain axis, it is still not clear whether manipulations through probiotics and prebiotics administration could be beneficial in the treatment of neurological problems. The understanding of the gut microbiota and its activities is essential for the generation of future personalized healthcare strategies. Here, we explore and summarize the potential beneficial effects of probiotics and prebiotics in the neurodevelopmental process and in the prevention and treatment of certain neurological human diseases, highlighting current and future perspectives in this topic.
Collapse
Affiliation(s)
- Tomás Cerdó
- Department of Paediatrics, School of Medicine, University of Granada, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
| | - Alicia Ruíz
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Biochemistry and Molecular Biology 2, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
| | - Antonio Suárez
- Department of Biochemistry and Molecular Biology 2, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, 18016 Granada, Spain.
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Carlos III Institute, 18016 Granada, Spain.
- Department of Paediatrics, Faculty of Medicine, University of Granada, Av. de la Investigación, 11, 18016 Granada, Spain.
| |
Collapse
|
46
|
Hidalgo-Cantabrana C, Delgado S, Ruiz L, Ruas-Madiedo P, Sánchez B, Margolles A. Bifidobacteria and Their Health-Promoting Effects. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0010-2016. [PMID: 28643627 PMCID: PMC11687494 DOI: 10.1128/microbiolspec.bad-0010-2016] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Indexed: 12/28/2022] Open
Abstract
Bifidobacteria are members of the intestinal microbiota of mammals and other animals, and some strains are able to exert health-promoting effects. The genus Bifidobacterium belongs to the Actinobacteria phylum. Firmicutes, Bacteroidetes, and Actinobacteria constitute the most abundant phyla in the human intestinal microbiota, Firmicutes and Bacteroidetes being predominant in adults, and Actinobacteria in breast-fed infants, where bifidobacteria can reach levels higher than 90% of the total bacterial population. They are among the first microbial colonizers of the intestines of newborns, and play key roles in the development of their physiology, including maturation of the immune system and use of dietary components. Indeed, some nutrients, such as human milk oligosaccharides, are important drivers of bifidobacterial development. Some Bifidobacterium strains are considered probiotic microorganisms because of their beneficial effects, and they have been included as bioactive ingredients in functional foods, mainly dairy products, as well as in food supplements and pharma products, alone, or together with, other microbes or microbial substrates. Well-documented scientific evidence of their activities is currently available for bifidobacteria-containing preparations in some intestinal and extraintestinal pathologies. In this review, we focus on the role of bifidobacteria as members of the human intestinal microbiota and their use as probiotics in the prevention and treatment of disease.
Collapse
Affiliation(s)
- Claudio Hidalgo-Cantabrana
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| | - Susana Delgado
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| | - Borja Sánchez
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Dairy Research Institute of Asturias, Spanish National Research Council (IPLA-CSIC), Paseo Río Linares s/n 33300, Villaviciosa, Asturias, Spain
| |
Collapse
|
47
|
Nallagatla H, Hemalatha R, Kondapalli NB, Mohammed S. Impact of a novel synbiotic supplementation during gestation and lactation on immune responses in the Swiss albino mice offspring. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:2453-2459. [PMID: 27696418 DOI: 10.1002/jsfa.8059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/14/2016] [Accepted: 09/25/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Prebiotics from various regularly consumed cereals and novel substrates are currently being utilised as functional foods. The aim of this study was to determine the effect of synbiotic, formulated with prebiotic extracted from natural resources like green gram (Vigna radiata) along with probiotic Lactobacillus rhamnosus GG (LGG) in modulating immune responses in the offspring when supplemented during gestation and lactation. RESULTS Synbiotic supplementation was effective in improving cell mediated immunity and humoral immunity among F0 dams. Among F1 pups (F1 Syn + and F1 Syn-), synbiotic supplementation showed significantly heightened (P < 0.05) splenocyte proliferation, increased interleukin-10, interferon gamma and interleukin-17 responses, leucocyte phagocytic ability and increased secretory-immunoglobulin A. However, four-fold increase in IgG titres to Hepatitis-B vaccine was observed only in those mice that were supplemented with synbiotic postweaning (F1 Syn+). CONCLUSION Synbiotic supplementation to pregnant dams affected the offspring's cellular and mucosal immunity favorably. However, IgG response to Hepatitis-B vaccine was influenced positively only when the supplementation was extended to the offsprings in the post weaning period. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Himaja Nallagatla
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Rajkumar Hemalatha
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Narendra Babu Kondapalli
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| | - Shujauddin Mohammed
- Department of Clinical Microbiology & Immunology, National Institute of Nutrition (ICMR), Jamai-Osmania, Hyderabad, 500 007, Telangana, India
| |
Collapse
|
48
|
Górska S, Schwarzer M, Srutkova D, Hermanova P, Brzozowska E, Kozakova H, Gamian A. Polysaccharides L900/2 and L900/3 isolated from Lactobacillus rhamnosus LOCK 0900 modulate allergic sensitization to ovalbumin in a mouse model. Microb Biotechnol 2017; 10:586-593. [PMID: 28165193 PMCID: PMC5404188 DOI: 10.1111/1751-7915.12606] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/08/2017] [Indexed: 01/01/2023] Open
Abstract
Here, we compared the abilities of polysaccharides L900/2 and L900/3, which were previously isolated from Lactobacillus rhamnosus LOCK 0900, to modulate the immune response to bystander antigens in a mouse model of ovalbumin (OVA) sensitization. In vivo, both polysaccharides reduced the levels of OVA‐specific IgE, IgE‐dependent basophil degranulation and IgG2a antibodies, but had no effect on the levels of OVA‐specific IgA or IgG1. Interestingly, both polysaccharides triggered recall cellular responses with distinct properties. L900/3 significantly suppressed the OVA‐induced upregulations of IL‐4, IL‐5, IL‐10 and IL‐13 in re‐stimulated spleen cells and mesenteric lymph nodes. Our findings support and expand on our previous in vitro studies by demonstrating that polymer L900/3 might modulate the Th1/Th2 balance and could be a promising candidate molecule for preventing allergic sensitization.
