1
|
Zhang M, Li D, Sun L, He Y, Liu Q, He Y, Li F. Lactobacillus reuteri Alleviates Hyperoxia-Induced BPD by Activating IL-22/STAT3 Signaling Pathway in Neonatal Mice. Mediators Inflamm 2024; 2024:4965271. [PMID: 39687635 PMCID: PMC11649352 DOI: 10.1155/mi/4965271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic respiratory disease in preterm infants. Little is known about the regulatory effect of lung Lactobacillus and its mechanism in BPD. This study explored the effect of L. reuteri on hyperoxia-induced mice lung injuries and examined whether L. reuteri played a role via the IL-22/STAT3 pathway. We found that the intranasal administration of L. reuteri and its tryptophan metabolite indole-3-aldehyde (3-IAld) ameliorated hyperoxia-induced mice lung BPD-like changes, deceased proinflammatory cytokines (IL-1β, IL-6, and TNF-α), and increased the levels of surfactant-associated protein C (SPC), aquaporin 5 (AQP5), and vascular endothelial growth factor receptor 2 (VEGFR2, also known as FLK-1). Furthermore, L. reuteri and 3-IAld increased the expression of IL-22. IL-22 was also confirmed to ameliorate hyperoxia-induced mice lung pathological changes, and the protective effects of L. reuteri could be inhibited by anti-IL-22 neutralizing antibody. Finally, we confirmed STAT3 activation by IL-22 in MLE-12 cells. In summary, our study confirmed L. reuteri alleviated hyperoxia-induced lung BPD-like changes in mice by activating the IL-22/STAT3 signaling pathway via IL-22 production. Probiotics Lactobacillus is a potential treatment for hyperoxia-induced lung injury in newborns.
Collapse
Affiliation(s)
- Meiyu Zhang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Department of Neonatology Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Decai Li
- Department of Pediatrics Chongqing Health Center for Women and Children, Department of Pediatrics Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Liujuan Sun
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Department of Neonatology Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Yu He
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Department of Neonatology Children's Hospital of Chongqing Medical University, Chongqing 400015, China
| | - Qingqing Liu
- Department of Pediatrics Chongqing Health Center for Women and Children, Department of Pediatrics Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Yi He
- Department of Pediatrics Chongqing Health Center for Women and Children, Department of Pediatrics Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Fang Li
- Department of Pediatrics Chongqing Health Center for Women and Children, Department of Pediatrics Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
2
|
Kleniewska P, Pawliczak R. Can probiotics be used in the prevention and treatment of bronchial asthma? Pharmacol Rep 2024; 76:740-753. [PMID: 38951480 PMCID: PMC11294272 DOI: 10.1007/s43440-024-00618-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Asthma is a lifelong condition with varying degrees of severity and susceptibility to symptom control. Recent studies have examined the effects of individual genus, species, and strains of probiotic microorganisms on the course of asthma. The present review aims to provide an overview of current knowledge on the use of probiotic microorganisms, mainly bacteria of the genus Lactobacillus and Bifidobacterium, in asthma prevention and treatment. Recent data from clinical trials and mouse models of allergic asthma indicate that probiotics have therapeutic potential in this condition. Animal studies indicate that probiotic microorganisms demonstrate anti-inflammatory activity, attenuate airway hyperresponsiveness (AHR), and reduce airway mucus secretion. A randomized, double-blind, placebo-controlled human trials found that combining multi-strain probiotics with prebiotics yielded promising outcomes in the treatment of clinical manifestations of asthma. It appears that probiotic supplementation is safe and significantly reduces the frequency of asthma exacerbations, as well as improved forced expiratory volume and peak expiratory flow parameters, and greater attenuation of inflammation. Due to the small number of available clinical trials, and the use of a wide range of probiotic microorganisms and assessment methods, it is not possible to draw clear conclusions regarding the use of probiotics as asthma treatments.
Collapse
Affiliation(s)
- Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9, Łódź, 90-752, Poland.
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9, Łódź, 90-752, Poland
| |
Collapse
|
3
|
Glieca S, Quarta E, Bottari B, Lal VC, Sonvico F, Buttini F. The role of airways microbiota on local and systemic diseases: a rationale for probiotics delivery to the respiratory tract. Expert Opin Drug Deliv 2024; 21:991-1005. [PMID: 39041243 DOI: 10.1080/17425247.2024.2380334] [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: 02/15/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Recent discoveries in the field of lung microbiota have enabled the investigation of new therapeutic interventions involving the use of inhaled probiotics. AREAS COVERED This review provides an overview of what is known about the correlation between airway dysbiosis and the development of local and systemic diseases, and how this knowledge can be exploited for therapeutic interventions. In particular, the review focused on attempts to formulate probiotics that can be deposited directly on the airways. EXPERT OPINION Despite considerable progress since the emergence of respiratory microbiota restoration as a new research field, numerous clinical implications and benefits remain to be determined. In the case of local diseases, once the pathophysiology is understood, manipulating the lung microbiota through probiotic administration is an approach that can be exploited. In contrast, the effect of pulmonary dysbiosis on systemic diseases remains to be clarified; however, this approach could represent a turning point in their treatment.
Collapse
Affiliation(s)
| | - Eride Quarta
- Food and Drug Department, University of Parma, Parma, Italy
| | | | | | - Fabio Sonvico
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parma, Italy
| |
Collapse
|
4
|
Chaudhary PP, Kaur M, Myles IA. Does "all disease begin in the gut"? The gut-organ cross talk in the microbiome. Appl Microbiol Biotechnol 2024; 108:339. [PMID: 38771520 PMCID: PMC11108886 DOI: 10.1007/s00253-024-13180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
The human microbiome, a diverse ecosystem of microorganisms within the body, plays pivotal roles in health and disease. This review explores site-specific microbiomes, their role in maintaining health, and strategies for their upkeep, focusing on oral, lung, vaginal, skin, and gut microbiota, and their systemic connections. Understanding the intricate relationships between these microbial communities is crucial for unraveling mechanisms underlying human health. Recent research highlights bidirectional communication between the gut and distant microbiome sites, influencing immune function, metabolism, and disease susceptibility. Alterations in one microbiome can impact others, emphasizing their interconnectedness and collective influence on human physiology. The therapeutic potential of gut microbiota in modulating distant microbiomes offers promising avenues for interventions targeting various disorders. Through interdisciplinary collaboration and technological advancements, we can harness the power of the microbiome to revolutionize healthcare, emphasizing microbiome-centric approaches to promote holistic well-being while identifying areas for future research.
Collapse
Affiliation(s)
- Prem Prashant Chaudhary
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Mahaldeep Kaur
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
5
|
Kim HS, Kim B, Holzapfel WH, Kang H. Lactiplantibacillusplantarum APsulloc331261 (GTB1 ™) promotes butyrate production to suppress mucin hypersecretion in a murine allergic airway inflammation model. Front Microbiol 2024; 14:1292266. [PMID: 38449878 PMCID: PMC10915089 DOI: 10.3389/fmicb.2023.1292266] [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: 09/11/2023] [Accepted: 12/15/2023] [Indexed: 03/08/2024] Open
Abstract
Introduction Allergic airway diseases are one of the serious health problems in worldwide and allergic airway inflammation is a prerequisite led to the exacerbated situation such as mucus hypersecretion, epithelial barrier damage and microbiota dysbiosis. Because of side effects and low efficiencies of current therapeutics, the need for novel alternatives has been urged. Probiotics in which have diverse and beneficial modulatory effects have been applied to the airway inflammation model and the underlying mechanism needs to be investigated. Methods We aimed to evaluate whether our target strain, Lactiplantibacillus plantarum APsulloc331261 (GTB1TM) isolated from green tea, can ameliorate allergic airway inflammation in mice and to figure out the mechanism. We induced allergic airway inflammation to mice by ovalbumin (OVA) and administered GTB1 orally and the immune and epithelial barrier markers were assessed. The gut metabolite and microbiota were also analysed, and the in vitro cell-line experiment was introduced to confirm the hypothesis of the study. Results GTB1 ameliorated type 2 inflammation and suppressed mucin hypersecretion with the inhibition of MUC5AC in inflamed mice. Moreover, GTB1 increased the butyrate production and the relative abundance of butyrate producer, Clostridium cluster IV. We assumed that butyrate may have a potential role and investigated the effect of butyrate in mucin regulation via human airway epithelial cell line, A549. Butyrate significantly reduced the gene expression of MUC5AC in A549 cells suggesting its regulatory role in mucus production. Conclusion Therefore, our study demonstrates that the oral administration of GTB1 can ameliorate allergic airway inflammation and mucin hypersecretion by butyrate production.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- HEM Pharma, Pohang, Republic of Korea
| | - Bobae Kim
- HEM Pharma, Pohang, Republic of Korea
| | - Wilhelm H. Holzapfel
- Department of Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- HEM Pharma, Pohang, Republic of Korea
| | | |
Collapse
|
6
|
Gupta N, Abd EL-Gawaad N, Osman Abdallah SA, Al-Dossari M. Possible modulating functions of probiotic Lactiplantibacillus plantarum in particulate matter-associated pulmonary inflammation. Front Cell Infect Microbiol 2024; 13:1290914. [PMID: 38264731 PMCID: PMC10803600 DOI: 10.3389/fcimb.2023.1290914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Pulmonary disease represents a substantial global health burden. Increased air pollution, especially fine particulate matter (PM2.5) is the most concerned proportion of air pollutants to respiratory health. PM2.5 may carry or combine with other toxic allergens and heavy metals, resulting in serious respiratory allergies and anaphylactic reactions in the host. Available treatment options such as antihistamines, steroids, and avoiding allergens/dust/pollutants could be limited due to certain side effects and immense exposure to air pollutants, especially in most polluted countries. In this mini-review, we summarized how PM2.5 triggers respiratory hyperresponsiveness and inflammation, and the probiotic Lactiplantibacillus plantarum supplementation could minimize the risk of the same. L. plantarum may confer beneficial effects in PM2.5-associated pulmonary inflammation due to significant antioxidant potential. We discussed L. plantarum's effect on PM2.5-induced reactive oxygen species (ROS), inflammatory cytokines, lipid peroxidation, and DNA damage. Available preclinical evidence shows L. plantarum induces gut-lung axis, SCFA, GABA, and other neurotransmitter signaling via gut microbiota modulation. SCFA signals are important in maintaining lung homeostasis and regulating intracellular defense mechanisms in alveolar cells. However, significant research is needed in this direction to contemplate L. plantarum's therapeutic potential in pulmonary allergies.
