1
|
Borrego-Ruiz A, Borrego JJ. Nutritional and Microbial Strategies for Treating Acne, Alopecia, and Atopic Dermatitis. Nutrients 2024; 16:3559. [PMID: 39458553 PMCID: PMC11510041 DOI: 10.3390/nu16203559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Diet is one of the major determinants of the composition and function of the gut microbiome, and diverse studies have established directional connections between gut microbiome dysbiosis and skin dyshomeostasis. Furthermore, a significant link between the gut and certain skin-related disorders has been reported. This work reviews the mechanisms underlying the relationship between nutritional factors, gut microbiome, and certain skin diseases such as acne vulgaris, alopecia, and atopic dermatitis. In addition, it explores how the modulation of the gut microbiome and human skin through diet and various microbial strategies, including probiotics, synbiotics, postbiotics, and fecal microbiota transplantation, may serve as future treatments for skin diseases, possibly replacing traditional methods such as antibiotic, topical corticosteroid, and laser therapies. RESULTS The adequate intake of certain foods can promote a balanced gut microbiome, potentially reducing skin inflammation and improving overall skin health, while poor dietary choices may lead to worse outcomes by disrupting gut homeostasis. In this regard, diets high in antioxidants, fiber, and phytonutrients appear to be beneficial for enhancing skin health and preventing associated comorbidities. In addition, the administration of probiotics, synbiotics, and postbiotics in the treatment of cutaneous diseases has been shown to restore skin dyshomeostasis and to improve the symptoms of the reviewed skin conditions. CONCLUSIONS Consuming a healthy, plant-based diet can reduce skin inflammation and enhance overall skin health. Although the application of probiotics, synbiotics, and postbiotics has demonstrated promise in modulating inflammation, enhancing tissue regeneration, and inhibiting pathogenic colonization, further research is required.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
2
|
Mennini M, Piccirillo M, Furio S, Valitutti F, Ferretti A, Strisciuglio C, De Filippo M, Parisi P, Peroni DG, Di Nardo G, Ferrari F. Probiotics and other adjuvants in allergen-specific immunotherapy for food allergy: a comprehensive review. FRONTIERS IN ALLERGY 2024; 5:1473352. [PMID: 39450374 PMCID: PMC11499231 DOI: 10.3389/falgy.2024.1473352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
This review delves into the potential of manipulating the microbiome to enhance oral tolerance in food allergy, focusing on food allergen-specific immunotherapy (FA-AIT) and the use of adjuvants, with a significant emphasis on probiotics. FA-AIT, including oral (OIT), sublingual (SLIT), and epicutaneous (EPIT) immunotherapy, has shown efficacy in desensitizing patients and achieving sustained unresponsiveness (SU). However, the long-term effectiveness and safety of FA-AIT are still under investigation. Probiotics, particularly strains of Lactobacillus, play a crucial role in enhancing immune tolerance by promoting regulatory T cells (Tregs) and modulating cytokine profiles. These probiotics can induce semi-mature dendritic cells, enhance CD40 expression, inhibit IL-4 and IL-5, and promote IL-10 and TGF-β, thus contributing to mucosal defense and immunological tolerance. Clinical trials combining probiotics with FA-AIT have demonstrated improved desensitization rates and immune tolerance in food-allergic patients. For example, the combination of Lactobacillus rhamnosus with peanut OIT resulted in a significantly higher rate of SU compared to the placebo group, along with notable immune changes such as reduced peanut-specific IgE and increased IgG4 levels. The review also explores other adjuvants in FA-AIT, such as biologic drugs, which target specific immune pathways to improve treatment outcomes. Additionally, nanoparticles and herbal therapies like food allergy herbal formula 2 (FAHF-2) are discussed for their potential to enhance allergen delivery and immunogenicity, reduce adverse events, and improve desensitization. In conclusion, integrating probiotics and other adjuvants into FA-AIT protocols could significantly enhance the safety and efficacy of FA-AIT, leading to better patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maurizio Mennini
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Marisa Piccirillo
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Silvia Furio
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Francesco Valitutti
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy
| | - Alessandro Ferretti
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Maria De Filippo
- Department of Maternal Infantile and Urological Sciences, AOU Policlinico Umberto I, Rome, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Pasquale Parisi
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Diego Giampietro Peroni
- Section of Pediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Di Nardo
- Pediatric Unit, NESMOS Department, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
3
|
Bhatt S, Patel A, Kesselman MM, Demory ML. Hand Sanitizer: Stopping the Spread of Infection at a Cost. Cureus 2024; 16:e61846. [PMID: 38975405 PMCID: PMC11227450 DOI: 10.7759/cureus.61846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
The recent rise in hand sanitizer use due to the COVID-19 pandemic has had a beneficial impact on stopping the spread of disease, but the potential negative implications of its overuse on the body and the microbiome have yet to be thoroughly reviewed. Epidermal layers absorb hand sanitizer from direct application to the skin, making them some of the most susceptible cells to the adverse effects of overuse. The increased usage of hand sanitizer can affect the variation, quantity, and diversity of the skin microflora, leading to conditions such as eczema, atopic dermatitis, and even systemic toxicity due to colonization of the skin with pathogenic bacteria. Due to the close-knit relationship between the skin and gut, the gastrointestinal system can also incur disruptions due to the negative effects on the skin as a result of excessive hand sanitizer use, leading to gut dysbiosis. Additionally, the accidental ingestion of hand sanitizer, and its abuse or misuse, can be toxic and lead to alcohol poisoning, which is an issue most commonly seen not only in the pediatric population but also in adolescents and adults due to aberrant recreational exposure. As a vulnerable body system, the eyes can also be negatively impacted by hand sanitizer misuse leading to chemical injury, visual impairment, and even blindness. In this review, we aim to highlight the variations in hand sanitizer formulation, the benefits, and how misuse or overuse may lead to adverse effects on the skin, gut, and eyes. In particular, we review the advantages and disadvantages of alcohol-based hand sanitizers (ABHSs) and non-alcohol-based hand sanitizers (NABHSs) and how the components and chemicals used in each can contribute to organ dysbiosis and systemic damage.
Collapse
Affiliation(s)
- Shreya Bhatt
- Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Aasha Patel
- Dentistry, Roseman University College of Dental Medicine, South Jordan, USA
| | - Marc M Kesselman
- Rheumatology, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Michelle L Demory
- Microbiology and Immunology, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, USA
| |
Collapse
|
4
|
Abdi A, Oroojzadeh P, Valivand N, Sambrani R, Lotfi H. Immunological aspects of probiotics for improving skin diseases: Influence on the Gut-Brain-Skin Axis. Biochem Biophys Res Commun 2024; 702:149632. [PMID: 38340656 DOI: 10.1016/j.bbrc.2024.149632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/27/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
The interplay between gut microbiota and human health, both mental and physical, is well-documented. This connection extends to the gut-brain-skin axis, linking gut microbiota to skin health. Recent studies have underscored the potential of probiotics and prebiotics to modulate gut microbiota, supported by in vivo and clinical investigations. In this comprehensive review, we explore the immunological implications of probiotics in influencing the gut-skin axis for the treatment and prevention of skin conditions, including psoriasis, acne, diabetic ulcers, atopic dermatitis, and skin cancer. Our analysis reveals that probiotics exert their effects by modulating cytokine production, whether administered orally or topically. Probiotics bolster skin defenses through the production of antimicrobial peptides and the induction of keratinocyte differentiation and regeneration. Yet, many questions surrounding probiotics remain unanswered, necessitating further exploration of their mechanisms of action in the context of skin diseases.
Collapse
Affiliation(s)
- Ali Abdi
- Medical Immunology, Aziz Sancar Institute of Experimental Medicine, İstanbul University, Istanbul, Turkey
| | - Parvin Oroojzadeh
- Student Research Committee, Department of Food Science and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nassim Valivand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Roshanak Sambrani
- Clinical Research Development Unit of Razi Educational and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
5
|
Feng F, Li R, Tian R, Wu X, Zhang N, Nie Z. The causal relationship between gut microbiota and immune skin diseases: A bidirectional Mendelian randomization. PLoS One 2024; 19:e0298443. [PMID: 38512926 PMCID: PMC10956797 DOI: 10.1371/journal.pone.0298443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/23/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that alterations in gut microbiota are associated with a variety of skin diseases. However, whether this association reflects a causal relationship remains unknown. We aimed to reveal the causal relationship between gut microbiota and skin diseases, including psoriasis, atopic dermatitis, acne, and lichen planus. METHODS We obtained full genetic association summary data for gut microbiota, psoriasis, atopic dermatitis, acne, and lichen planus from public databases and used three methods, mainly inverse variance weighting, to analyze the causal relationships between gut microbiota and these skin diseases using bidirectional Mendelian randomization, as well as sensitivity and stability analysis of the results using multiple methods. RESULTS The results showed that there were five associated genera in the psoriasis group, seven associated genera were obtained in the atopic dermatitis group, a total of ten associated genera in the acne group, and four associated genera in the lichen planus group. The results corrected for false discovery rate showed that Eubacteriumfissicatenagroup (P = 2.20E-04, OR = 1.24, 95%CI:1.11-1.40) and psoriasis still showed a causal relationship. In contrast, in the reverse Mendelian randomization results, there was no evidence of an association between these skin diseases and gut microbiota. CONCLUSION We demonstrated a causal relationship between gut microbiota and immune skin diseases and provide a new therapeutic perspective for the study of immune diseases: targeted modulation of dysregulation of specific bacterial taxa to prevent and treat psoriasis, atopic dermatitis, acne, and lichen planus.
Collapse
Affiliation(s)
- Fei Feng
- Tianjin Medical University, Tianjin, China
| | - Ruicheng Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Tian
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueyi Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nannan Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhenhua Nie
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
6
|
Xie Q, Yuan J, Wang Y. Treating asthma patients with probiotics: a systematic review and meta-analysis. NUTR HOSP 2023; 40:829-838. [PMID: 37073761 DOI: 10.20960/nh.04360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
Introduction Objective: To evaluate the role of probiotics in the treatment of asthma patients by meta-analysis. Methods: PubMed, Embase, The Cochrane Library, Web of Science, and other databases were searched by computer, and the relevant literature on the treatment of asthma by probiotics that met the inclusion criteria was screened by manual retrieval. Meta-analysis was performed using Revman 5.4 software and the combined effect was evaluated by odds ratio (OR) or mean difference (MD) and 95 % confidence interval (CI). Results: a total of ten references were included, all of which were randomized controlled studies, and a total of 1,101 people were investigated. Fractional exhaled nitric oxide (FeNO) (MD = -7.17, 95 % CI: -12.81, -1.54), asthma symptom severity (MD = -0.07, 95 % CI: -0.10, -0.04), Childhood Asthma Control Test (CACT) (MD = 2.26, 95 % CI: 1.14, 3.39), and the number of acute episodes of asthma (OR = 0.30, 95 % CI: 0.19, 0.47) in the probiotics group were better than those in the control group. There was no significant difference in forced expiratory volume in the first second (FEV1) (MD = 0.11, 95 % CI: -0.05, 0.26) and FEV1/FVC (%) (MD = 0.32, 95 % CI: -1.48, 2.12). Conclusion: the use of probiotics in patients with asthma can improve lung inflammation and asthma symptoms, reduce the number of asthma attacks, and have no effect on lung function.