Collapse
Affiliation(s)
- Sabina Górska
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Martin Schwarzer
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Dagmar Srutkova
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Petra Hermanova
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Ewa Brzozowska
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Hana Kozakova
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Andrzej Gamian
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| |
Collapse
|
49
|
张 惠, 樊 蕊, 张 静, 陶 小, 孙 新. [Association between risk factors during maternal pregnancy and the neonatal period and childhood bronchial asthma]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:49-53. [PMID: 28100322 PMCID: PMC7390120 DOI: 10.7499/j.issn.1008-8830.2017.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/16/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To study the association of the risk factors during maternal pregnancy and the neonatal period with childhood bronchial asthma. METHODS A total of 306 children with asthma (asthma group) and 250 healthy children (control group) were enrolled. Their clinical data during the neonatal period and the maternal data during pregnancy were retrospectively studied. RESULTS The univariate analysis showed that there were significant differences in the rates of maternal use of antibiotics during pregnancy, use of antibiotics and probiotics during the neonatal period, preterm birth, cesarean section, low birth weight, and breast feeding (>6 months) between the asthma and control groups (P<0.05). The multivariate logistic regression analysis showed that use of antibiotics during pregnancy (OR=3.908, 95%CI: 1.277-11.962), use of antibiotics during neonatal period (OR=24.154, 95%CI: 7.864-74.183), preterm birth (OR=8.535, 95%CI: 2.733-26.652), and cesarean section (OR=4.588, 95%CI: 2.887-7.291) were independent risk factors for childhood asthma. The use of probiotics during the neonatal period (OR=0.014, 95%CI: 0.004-0.046) and breast feeding (>6 months) (OR=0.161, 95%CI: 0.103-0.253) were protective factors for childhood asthma. CONCLUSIONS The early prevention of childhood asthma can be improved by reducing the use of antibiotics during pregnancy, reducing cesarean section, avoiding abuse of antibiotics during the neonatal period, trying breast feeding and taking probiotics in early stage.
Collapse
Affiliation(s)
- 惠琴 张
- />第四军医大学西京医院儿科, 陕西 西安 710032Department of Pediatrics, Xijing Hospital of Fourth Military Medical University, Xi'an 710032, China
| | - 蕊 樊
- />第四军医大学西京医院儿科, 陕西 西安 710032Department of Pediatrics, Xijing Hospital of Fourth Military Medical University, Xi'an 710032, China
| | - 静静 张
- />第四军医大学西京医院儿科, 陕西 西安 710032Department of Pediatrics, Xijing Hospital of Fourth Military Medical University, Xi'an 710032, China
| | - 小娟 陶
- />第四军医大学西京医院儿科, 陕西 西安 710032Department of Pediatrics, Xijing Hospital of Fourth Military Medical University, Xi'an 710032, China
| | - 新 孙
- />第四军医大学西京医院儿科, 陕西 西安 710032Department of Pediatrics, Xijing Hospital of Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
50
|
Zheng H, Liang H, Wang Y, Miao M, Shi T, Yang F, Liu E, Yuan W, Ji ZS, Li DK. Altered Gut Microbiota Composition Associated with Eczema in Infants. PLoS One 2016; 11:e0166026. [PMID: 27812181 PMCID: PMC5094743 DOI: 10.1371/journal.pone.0166026] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/23/2016] [Indexed: 12/20/2022] Open
Abstract
Eczema is frequently the first manifestation of an atopic diathesis and alteration in the diversity of gut microbiota has been reported in infants with eczema. To identify specific bacterial communities associated with eczema, we conducted a case-control study of 50 infants with eczema (cases) and 51 healthy infants (controls). We performed high-throughput sequencing for V3–V4 hypervariable regions of the 16S rRNA genes from the gut fecal material. A total of 12,386 OTUs (operational taxonomic units) at a 97% similarity level were obtained from the two groups, and we observed a difference in taxa abundance, but not the taxonomic composition, of gut microbiota between the two groups. We identified four genera enriched in healthy infants: Bifidobacterium, Megasphaera, Haemophilus and Streptococcus; and five genera enriched in infants with eczema: Escherichia/Shigella, Veillonella, Faecalibacterium, Lachnospiraceae incertae sedis and Clostridium XlVa. Several species, such as Faecalibacterium prausnitzii and Ruminococcus gnavus, that are known to be associated with atopy or inflammation, were found to be significantly enriched in infants with eczema. Higher abundance of Akkermansia muciniphila in eczematous infants might reduce the integrity of intestinal barrier function and therefore increase the risk of developing eczema. On the other hand, Bacteroides fragilis and Streptococcus salivarius, which are known for their anti-inflammatory properties, were less abundant in infants with eczema. The observed differences in genera and species between cases and controls in this study may provide insight into the link between the microbiome and eczema risk.
Collapse
Affiliation(s)
- Huajun Zheng
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Hong Liang
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Yuezhu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai 201203, China
| | - Maohua Miao
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Tao Shi
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Fen Yang
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Enuo Liu
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
| | - Wei Yuan
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
- * E-mail: (WY); (ZSJ)
| | - Zai-Si Ji
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai 200032, China
- * E-mail: (WY); (ZSJ)
| | - De-Kun Li
- Division of Research, Kaiser Foundation Research Institute, 2000 Broadway, Oakland, CA 94612, United States of America
| |
Collapse
|