Collapse
Affiliation(s)
- Nishant Gupta
- Medical Research and Development, River Engineering, Greater Noida, India
| | - N.S. Abd EL-Gawaad
- Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - M. Al-Dossari
- Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
7
|
Kim HS, Oh H, Kim B, Ji Y, Holzapfel WH, Kang H, Arellano-Ayala K. Multifunctional effects of Lactobacillus sakei HEM 224 on the gastrointestinal tract and airway inflammation. Sci Rep 2023; 13:17918. [PMID: 37864021 PMCID: PMC10589218 DOI: 10.1038/s41598-023-45043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/15/2023] [Indexed: 10/22/2023] Open
Abstract
Mucosal tissues serve as the first defense line and their commensal microbiota play a role in sustaining of host health. This study aimed to isolate and evaluate a putative probiotic strain on various mucosal regions. Lactobacillus sakei HEM 224 was isolated from traditional Korean kimchi and identified. In the safety assessment L. sakei HEM 224 showed negative results for hemolysis, biogenic amine production and transferable antibiotic resistance. The probiotic potential of strain HEM 224 in diverse mucosal areas was shown in two different models, viz. a murine model with colitis induced by dextran sulfate sodium (DSS) and an allergic airway inflammation model induced by ovalbumin (OVA). In the colitis model, oral administration of L. sakei HEM 224 improved colitis physiology with immunomodulation, enhancing barrier components and gut microbiota alteration. In the allergic airway inflammation model, the intranasal administration of the strain decreased type 2 inflammation and enhanced epithelial barrier integrity from the airways. These results demonstrate that L. sakei HEM 224 can ameliorate inflammatory conditions in both the gastrointestinal and respiratory tracts through the reinforcement of the epithelial barrier and immunomodulation.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Hanna Oh
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Yosep Ji
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Convergence, Handong Global University, 558, Handong-ro, Pohang, Gyeongbuk, 37554, Republic of Korea.
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea.
| | - Hyeji Kang
- HEM Pharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea.
- Global Green Research and Development Institute, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
| | - Karina Arellano-Ayala
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, Vienna, 1030, Austria
| |
Collapse
|
8
|
Niu X, Yin X, Wu X, Zhang Q, Jiang Y, He J, Zhao Y, Zhang C, Ren Y, Lai M, Sang Y, Wang R. Heat-Killed Bifidobacterium longum BBMN68 in Pasteurized Yogurt Alleviates Mugwort Pollen-Induced Allergic Airway Responses through Gut Microbiota Modulation in a Murine Model. Foods 2023; 12:2049. [PMID: 37238867 DOI: 10.3390/foods12102049if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 07/26/2024] Open
Abstract
Many probiotic bacteria have been proven to prevent allergic airway responses through immunomodulation. This study was conducted to evaluate the effects of heat-killed Bifidobacterium longum BBMN68 (BBMN68) in pasteurized yogurt on the alleviation of mugwort pollen (MP)-induced allergic inflammation. BALB/c mice aged 5-6 weeks were randomly assigned and fed pasteurized yogurt containing heat-killed BBMN68 for 27 days, followed by allergic sensitization and challenge with MP extract. The allergic mice that received pasteurized yogurt containing heat-killed BBMN68 had improved immune status, including a lower serum IgE level, decreased serum interleukin (IL)-4, IL-5, and IL-13 concentrations, and alleviated airway inflammation manifested by increased macrophage and decreased eosinophil and neutrophil counts in BALF, as well as airway remodeling and suppressed peribronchial cellular infiltration. Moreover, oral administration of pasteurized yogurt containing heat-killed BBMN68 significantly modulated gut microbiota composition by influencing the proportion of beneficial genera associated with inflammation and immunity, such as Lactobacillus, Candidatus_Saccharimonas, Odoribacter, and Parabacteroides, which also negatively correlated with serum IgE and Th2 cytokine levels. These results demonstrated that pasteurized yogurt containing heat-killed BBMN68 had mitigative effects on allergic airway inflammation, likely through maintaining the systemic Th1/Th2 immune balance by altering the structure and function of the gut microbiota.
Collapse
Affiliation(s)
- Xiaokang Niu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xindi Yin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xiuying Wu
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yunyun Jiang
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Jingjing He
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yuyang Zhao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chao Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yimei Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Mengxuan Lai
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Yue Sang
- Hebei Engineering Research Center of Animal Product, Sanhe 065200, China
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| |
Collapse
|
9
|
Bernardo G, Le Noci V, Ottaviano E, De Cecco L, Camisaschi C, Guglielmetti S, Di Modica M, Gargari G, Bianchi F, Indino S, Sartori P, Borghi E, Sommariva M, Tagliabue E, Triulzi T, Sfondrini L. Reduction of Staphylococcus epidermidis in the mammary tumor microbiota induces antitumor immunity and decreases breast cancer aggressiveness. Cancer Lett 2023; 555:216041. [PMID: 36565918 DOI: 10.1016/j.canlet.2022.216041] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
The mammary gland hosts a microbiota, which differs between malignant versus normal tissue. We found that aerosolized antibiotics decrease murine mammary tumor growth and strongly limit lung metastasis. Oral absorbable antibiotics also reduced mammary tumors. In ampicillin-treated nodules, the immune microenvironment consisted of an M1 profile and improved T cell/macrophage infiltration. In these tumors, we noted an under-representation of microbial recognition and complement pathways, supported by TLR2/TLR7 protein and C3-fragment deposition reduction. By 16S rRNA gene profiling, we observed increased Staphylococcus levels in untreated tumors, among which we isolated Staphylococcus epidermidis, which had potent inflammatory activity and increased Tregs. Conversely, oral ampicillin lowered Staphylococcus epidermidis in mammary tumors and expanded bacteria promoting an M1 phenotype and reducing MDSCs and tumor growth. Ampicillin/paclitaxel combination improved the chemotherapeutic efficacy. Notably, an Amp-like signature, based on genes differentially expressed in murine tumors, identified breast cancer patients with better prognosis and high immune infiltration that correlated with a bacteria response signature. This study highlights the significant influence of mammary tumor microbiota on local immune status and the relevance of its treatment with antibiotics, in combination with breast cancer therapies.
Collapse
Affiliation(s)
- Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
| | - Emerenziana Ottaviano
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy.
| | - Loris De Cecco
- Molecular Mechanisms Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Chiara Camisaschi
- Biomarkers Unit, Department of Applied Research and Technical Development, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Simone Guglielmetti
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l'Ambiente (DeFENS), Università degli Studi di Milano, Via Mangiagalli 25, 20133, Milan, Italy.
| | - Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Giorgio Gargari
- Dipartimento di Scienze per gli Alimenti, la Nutrizione e l'Ambiente (DeFENS), Università degli Studi di Milano, Via Mangiagalli 25, 20133, Milan, Italy.
| | - Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy; U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, Piazza Edmondo Malan 2, 20097, San Donato Milanese, Milan, Italy.
| | - Serena Indino
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
| | - Patrizia Sartori
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
| | - Elisa Borghi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via di Rudinì 8, 20142, Milan, Italy.
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy; Molecular Targeting Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy; Molecular Targeting Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
10
|
Spacova I, De Boeck I, Cauwenberghs E, Delanghe L, Bron PA, Henkens T, Simons A, Gamgami I, Persoons L, Claes I, van den Broek MFL, Schols D, Delputte P, Coenen S, Verhoeven V, Lebeer S. Development of a live biotherapeutic throat spray with lactobacilli targeting respiratory viral infections. Microb Biotechnol 2022; 16:99-115. [PMID: 36468246 PMCID: PMC9803329 DOI: 10.1111/1751-7915.14189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/09/2022] Open
Abstract
Respiratory viruses such as influenza viruses, respiratory syncytial virus (RSV), and coronaviruses initiate infection at the mucosal surfaces of the upper respiratory tract (URT), where the resident respiratory microbiome has an important gatekeeper function. In contrast to gut-targeting administration of beneficial bacteria against respiratory viral disease, topical URT administration of probiotics is currently underexplored, especially for the prevention and/or treatment of viral infections. Here, we report the formulation of a throat spray with live lactobacilli exhibiting several in vitro mechanisms of action against respiratory viral infections, including induction of interferon regulatory pathways and direct inhibition of respiratory viruses. Rational selection of Lactobacillaceae strains was based on previously documented beneficial properties, up-scaling and industrial production characteristics, clinical safety parameters, and potential antiviral and immunostimulatory efficacy in the URT demonstrated in this study. Using a three-step selection strategy, three strains were selected and further tested in vitro antiviral assays and in formulations: Lacticaseibacillus casei AMBR2 as a promising endogenous candidate URT probiotic with previously reported barrier-enhancing and anti-pathogenic properties and the two well-studied model strains Lacticaseibacillus rhamnosus GG and Lactiplantibacillus plantarum WCFS1 that display immunomodulatory capacities. The three strains and their combination significantly reduced the cytopathogenic effects of RSV, influenza A/H1N1 and B viruses, and HCoV-229E coronavirus in co-culture models with bacteria, virus, and host cells. Subsequently, these strains were formulated in a throat spray and human monocytes were employed to confirm the formulation process did not reduce the interferon regulatory pathway-inducing capacity. Administration of the throat spray in healthy volunteers revealed that the lactobacilli were capable of temporary colonization of the throat in a metabolically active form. Thus, the developed spray with live lactobacilli will be further explored in the clinic as a potential broad-acting live biotherapeutic strategy against respiratory viral diseases.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | - Ilke De Boeck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | - Eline Cauwenberghs
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | - Lize Delanghe
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | - Peter A. Bron
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | | | | | | | - Leentje Persoons
- Laboratory of Virology and Chemotherapy, KU Leuven Department of Microbiology, Immunology and TransplantationRega InstituteLeuvenBelgium
| | | | - Marianne F. L. van den Broek
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, KU Leuven Department of Microbiology, Immunology and TransplantationRega InstituteLeuvenBelgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Samuel Coenen
- Family Medicine and Population Health (FAMPOP)University of AntwerpAntwerpBelgium,Vaccine & Infectious Disease Institute (VAXINFECTIO)University of AntwerpAntwerpBelgium
| | - Veronique Verhoeven
- Family Medicine and Population Health (FAMPOP)University of AntwerpAntwerpBelgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience EngineeringUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
11
|
Du T, Lei A, Zhang N, Zhu C. The Beneficial Role of Probiotic Lactobacillus in Respiratory Diseases. Front Immunol 2022; 13:908010. [PMID: 35711436 PMCID: PMC9194447 DOI: 10.3389/fimmu.2022.908010] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 12/24/2022] Open
Abstract
Respiratory diseases cause a high incidence and mortality worldwide. As a natural immunobiotic, Lactobacillus has excellent immunomodulatory ability. Administration of some Lactobacillus species can alleviate the symptoms of respiratory diseases such as respiratory tract infections, asthma, lung cancer and cystic fibrosis in animal studies and clinical trials. The beneficial effect of Lactobacillus on the respiratory tract is strain dependent. Moreover, the efficacy of Lactobacillus may be affected by many factors, such as bacteria dose, timing and host background. Here, we summarized the beneficial effect of administered Lactobacillus on common respiratory diseases with a focus on the mechanism and safety of Lactobacillus in regulating respiratory immunity.