Collapse
Affiliation(s)
- Qiwei Xie
- The Affiliated Hospital of Medical School. Ningbo University
| | - Jiatian Yuan
- The Affiliated Hospital of Medical School. Ningbo University
| | - Yaoweng Wang
- The Affiliated Hospital of Medical School. Ningbo University
| |
Collapse
|
7
|
Hyseni E, Glavas Dodov M. Probiotics in dermatological and cosmetic products – application and efficiency. MAKEDONSKO FARMACEVTSKI BILTEN 2023. [DOI: 10.33320/maced.pharm.bull.2022.68.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The term “probiotics” has first been used in 1907 by Elie Metchnikoff. Since then, probiotics have been part of research not only in regards of digestive health, but also inflammatory diseases. Lately, there has been an increased interest of probiotic’s effects in skincare. The management of atopic dermatitis, acne, psoriasis, photo aging, skin cancer, intimate care, oral care, wound healing is getting harder each passing day, due to increased antibiotic resistance and other side effects of conventional therapy. Therefore, new ingredients have been investigated and probiotics have been proved to be effective in treating various skin conditions.
This review aims to evaluate the scientific evidence on topical and oral probiotics, and to evaluate the efficacy of cosmetic and dermatological products containing probiotics. Many studies have shown that skin and gut microbiome alterations have an important role in skin health. Although this is a new topic in dermatology and cosmetology, there have been some promising results in lots of research studies that the use of probiotics in cosmetic products may help improve the patient’s outcome. While oral probiotics have been shown to promote gut health, which influences the host immune system and helps treat different skin diseases, the mechanism of action of topical probiotics is not yet fully understood. Although the number of commercial probiotic cosmetic products released in the market is increasing and most of the studies have not shown any serious side effect of probiotics, further studies, in larger and heterogeneous groups are needed.
Collapse
Affiliation(s)
- Edita Hyseni
- Center of Pharmaceutical nanotechnology, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, N. Macedonia
| | - Marija Glavas Dodov
- Center of Pharmaceutical nanotechnology, Faculty of Pharmacy, Ss Cyril and Methodius University in Skopje, Majka Tereza 47, 1000 Skopje, N. Macedonia
| |
Collapse
|
8
|
Sadrifar S, Abbasi-Dokht T, Forouzandeh S, Malek F, Yousefi B, Salek Farrokhi A, Karami J, Baharlou R. Immunomodulatory effects of probiotic supplementation in patients with asthma: a randomized, double-blind, placebo-controlled trial. Allergy Asthma Clin Immunol 2023; 19:1. [PMID: 36593510 PMCID: PMC9806812 DOI: 10.1186/s13223-022-00753-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/12/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Asthma is considered to be a chronic inflammatory disorder of the airways. Probiotics are living microorganisms that are found in the human gut and have protective effects against a wide range of diseases such as allergies. The aim of this study was to investigate the improvement of clinical asthma symptoms and changes in the expression pattern of selective microRNAs in patients with asthma and the changes in IL-4 and IFN-γ plasma levels after receiving probiotics. MATERIALS AND METHODS The present study was a randomized, double-blind, placebo-controlled trial that enrolled 40 asthmatic patients. They were treated with probiotics or placebo: 1 capsule/day for 8 weeks. Pulmonary function tests, IL-4 and IFN-γ levels, and expression of microRNAs were assessed at baseline and after treatment. RESULTS The results showed that the expression of miR-16, miR146-a and IL-4 levels in patients with asthma after receiving probiotic supplementation was significantly reduced and miR-133b expression was increased. In addition, pulmonary function tests showed a significant improvement in Forced Expiratory Volume in 1 s and Forced Vital Capacity after receiving probiotics. CONCLUSION In our study, 8-week treatment with probiotic supplementation led to reduced Th2 cells-associated IL-4 and improved Forced Expiratory Volume and Forced Vital Capacity. It appears probiotics can be used in addition to common asthma treatments.
Collapse
Affiliation(s)
- Sina Sadrifar
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Tannaz Abbasi-Dokht
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Sarvenaz Forouzandeh
- grid.486769.20000 0004 0384 8779Department of Internal medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Farhad Malek
- grid.486769.20000 0004 0384 8779Department of Internal medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Amir Salek Farrokhi
- grid.420169.80000 0000 9562 2611Department of Immunology, Pasteur Institute, Tehran, Iran
| | - Jafar Karami
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
| | - Rasoul Baharlou
- grid.486769.20000 0004 0384 8779Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran ,grid.486769.20000 0004 0384 8779Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
9
|
Jin Q, Ren F, Dai D, Sun N, Qian Y, Song P. The causality between intestinal flora and allergic diseases: Insights from a bi-directional two-sample Mendelian randomization analysis. Front Immunol 2023; 14:1121273. [PMID: 36969260 PMCID: PMC10033526 DOI: 10.3389/fimmu.2023.1121273] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
Background Growing evidence shows a significant association between intestinal flora and allergic diseases, specifically atopic dermatitis (AD), allergic rhinitis (AR), and allergic asthma (AA). However, the causality has not yet been clarified. Objective We conducted a bidirectional two-sample Mendelian randomization (TSMR) analysis to study the causal relationships between intestinal flora classification and AD, AR, or AA. Materials and methods We obtained summary data of intestinal flora, AD, AR, and AA from a genome-wide association research. The inverse-variance weighted method is the primary method for analyzing causality in the TSMR analysis. Several sensitivity analyses were conducted to examine the stability of TSMR results. Reverse TSMR analysis was also performed to assess whether there was a reverse causality. Results A total of 7 bacterial taxa associated with AD, AR, and AA were identified by the current TSMR analysis. Specifically, the genus Dialister(P=0.034)and genus Prevotella(P=0.047)were associated with a higher risk of AD, whereas class Coriobacteriia (P=0.034) and its child taxon, order Coriobacteriales (P=0.034) and family Coriobacteriaceae (P=0.034), all had a protective effect on AR. In addition, the family Victivallaceae (P=0.019) was identified as a risk factor for AR. We also noticed a positive association between the genus Holdemanella (P=0.046) and AA. The reverse TSMR analysis didn't suggest any evidence of reverse causality from allergic diseases to the intestinal flora. Conclusion We confirmed the causal relationship between intestinal flora and allergic diseases and provided an innovative perspective for research on allergic diseases: targeted regulation of dysregulation of specific bacterial taxa to prevent and treat AD, AR, and AA.
Collapse
Affiliation(s)
- Qiubai Jin
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feihong Ren
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Dai
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiyun Qian
- Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Ping Song
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Ping Song,
| |
Collapse
|
10
|
Sadrifar S, Abbasi-Dokht T, Forouzandeh S, Malek F, Baharlou R. The impact of multistrains of probiotics on Th17-related cytokines in patients with asthma: a randomized, double-blind, placebo-controlled trial. J Asthma 2022; 60:1306-1315. [PMID: 36332136 DOI: 10.1080/02770903.2022.2144353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Asthma is known as one of the most common chronic inflammatory diseases characterized by recurrent obstruction and inflammation of the airways. Probiotics are defined as a group of beneficial living microorganisms that are beneficial in many disorders, including allergies. The aim of this study was to investigate the probiotic supplement effects on improvement of clinical asthma symptom and changes in the pattern of Th17-related inflammatory cytokines in asthmatic patients. METHODS This was a randomized controlled clinical trial with parallel, double-blind groups. Forty patients with asthma were enrolled and received 1 capsule/day of a probiotic supplement for 8 weeks. Respiratory function tests; and the level of IL-6, IL-17, IL-21 and TGF-β were evaluated at the baseline and end of intervention. RESULTS The results showed that the level of IL-6 and IL-17 in patients after receiving probiotics was reduced and expression of TGF-β was increased as compared to the baseline. Also, the expression of IL-17 and IL-21 in the probiotic group was significantly lower than the placebo group at the end of the intervention. In addition, an improvement in pulmonary function tests and clinical symptoms was observed after receiving probiotics. CONCLUSIONS Eight-weeks treatment with a probiotic supplementation suggests that it may effect on Th17 cells-associated IL-6, IL-17 and TGF-β; and Forced Expiratory Volume in 1 s and Forced Vital Capacity. Taken together, these results suggest that probiotics may have the ability to affect neutrophilic asthma and they can possibly be used besides common treatments for patients with neutrophilic asthma.
Collapse
Affiliation(s)
- Sina Sadrifar
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Tannaz Abbasi-Dokht
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Sarvenaz Forouzandeh
- Department of Internal Medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Farhad Malek
- Department of Internal Medicine, Kosar Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
11
|
Varela-Trinidad GU, Domínguez-Díaz C, Solórzano-Castanedo K, Íñiguez-Gutiérrez L, Hernández-Flores TDJ, Fafutis-Morris M. Probiotics: Protecting Our Health from the Gut. Microorganisms 2022; 10:1428. [PMID: 35889147 PMCID: PMC9316266 DOI: 10.3390/microorganisms10071428] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota (GM) comprises billions of microorganisms in the human gastrointestinal tract. This microbial community exerts numerous physiological functions. Prominent among these functions is the effect on host immunity through the uptake of nutrients that strengthen intestinal cells and cells involved in the immune response. The physiological functions of the GM are not limited to the gut, but bidirectional interactions between the gut microbiota and various extraintestinal organs have been identified. These interactions have been termed interorganic axes by several authors, among which the gut-brain, gut-skin, gut-lung, gut-heart, and gut-metabolism axes stand out. It has been shown that an organism is healthy or in homeostasis when the GM is in balance. However, altered GM or dysbiosis represents a critical factor in the pathogenesis of many local and systemic diseases. Therefore, probiotics intervene in this context, which, according to various published studies, allows balance to be maintained in the GM, leading to an individual's good health.
Collapse
Affiliation(s)
- Gael Urait Varela-Trinidad
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Carolina Domínguez-Díaz
- Doctorado en Ciencias Biomédicas, Con Orientaciones en Inmunología y Neurociencias, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico; (G.U.V.-T.); (C.D.-D.)
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
| | - Karla Solórzano-Castanedo
- Doctorado en Ciencias de la Nutrición Traslacional, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico;
| | - Liliana Íñiguez-Gutiérrez
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
| | - Teresita de Jesús Hernández-Flores
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara, Coronel Calderón 777, Guadalajara 44280, Mexico; (L.Í.-G.); (T.d.J.H.-F.)
- Departamento de Disciplinas Filosóficas Metodológicas e Intrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| | - Mary Fafutis-Morris
- Centro de Investigación en Inmunología y Dermatología (CIINDE), Calzada del Federalismo Nte 3102, Zapopan 45190, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico
| |
Collapse
|
12
|
Chen N, Liu F, Gao Q, Wang R, Zhang L, Li Y. A Meta-Analysis of Probiotics for the Treatment of Allergic Airway Diseases in Children and Adolescents. Am J Rhinol Allergy 2022; 36:480-490. [PMID: 35238209 DOI: 10.1177/19458924221080159] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Allergic airway disease is a chronic airway allergic inflammatory disease including allergic rhinitis (AR) and allergic asthma which is common in children and adolescents. Recently the probiotics has been becoming a supplementary or alternative therapy to allergic diseases, however the effect of them has not been clearly established. OBJECTIVE The purpose of the present meta-analysis was to evaluate the effectiveness of probiotics on allergic airway disease including AR and allergic asthma in children and adolescents. METHODS We performed a comprehensive search on PubMed, Cochrane Library, EMBASE for relevant publications from 1 Jan 2000 to 1 July 2021. Physical examinations, Pediatric Rhinoconjunctivitis Quality of Life Questionnaires (PRQLQs), Total Nasal Symptom Score (TNSS), Nasal or Eye Symptom Score (NSS or ESS), serum allergen-specific IgE, and eosinophil were used as evaluating indicators for AR and allergic asthma in children and adolescents. The meta-analysis was performed using Review Manager (RevMan, Version 5.3). RESULTS 15 randomized controlled trials (RCTs) with a total of 1388 participants were included for the meta-analysis. Among them, 729 patients treated with probiotics served as the probiotics group, and 659 patients with placebo as control group. Significantly greater reduction in PRQLQs from baseline to endpoint (SMD = -2.57, 95% CI [ - 4.66, -0.48] P < 0.01), NSS (SMD = -1.43, 95% CI [ - 1.63, - 1.23], P < 0.01) and ESS (total MD = -1.67, 95% CI [ - 1.79, - 1.55], P < 0.01) were observed in probiotics group compared to control group. Probiotics have no significant effect to serum IgE and eosinophils (P > 0.01). CONCLUSION The results of this meta-analysis indicated that probiotics treatment may reduce PRQLQs, NSS, ESS in patients with allergic airway disease. More research involving the mechanism of probiotics are needed to clarify the role of probiotics in AR and allergic asthma in children and adolescents.
Collapse
Affiliation(s)
- Na Chen
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Fang Liu
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Qiang Gao
- Department of Gastroenterology and Hepatology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lei Zhang
- Department of Otolaryngology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Youwei Li
- Department of Radiology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Chen L, Ni Y, Wu X, Chen G. Probiotics for the prevention of atopic dermatitis in infants from different geographic regions: a systematic review and meta-analysis. J DERMATOL TREAT 2022; 33:2931-2939. [PMID: 35708329 DOI: 10.1080/09546634.2022.2091101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BackgroundAtopic dermatitis (AD) is a common skin disease during infancy, but the preventive effect of probiotics on AD remains unclear.AimTo evaluate the efficacy of probiotic supplementation for the prevention of AD in infants.MethodsPubMed, Cochrane Library, and Embase databases were reviewed for relevant randomized controlled trials (RCTs). Two authors independently extracted the data. The primary endpoint was the risk of AD in infants. Risk ratio (RR) and 95% confidence interval (CI) were calculated.ResultsA total of 20 related articles including 22 studies were included. A significant reduction in AD risk was revealed for the probiotic group compared to the control group (RR 0.76; 95% CI 0.67-0.87). Subgroup analyses by participant receiving intervention revealed that not only probiotics given to mother (RR 0.70; 95% CI 0.57-0.85) or given to mother and infant (RR 0.76; 95% CI 0.61-0.95) were effective in preventing AD in infants, and probiotics given to infants alone (RR 0.76; 95% CI 0.60-0.96) still effectively decreased the risk of AD.ConclusionProbiotic supplementation reduced the risk of developing AD in infants. Furthermore, probiotic supplementation given to mother or to mother and infant could effectively prevent AD in infants.
Collapse
Affiliation(s)
- Lifeng Chen
- Pritzker School of Molecular Engineering, University of Chicago, USA
| | - Yongshan Ni
- Fujian Medical University Union Hospital, Fujian, China
| | - Xingdong Wu
- Xiamen Children's Hospital, Children's Hospital of Fudan University Xiamen Branch, Xiamen, China
| | - Guixia Chen
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
14
|
Saeed NK, Al-Beltagi M, Bediwy AS, El-Sawaf Y, Toema O. Gut microbiota in various childhood disorders: Implication and indications. World J Gastroenterol 2022; 28:1875-1901. [PMID: 35664966 PMCID: PMC9150060 DOI: 10.3748/wjg.v28.i18.1875] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/08/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota has a significant role in gut development, maturation, and immune system differentiation. It exerts considerable effects on the child's physical and mental development. The gut microbiota composition and structure depend on many host and microbial factors. The host factors include age, genetic pool, general health, dietary factors, medication use, the intestine's pH, peristalsis, and transit time, mucus secretions, mucous immunoglobulin, and tissue oxidation-reduction potentials. The microbial factors include nutrient availability, bacterial cooperation or antagonism, and bacterial adhesion. Each part of the gut has its microbiota due to its specific characteristics. The gut microbiota interacts with different body parts, affecting the pathogenesis of many local and systemic diseases. Dysbiosis is a common finding in many childhood disorders such as autism, failure to thrive, nutritional disorders, coeliac disease, Necrotizing Enterocolitis, helicobacter pylori infection, functional gastrointestinal disorders of childhood, inflammatory bowel diseases, and many other gastrointestinal disorders. Dysbiosis is also observed in allergic conditions like atopic dermatitis, allergic rhinitis, and asthma. Dysbiosis can also impact the development and the progression of immune disorders and cardiac disorders, including heart failure. Probiotic supplements could provide some help in managing these disorders. However, we are still in need of more studies. In this narrative review, we will shed some light on the role of microbiota in the development and management of common childhood disorders.
Collapse
Affiliation(s)
- Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Manama 12, Bahrain
- Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Busaiteen 15503, Bahrain
| | - Mohammed Al-Beltagi
- Department of Pediatrics, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Adel Salah Bediwy
- Department of Chest Disease, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
| | - Yasser El-Sawaf
- Department of Tropical Medicine, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Gastroenterology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al-Habib Medical Group, Manama 26671, Bahrain
| | - Osama Toema
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| |
Collapse
|
15
|
Mahmud MR, Akter S, Tamanna SK, Mazumder L, Esti IZ, Banerjee S, Akter S, Hasan MR, Acharjee M, Hossain MS, Pirttilä AM. Impact of gut microbiome on skin health: gut-skin axis observed through the lenses of therapeutics and skin diseases. Gut Microbes 2022; 14:2096995. [PMID: 35866234 PMCID: PMC9311318 DOI: 10.1080/19490976.2022.2096995] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 02/08/2023] Open
Abstract
The human intestine hosts diverse microbial communities that play a significant role in maintaining gut-skin homeostasis. When the relationship between gut microbiome and the immune system is impaired, subsequent effects can be triggered on the skin, potentially promoting the development of skin diseases. The mechanisms through which the gut microbiome affects skin health are still unclear. Enhancing our understanding on the connection between skin and gut microbiome is needed to find novel ways to treat human skin disorders. In this review, we systematically evaluate current data regarding microbial ecology of healthy skin and gut, diet, pre- and probiotics, and antibiotics, on gut microbiome and their effects on skin health. We discuss potential mechanisms of the gut-skin axis and the link between the gut and skin-associated diseases, such as psoriasis, atopic dermatitis, acne vulgaris, rosacea, alopecia areata, and hidradenitis suppurativa. This review will increase our understanding of the impacts of gut microbiome on skin conditions to aid in finding new medications for skin-associated diseases.
Collapse
Affiliation(s)
- Md. Rayhan Mahmud
- Department of Production Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sharmin Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Lincon Mazumder
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Israt Zahan Esti
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Sumona Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | - Mrityunjoy Acharjee
- Department of Bioscience, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | | | | |
Collapse
|
16
|
Kazemi A, Soltani S, Nasri F, Clark CCT, Kolahdouz-Mohammadi R. The effect of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin production: a systematic review and meta-analysis of clinical trials. Int J Food Sci Nutr 2020; 72:632-649. [PMID: 33307904 DOI: 10.1080/09637486.2020.1857710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this systematic review and meta-analysis was to evaluate the effect of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin production. We searched PubMed, Scopus, Web of Science, National Institute of Health Clinical Trials Register, and Cochrane Central Register of Clinical Trials, up to February 2020. All clinical trials that investigated the effects of oral intake of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin (Ig)A, IgE, Japanese cedar pollen (JCP)-specific IgE, IgG, and IgM, for a duration of >7 days were included. Fifty-nine studies met the inclusion criteria, of these 54 studies were included in the analysis. The results indicated a significant increase in salivary IgA secretion rate (SMD = 0.21, 95% CI 0.02-0.39), while no significant effect was observed on other Igs. In conclusion, mentioned supplementation induced a small but significant effect on salivary secretion rate of IgA.