Collapse
|
12
|
Mårtensson A, Nordström F, Cervin-Hoberg C, Lindstedt M, Sakellariou C, Cervin A, Greiff L. Nasal administration of a probiotic assemblage in allergic rhinitis: a placebo-controlled crossover experimental study. Clin Exp Allergy 2022; 52:774-783. [PMID: 35075723 PMCID: PMC9314659 DOI: 10.1111/cea.14098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Topical probiotics have been suggested as a treatment option for allergic rhinitis, as they may skew the immune response towards a beneficial type-1 non-allergic profile. So far observations in man have exclusively involved oral intake. The aim of this study was to examine if a topical/nasal administration of a probiotic assemblage (PA) affects quality of life, symptoms, and signs of allergic rhinitis in a nasal allergen challenge (NAC) model. METHODS In a placebo-controlled and crossover design, 24 patients with seasonal allergic rhinitis were subjected to topical/nasal administration with a PA of Lactobacillus rhamnosus SP1, Lactobacillus paracasei 101/37, and Lactococcus lactis L1A for three weeks. The last week of each treatment period was combined with a NAC-series. Efficacy variables were "Mini-Rhinoconjunctivitis Quality of Life Questionnaire" (Mini-RQLQ), "Total Nasal Symptom Score" (TNSS), "Peak Nasal Inspiratory Flow" (PNIF), and "Fractional Exhaled Nitric Oxide" (FeNO). In addition, to assess whether or not the PA produced any pro-inflammatory effect per se, soluble analytes were monitored in nasal lavage fluids. Finally, bacterial cultures, sampled using swabs from the middle nasal meatus, were assessed for presence of the PA by MALDI-TOF analysis. RESULTS Administration of the PA did not produce any nasal symptoms (cf. placebo). An innate response was discerned within the PA-run (cf. baseline), but no change in nasal lavage fluid levels of cytokines/mediators were observed cf. placebo except for IL-17/IL-17A (a minor increase in the PA run). Administration of the PA did neither affect Mini-RQLQ, TNSS, PNIF, nor FeNO. No evidence of persistent colonization was observed. CONCLUSION Topical/nasal administration of a PA comprising Lactobacillus rhamnosus SP1, Lactobacillus paracasei 101/37, Lactococcus lactis L1A, while likely evoking a minor innate immune response yet being safe, does not affect quality of life, symptoms, or signs of allergic rhinitis.
Collapse
Affiliation(s)
- Anders Mårtensson
- Department of ORL, Head and Neck Surgery, Helsingborg Hospital, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Franziska Nordström
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of ORL, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| | - Charlotte Cervin-Hoberg
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of ORL, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Anders Cervin
- Department of ORL, Royal Brisbane & Women's Hospital, University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Lennart Greiff
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of ORL, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
13
|
Bernard-Raichon L, Colom A, Monard SC, Namouchi A, Cescato M, Garnier H, Leon-Icaza SA, Métais A, Dumas A, Corral D, Ghebrendrias N, Guilloton P, Vérollet C, Hudrisier D, Remot A, Langella P, Thomas M, Cougoule C, Neyrolles O, Lugo-Villarino G. A Pulmonary Lactobacillus murinus Strain Induces Th17 and RORγt + Regulatory T Cells and Reduces Lung Inflammation in Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2021; 207:1857-1870. [PMID: 34479945 DOI: 10.4049/jimmunol.2001044] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 07/24/2021] [Indexed: 12/31/2022]
Abstract
The lungs harbor multiple resident microbial communities, otherwise known as the microbiota. There is an emerging interest in deciphering whether the pulmonary microbiota modulate local immunity, and whether this knowledge could shed light on mechanisms operating in the response to respiratory pathogens. In this study, we investigate the capacity of a pulmonary Lactobacillus strain to modulate the lung T cell compartment and assess its prophylactic potential upon infection with Mycobacterium tuberculosis, the etiological agent of tuberculosis. In naive mice, we report that a Lactobacillus murinus (Lagilactobacillus murinus) strain (CNCM I-5314) increases the presence of lung Th17 cells and of a regulatory T cell (Treg) subset known as RORγt+ Tregs. In particular, intranasal but not intragastric administration of CNCM I-5314 increases the expansion of these lung leukocytes, suggesting a local rather than systemic effect. Resident Th17 and RORγt+ Tregs display an immunosuppressive phenotype that is accentuated by CNCM I-5314. Despite the well-known ability of M. tuberculosis to modulate lung immunity, the immunomodulatory effect by CNCM I-5314 is dominant, as Th17 and RORγt+ Tregs are still highly increased in the lung at 42-d postinfection. Importantly, CNCM I-5314 administration in M. tuberculosis-infected mice results in reduction of pulmonary inflammation, without increasing M. tuberculosis burden. Collectively, our findings provide evidence for an immunomodulatory capacity of CNCM I-5314 at steady state and in a model of chronic inflammation in which it can display a protective role, suggesting that L. murinus strains found in the lung may shape local T cells in mice and, perhaps, in humans.
Collapse
Affiliation(s)
- Lucie Bernard-Raichon
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France;
| | - André Colom
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sarah C Monard
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Amine Namouchi
- Centre for Integrative Genetics, Norwegian University of Life Sciences, As, Norway; and
| | - Margaux Cescato
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Hugo Garnier
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Stephen A Leon-Icaza
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Arnaud Métais
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Alexia Dumas
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Dan Corral
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Natsinet Ghebrendrias
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Pauline Guilloton
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Denis Hudrisier
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Aude Remot
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Thomas
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Céline Cougoule
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France;
| |
Collapse
|
14
|
Pyclik MJ, Srutkova D, Razim A, Hermanova P, Svabova T, Pacyga K, Schwarzer M, Górska S. Viability Status-Dependent Effect of Bifidobacterium longum ssp . longum CCM 7952 on Prevention of Allergic Inflammation in Mouse Model. Front Immunol 2021; 12:707728. [PMID: 34354710 PMCID: PMC8329652 DOI: 10.3389/fimmu.2021.707728] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/20/2022] Open
Abstract
The classical definition of probiotics states that bacteria must be alive to be beneficial for human organism. However, recent reports show that inactivated bacteria or their effector molecules can also possess such properties. In this study, we investigated the physical and immunomodulatory properties of four Bifidobacterium strains in the heat-treated (HT) and untreated (UN) forms. We showed that temperature treatment of bacteria changes their size and charge, which affects their interaction with epithelial and immune cells. Based on the in vitro assays, we observed that all tested strains reduced the level of OVA-induced IL-4, IL-5, and IL-13 in the spleen culture of OVA-sensitized mice. We selected Bifidobacterium longum ssp. longum CCM 7952 (Bl 7952) for further analysis. In vivo experiments confirmed that untreated Bl 7952 exhibited allergy-reducing properties when administered intranasally to OVA-sensitized mice, which manifested in significant suppression of airway inflammation. Untreated Bl 7952 decreased local and systemic levels of Th2 related cytokines, OVA-specific IgE antibodies and simultaneously inhibited airway eosinophilia. In contrast, heat-treated Bl 7952 was only able to reduce IL-4 levels in the lungs and eosinophils in bronchoalveolar lavage, but increased neutrophil and macrophage numbers. We demonstrated that the viability status of Bl 7952 is a prerequisite for the beneficial effects of bacteria, and that heat treatment reduces but does not completely abolish these properties. Further research on bacterial effector molecules to elucidate the beneficial effects of probiotics in the prevention of allergic diseases is warranted.
Collapse
Affiliation(s)
- Marcelina Joanna Pyclik
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Agnieszka Razim
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Petra Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Tereza Svabova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Katarzyna Pacyga
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
15
|
Jakubczyk D, Górska S. Impact of Probiotic Bacteria on Respiratory Allergy Disorders. Front Microbiol 2021; 12:688137. [PMID: 34234762 PMCID: PMC8256161 DOI: 10.3389/fmicb.2021.688137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory allergy is a common disease with an increased prevalence worldwide. The effective remedy is still unknown, and a new therapeutic approach is highly desirable. The review elaborates the influence of probiotic bacteria on respiratory allergy prevention and treatment with particular emphasis on the impact of the current methods of their administration – oral and intranasal. The background of the respiratory allergy is complex thus, we focused on the usefulness of probiotics in the alleviation of different allergy factors, in particular involved in pathomechanism, local hypersensitive evidence and the importance of epithelial barrier. In this review, we have shown that (1) probiotic strains may vary in modulatory potential in respiratory allergy, (2) probiotic bacteria are beneficial in oral and intranasal administration, (3) recombinant probiotic bacteria can modulate the course of respiratory allergy.
Collapse
Affiliation(s)
- Dominika Jakubczyk
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
16
|
Nam W, Kim H, Kim J, Nam B, Bae C, Kim J, Park S, Lee J, Sim J. Lactic Acid Bacteria and Natural Product Complex Ameliorates Ovalbumin-Induced Airway Hyperresponsiveness in Mice. J Med Food 2021; 24:517-526. [PMID: 34009021 DOI: 10.1089/jmf.2020.4853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The incidence of respiratory diseases, such as asthma, has substantially increased in recent times owing to environmental changes, such as air pollution. Induction of a chronic inflammatory response begins with production of biologically active mediators from the airway epithelium, which attracts and recruits inflammatory cells into the lung airway. In our previous study, we confirmed that Lactobacillus casei HY2782 and Bifidobacterium animalis spp. lactis HY8002 could improve lung inflammation in the COPD animal model. In this study, we investigated the effect of the HY2782 complex against airway hyperresponsiveness by using an ovalbumin (OVA)-induced animal model. An orally administered HY2782 complex on OVA-induced allergic asthma in a BALB/c mouse model was used. The present results showed that the HY2782 complex suppressed total immunoglobulin E in serum and bronchoalveolar lavage fluid (BALF). The cytokine production profile in BALF and serum revealed that the HY2782 complex showed reduced levels of Th2 cytokines among immune factors released due to the elevated allergic response. Levels of inflammatory mediators in BALF, MCP-1, MIP-2, and CXCL-9 were decreased by oral administration of the HY2782 complex. Lower numbers of eosinophils and neutrophils in BALF suggested that inflammation was ameliorated by the HY2782 complex. Histological observation of lung sections also showed infiltration of fewer cells. From results, we suggested that the HY2782 complex effectively responds to improvement of the immune response and airway hypersensitivity reaction because of the anti-inflammatory effect of the Pueraria lobata root extract and antioxidant effect of HY2782.