Collapse
Affiliation(s)
- Asma Kazemi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Nasri
- Department Immunology, School of medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cain C T Clark
- Centre Intelligent Healthcare, Coventry University, Coventry, CV15FB, UK
| | - Roya Kolahdouz-Mohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Kim WK, Jang YJ, Han DH, Jeon K, Lee C, Han HS, Ko G. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes 2020; 12:1-14. [PMID: 33016202 PMCID: PMC7553742 DOI: 10.1080/19490976.2020.1819156] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 02/03/2023] Open
Abstract
Administration of probiotics has been linked to immune regulation and changes in gut microbiota composition, with effects on atopic dermatitis (AD). In this study, we investigated amelioration of the symptoms of AD using Lactobacillus paracasei KBL382 isolated from the feces of healthy Koreans. Mice with Dermatophagoides farinae extract (DFE)-induced AD were fed 1 × 109 CFU d-1 of L. paracasei KBL382 for 4 weeks. Oral administration of L. paracasei KBL382 significantly reduced AD-associated skin lesions, epidermal thickening, serum levels of immunoglobulin E, and immune cell infiltration. L. paracasei KBL382-treated mice showed decreased production of T helper (Th)1-, Th2-, and Th17-type cytokines, including thymic stromal lymphopoietin, thymus, and activation-regulated chemokine, and macrophage-derived chemokine, and increased production of the anti-inflammatory cytokine IL-10 and transforming growth factor-β in skin tissue. Intake of L. paracasei KBL382 also increased the proportion of CD4+ CD25+ Foxp3+ regulatory T cells in mesenteric lymph nodes. In addition, administration of L. paracasei KBL382 dramatically changed the composition of gut microbiota in AD mice. Administration of KBL382 significantly ameliorates AD-like symptoms by regulating the immune response and altering the composition of gut microbiota.
Collapse
Affiliation(s)
- Woon-Ki Kim
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - You Jin Jang
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Dae Hee Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Kyungchan Jeon
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Cheonghoon Lee
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Hyuk Seung Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - GwangPyo Ko
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- N-Bio, Seoul National University, Seoul, Republic of Korea
- KoBioLabs, Inc., Seoul, Republic of Korea
- Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Kim WK, Jang YJ, Han DH, Jeon K, Lee C, Han HS, Ko G. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes 2020; 12:1819156. [PMID: 33016202 PMCID: PMC7553742 DOI: 10.1080/19490976.2020.1819156 10.1080/19490976.2020.1819156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Administration of probiotics has been linked to immune regulation and changes in gut microbiota composition, with effects on atopic dermatitis (AD). In this study, we investigated amelioration of the symptoms of AD using Lactobacillus paracasei KBL382 isolated from the feces of healthy Koreans. Mice with Dermatophagoides farinae extract (DFE)-induced AD were fed 1 × 109 CFU d-1 of L. paracasei KBL382 for 4 weeks. Oral administration of L. paracasei KBL382 significantly reduced AD-associated skin lesions, epidermal thickening, serum levels of immunoglobulin E, and immune cell infiltration. L. paracasei KBL382-treated mice showed decreased production of T helper (Th)1-, Th2-, and Th17-type cytokines, including thymic stromal lymphopoietin, thymus, and activation-regulated chemokine, and macrophage-derived chemokine, and increased production of the anti-inflammatory cytokine IL-10 and transforming growth factor-β in skin tissue. Intake of L. paracasei KBL382 also increased the proportion of CD4+ CD25+ Foxp3+ regulatory T cells in mesenteric lymph nodes. In addition, administration of L. paracasei KBL382 dramatically changed the composition of gut microbiota in AD mice. Administration of KBL382 significantly ameliorates AD-like symptoms by regulating the immune response and altering the composition of gut microbiota.
Collapse
Affiliation(s)
- Woon-Ki Kim
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea,Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - You Jin Jang
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Dae Hee Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Kyungchan Jeon
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Cheonghoon Lee
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea,Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Hyuk Seung Han
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - GwangPyo Ko
- Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea,Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea,N-Bio, Seoul National University, Seoul, Republic of Korea,KoBioLabs, Inc., Seoul, Republic of Korea,Center for Human and Environmental Microbiome, Seoul National University, Seoul, Republic of Korea,CONTACT GwangPyo Ko Graduate School of Public Health, Seoul National University, Seoul08826, Republic of Korea
| |
Collapse
|
19
|
Mann EA, Bae E, Kostyuchek D, Chung HJ, McGee JS. The Gut Microbiome: Human Health and Inflammatory Skin Diseases. Ann Dermatol 2020; 32:265-272. [PMID: 33911753 PMCID: PMC7992658 DOI: 10.5021/ad.2020.32.4.265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/16/2019] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
The human microbiome is a rich environment consisting of bacteria, fungi and other commensal microorganisms of the gut, mucosa and skin. The functional role of the gut microbiome includes facilitation in metabolism of macronutrients, maturation of the immune system, and production of pro- or anti-inflammatory signaling molecules and peptides. The identification of these resident organisms has brought about a new understanding of disease processes. Nevertheless, more questions remain regarding the interactions within the microbiome, its interactions with the host, and its contributions to the pathophysiology of disease. The purpose of this review is to examine the existing medical literature to highlight the role of the gut microbiome in human health, also paying attention to its role in several inflammatory skin diseases, namely atopic dermatitis, psoriasis, and rosacea.
Collapse
Affiliation(s)
- Emily A Mann
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Edward Bae
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Darya Kostyuchek
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Hye Jin Chung
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Jean S McGee
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
20
|
Amalia N, Orchard D, Francis KL, King E. Systematic review and meta‐analysis on the use of probiotic supplementation in pregnant mother, breastfeeding mother and infant for the prevention of atopic dermatitis in children. Australas J Dermatol 2019; 61:e158-e173. [DOI: 10.1111/ajd.13186] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Nasya Amalia
- Faculty of Medicine Universitas Indonesia Jakarta Indonesia
- Faculty of Medical Science University of Melbourne Melbourne Australia
| | - David Orchard
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| | - Kate Louise Francis
- Clinical Epidemiology and Biostatistics Unit Murdoch Children's Research Institute The Royal Children's Hospital Melbourne Australia
| | - Emma King
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| |
Collapse
|
21
|
Indira M, Venkateswarulu TC, Abraham Peele K, Nazneen Bobby M, Krupanidhi S. Bioactive molecules of probiotic bacteria and their mechanism of action: a review. 3 Biotech 2019; 9:306. [PMID: 31355115 PMCID: PMC6656846 DOI: 10.1007/s13205-019-1841-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
The bacteria residing in the gut environment do play a pivotal role in metabolic activities of the host. The metabolites produced by these bacteria affect the physiology and health of the host. The gut bacteria are exposed to environmental conditions where multiple factors such as lifestyle, stress, antibiotics, host genetics and infections have an influence on them. In case of pathogenesis of a disease, the gut bacterial composition is altered which leads to a diseased state. This stage is due to colonization of bacterial pathogens in the gut environment. The pathological condition can be alleviated by administering probiotic strains into the gut environment. The probiotic strains produce therapeutic molecules such as amino acids, vitamins, bacteriocins, enzymes, immunomodulatory compounds and short-chain fatty acids. This review discusses recent evidences of the impact of bioactive molecules produced by probiotic bacteria and their mechanism of action in the gut environment to maintain homeostasis and health of the host without any effect on beneficial bacteria sharing the same niche. In addition, the manufacturing challenges of probiotic products for various applications are discussed here.
Collapse
Affiliation(s)
- M. Indira
- Department of Bio-Technology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Andhra Pradesh 522213 India
| | - T. C. Venkateswarulu
- Department of Bio-Technology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Andhra Pradesh 522213 India
| | - K. Abraham Peele
- Department of Bio-Technology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Andhra Pradesh 522213 India
| | - Md. Nazneen Bobby
- Department of Bio-Technology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Andhra Pradesh 522213 India
| | - S. Krupanidhi
- Department of Bio-Technology, Vignan’s Foundation for Science, Technology and Research, Vadlamudi, Andhra Pradesh 522213 India
| |
Collapse
|
22
|
Rusu E, Enache G, Cursaru R, Alexescu A, Radu R, Onila O, Cavallioti T, Rusu F, Posea M, Jinga M, Radulian G. Prebiotics and probiotics in atopic dermatitis. Exp Ther Med 2019; 18:926-931. [PMID: 31384325 DOI: 10.3892/etm.2019.7678] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Recent years have brought additional data on the benefits of prebiotics and probiotics treatment in patients with atopic dermatitis. This review includes all the articles published in PubMed, Scopus, Embase and Cochrane databases until 05.06.2018. The terms used for the search were 'prebiotic', 'probiotic', 'atopic dermatitis', 'Severity Scoring of Atopic Dermatitis', and 'SCORAD'. There was an increase of the intestinal permeability reported in patients with atopic dermatitis and a reduction of the cutaneous microbiome diversity. Probiotics modulate the general microbiome and immune status by improving the intestinal barrier; these effects can be responsible for reducing allergic phenomenon and atopic dermatitis severity. We have structured the results by age groups as infants, 1-18 years, adults, and also pregnancy and lactation. Literature does not offer yet answers on the issues such as the optimal dosing, duration needed to see beneficial effects, the optimal time to start the treatment; the personalized use of probiotics according to colonic dysbiosis may be associated with better results. However, most randomized controlled trials and meta-analyzes support the administration of probiotics for at least 8 weeks in order to obtain beneficial effects in improving severity scoring of atopic dermatitis.
Collapse
Affiliation(s)
- Emilia Rusu
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Georgiana Enache
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania.,Department of Diabetes, Nutrition and Metabolic Diseases, 'Dr. Pompei Samarian' County Emergency Hospital, 910071 Calarasi, Romania
| | - Raluca Cursaru
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Alexandra Alexescu
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Raluca Radu
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Oana Onila
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Teodora Cavallioti
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| | - Florin Rusu
- Department of Urology, 'Dr. Carol Davila' Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Mihaela Posea
- Department of Diabetes, Nutrition and Metabolic Diseases, Smart Nutrition Clinic, 051075 Bucharest, Romania
| | - Mariana Jinga
- Department of Gastroenterology, 'Dr. Carol Davila' Central Military Emergency University Hospital, 051075 Bucharest, Romania
| | - Gabriela Radulian
- Department 2 Infectious Diseases, Epidemiology, Microbiology, Parasitology, Virology, Diabetes, Endocrinology, 'Carol Davila' University of Medicine and Farmacy, 010458 Bucharest, Romania
| |
Collapse
|
23
|
Microbiome of the Skin and Gut in Atopic Dermatitis (AD): Understanding the Pathophysiology and Finding Novel Management Strategies. J Clin Med 2019; 8:jcm8040444. [PMID: 30987008 PMCID: PMC6518061 DOI: 10.3390/jcm8040444] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a long-standing inflammatory skin disease that is highly prevalent worldwide. Multiple factors contribute to AD, with genetics as well as the environment affecting disease development. Although AD shows signs of skin barrier defect and immunological deviation, the mechanism underlying AD is not well understood, and AD treatment is often very difficult. There is substantial data that AD patients have a disturbed microbial composition and lack microbial diversity in their skin and gut compared to controls, which contributes to disease onset and atopic march. It is not clear whether microbial change in AD is an outcome of barrier defect or the cause of barrier dysfunction and inflammation. However, a cross-talk between commensals and the immune system is now noticed, and their alteration is believed to affect the maturation of innate and adaptive immunity during early life. The novel concept of modifying skin and gut microbiome by applying moisturizers that contain nonpathogenic biomass or probiotic supplementation during early years may be a preventive and therapeutic option in high risk groups, but currently lacks evidence. This review discusses the nature of the skin and gut flora in AD, possible mechanisms of skin-gut interaction, and the therapeutic implications of microbiome correction in AD.