Collapse
Affiliation(s)
- Woo Nam
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Hyeonji Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Jisoo Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Bora Nam
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Chuhyun Bae
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Jooyun Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Soodong Park
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | | | - Jaehun Sim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| |
Collapse
|
17
|
Allam VSRR, Chellappan DK, Jha NK, Shastri MD, Gupta G, Shukla SD, Singh SK, Sunkara K, Chitranshi N, Gupta V, Wich PR, MacLoughlin R, Oliver BGG, Wernersson S, Pejler G, Dua K. Treatment of chronic airway diseases using nutraceuticals: Mechanistic insight. Crit Rev Food Sci Nutr 2021; 62:7576-7590. [PMID: 33977840 DOI: 10.1080/10408398.2021.1915744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Respiratory diseases, both acute and chronic, are reported to be the leading cause of morbidity and mortality, affecting millions of people globally, leading to high socio-economic burden for the society in the recent decades. Chronic inflammation and decline in lung function are the common symptoms of respiratory diseases. The current treatment strategies revolve around using appropriate anti-inflammatory agents and bronchodilators. A range of anti-inflammatory agents and bronchodilators are currently available in the market; however, the usage of such medications is limited due to the potential for various adverse effects. To cope with this issue, researchers have been exploring various novel, alternative therapeutic strategies that are safe and effective to treat respiratory diseases. Several studies have been reported on the possible links between food and food-derived products in combating various chronic inflammatory diseases. Nutraceuticals are examples of such food-derived products which are gaining much interest in terms of its usage for the well-being and better human health. As a consequence, intensive research is currently aimed at identifying novel nutraceuticals, and there is an emerging notion that nutraceuticals can have a positive impact in various respiratory diseases. In this review, we discuss the efficacy of nutraceuticals in altering the various cellular and molecular mechanisms involved in mitigating the symptoms of respiratory diseases.
Collapse
Affiliation(s)
- Venkata Sita Rama Raju Allam
- Department of Medical Biochemistry and Microbiology, Biomedical Centre (BMC), Uppsala University, Uppsala, Sweden
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, New South Wales, Australia
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Krishna Sunkara
- Emergency Clinical Management, Intensive Care Unit, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Australia.,Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, Galway, Ireland.,School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Brian Gregory George Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia.,Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
| | - Sara Wernersson
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Biomedical Centre (BMC), Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
18
|
Kim HS, Arellano K, Park H, Todorov SD, Kim B, Kang H, Park YJ, Suh DH, Jung ES, Ji Y, Holzapfel WH. Assessment of the safety and anti-inflammatory effects of three Bacillus strains in the respiratory tract. Environ Microbiol 2021; 23:3077-3098. [PMID: 33899316 DOI: 10.1111/1462-2920.15530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022]
Abstract
Chronic respiratory diseases are part of accumulating health problems partly due to worldwide increase in air pollution. By their antimicrobial and immunomodulatory properties, some probiotics constitute promising alternatives for the prevention and treatment of chronic respiratory diseases. We have isolated Bacillus strains from Korean fermented foods and selected three potentially probiotic strains (two Bacillus subtilis and one Bacillus amyloliquefaciens) based on safety, antimicrobial efficacy, activity against airborne pathogens and their immunomodulatory properties in vivo. Safety evaluation included in silico analysis for confirming absence of virulence genes. Safety for the respiratory tract was confirmed by an in vivo pathogenicity test using a murine model. Antimicrobial activity was displayed against several airborne pathogens. Potential antimicrobial metabolites such as 2,3-butanediol and propylene glycol were identified as possible antagonistic agents. Immunomodulatory properties in vitro were confirmed by upregulation of IL-10 expression in a macrophage cell line. Intranasal instillation and inhalation in an ovalbumin (OVA)-induced lung inflammation murine model reduced T helper type 2 (Th2) cytokines at transcriptional and protein levels in the lungs. The safety and potentially beneficial role of these Bacillus strains could be demonstrated for the respiratory tract of a murine model.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Karina Arellano
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Haryung Park
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Svetoslav D Todorov
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Hyeji Kang
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| | - Yu Jin Park
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Dong Ho Suh
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Eun Sung Jung
- HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Yosep Ji
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., 77, Changnyong-daero 256 Beon-gil, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Convergence, Handong Global University, Pohang, Gyungbuk, 37554, Republic of Korea.,HEM Inc., Pohang, Gyungbuk, 37554, Republic of Korea
| |
Collapse
|
19
|
Meirlaen L, Levy EI, Vandenplas Y. Prevention and Management with Pro-, Pre and Synbiotics in Children with Asthma and Allergic Rhinitis: A Narrative Review. Nutrients 2021; 13:934. [PMID: 33799367 PMCID: PMC7999316 DOI: 10.3390/nu13030934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/04/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Allergic diseases including allergic rhinitis and asthma are increasing in the developing world, related to a westernizing lifestyle, while the prevalence is stable and decreasing in the industrialized world. This paper aims to answer the question if prevention and/or treatment of allergic rhinitis and asthma can be achieved by administrating pro-, pre- and/or synbiotics that might contribute to stabilizing the disturbed microbiome that influences the immune system through the gut-lung axis. We searched for relevant English articles in PubMed and Google Scholar. Articles interesting for the topic were selected using subject heading and key words. Interesting references in included articles were also considered. While there is substantial evidence from animal studies in well controlled conditions that selected probiotic strains may offer benefits in the prevention of wheezing and asthma, outcomes from clinical studies in infants (including as well pre- and postnatal administration) are disappointing. The latter may be related to the multiple confounding factors such as environment, strain selection and dosage, moment of administration and genetic background. There is little evidence to recommend administration of pro, pre- or synbiotics in the prevention of asthma and allergic rhinitis in children.
Collapse
Affiliation(s)
| | | | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (L.M.); (E.I.L.)
| |
Collapse
|
20
|
Mennini M, Tambucci R, Riccardi C, Rea F, De Angelis P, Fiocchi A, Assa'ad A. Eosinophilic Esophagitis and Microbiota: State of the Art. Front Immunol 2021; 12:595762. [PMID: 33679739 PMCID: PMC7933523 DOI: 10.3389/fimmu.2021.595762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic, food-triggered, immune-mediated disease of the oesophagus, clinically characterized by symptoms referred to oesophagal dysfunction, and histologically defined by an eosinophil productive inflammation of the oesophagal mucosa, among other cell types. The involvement of an adaptive Th2-type response to food antigens in EoE was known since 2000; several cytokines and chemokines promote food-specific responses, during which local production of IgE, but also IgG4 derived from plasma cells in lamina propria of oesophagal mucosa might play an important role. Evidence pointing towards a possible role for the innate immunity in EoE has arisen recently. Together, this evidence gives rise to a potential role that the innate immune system in general, and also the microbial pattern recognition receptors (PRRs) might play in EoE pathogenesis. Among PRRs, Toll-like receptors (TLRs) are type-I transmembrane receptors expressed both on epithelial and lamina propria cells with the capacity to distinguish between pathogen and commensal microbes. As TLRs in the different intestinal epithelia represent the primary mechanism of epithelial recognition of bacteria, this evidence underlines that oesophagal TLR-dependent signaling pathways in EoE support the potential implication of microbiota and the innate immune system in the pathogenesis of this disease. The oesophagal mucosa hosts a resident microbiota, although in a smaller population as compared with other districts of the gastrointestinal tract. Few studies have focused on the composition of the microbiota of the normal oesophagus alone. Still, additional information has come from studies investigating the oesophagal microbiota in disease and including healthy patients as controls. Our review aims to describe all the evidence on the oesophagal and intestinal microbiota in patients with EoE to identify the specific features of dysbiosis in this condition.
Collapse
Affiliation(s)
- Maurizio Mennini
- Division of Allergy, Bambino Gesù Children's Hospital-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Renato Tambucci
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Carla Riccardi
- Division of Allergy, Bambino Gesù Children's Hospital-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesca Rea
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| | - Alessandro Fiocchi
- Division of Allergy, Bambino Gesù Children's Hospital-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Amal Assa'ad
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
21
|
Sun T, Yu H, Fu J. Respiratory Tract Microecology and Bronchopulmonary Dysplasia in Preterm Infants. Front Pediatr 2021; 9:762545. [PMID: 34966701 PMCID: PMC8711720 DOI: 10.3389/fped.2021.762545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe respiratory complication in preterm infants. Although the etiology and pathogenesis of BPD are complex and remain to be clarified, recent studies have reported a certain correlation between the microecological environment of the respiratory tract and BPD. Changes in respiratory tract microecology, such as abnormal microbial diversity and altered evolutional patterns, are observed prior to the development of BPD in premature infants. Therefore, research on the colonization and evolution of neonatal respiratory tract microecology and its relationship with BPD is expected to provide new ideas for its prevention and treatment. In this paper, we review microecological changes in the respiratory tract and the mechanisms by which they can lead to BPD in preterm infants.
Collapse
Affiliation(s)
- Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Yildiz S, Pereira Bonifacio Lopes JP, Bergé M, González-Ruiz V, Baud D, Kloehn J, Boal-Carvalho I, Schaeren OP, Schotsaert M, Hathaway LJ, Rudaz S, Viollier PH, Hapfelmeier S, Francois P, Schmolke M. Respiratory tissue-associated commensal bacteria offer therapeutic potential against pneumococcal colonization. eLife 2020; 9:53581. [PMID: 33287959 PMCID: PMC7723408 DOI: 10.7554/elife.53581] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Under eubiotic conditions commensal microbes are known to provide a competitive barrier against invading bacterial pathogens in the intestinal tract, on the skin or on the vaginal mucosa. Here, we evaluate the role of lung microbiota in Pneumococcus colonization of the lungs. In eubiosis, the lungs of mice were dominantly colonized by Lactobacillus murinus. Differential analysis of 16S rRNA gene sequencing or L. murinus-specific qPCR of DNA from total organ homogenates vs.broncho alveolar lavages implicated tight association of these bacteria with the host tissue. Pure L. murinus conditioned culture medium inhibited growth and reduced the extension of pneumococcal chains. Growth inhibition in vitro was likely dependent on L. murinus-produced lactic acid, since pH neutralization of the conditioned medium aborted the antibacterial effect. Finally, we demonstrate that L. murinus provides a barrier against pneumococcal colonization in a respiratory dysbiosis model after an influenza A virus infection, when added therapeutically.
Collapse
Affiliation(s)
- Soner Yildiz
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Matthieu Bergé
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Víctor González-Ruiz
- Analytical Sciences, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Damian Baud
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Joachim Kloehn
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Inês Boal-Carvalho
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Olivier P Schaeren
- Institute for Infectious Disease (IFIK), University of Bern, Bern, Switzerland.,Graduate School GCB, University of Bern, Bern, Switzerland
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Lucy J Hathaway
- Institute for Infectious Disease (IFIK), University of Bern, Bern, Switzerland
| | - Serge Rudaz
- Analytical Sciences, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Patrick H Viollier
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Patrice Francois
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
23
|
Spacova I, Van Beeck W, Seys S, Devos F, Vanoirbeek J, Vanderleyden J, Ceuppens J, Petrova M, Lebeer S. Lactobacillus rhamnosus probiotic prevents airway function deterioration and promotes gut microbiome resilience in a murine asthma model. Gut Microbes 2020; 11:1729-1744. [PMID: 32522072 PMCID: PMC7524350 DOI: 10.1080/19490976.2020.1766345] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Allergic asthma is a highly prevalent inflammatory disease of the lower airways, clinically characterized by airway hyperreactivity and deterioration of airway function. Immunomodulatory probiotic bacteria are increasingly being explored to prevent asthma development, alone or in combination with other treatments. In this study, wild-type and recombinant probiotic Lactobacillus rhamnosus GR-1 were tested as preventive treatment of experimental allergic asthma in mice. Recombinant L. rhamnosus GR-1 was designed to produce the major birch pollen allergen Bet v 1, to promote allergen-specific immunomodulation. Administration of wild-type and recombinant L. rhamnosus GR-1 prevented the development of airway hyperreactivity. Recombinant L. rhamnosus GR-1 also prevented elevation of airway total cell counts, lymphocyte counts and lung IL-1β levels, while wild-type L. rhamnosus GR-1 inhibited airway eosinophilia. Of note, a shift in gut microbiome composition was observed after asthma development, which correlated with the severity of airway inflammation and airway hyperreactivity. In the groups that received L. rhamnosus GR-1, this asthma-associated shift in gut microbiome composition was not observed, indicating microbiome-modulating effects of this probiotic. These data demonstrate that L. rhamnosus GR-1 can prevent airway function deterioration in allergic asthma. Bet v 1 expression by L. rhamnosus GR-1 further contributed to lower airway inflammation, although not solely through the expected reduction in T helper 2-associated responses, suggesting involvement of additional mechanisms. The beneficial effects of L. rhamnosus GR-1 correlate with increased gut microbiome resilience, which in turn is linked to protection of airway function, and thus further adds support to the existence of a gut-lung axis.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M2S), KU Leuven, Leuven, Belgium
| | - Wannes Van Beeck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sven Seys
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Fien Devos
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jeroen Vanoirbeek
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jozef Vanderleyden
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M2S), KU Leuven, Leuven, Belgium
| | - Jan Ceuppens
- Allergy and Clinical Immunology Research Group, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | - Mariya Petrova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M2S), KU Leuven, Leuven, Belgium,Microbiome Insights and Probiotics Consultancy, Karlovo, Bulgaria,Mariya Petrova Microbiome insights and Probiotics Consultancy,Karlovo4300, Bulgaria
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M2S), KU Leuven, Leuven, Belgium,CONTACT Sarah Lebeer University of Antwerp, Department of Bioscience Engineering,AntwerpB-2020, Belgium
| |
Collapse
|
24
|
Falsaperla R, Lombardo F, Filosco F, Romano C, Saporito MAN, Puglisi F, Piro E, Ruggieri M, Pavone P. Oxidative Stress in Preterm Infants: Overview of Current Evidence and Future Prospects. Pharmaceuticals (Basel) 2020; 13:E145. [PMID: 32645921 PMCID: PMC7408528 DOI: 10.3390/ph13070145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022] Open
Abstract
Preterm birth (PTB), defined as parturition prior to 37 weeks of gestation, is the leading cause of morbidity and mortality in the neonatal population. The incidence and severity of complications of prematurity increase with decreasing gestational age and birthweight. The aim of this review study is to select the most current evidence on the role of oxidative stress in the onset of preterm complication prevention strategies and treatment options with pre-clinical and clinical trials. We also provide a literature review of primary and secondary studies on the role of oxidative stress in preterm infants and its eventual treatment in prematurity diseases. We conducted a systematic literature search of the Medline (Pubmed), Scholar, and ClinicalTrials.gov databases, retroactively, over a 7-year period. From an initial 777 articles identified, 25 articles were identified that met the inclusion and exclusion criteria. Of these, there were 11 literature reviews: one prospective cohort study, one experimental study, three case-control studies, three pre-clinical trials, and six clinical trials. Several biomarkers were identified as particularly promising, such as the products of the peroxidation of polyunsaturated fatty acids, those of the oxidation of phenylalanine, and the hydroxyl radicals that can attack the DNA chain. Among the most promising drugs, there are those for the prevention of neurological damage, such as melatonin, retinoid lactoferrin, and vitamin E. The microbiome also has an important role in oxidative stress. In conclusion, the most recent studies show that a strong relationship between oxidative stress and prematurity exists and that, unfortunately, there is still little therapeutic evidence reported in the literature.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care, AUO San Marco-Policlinico, University of Catania, 95123 Catania, Italy; (R.F.); (M.A.N.S.); (F.P.)
| | - Filadelfo Lombardo
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania street Santa Sofia 78, 95123 Catania, Italy; (F.L.); (F.F.)
| | - Federica Filosco
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania street Santa Sofia 78, 95123 Catania, Italy; (F.L.); (F.F.)
| | - Catia Romano
- Child and Adolescent Neuropsychiatry, Department Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy;
| | - Marco Andrea Nicola Saporito
- Neonatal Intensive Care, AUO San Marco-Policlinico, University of Catania, 95123 Catania, Italy; (R.F.); (M.A.N.S.); (F.P.)
| | - Federica Puglisi
- Neonatal Intensive Care, AUO San Marco-Policlinico, University of Catania, 95123 Catania, Italy; (R.F.); (M.A.N.S.); (F.P.)
| | - Ettore Piro
- University Hospital “P. Giaccone”, Department of Sciences for Health Promotion, Maternal Infant Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, Neonatal Intensive Care Unit, 90121 Palermo, Italy;
| | - Martino Ruggieri
- Department of Clinical and Experimental Medicine Section of Pediatrics and Child Neuropsychiatry, AUO San Marco-Policlinco, University of Catania, 95123 Catania, Italy;
| | - Piero Pavone
- Department of Clinical and Experimental Medicine Section of Pediatrics and Child Neuropsychiatry, AUO San Marco-Policlinco, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
25
|
Ghiamati Yazdi F, Zakeri A, van Ark I, Leusink-Muis T, Braber S, Soleimanian-Zad S, Folkerts G. Crude Turmeric Extract Improves the Suppressive Effects of Lactobacillus rhamnosus GG on Allergic Inflammation in a Murine Model of House Dust Mite-Induced Asthma. Front Immunol 2020; 11:1092. [PMID: 32582180 PMCID: PMC7287160 DOI: 10.3389/fimmu.2020.01092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
There is a strong correlation between dysregulation of the gastrointestinal microbiota and development of allergic diseases. The most prevalent therapies for relieving asthma symptoms are associated with serious side effects, and therefore novel approaches are needed. Our objective was to elucidate whether oral administration of Lactobacillus rhamnosus GG (LGG) as a probiotic or turmeric powder (TP) as a prebiotic or both as a synbiotic mitigate allergic inflammation including lung function, airway inflammatory cell infiltration, Th2 cytokines/chemokine in a murine model of house dust mite (HDM)-induced asthma. BALB/c mice were intranasally sensitized and challenged with HDM received TP (20 mg/Kg mouse), or/and LGG (105 or 107 cfu/ml), or both orally. Interestingly, the synbiotic intervention (HDM-TP-LGG E7) specifically suppress the developement of airway hyperresponsiveness in response to methacholine. Besides, our synbiotic, TP, and LGG strongly down-regulated eosinophilia, IL-5, CCL17, IL-13. In terms of T cell response, CD4+ Th2 cells and CD4+ Th17 population were reduced in the splenocytes of the treatment groups compared to control. The synbiotic group not only elevated CD25+Foxp3+Treg frequency compared to asthmatic group, but also increased T reg cells compared to the probiotic group. The synbiotic also indicated the superior effect in suppressing Th2 cells compared to probiotic. Although, TP and LGG alone displayed suppressive effects, this study showed that the combination therapy consisting of TP and LGG (synbiotic) is more effective in some of the parameters than either of the treatments alone. This novel synbiotic, might be considered as a potential food-based drug for translational medicine and can possibly be used along with corticosteroid treatment.
Collapse
Affiliation(s)
- Fariba Ghiamati Yazdi
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology (IUT), Isfahan, Iran.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Amin Zakeri
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Sabihe Soleimanian-Zad
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology (IUT), Isfahan, Iran
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
26
|
Goto T. Airway Microbiota as a Modulator of Lung Cancer. Int J Mol Sci 2020; 21:ijms21093044. [PMID: 32357415 PMCID: PMC7246469 DOI: 10.3390/ijms21093044] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
Recent research on cancer-associated microbial communities has elucidated the interplay between bacteria, immune cells, and tumor cells; the bacterial pathways involved in the induction of carcinogenesis; and their clinical significance. Although accumulating evidence shows that a dysbiotic condition is associated with lung carcinogenesis, the underlying mechanisms remain unclear. Microorganisms possibly trigger tumor initiation and progression, presumably via the production of bacterial toxins and other pro-inflammatory factors. The purpose of this review is to discuss the basic role of the airway microbiome in carcinogenesis and the underlying molecular mechanisms, with the aim of developing anticancer strategies involving the airway microbiota. In addition, the mechanisms via which the microbiome acts as a modulator of immunotherapies in lung cancer are summarized.