Collapse
|
24
|
|
25
|
Swartwout B, Luo XM. Implications of Probiotics on the Maternal-Neonatal Interface: Gut Microbiota, Immunomodulation, and Autoimmunity. Front Immunol 2018; 9:2840. [PMID: 30559747 PMCID: PMC6286978 DOI: 10.3389/fimmu.2018.02840] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022] Open
Abstract
Probiotics are being investigated for the treatment of autoimmune disease by re-balancing dysbiosis induced changes in the immune system. Pregnancy is a health concern surrounding autoimmune disease, both for the mother and her child. Probiotics for maternity are emerging on the market and have gained significant momentum in the literature. Thus far, evidence supports that probiotics alter the structure of the normal microbiota and the microbiota changes significantly during pregnancy. The interaction between probiotics-induced changes and normal changes during pregnancy is poorly understood. Furthermore, there is emerging evidence that the maternal gut microbiota influences the microbiota of offspring, leading to questions on how maternal probiotics may influence the health of neonates. Underpinning the development and balance of the immune system, the microbiota, especially that of the gut, is significantly important, and dysbiosis is an agent of immune dysregulation and autoimmunity. However, few studies exist on the implications of maternal probiotics for the outcome of pregnancy in autoimmune disease. Is it helpful or harmful for mother with autoimmune disease to take probiotics, and would this be protective or pathogenic for her child? Controversy surrounds whether probiotics administered maternally or during infancy are healthful for allergic disease, and their use for autoimmunity is relatively unexplored. This review aims to discuss the use of maternal probiotics in health and autoimmune disease and to investigate their immunomodulatory properties.
Collapse
Affiliation(s)
- Brianna Swartwout
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, VA, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
26
|
Efficacy of Lactobacillus Administration in School-Age Children with Asthma: A Randomized, Placebo-Controlled Trial. Nutrients 2018; 10:nu10111678. [PMID: 30400588 PMCID: PMC6265750 DOI: 10.3390/nu10111678] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 01/01/2023] Open
Abstract
Probiotics may have immunomodulatory effects. However, these effects in asthma remain unclear and warrant clinical trials. Here, we evaluated the effects of Lactobacillus paracasei (LP), Lactobacillus fermentum (LF), and their combination (LP + LF) on the clinical severity, immune biomarkers, and quality of life in children with asthma. This double-blind, prospective, randomized, placebo-controlled trial included 160 children with asthma aged 6–18 years (trial number: NCT01635738), randomized to receive LP, LF, LP + LF, or a placebo for 3 months. Their Global Initiative for Asthma–based asthma severity, Childhood Asthma Control Test (C-ACT) scores, Pediatric Asthma Severity Scores, Pediatric Asthma Quality of Life Questionnaire scores, peak expiratory flow rates (PEFRs), medication use, the levels of immune biomarkers (immunoglobulin E (IgE), interferon γ, interleukin 4, and tumor necrosis factor α) at different visits, and the associated changes were evaluated. Compared with the placebo group by generalized estimating equation model, children receiving LP, LF, and LP + LF had lower asthma severity (p = 0.024, 0.038, and 0.007, respectively) but higher C-ACT scores (p = 0.005, < 0.001, and < 0.001, respectively). The LP + LF group demonstrated increased PEFR (p < 0.01) and decreased IgE levels (p < 0.05). LP, LF, or their combination (LP + LF) can aid clinical improvement in children with asthma.
Collapse
|
27
|
Yousefi B, Eslami M, Ghasemian A, Kokhaei P, Salek Farrokhi A, Darabi N. Probiotics importance and their immunomodulatory properties. J Cell Physiol 2018; 234:8008-8018. [DOI: 10.1002/jcp.27559] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/13/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Bahman Yousefi
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | - Majid Eslami
- Department of Bacteriology and Virology Semnan University of Medical Sciences Semnan Iran
| | | | - Parviz Kokhaei
- Cancer Research Center, and Department of Immunology Semnan University of Medical Sciences Semnan Iran
- Immune and Gene Therapy Lab Cancer Centre Karolinska, Karolinska University Hospital Stockholm Sweden
| | | | - Narges Darabi
- Department of Bacteriology and Virology Semnan University of Medical Sciences Semnan Iran
| |
Collapse
|
28
|
Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol 2018; 9:1459. [PMID: 30042740 PMCID: PMC6048199 DOI: 10.3389/fmicb.2018.01459] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The adult intestine hosts a myriad of diverse bacterial species that reside mostly in the lower gut maintaining a symbiosis with the human habitat. In the current review, we describe the neoteric advancement in our comprehension of how the gut microbiota communicates with the skin as one of the main regulators in the gut-skin axis. We attempted to explore how this potential link affects skin differentiation and keratinization, its influence on modulating the cutaneous immune response in various diseases, and finally how to take advantage of this communication in the control of different skin conditions.
Collapse
Affiliation(s)
- Iman Salem
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Amy Ramser
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nancy Isham
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
29
|
Chen M, Welch M, Laubach S. Preventing Peanut Allergy. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2018; 31:2-8. [PMID: 29588872 DOI: 10.1089/ped.2017.0826] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
The rising prevalence of food allergy and specifically peanut allergy has had significant implications for affected patients, families, and society. The current standard of care remains strict avoidance and the use of emergency medications for accidental ingestions. There is recent evidence-based information to suggest that one approach to preventing peanut allergy lies in early introduction of peanut. This represents a paradigm shift from previous recommendations and has led to updated guidelines in the United States, Europe, and Australasia on the introduction of potentially allergenic foods in the infant diet. This new approach to prevention has some practical obstacles and challenges associated with its implementation. There is also growing interest in the role of maintaining a healthy skin barrier in prevention of sensitization and food allergy. Other approaches, including pro- and prebiotics, prenatal maternal dietary avoidance, breastfeeding, and the use of specific formulas, have not shown reproducibly favorable results. As children with peanut allergy are unlikely to outgrow their food allergy, early oral immunotherapy in those with established peanut allergy is being investigated with the hopes of altering the natural history of an otherwise lifelong disease.
Collapse
Affiliation(s)
- Meng Chen
- Department of Medicine, University of California, San Diego, San Diego, California
| | - Michael Welch
- Division of Allergy, Immunology, and Rheumatology, Rady Children's Hospital, San Diego, California.,Allergy & Asthma Medical Group & Research Center, San Diego, California
| | - Susan Laubach
- Department of Pediatrics, University of California, San Diego, San Diego, California.,Division of Allergy, Immunology, and Rheumatology, Rady Children's Hospital, San Diego, California.,Allergy & Asthma Medical Group & Research Center, San Diego, California
| |
Collapse
|
30
|
Bauer SM. Atopic Eczema: Genetic Associations and Potential Links to Developmental Exposures. Int J Toxicol 2017; 36:187-198. [DOI: 10.1177/1091581817701075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Atopic eczema (AE), or atopic dermatitis (AD), is a common inflammatory skin disease with a disrupted epidermal barrier and an allergic immune response. AD/AE is prominently characterized by a symptomatic itch and transient skin lesions. Infants compose a significant percentage affected. Two models have been proposed to explain AD/AE skin pathology: the gut microbiome-focused inside-outside model and the outside-inside model concentrating on the disrupted skin barrier/skin microbiome. Gene disruptions contributing to epidermal structure, as well as those in immune system genes, are implicated. Over 30 genes have been linked to AD/AE with Flg and Tmem79/Matt alterations being common. Other linked disruptions are in the interleukin-1 family of cytokines/receptors and the TH2 gene family of cytokines. Inheritable epigenetic modifications of the genes or associated proteins may also be involved. Skin barrier disruption and the allergic immune response have been the main foci in mechanistic studies of AD/AE, but the role of the environment is becoming more apparent. Thus, an examination of in utero exposures could be very helpful in understanding the heterogeneity of AD/AE. Although research is limited, there is evidence that developmental exposure to environmental tobacco smoke or phthalates may impact disease. Management for AD/AE includes topical corticosteroids and calcineurin inhibitors, which safely facilitate improvements in select individuals. Disease heterogeneity warrants continued research not only into elucidating disease mechanism(s), via identification of contributing genetic alterations, but also research to understand how/when these genetic alterations occur. This may lead to the cure that those affected by AD/AE eagerly await.
Collapse
Affiliation(s)
- Stephen M. Bauer
- Assistant Professor of Biology, Department of Biology, Belmont Abbey College, Belmont, NC, USA
| |
Collapse
|
31
|
Yang B, Xiao L, Liu S, Liu X, Luo Y, Ji Q, Yang P, Liu Z. Exploration of the effect of probiotics supplementation on intestinal microbiota of food allergic mice. Am J Transl Res 2017; 9:376-385. [PMID: 28337267 PMCID: PMC5340674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/29/2016] [Indexed: 06/06/2023]
Abstract
Environmental factor-induced alterations in intestinal microbiota have been demonstrated to be associated with increasing prevalence of food allergy. However, it is not clear to what extent oral administration of probiotics can affect gut microbiota composition, thus inhibiting food allergy development. Using ovalbumin (OVA)-sensitized murine model, it was demonstrated that probiotics ameliorated allergic symptoms, including reducing OVA specific-IgE, and -IgG1 levels in the serum, Th2 cytokines release in spleen, and occurrence of diarrhea. Moreover, 16S rRNA analysis showed that the probiotics-mediated protection was conferred by an enrichment of Coprococcus and Rikenella. The present study supports the theory that probiotics can treat food allergy by modulating specific genera of the gut microbiota.
Collapse
Affiliation(s)
- Bo Yang
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen UniversityShenzhen 518060, China
| | | | | | - Xiaoyu Liu
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
| | - Yan Luo
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
| | - Qiongmei Ji
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
| | - Pingchang Yang
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen UniversityShenzhen 518060, China
| | - Zhigang Liu
- Institute of Allergy and Immunology, School of Medicine, Shenzhen UniversityShenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen UniversityShenzhen 518060, China
| |
Collapse
|
32
|
Alvi S, Javeed A, Akhtar B, Sharif A, Akhtar MF. Probiotics for cure of Helicobacter pylori infection: A review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2017. [DOI: 10.1080/10942912.2016.1233432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sadia Alvi
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Aqeel Javeed
- Department of Pharmacology and Toxicology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Bushra Akhtar
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Ali Sharif
- Department of Pharmacy, University of Lahore, Lahore, Pakistan
| | | |
Collapse
|
33
|
Juan Z, Zhao-Ling S, Ming-Hua Z, Chun W, Hai-Xia W, Meng-Yun L, Jian-Qiong H, Yue-Jie Z, Xin S. Oral administration of Clostridium butyricum CGMCC0313-1 reduces ovalbumin-induced allergic airway inflammation in mice. Respirology 2017; 22:898-904. [PMID: 28122397 DOI: 10.1111/resp.12985] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Probiotic bacteria can induce immune regulation or immune tolerance in patients with allergic diseases, but the underlying mechanisms are still unclear. There has been a growing interest in the use of beneficial bacteria for allergic diseases recently. This study aimed at exploring whether Clostridium butyricum CGMCC0313-1 (C. butyricum) can reduce ovalbumin (OVA)-induced allergic airway inflammation in a mouse model. METHODS Mouse model of allergic airway inflammation induced via OVA was used in this study. C. butyricum was administered daily by the oral route during or after the sensitization. Airway function, pulmonary airway inflammation, mast cell degranulation, T helper (Th)-specific and anti-inflammatory cytokines, OVA-specific Ig, matrix metalloproteinase 9 (MMP-9) and histopathological alterations were examined. RESULTS C. butyricum significantly reduced lung resistance in the asthmatic mice. Pulmonary airway inflammation, mast cell degranulation, airway remodelling and the expression of OVA-specific IgE/G1 were suppressed by oral C. butyricum. It also reversed the imbalance of Th1/Th2 and increased the anti-inflammatory cytokine IL-10. CONCLUSION C. butyricum reduces OVA-induced allergic airway inflammation in mice and might be an additional or supplementary therapy for allergic asthma.
Collapse
Affiliation(s)
- Zhang Juan
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shi Zhao-Ling
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zeng Ming-Hua
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Chun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wu Hai-Xia
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Liu Meng-Yun
- Respiratory Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Huang Jian-Qiong
- Respiratory Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Zheng Yue-Jie
- Respiratory Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Sun Xin
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Understanding probiotics' role in allergic children: the clue of gut microbiota profiling. Curr Opin Allergy Clin Immunol 2016; 15:495-503. [PMID: 26258924 DOI: 10.1097/aci.0000000000000203] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW To investigate the functional role of gut microbiota in diet-modulated diseases, evaluating probiotic administration effects by systems biology-driven approaches. Understanding the role of host-gut microbial and gut microbe-microbe interactions in either allergic and healthy children may assist in selecting effective and targeted probiotics for personalized therapies. RECENT FINDINGS Food allergy shows a significant increase, especially in Western countries where growing epidemiological data indicate prevalence of small family groups, limited rate of infections in childhood compared with low-income countries, high consumption of sterile foods, hence stimulating a poor trigger of the gut immune system. Therefore, new therapeutic strategies to treat food allergy consist of probiotic administration since early life, thus modulating gut microbiota through immune system stimulation at the mucosal level. SUMMARY Currently, new insights for probiotic selection should take into consideration both phenotyping and genotyping bacterial features and host-microbial cross-talk at gut level, by employing multicomponent systems biology approaches to unveil gut ecosystem dynamics in terms of bacteria phylotypes and their metabolic activities. Moreover, new food processes need to be considered to assess the actual performance of probiotic strains administered to allergic patients. The advent of high-performance platforms employing genomic- and mass spectrometry-based techniques has opened new perspectives on the gut microbiota field, and may now serve as advanced tool to dynamically investigate the interplay between probiotics and gut microbiota ecology under allergic conditions.
Collapse
|
35
|
Peng Y, Li A, Yu L, Qin G. The role of probiotics in prevention and treatment for patients with allergic rhinitis: A systematic review. Am J Rhinol Allergy 2016; 29:292-8. [PMID: 26163249 DOI: 10.2500/ajra.2015.29.4192] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) is a disease of respiratory allergy, and probiotics can provide a potential strategy for its management. The purpose of this study was to carry out a systematic review to investigate the role of probiotics in the prevention and treatment of AR. METHODS We searched for randomized controlled trials (RCTs) of the use of probiotics for the prevention and treatment of AR in the major electronic databases up to March 2014. The quality of the included RCTs was evaluated, and the data were independently extracted by two assessors. Meta-analyses were performed. Continuous data were expressed as the mean difference (MD) or standardized MD with 95% confidence interval (CI). Dichotomous data were expressed as odds ratio with the 95% CI. A p value <0.05 was considered significant. RESULTS A total of 11 RCTs were included in the analysis. Probiotic intake was associated with a significant overall improvement of the quality of life scores and nasal symptom scores of patients with AR (MD -2.97 [95% CI, -4.77 to -1.16)]; p = 0.001). No improvements with regard to prevention or immunologic parameters were noted in the patients with AR. CONCLUSIONS The current evidence is not sufficiently strong to verify a preventive role of probiotics in AR, but probiotics may improve the overall quality of life and nasal symptom scores. Because the available data were generated from only a few trials with a high degree of heterogeneity, routine use of probiotics for prevention and treatment in patients with AR cannot be recommended.
Collapse
Affiliation(s)
- Yi Peng
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan, China
| | | | | | | |
Collapse
|
36
|
Guangorena-Gómez JO, Maravilla-Domínguez A, García-Arenas G, Cervantes-Flores M, Meza-Velázquez R, Rivera-Guillén M, Acosta-Saavedra LC, Goytia-Acevedo RC. Modulation of the immune response by infection with Cryptosporidium spp. in children with allergic diseases. Parasite Immunol 2016; 38:468-80. [PMID: 27150641 DOI: 10.1111/pim.12334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/03/2016] [Indexed: 01/06/2023]
Abstract
It has been demonstrated that the allergic response can be ameliorated by the administration of pathogen derivatives that activate Toll-like receptors and induce a Th1-type immune response (IR). Cryptosporidium is a parasite that promotes an IR via Toll-like receptors and elicits the production of Th1-type cytokines, which limit cryptosporidiosis. The aim of this study was to investigate allergy-related immune markers in children naturally infected with Cryptosporidium. In a cross-sectional study, 49 children with or without clinical diagnosis of allergies, oocysts of Cryptosporidium spp. in the faeces were screened microscopically. We microscopically screened for leucocytes, examined T and B cells for allergy-related activation markers using flow cytometry and evaluated serum for total IgE using chemiluminescence. Children with allergies and Cryptosporidium in the faeces had significantly lower levels of total IgE, B cells, CD19(+) CD23(+) and CD19(+) CD124(+) cells as well as a greater percentage of interferon-gamma (IFN-γ(+) ) and IL-4(+) CD4(+) cells than children with allergies without Cryptosporidium. This is the first description of the modulation of the IR in children with allergic diseases in the setting of natural Cryptosporidium infection. Our findings suggest the involvement of CD4(+) cells producing IL-4 and IFN-γ in the IR to Cryptosporidium in naturally infected children.
Collapse
Affiliation(s)
- J O Guangorena-Gómez
- Facultad de Medicina, Universidad Juárez del Estado de Durango (UJED), Gómez Palacio, Durango, México
| | - A Maravilla-Domínguez
- Facultad de Medicina, Universidad Juárez del Estado de Durango (UJED), Gómez Palacio, Durango, México
| | - G García-Arenas
- Facultad de Medicina, Universidad Juárez del Estado de Durango (UJED), Gómez Palacio, Durango, México
| | | | - R Meza-Velázquez
- Facultad de Medicina, Universidad Juárez del Estado de Durango (UJED), Gómez Palacio, Durango, México.,Centro de Atención a Metales Pesados, Torreón, Coahuila, México
| | - M Rivera-Guillén
- Centro de Atención a Metales Pesados, Torreón, Coahuila, México.,Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | | | - R C Goytia-Acevedo
- Facultad de Medicina, Universidad Juárez del Estado de Durango (UJED), Gómez Palacio, Durango, México
| |
Collapse
|
37
|
Rather IA, Bajpai VK, Kumar S, Lim J, Paek WK, Park YH. Probiotics and Atopic Dermatitis: An Overview. Front Microbiol 2016; 7:507. [PMID: 27148196 PMCID: PMC4828648 DOI: 10.3389/fmicb.2016.00507] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
Abstract
Atopic dermatitis (AD) is a common, recurrent, chronic inflammatory skin disease that is a cause of considerable economic and social burden. Its prevalence varies substantially among different countries with an incidence rate proclaimed to reach up to 20% of children in developed countries and continues to escalate in developing nations. This increased rate of incidence has changed the focus of research on AD toward epidemiology, prevention, and treatment. The effects of probiotics in the prevention and treatment of AD remain elusive. However, evidence from different research groups show that probiotics could have positive effect on AD treatment, if any, that depend on multiple factors, such as specific probiotic strains, time of administration (onset time), duration of exposure, and dosage. However, till date we still lack strong evidence to advocate the use of probiotics in the treatment of AD, and questions remain to be answered considering its clinical use in future. Based on updated information, the processes that facilitate the development of AD and the topic of the administration of probiotics are addressed in this review.
Collapse
Affiliation(s)
- Irfan A Rather
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University Gyeongsan, South Korea
| | - Vivek K Bajpai
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University Gyeongsan, South Korea
| | - Sanjay Kumar
- Department of Clinical Studies, College of Veterinary Medicine, New Bolton Center University of Pennsylvania, Pennsylvania, PA USA
| | - Jeongheui Lim
- National Science Museum, Ministry of Science, ICT and Future Planning, Daejeon South Korea
| | - Woon K Paek
- National Science Museum, Ministry of Science, ICT and Future Planning, Daejeon South Korea
| | - Yong-Ha Park
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University Gyeongsan, South Korea
| |
Collapse
|
38
|
Akelma AZ, Kılıç Topçu Zİ. Probiotics and allergic diseases. World J Immunol 2016; 6:75-82. [DOI: 10.5411/wji.v6.i1.75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Accepted: 01/22/2016] [Indexed: 02/05/2023] Open
Abstract
The prevalence of allergic diseases including atopic dermatitis, asthma, allergic rhinitis (AR) and food allergy is increasing worldwide and they cause a big economic and social burden. Understanding of reasons that contribute to the etiology of allergic diseases as well as new treatment approaches are very important for the follow-up and prevention of these diseases. In recent years, probiotics seem to be promising for allergic diseases. The effect of probiotics in the prevention and treatment of eczema is more extensively studied, but little is known about the association of the microbial flora of the host and allergic airway diseases and the efficacy of probiotics in decreasing the symptoms of patients with asthma and rhinitis. Hitherto, there is no strong evidence for use of probiotics in the treatment of eczema; however, administration of probiotics in breastfeeding mothers in the prenatal period and infants in the postnatal period can be accepted as a safe and helpful option in the prevention of eczema. In contrast, there is not yet reliable evidence or recommendations on use of probiotics for the prevention or treatment of asthma, AR, food allergy, and anaphylaxis currently. More standardized studies should be performed with different strains of probiotics to evaluate the protective and therapeutic effects of probiotics on other allergic diseases as well as eczema. In this review, the relationship between allergy and probiotics is handled in the light of current literature.