Collapse
Affiliation(s)
- Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Kofu, Yamanashi 4008506, Japan
| |
Collapse
|
27
|
Oral and nasal probiotic administration for the prevention and alleviation of allergic diseases, asthma and chronic obstructive pulmonary disease. Nutr Res Rev 2020; 34:1-16. [PMID: 32281536 DOI: 10.1017/s0954422420000116] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interaction between a healthy microbiome and the immune system leads to body homeostasis, as dysbiosis in microbiome content and loss of diversity may result in disease development. Due to the ability of probiotics to help and modify microbiome constitution, probiotics are now widely used for the prevention and treatment of different gastrointestinal, inflammatory, and, more recently, respiratory diseases. In this regard, chronic respiratory diseases including chronic obstructive pulmonary disease (COPD), asthma and allergic rhinitis are among the most common and complicated respiratory diseases with no specific treatment until now. Accordingly, many studies have evaluated the therapeutic efficacy of probiotic administration (mostly via the oral route and much lesser nasal route) on chronic respiratory diseases. We tried to summarise and evaluate these studies to give a perspective of probiotic therapy via both the oral and nasal routes for respiratory infections (in general) and chronic respiratory diseases (specifically). We finally concluded that probiotics might be useful for allergic diseases. For asthmatic patients, probiotics can modulate serum cytokines and IgE and decrease eosinophilia, but with no significant reduction in clinical symptoms. For COPD, only limited studies were found with uncertain clinical efficacy. For intranasal administration, although some studies propose more efficiency than the oral route, more clinical evaluations are warranted.
Collapse
|
28
|
Chen LH, Huang SY, Huang KC, Hsu CC, Yang KC, Li LA, Chan CH, Huang HY. Lactobacillus paracasei PS23 decelerated age-related muscle loss by ensuring mitochondrial function in SAMP8 mice. Aging (Albany NY) 2020; 11:756-770. [PMID: 30696799 PMCID: PMC6366975 DOI: 10.18632/aging.101782] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022]
Abstract
Sarcopenia is a common impairment in the elderly population responsible for poor outcomes later in life; it can be caused by age-related alternations. Only a few strategies have been reported to reduce sarcopenia. Lactobacillus paracasei PS23 (LPPS23) has been reported to delay some age-related disorders. Therefore, here we investigated whether LPPS23 decelerates age-related muscle loss and its underlying mechanism. Female senescence-accelerated mouse prone-8 (SAMP8) mice were divided into three groups (n=6 each): non-aging (16-week-old), control (28-week-old), and PS23 (28-week-old) groups. The control and PS23 groups were given saline and LPPS23, respectively. We evaluated the effects of LPPS23 by analyzing body weight and composition, muscle strength, protein uptake, mitochondrial function, reactive oxygen species (ROS), antioxidant enzymes, and inflammation-related cytokines. LPPS23 significantly attenuated age-related decreases of muscle mass and strength. Compared to the control group, the non-aging and PS23 groups exhibited higher mitochondrial function, IL10, antioxidant enzymes, and protein uptake. Moreover, inflammatory cytokines and ROS were lower in the non-aging and PS23 groups than the control group. Taken together, LPPS23 extenuated sarcopenia progression during aging; this effect might have been enacted by preserving the mitochondrial function via reducing age-related inflammation and ROS and by retaining protein uptake in the SAMP8 mice.
Collapse
Affiliation(s)
- Li-Han Chen
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei, Taiwan
| | - Shih-Yi Huang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Family Medicine, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Chih-Chieh Hsu
- Research and Development Department, Bened Biomedical Co., Ltd., Taipei, Taiwan
| | - Kuen-Cheh Yang
- Department of Family Medicine, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Lin-Ai Li
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei, Taiwan
| | - Ching-Hung Chan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei, Taiwan
| | - Hui-Yu Huang
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
29
|
Horve PF, Lloyd S, Mhuireach GA, Dietz L, Fretz M, MacCrone G, Van Den Wymelenberg K, Ishaq SL. Building upon current knowledge and techniques of indoor microbiology to construct the next era of theory into microorganisms, health, and the built environment. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2020; 30:219-235. [PMID: 31308484 PMCID: PMC7100162 DOI: 10.1038/s41370-019-0157-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 05/06/2023]
Abstract
In the constructed habitat in which we spend up to 90% of our time, architectural design influences occupants' behavioral patterns, interactions with objects, surfaces, rituals, the outside environment, and each other. Within this built environment, human behavior and building design contribute to the accrual and dispersal of microorganisms; it is a collection of fomites that transfer microorganisms; reservoirs that collect biomass; structures that induce human or air movement patterns; and space types that encourage proximity or isolation between humans whose personal microbial clouds disperse cells into buildings. There have been recent calls to incorporate building microbiology into occupant health and exposure research and standards, yet the built environment is largely viewed as a repository for microorganisms which are to be eliminated, instead of a habitat which is inexorably linked to the microbial influences of building inhabitants. Health sectors have re-evaluated the role of microorganisms in health, incorporating microorganisms into prevention and treatment protocols, yet no paradigm shift has occurred with respect to microbiology of the built environment, despite calls to do so. Technological and logistical constraints often preclude our ability to link health outcomes to indoor microbiology, yet sufficient study exists to inform the theory and implementation of the next era of research and intervention in the built environment. This review presents built environment characteristics in relation to human health and disease, explores some of the current experimental strategies and interventions which explore health in the built environment, and discusses an emerging model for fostering indoor microbiology rather than fearing it.
Collapse
Affiliation(s)
- Patrick F Horve
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
| | - Savanna Lloyd
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
| | - Gwynne A Mhuireach
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
| | - Leslie Dietz
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
| | - Mark Fretz
- Institute for Health and the Built Environment, University of Oregon, Portland, OR, 97209, USA
| | - Georgia MacCrone
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
| | - Kevin Van Den Wymelenberg
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA
- Institute for Health and the Built Environment, University of Oregon, Portland, OR, 97209, USA
| | - Suzanne L Ishaq
- Biology and the Built Environment Center, University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
30
|
Lung Microbiome in Asthma: Current Perspectives. J Clin Med 2019; 8:jcm8111967. [PMID: 31739446 PMCID: PMC6912699 DOI: 10.3390/jcm8111967] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence implicates the human microbiome as a potentially influential player actively engaged in shaping the pathogenetic processes underlying the endotypes and phenotypes of chronic respiratory diseases, particularly of the airways. In this article, we specifically review current evidence on the characteristics of lung microbiome, and specifically the bacteriome, the modes of interaction between lung microbiota and host immune system, the role of the “lung–gut axis”, and the functional effects thereof on asthma pathogenesis. We also attempt to explore the possibilities of therapeutic manipulation of the microbiome, aiming at the establishment of asthma prevention strategies and the optimization of asthma treatment.
Collapse
|
31
|
Kumpitsch C, Koskinen K, Schöpf V, Moissl-Eichinger C. The microbiome of the upper respiratory tract in health and disease. BMC Biol 2019; 17:87. [PMID: 31699101 PMCID: PMC6836414 DOI: 10.1186/s12915-019-0703-z] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 02/08/2023] Open
Abstract
The human upper respiratory tract (URT) offers a variety of niches for microbial colonization. Local microbial communities are shaped by the different characteristics of the specific location within the URT, but also by the interaction with both external and intrinsic factors, such as ageing, diseases, immune responses, olfactory function, and lifestyle habits such as smoking. We summarize here the current knowledge about the URT microbiome in health and disease, discuss methodological issues, and consider the potential of the nasal microbiome to be used for medical diagnostics and as a target for therapy.
Collapse
Affiliation(s)
- Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Kaisa Koskinen
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Veronika Schöpf
- Institute of Psychology, University of Graz, Universitaetsplatz 2, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
- Present address: Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
32
|
Kumova OK, Fike AJ, Thayer JL, Nguyen LT, Mell JC, Pascasio J, Stairiker C, Leon LG, Katsikis PD, Carey AJ. Lung transcriptional unresponsiveness and loss of early influenza virus control in infected neonates is prevented by intranasal Lactobacillus rhamnosus GG. PLoS Pathog 2019; 15:e1008072. [PMID: 31603951 PMCID: PMC6808501 DOI: 10.1371/journal.ppat.1008072] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/23/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Respiratory viral infections contribute substantially to global infant losses and disproportionately affect preterm neonates. Using our previously established neonatal murine model of influenza infection, we demonstrate that three-day old mice are exceptionally sensitive to influenza virus infection and exhibit high mortality and viral load. Intranasal pre- and post-treatment of neonatal mice with Lactobacillus rhamnosus GG (LGG), an immune modulator in respiratory viral infection of adult mice and human preterm neonates, considerably improves neonatal mice survival after influenza virus infection. We determine that both live and heat-killed intranasal LGG are equally efficacious in protection of neonates. Early in influenza infection, neonatal transcriptional responses in the lung are delayed compared to adults. These responses increase by 24 hours post-infection, demonstrating a delay in the kinetics of the neonatal anti-viral response. LGG pretreatment improves immune gene transcriptional responses during early infection and specifically upregulates type I IFN pathways. This is critical for protection, as neonatal mice intranasally pre-treated with IFNβ before influenza virus infection are also protected. Using transgenic mice, we demonstrate that the protective effect of LGG is mediated through a MyD88-dependent mechanism, specifically via TLR4. LGG can improve both early control of virus and transcriptional responsiveness and could serve as a simple and safe intervention to protect neonates.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Adam J. Fike
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Jillian L. Thayer
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Linda T. Nguyen
- Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Joshua Chang Mell
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Christopher Stairiker
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Leticia G. Leon
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
33
|
Spacova I, Petrova MI, Fremau A, Pollaris L, Vanoirbeek J, Ceuppens JL, Seys S, Lebeer S. Intranasal administration of probiotic Lactobacillus rhamnosus GG prevents birch pollen-induced allergic asthma in a murine model. Allergy 2019; 74:100-110. [PMID: 29888398 DOI: 10.1111/all.13502] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND There is an increasing interest in targeted application of probiotic bacteria for prevention and treatment of airway diseases, including allergies. Here, we investigated the beneficial effects of preventive intranasal treatment with probiotics Lactobacillus rhamnosus GG and L. rhamnosus GR-1 in a mouse model of allergic asthma. METHODS Lactobacillus rhamnosus was administered intranasally eight times on days 1-4 and 8-11 at 5 × 108 CFU/dose, followed by a 2-week asthma induction protocol with birch pollen extract on alternating days. Effects of preventive treatment were analyzed based on serum antibody levels, bronchoalveolar lavage cell counts, lung histology, lung cytokine levels, and airway hyperreactivity. Colonization and translocation of L. rhamnosus were assessed by bacterial cell counts in nasal mucosa, fecal samples, cervical lymph nodes, and blood. Binding of fluorescent L. rhamnosus to fixed murine nasal mucosal cells and airway macrophages was visualized by fluorescence microscopy. RESULTS Transient colonization of the murine upper airways by L. rhamnosus GG was demonstrated and was approximately ten times higher compared to L. rhamnosus GR-1. Marked binding of fluorescent L. rhamnosus GG to murine nasal mucosal cells and airway macrophages was visualized. Preventive treatment with L. rhamnosus GG (but not L. rhamnosus GR-1) resulted in a significant decrease in bronchoalveolar lavage eosinophil counts, lung interleukin-13 and interleukin-5 levels, and airway hyperreactivity. A tendency toward a decrease in serum Bet v 1-specific immunoglobulin G1 was likewise observed. CONCLUSION Intranasally administered L. rhamnosus GG prevents the development of cardinal features of birch pollen-induced allergic asthma in a strain-specific manner.