Collapse
|
39
|
Mariman R, Reefman E, Tielen F, Persoon-Deen C, van de Mark K, Worms N, Koning F, Nagelkerken L. Lactobacillus plantarum NCIMB8826 ameliorates inflammation of colon and skin in human APOC1 transgenic mice. Benef Microbes 2016; 7:215-25. [DOI: 10.3920/bm2015.0074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genetic predisposition and environmental factors, including the gut microbiota, have been suggested as major factors in the development and progression of atopic dermatitis. Hyperlipidemic human APOC1+/+ transgenic mice display many features of human atopic dermatitis, such as scaling, lichenification, excoriations, and pruritus, along with a disturbed skin barrier function. Cytokine analysis of serum shows an increase of various pro-inflammatory cytokines, including interleukin (IL)-12p40, IL-6, and IL-1α, but lower levels of interferon-γ. These mice also display aspects of colitis evident from macroscopic and histological abnormalities. Genome-wide transcriptome analysis of the intestine shows up-regulation of several genes associated with mast cells and eosinophils and this observation was confirmed by demonstrating increased numbers of IgE+ and FcRε+ mast cells in the colon and in the skin. Oral treatment with Lactobacillus plantarum NCIMB8826 resulted in decreased numbers of mast cells in the colon. Moreover, this L. plantarum strain ameliorated skin pathology, evident from improved skin barrier integrity, absence of skin thickening, and less excoriations. These results suggest that modulation of intestinal immune homeostasis contributes to the suppression of atopic dermatitis.
Collapse
Affiliation(s)
- R. Mariman
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
- Leiden University Medical Centrum, Department of Immunohematology and Bloodtransfusion, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - E. Reefman
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| | - F. Tielen
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| | - C. Persoon-Deen
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| | - K. van de Mark
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| | - N. Worms
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| | - F. Koning
- Leiden University Medical Centrum, Department of Immunohematology and Bloodtransfusion, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - L. Nagelkerken
- TNO, Metabolic Health Research, Zernikedreef 9, 2333 CK Leiden, the Netherlands
| |
Collapse
|
40
|
Cosenza L, Nocerino R, Di Scala C, di Costanzo M, Amoroso A, Leone L, Paparo L, Pezzella C, Aitoro R, Berni Canani R. Bugs for atopy: the Lactobacillus rhamnosus GG strategy for food allergy prevention and treatment in children. Benef Microbes 2016; 6:225-32. [PMID: 25851101 DOI: 10.3920/bm2014.0158] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Food allergy (FA) is a major health issue for children living in Western countries. At this time the only proven treatment for FA is elimination of offender antigen from the diet. It is becoming clear that the development of gut microbiota exerts a profound influence on immune system maturation and tolerance acquisition. Increasing evidence suggests that perturbations in gut microbiota composition of infants are implicated in the pathogenesis of FA. These findings have unveiled new strategies to prevent and treat FA using probiotics bacteria or bacterial substance to limit T-helper (Th)/Th2 bias, which changes during the disease course. Selected probiotics administered during infancy may have a role in the prevention and treatment of FA. Lactobacillus rhamnosus GG (LGG) is the most studied probiotic in this field. Administration of LGG in early life have a role in FA prevention. Preliminary evidence shows that LGG accelerates oral tolerance acquisition in cow's milk allergic infants. We are understanding the mechanisms elicited by LGG and metabolites in influencing food allergen sensitization. A deeper definition of these mechanisms is opening the way to new immunotherapeutics for children affected by FA that can efficiently limit the disease burden.
Collapse
Affiliation(s)
- L Cosenza
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - R Nocerino
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - C Di Scala
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - M di Costanzo
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - A Amoroso
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - L Leone
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - L Paparo
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - C Pezzella
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - R Aitoro
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| | - R Berni Canani
- Department of Translational Medical Science, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy European Laboratory for The Investigation of Food Induced Diseases, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy CEINGE Advanced Biotechnologies, University of Naples 'Federico II', Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
41
|
Xia LP, Jiang Y. [Effect of probiotics in prevention and treatment of allergic diseases in children]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:189-194. [PMID: 26903069 PMCID: PMC7403039 DOI: 10.7499/j.issn.1008-8830.2016.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/15/2015] [Indexed: 06/05/2023]
Abstract
The increasing incidence rate of allergic diseases has attracted global attention, and these diseases greatly threaten children′s health. The common pathogenesis of allergic diseases is the specific IgE- or cell-mediated immune response to common inhalant or food allergens. Epidemiological investigation, analysis of fecal flora, and clinical studies all suggest that the development and progression of allergic diseases are closely related to the early disturbance of intestinal flora. Probiotics can regulate intestinal immune response, increase the barrier function of epithelial cells, inhibit the adhesion and colonization of pathogenic bacteria, and thus restore or reconstruct normal intestinal flora. With the increasing understanding of allergic diseases, the effect of probiotics in the prevention and treatment of such diseases will be taken more and more seriously.
Collapse
Affiliation(s)
- Li-Ping Xia
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | | |
Collapse
|
42
|
Xia LP, Jiang Y. [Effect of probiotics in prevention and treatment of allergic diseases in children]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:189-94. [PMID: 26903069 PMCID: PMC7403039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/15/2015] [Indexed: 11/04/2023]
Abstract
The increasing incidence rate of allergic diseases has attracted global attention, and these diseases greatly threaten children′s health. The common pathogenesis of allergic diseases is the specific IgE- or cell-mediated immune response to common inhalant or food allergens. Epidemiological investigation, analysis of fecal flora, and clinical studies all suggest that the development and progression of allergic diseases are closely related to the early disturbance of intestinal flora. Probiotics can regulate intestinal immune response, increase the barrier function of epithelial cells, inhibit the adhesion and colonization of pathogenic bacteria, and thus restore or reconstruct normal intestinal flora. With the increasing understanding of allergic diseases, the effect of probiotics in the prevention and treatment of such diseases will be taken more and more seriously.
Collapse
Affiliation(s)
- Li-Ping Xia
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | | |
Collapse
|
43
|
Three Novel Candidate Probiotic Strains with Prophylactic Properties in a Murine Model of Cow's Milk Allergy. Appl Environ Microbiol 2016; 82:1722-1733. [PMID: 26729723 DOI: 10.1128/aem.03440-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/29/2015] [Indexed: 01/19/2023] Open
Abstract
Food allergies can have significant effects on morbidity and on quality of life. Therefore, the development of efficient approaches to reduce the risk of developing food allergies is of considerable interest. The aim of this study was to identify and select probiotic strains with preventive properties against allergies using a combination of in vitro and in vivo approaches. To that end, 31 strains of bifidobacteria and lactic acid bacteria were screened for their immunomodulatory properties in two cellular models, namely, human peripheral blood mononuclear cells (PBMCs) and T helper 2 (Th2)-skewed murine splenocytes. Six strains inducing a high interleukin-10 (IL-10)/IL-12p70 ratio and a low secretion of IL-4 on the two cellular models were selected, and their protective impact was tested in vivo in a murine model of food allergy to β-lactoglobulin. Three strains showed a protective impact on sensitization, with a decrease in allergen-specific IgE, and on allergy, with a decrease in mast cell degranulation. Analysis of the impact of these three strains on the T helper balance revealed different mechanisms of action. The Lactobacillus salivarius LA307 strain proved to block Th1 and Th2 responses, while the Bifidobacterium longum subsp. infantis LA308 strain induced a pro-Th1 profile and the Lactobacillus rhamnosus LA305 strain induced pro-Th1 and regulatory responses. These results demonstrate that a combination of in vitro and in vivo screening is effective in probiotic strain selection and allowed identification of three novel probiotic strains that are active against sensitization in mice.
Collapse
|
44
|
Besseling-van der Vaart I, Heath MD, Guagnini F, Kramer MF. In vitro evidence for efficacy in food intolerance for the multispecies probiotic formulation Ecologic® Tolerance (Syngut™). Benef Microbes 2015; 7:111-118. [PMID: 26565083 DOI: 10.3920/bm2015.0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The beneficial effects of probiotics are currently the subject of extensive studies in health and medical research. The aim of this research was to specifically design a new probiotic formulation for supplementation in people suffering from food intolerance. The selection of strains was focussed on the capacity to influence mechanisms of action that are important in development of food intolerance with the following parameters measure: in vitro capacity to produce β-galactosidase, in vitro strengthening of the epithelial barrier, in vitro stimulation of cytokines produced by regulatory T cells, in addition to assessing fundamental quality criteria (stability, gastrointestinal (GI)-survival, multispecies concept, allergen-free). Ecologic®Tolerance/Syngut™ was subsequently developed consisting of a multispecies concept using 4 different probiotic strains (Bifidobacterium lactis W51, Lactobacillus acidophilus W22, Lactobacillus plantarum W21 and Lactococcus lactis W19). Each of these strains demonstrated ability to survive the GI-tract and strain specific effects in producing β-galactosidase, strengthening the gut barrier function after immunological-induced stress and inhibiting Th2 cytokines (IL-4, IL-5 and IL-13 (≥50%), in addition to stimulating interleukin-10 levels; thus, providing in vitro evidence for the efficacy of the selected strains to provide beneficial effects in patients suffering from food intolerance.