Collapse
Affiliation(s)
- I. Spacova
- Centre of Microbial and Plant Genetics Department of Microbial and Molecular Systems (M²S) KU Leuven Leuven Belgium
- Research Group Environmental Ecology and Applied Microbiology Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - M. I. Petrova
- Centre of Microbial and Plant Genetics Department of Microbial and Molecular Systems (M²S) KU Leuven Leuven Belgium
- Research Group Environmental Ecology and Applied Microbiology Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - A. Fremau
- Centre of Microbial and Plant Genetics Department of Microbial and Molecular Systems (M²S) KU Leuven Leuven Belgium
| | - L. Pollaris
- Centre for Environment and Health Department of Public Health and Primary Care KU Leuven Leuven Belgium
| | - J. Vanoirbeek
- Centre for Environment and Health Department of Public Health and Primary Care KU Leuven Leuven Belgium
| | - J. L. Ceuppens
- Laboratory of Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. Seys
- Laboratory of Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. Lebeer
- Centre of Microbial and Plant Genetics Department of Microbial and Molecular Systems (M²S) KU Leuven Leuven Belgium
- Research Group Environmental Ecology and Applied Microbiology Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| |
Collapse
|
34
|
Capasso L, Vento G, Loddo C, Tirone C, Iavarone F, Raimondi F, Dani C, Fanos V. Oxidative Stress and Bronchopulmonary Dysplasia: Evidences From Microbiomics, Metabolomics, and Proteomics. Front Pediatr 2019; 7:30. [PMID: 30815432 PMCID: PMC6381008 DOI: 10.3389/fped.2019.00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/24/2019] [Indexed: 01/02/2023] Open
Abstract
Bronchopulmonary dysplasia is a major issue affecting morbidity and mortality of surviving premature babies. Preterm newborns are particularly susceptible to oxidative stress and infants with bronchopulmonary dysplasia have a typical oxidation pattern in the early stages of this disease, suggesting the important role of oxidative stress in its pathogenesis. Bronchopulmonary dysplasia is a complex disease where knowledge advances as new investigative tools become available. The explosion of the "omics" disciplines has recently affected BPD research. This review focuses on the new evidence coming from microbiomics, metabolomics and proteomics in relation to oxidative stress and pathogenesis of bronchopulmonary dysplasia. Since the pathogenesis is not yet completely understood, information gained in this regard would be important for planning an efficacious prevention and treatment strategy for the future.
Collapse
Affiliation(s)
- Letizia Capasso
- Neonatology, Section of Pediatrics, Department of Translational Sciences, University of Naples Federico II, Naples, Italy
| | - Giovanni Vento
- Division of Neonatology, Department of Woman and Child Health, Pediatrics area, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Loddo
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Azienda Ospedaliero-Universitaria Cagliari and University of Cagliari, Cagliari, Italy
| | - Chiara Tirone
- Division of Neonatology, Department of Woman and Child Health, Pediatrics area, Fondazione Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Iavarone
- Institute of Biochemistry, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Raimondi
- Neonatology, Section of Pediatrics, Department of Translational Sciences, University of Naples Federico II, Naples, Italy
| | - Carlo Dani
- Neonatology, University Hospital Careggi, Firenze, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, Azienda Ospedaliero-Universitaria Cagliari and University of Cagliari, Cagliari, Italy
| |
Collapse
|
35
|
Martens K, Pugin B, De Boeck I, Spacova I, Steelant B, Seys SF, Lebeer S, Hellings PW. Probiotics for the airways: Potential to improve epithelial and immune homeostasis. Allergy 2018; 73:1954-1963. [PMID: 29869783 DOI: 10.1111/all.13495] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2018] [Indexed: 12/30/2022]
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefit on the host. The therapeutic effects of probiotics have been mostly studied in the gastrointestinal tract, but recent evidence points toward the potential of these bacteria to prevent and/or treat chronic airway diseases. In this review, possible mechanisms of action of probiotics in the airways are described, with a particular focus on their capacity to modulate the epithelial barrier function and their mode of interaction with the immune system. Indeed, probiotic bacteria, mostly lactobacilli, can promote the expression and regulation of tight junctions and adherence junctions, resulting in the restoration of a defective epithelial barrier. These bacteria interact with the epithelial barrier and immune cells through pattern recognition receptors, such as Toll-like receptors, which upon activation can stimulate or suppress various immune responses. Finally, the clinical potential of probiotics to treat inflammatory diseases of the upper and lower respiratory tract, and the difference between their mode of application (eg, oral or nasal) are discussed here.
Collapse
Affiliation(s)
- K. Martens
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - B. Pugin
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - I. De Boeck
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - I. Spacova
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - B. Steelant
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. F. Seys
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - S. Lebeer
- Department of Bioscience Engineering University of Antwerp Antwerp Belgium
| | - P. W. Hellings
- Clinical Immunology Department of Microbiology and Immunology KU Leuven Leuven Belgium
- Clinical Division of Otorhinolaryngology, Head and Neck Surgery University Hospitals Leuven Leuven Belgium
- Department of Otorhinolaryngology University Hospitals Ghent Ghent Belgium
- Department of Otorhinolaryngology Academic Medical Center University of Amsterdam Amsterdam the Netherlands
| |
Collapse
|
36
|
Spacova I, Ceuppens JL, Seys SF, Petrova MI, Lebeer S. Probiotics against airway allergy: host factors to consider. Dis Model Mech 2018; 11:11/7/dmm034314. [PMID: 30037806 PMCID: PMC6078401 DOI: 10.1242/dmm.034314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The worldwide prevalence of allergic diseases has drastically increased in the past decades. Recent studies underline the importance of microbial exposure for the development of a balanced immune system. Consequently, probiotic bacteria are emerging as a safe and natural strategy for allergy prevention and treatment. However, clinical probiotic intervention studies have so far yielded conflicting results. There is increasing awareness about the importance of host-associated factors that determine whether an individual will respond to a specific probiotic treatment, and it is therefore crucial to promote a knowledge-based instead of an empirical selection of promising probiotic strains and their administration regimen.In this Review, we summarize the insights from animal model studies of allergic disease, which reveal how host-related factors - such as genetic makeup, sex, age and microbiological status - can impact the outcomes of preventive or curative probiotic treatment. We explore why and how these factors can influence the results of probiotic studies and negatively impact the reproducibility in animal experiments. These same factors might profoundly influence the outcomes of human clinical trials, and can potentially explain the conflicting results from probiotic intervention studies. Therefore, we also link these host-related factors to human probiotic study outcomes in the context of airway allergies.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium.,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Jan L Ceuppens
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Sven F Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Mariya I Petrova
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium .,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW In terms of immune regulating functions, analysis of the microbiome has led the development of therapeutic strategies that may be applicable to asthma management. This review summarizes the current literature on the gut and lung microbiota in asthma pathogenesis with a focus on the roles of innate molecules and new microbiome-mediated therapeutics. RECENT FINDINGS Recent clinical and basic studies to date have identified several possible therapeutics that can target innate immunity and the microbiota in asthma. Some of these drugs have shown beneficial effects in the treatment of certain asthma phenotypes and for protection against asthma during early life. Current clinical evidence does not support the use of these therapies for effective treatment of asthma. The integration of the data regarding microbiota with technologic advances, such as next generation sequencing and omics offers promise. Combining comprehensive bioinformatics, new molecules and approaches may shape future asthma treatment.
Collapse
|
38
|
Abstract
The mucosal surfaces of the human body are typically colonized by polymicrobial communities seeded in infancy and are continuously shaped by environmental exposures. These communities interact with the mucosal immune system to maintain homeostasis in health, but perturbations in their composition and function are associated with lower airway diseases, including asthma, a developmental and heterogeneous chronic disease with various degrees and types of airway inflammation. This review will summarize recent studies examining airway microbiota dysbioses associated with asthma and their relationship with the pathophysiology of this disease.
Collapse
|
39
|
Collins JJP, Tibboel D, de Kleer IM, Reiss IKM, Rottier RJ. The Future of Bronchopulmonary Dysplasia: Emerging Pathophysiological Concepts and Potential New Avenues of Treatment. Front Med (Lausanne) 2017; 4:61. [PMID: 28589122 PMCID: PMC5439211 DOI: 10.3389/fmed.2017.00061] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
Yearly more than 15 million babies are born premature (<37 weeks gestational age), accounting for more than 1 in 10 births worldwide. Lung injury caused by maternal chorioamnionitis or preeclampsia, postnatal ventilation, hyperoxia, or inflammation can lead to the development of bronchopulmonary dysplasia (BPD), one of the most common adverse outcomes in these preterm neonates. BPD patients have an arrest in alveolar and microvascular development and more frequently develop asthma and early-onset emphysema as they age. Understanding how the alveoli develop, and repair, and regenerate after injury is critical for the development of therapies, as unfortunately there is still no cure for BPD. In this review, we aim to provide an overview of emerging new concepts in the understanding of perinatal lung development and injury from a molecular and cellular point of view and how this is paving the way for new therapeutic options to prevent or treat BPD, as well as a reflection on current treatment procedures.