Collapse
Affiliation(s)
| | - M D Heath
- 2 Allergy Therapeutics plc., Dominion Way, Worthing, West Sussex BN14 8SA, United Kingdom
| | - F Guagnini
- 3 Allergy Therapeutics Italia, Via IV Novembre 76, 20019 Settimo Milanese, Italy
| | - M F Kramer
- 4 Bencard Allergy GmbH, Messerschmitstrasse 4, 80992 München, Germany
| |
Collapse
|
45
|
Akelma AZ, Biten AA. Probiotics and infantile atopic eczema. PEDIATRIC HEALTH MEDICINE AND THERAPEUTICS 2015; 6:147-151. [PMID: 29388601 PMCID: PMC5683264 DOI: 10.2147/phmt.s50476] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pediatric eczema is a common disease which causes economic and social burden. Its incidence differs among the societies, with an incidence reported to reach up to 20% in developed countries. Eczema is the first allergic disease seen in the childhood, and it is recognized as a precursor for the development of atopic diseases such as asthma, allergic rhinitis, and food allergy in the forthcoming years of children. Increased incidence of eczema in recent years has led to new research in epidemiology, prevention, and intervention of this disease. It is no doubt important to treat itching, rash, and excoriation of the skin; however, treatment of pediatric eczema should not be considered only as a treatment of skin lesions. Considering skin treatment as the tip of the iceberg, proper management of the allergic processes can be accepted as the rest of the iceberg. The role of probiotics in the prevention of atopic eczema is yet to be clarified. Evidence presented by existing studies suggesting that probiotics may prevent pediatric eczema is not strong enough. A positive effect, if any, may be related with onset time, dose, duration, and use of specific probiotics. To date, there is no strong evidence for use of probiotics in the treatment of eczema; however, administration of probiotics in breast-feeding mothers in the prenatal period and in infants in the postnatal period can be accepted as a safe and helpful option in the prevention of eczema. Nevertheless, there are still questions to be answered in the future about probiotic administration for eczema. Clinical use of probiotics will gradually become more widespread when these questions are answered. Based on current information, the administration of probiotics for pediatric eczema topic is addressed in this review.
Collapse
Affiliation(s)
- Ahmet Zülfikar Akelma
- Pediatric Allergy and Immunology Unit, Ankara Kecioren Teaching and Research Hospital, Ankara, Turkey
| | - Aziz Alper Biten
- General Directorate of Management Services, Republic of Turkey Ministry of Health, Ankara, Turkey
| |
Collapse
|
46
|
Regulation of TWIK-related potassium channel-1 (Trek1) restitutes intestinal epithelial barrier function. Cell Mol Immunol 2015; 13:110-8. [PMID: 25683610 DOI: 10.1038/cmi.2014.137] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/26/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022] Open
Abstract
The disruption of epithelial barrier integrity is an important factor in the pathogenesis of various immune disorders. However, the restitution of the compromised barrier functions is difficult. This study investigates the regulation of TWIK-related potassium channel-1 (Trek1) in the restitution of intestinal epithelial barrier functions. The human colon epithelial cell line T84 was cultured in monolayers and used to observe epithelial barrier functions in vitro. An intestinal allergy mouse model was created. Cytokine levels were determined by enzyme-linked immunosorbent assay and western blotting. The results showed that Trek1 deficiency induced T84 monolayer barrier disruption. Allergic responses markedly suppressed the expression of Trek1 in the intestinal epithelia via activating the mitogen-activated protein kinase pathways and increasing the expression of histone deacetylase-1. The inhibition of histone deacetylase-1 by sodium butyrate or the administration of a butyrate-producing probiotic (Clostridium butyricum) restored the intestinal epithelial barrier functions and markedly enhanced the effect of antigen-specific immunotherapy. The data suggest that Trek1 is required for the maintenance of intestinal epithelial barrier integrity. Allergic responses induce an insufficiency of Trek1 expression in the intestinal epithelia. Trek1 expression facilitates the restoration of intestinal epithelial barrier functions in an allergic environment.
Collapse
|
47
|
Wu CT, Chen PJ, Lee YT, Ko JL, Lue KH. Effects of immunomodulatory supplementation with Lactobacillus rhamnosus on airway inflammation in a mouse asthma model. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2014; 49:625-635. [PMID: 25440975 DOI: 10.1016/j.jmii.2014.08.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/15/2014] [Accepted: 08/07/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Asthma is a common allergic disease. In previous studies, probiotics improved the balance of intestinal microbes, reduced inflammation, and promoted mucosal tolerance. This study investigated whether oral administrations of Lactobacillus rhamnosus GG (LGG) inhibited allergen (ovalbumin or OVA)-induced airway inflammation in a mouse asthma model. METHODS The allergy/asthma animal model in this study was sensitization with OVA. After intranasal challenge with OVA, the airway inflammation and hyper-responsiveness were determined by a Buxco system, bronchoalveolar lavage fluid analysis with Liu stain, and enzyme-linked immunosorbent assay. Histopathologic changes in the lung were detected by hematoxylin and eosin staining and immunohistochemistry staining. RESULTS Both pre- and post-treatment with LGG suppressed the airway hyper-responsiveness to methacholine and significantly decreased the number of infiltrating inflammatory cells and Th2 cytokines in bronchoalveolar lavage fluid and serum compared with the OVA-sensitized mice. In addition, LGG reduced OVA-specific IgE levels in serum. Oral LGG decreased matrix metalloproteinase 9 expression in lung tissue and inhibited inflammatory cell infiltration. CONCLUSION LGG had an anti-inflammatory effect on OVA-induced airway inflammation and might be an additional or supplementary therapy for allergic airway diseases.
Collapse
Affiliation(s)
- Chia-Ta Wu
- Department of Emergency Medicine, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Peng-Jung Chen
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Tzu Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ko-Haung Lue
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
48
|
Treatment of atopic dermatitis eczema with a high concentration of Lactobacillus salivarius LS01 associated with an innovative gelling complex: a pilot study on adults. J Clin Gastroenterol 2014; 48 Suppl 1:S47-51. [PMID: 25291127 DOI: 10.1097/mcg.0000000000000249] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
GOALS To evaluate the efficacy of a highly concentrated Lactobacillus salivarius preparation containing a gelling complex formed by Streptococcus thermophilus ST10 and tara gum in the treatment of atopic dermatitis (AD). BACKGROUND Previous studies have demonstrated an improvement in AD symptoms after administration of the probiotic strain L. salivarius LS01. S. thermophilus ST10 and tara gum create a gelling complex that adheres to intestinal mucus and improves barrier function. STUDY A prospective, controlled pilot trial was carried out to evaluate how the association of S. thermophilus ST10 and tara gum could improve the activity of L. salivarius LS01 administered at high doses to adults with AD. Twenty-five patients were included into the study: 13 were treated for 1 month with the active formulation, whereas 12 represented the placebo group. Scoring Atopic Dermatitis index was determined before and at the end of probiotic administration. Fecal samples were also collected to evaluate changes in bacterial counts of Staphylococcus aureus and clostridia. RESULTS A significant improvement in SCORAD index was observed in the probiotic group after 1 month of treatment, whereas no significant changes occurred in placebo patients. A slight decrease in fecal S. aureus count was observed in probiotic-treated patients. CONCLUSIONS Data obtained in this study suggest a potential role for L. salivarius LS01 in the treatment of AD. The addition of tara gum and S. thermophilus ST10 seems to improve the overall efficacy of the probiotic strain, in particular shortening the time required for the onset of the positive effects. Further studies to investigate the activity of this preparation are advisable.
Collapse
|
49
|
Inoue Y, Shimojo N. Microbiome/microbiota and allergies. Semin Immunopathol 2014; 37:57-64. [DOI: 10.1007/s00281-014-0453-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/02/2014] [Indexed: 02/07/2023]
|
50
|
Yang SI, Lee E, Jung YH, Kim HY, Seo JH, Kwon JW, Kim BJ, Kim HB, Lee SY, Jang GC, Kim WK, Shim JY, Kang MJ, Yu HS, Hong SJ. Effect of antibiotic use and mold exposure in infancy on allergic rhinitis in susceptible adolescents. Ann Allergy Asthma Immunol 2014; 113:160-165.e1. [PMID: 24973272 DOI: 10.1016/j.anai.2014.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 04/29/2014] [Accepted: 05/21/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Antibiotic use in infancy induces alteration in intestinal microbiota and is associated with the development of allergic diseases. Mold exposure is also associated with allergic diseases. Genetic susceptibility may interact with specific environmental factors in allergic disease development. OBJECTIVE To investigate independent and combined effects of antibiotic use and mold exposure in infancy on the risk of allergic rhinitis (AR) in adolescents. METHODS Data on AR and environmental factors were collected using the International Study of Asthma and Allergies in Childhood questionnaire from 7,389 adolescents from Seoul, Korea. TaqMan genotyping was performed for interleukin 13 (IL-13) (rs20541) and Toll-like receptor 4 (rs1927911) polymorphisms in 1,395 adolescents. RESULTS Age, parental history of AR, antibiotic use in infancy, and pet ownership during pregnancy or infancy were associated with an increased risk of current AR (diagnosis of AR and symptoms of AR within the preceding 12 months). Having older siblings was a protective effect. The adjusted odds ratio (aOR) for current AR for combined antibiotic use and mold exposure in infancy was 1.45 (95% confidence interval [CI], 1.01-2.09). For each factor separately, aORs were 1.25 (95% CI, 1.04-1.50) and 0.99 (95% CI, 0.75-1.31), respectively. Antibiotic and mold exposure in infancy, GA or AA genotypes of IL-13 (rs20541) (aOR 4.53; 95% CI, 1.66-12.38; P for interaction = .05), and CT+TT genotype of Toll-like receptor 4 (rs1927911) (aOR, 3.20; 95% CI, 1.24-8.26; P for interaction = .18) increased the risk of current AR. CONCLUSION Antibiotic use and mold exposure in infancy have additive effects on the risk of current AR in genetically susceptible adolescents. Gene-environment interactions between IL-13 (rs20541) and antibiotics or mold may play a role in AR.
Collapse
Affiliation(s)
- Song-I Yang
- Department of Pediatrics, Childhood Asthma Atopy Center, Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Ho Jung
- Department of Pediatrics, Childhood Asthma Atopy Center, Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung Young Kim
- Department of Pediatrics, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Ju-Hee Seo
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| | - Ji-Won Kwon
- Department of Pediatrics, Seoul National University Bundang Hospital, Seungnam, Korea
| | - Byoung-Ju Kim
- Department of Pediatrics, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Hyo-Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Hallym Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Gwang Cheon Jang
- Department of Pediatrics, National Health Insurance Corporation Ilsan Hospital, Goyang, Korea
| | - Woo-Kyung Kim
- Department of Pediatrics and the Allergy and Respiratory Research Laboratory, Inje University Seoul Paik Hospital, Seoul, Korea
| | - Jung Yeon Shim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mi-Jin Kang
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho-Sung Yu
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Research Center for Standardization of Allergic Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|