Collapse
Affiliation(s)
- Jennifer J P Collins
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Ismé M de Kleer
- Division of Pediatric Pulmonology, Department of Pediatrics, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
40
|
Holvoet S, Doucet-Ladevèze R, Perrot M, Barretto C, Nutten S, Blanchard C. Beneficial effect of Lactococcus lactis NCC 2287 in a murine model of eosinophilic esophagitis. Allergy 2016; 71:1753-1761. [PMID: 27259693 DOI: 10.1111/all.12951] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a severe inflammatory disease of the esophagus which is characterized histologically by an eosinophilic infiltration into the esophageal tissue. The efficacy of probiotics in the context of atopic diseases has been well investigated but, to date, there has been no study which has evaluated probiotic effects on EoE inflammation. This study sought to identify a probiotic which improves esophageal inflammation in experimental EoE. METHODS Two candidate probiotics, Lactococcus lactis NCC 2287 and Bifidobacterium lactis NCC 2818, were tested in a murine model of EoE elicited by epicutaneous sensitization with Aspergillus fumigatus protein extract. Administration of bacterial strains in drinking water was used, respectively, as a preventive or treatment measure, or continuously throughout the study. Inflammatory parameters were assessed in the esophagus, skin, and lungs after allergen challenge. RESULTS In this EoE model, supplementation with L. lactis NCC 2287 significantly decreased esophageal and bronchoalveolar eosinophilia but only when given as a therapeutic treatment. No significant effect on eosinophilia was observed when NCC 2287 was given as a preventive or a continuous intervention. NCC 2287 supplementation had no significant effect on immunoglobulin levels, skin symptom scores, or on transepidermal water loss. Supplementation with another probiotic, B. lactis NCC 2818, had no significant effect on esophageal eosinophilia. CONCLUSION We identified a L. lactis strain, able to attenuate esophageal eosinophilic inflammation in a preclinical model of EoE. This effect is strain specific and depends on the timing and duration of bacterial supplementation. Confirmation of these observations in human clinical trials is warranted.
Collapse
Affiliation(s)
- S. Holvoet
- Nutrition and Health Research; Allergy Group; Nestle Research Center; Lausanne Switzerland
| | - R. Doucet-Ladevèze
- Nutrition and Health Research; Allergy Group; Nestle Research Center; Lausanne Switzerland
| | - M. Perrot
- Data Science Group; Nestle Research Center; Lausanne Switzerland
| | - C. Barretto
- Data Science Group; Nestle Research Center; Lausanne Switzerland
| | - S. Nutten
- Nutrition and Health Research; Allergy Group; Nestle Research Center; Lausanne Switzerland
| | - C. Blanchard
- Nutrition and Health Research; Allergy Group; Nestle Research Center; Lausanne Switzerland
| |
Collapse
|
41
|
Lal CV, Travers C, Aghai ZH, Eipers P, Jilling T, Halloran B, Carlo WA, Keeley J, Rezonzew G, Kumar R, Morrow C, Bhandari V, Ambalavanan N. The Airway Microbiome at Birth. Sci Rep 2016; 6:31023. [PMID: 27488092 PMCID: PMC4973241 DOI: 10.1038/srep31023] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/13/2016] [Indexed: 12/29/2022] Open
Abstract
Alterations of pulmonary microbiome have been recognized in multiple respiratory disorders. It is critically important to ascertain if an airway microbiome exists at birth and if so, whether it is associated with subsequent lung disease. We found an established diverse and similar airway microbiome at birth in both preterm and term infants, which was more diverse and different from that of older preterm infants with established chronic lung disease (bronchopulmonary dysplasia). Consistent temporal dysbiotic changes in the airway microbiome were seen from birth to the development of bronchopulmonary dysplasia in extremely preterm infants. Genus Lactobacillus was decreased at birth in infants with chorioamnionitis and in preterm infants who subsequently went on to develop lung disease. Our results, taken together with previous literature indicating a placental and amniotic fluid microbiome, suggest fetal acquisition of an airway microbiome. We speculate that the early airway microbiome may prime the developing pulmonary immune system, and dysbiosis in its development may set the stage for subsequent lung disease.
Collapse
Affiliation(s)
- Charitharth Vivek Lal
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA.,Translational Research in Normal and Disordered Development Program (TReNDD) University of Alabama at Birmingham, AL, USA.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, AL, USA
| | - Colm Travers
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA
| | - Zubair H Aghai
- Department of Pediatrics, Thomas Jefferson University/Nemours, Philadelphia, PA, USA
| | - Peter Eipers
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA.,Translational Research in Normal and Disordered Development Program (TReNDD) University of Alabama at Birmingham, AL, USA
| | - Brian Halloran
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA.,Translational Research in Normal and Disordered Development Program (TReNDD) University of Alabama at Birmingham, AL, USA
| | - Waldemar A Carlo
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA
| | - Jordan Keeley
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA
| | - Gabriel Rezonzew
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA
| | - Ranjit Kumar
- Center for Clinical and Translational Sciences, University of Alabama at Birmingham, AL, USA
| | - Casey Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, AL, USA.,Translational Research in Normal and Disordered Development Program (TReNDD) University of Alabama at Birmingham, AL, USA.,Center for Clinical and Translational Sciences, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
42
|
Abstract
The increase in allergic airways disease has been linked to modern urbanization and lifestyle. Recent evidence suggests that the associated reduction in microbial exposure, reduction in dietary fibre intake and increased antibiotic use may cause early dysbiosis in infancy, which predisposes to immune dysregulation and allergic airways disease later in life. This implies that there may be a window of opportunity for primary prevention strategies aimed to protect or restore the microbiome early in life and thereby decrease the risk of developing allergic airways disease. Alternatively, strategies that correct dysbiosis may aid in the treatment of established allergic airways disease.
Collapse
|
43
|
Bugging inflammation: role of the gut microbiota. Clin Transl Immunology 2016; 5:e72. [PMID: 27195115 PMCID: PMC4855262 DOI: 10.1038/cti.2016.12] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 12/12/2022] Open
Abstract
The advent of vaccination and improved hygiene have eliminated many of the deadly infectious pathogens in developed nations. However, the incidences of inflammatory diseases, such as inflammatory bowel disease, asthma, obesity and diabetes are increasing dramatically. Research in the recent decades revealed that it is indeed the lack of early childhood microbial exposure, increase use of antibiotics, as well as increase consumption of processed foods high in carbohydrates and fats, and lacking fibre, which wreak havoc on the proper development of immunity and predispose the host to elevated inflammatory conditions. Although largely unexplored and under-appreciated until recent years, these factors impact significantly on the composition of the gut microbiota (a collection of microorganisms that live within the host mucosal tissue) and inadvertently play intricate and pivotal roles in modulating an appropriate host immune response. The suggestion that shifts in the composition of host microbiota is a risk factor for inflammatory disease raises an exciting opportunity whereby the microbiota may also present as a potential modifiable component or therapeutic target for inflammatory diseases. This review provides insights into the interactions between the microbiota and the immune system, how these affect disease phenotypes, and explore current and emerging therapies that target the gut microbiota as potential treatment for inflammatory diseases.
Collapse
|
44
|
PEERAJAN S, CHAIYASUT C, SIRILUN S, CHAIYASUT K, KESIKA P, SIVAMARUTHI BS. Enrichment of nutritional value of Phyllanthus emblica fruit juice using the probiotic bacterium, Lactobacillus paracasei HII01 mediated fermentation. FOOD SCIENCE AND TECHNOLOGY 2016. [DOI: 10.1590/1678-457x.0064] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Abstract
The human microbiome, i.e. the collection of microbes that live on, in and interact with the human body, is extraordinarily diverse; microbiota have been detected in every tissue of the human body interrogated to date. Resident microbiota interact extensively with immune cells and epithelia at mucosal surfaces including the airways, and chronic inflammatory and allergic respiratory disorders are associated with dysbiosis of the airway microbiome. Chronic rhinosinusitis (CRS) is a heterogeneous disease with a large socioeconomic impact, and recent studies have shown that sinus inflammation is associated with decreased sinus bacterial diversity and the concomitant enrichment of specific sinus pathogens. Here, we discuss the potential role for probiotic supplementation for CRS in light of this increasing understanding of the airway microbiome and microbial interactions with the host. We focus on the ecological significance of microbiome-based probiotic supplementation and potential interactions with the gastrointestinal tract and consider microbial administration methods for treatment of CRS.
Collapse
|
46
|
The effect of probiotics on immune regulation, acne, and photoaging. Int J Womens Dermatol 2015; 1:85-89. [PMID: 28491964 PMCID: PMC5418745 DOI: 10.1016/j.ijwd.2015.02.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 02/06/2023] Open
Abstract
Probiotics are live micro-organisms that provide a health benefit to the host. The role of probiotics in the management of disease, as well as immune modification, has recently experienced a renewed interest in society, as probiotics can be found in products ranging from yogurt to facial creams. In this article, we discuss the role of probiotics in the development of the immune system, the treatment of acne and rosacea, and protection against aging and photodamage.
Collapse
|
47
|
Vissers M, de Groot R, Ferwerda G. Severe viral respiratory infections: are bugs bugging? Mucosal Immunol 2014; 7:227-38. [PMID: 24220300 DOI: 10.1038/mi.2013.93] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/09/2013] [Indexed: 02/07/2023]
Abstract
Viral respiratory tract infections (RTI) pose a high burden on the youngest members of our society. Several risk factors are known for severe viral respiratory disease. However, a large proportion of the severe RTI cannot be explained by these risk factors. A growing body of evidence shows that the composition of the microbiota has a major influence on the training of both the mucosal and the systemic immune response and can thus potentially determine susceptibility for severe viral infections. In this review, we discuss the current evidence regarding the influence of bacterial colonization on the severity of viral respiratory infections.
Collapse
Affiliation(s)
- M Vissers
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| | - R de Groot
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| | - G Ferwerda
- 1] Department of Pediatrics, Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands [2] Nijmegen Institute for Infection, Inflammation and Immunity, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Perrin Y, Nutten S, Audran R, Berger B, Bibiloni R, Wassenberg J, Barbier N, Aubert V, Moulin J, Singh A, Magliola C, Mercenier A, Spertini F. Comparison of two oral probiotic preparations in a randomized crossover trial highlights a potentially beneficial effect of Lactobacillus paracasei NCC2461 in patients with allergic rhinitis. Clin Transl Allergy 2014; 4:1. [PMID: 24393277 PMCID: PMC3925289 DOI: 10.1186/2045-7022-4-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 12/17/2013] [Indexed: 12/14/2022] Open
Abstract
Background There is promising but conflicting evidence to recommend the addition of probiotics to foods for prevention and treatment of allergy. Based on previous studies with fermented milk containing Lactobacillus paracasei NCC2461, we aimed to compare the effect of a powder form of the latter probiotic with the effect of a blend of Lactobacillus acidophilus ATCC SD5221 and Bifidobacterium lactis ATCC SD5219 in patients with allergic rhinitis. Methods A double-blind, randomized, cross-over study, involving 31 adults with allergic rhinitis to grass pollen, was performed outside the grass pollen season (registration number: NCT01233154). Subjects received each product for 4-weeks in two phases separated by a wash-out period of 6 to 8 weeks. A nasal provocation test was performed before and after each 4-week product intake period, and outcome parameters (objective and subjective clinical symptoms; immune parameters) were measured during and/or 24 hours after the test. Results Out of the 31 subject enrolled, 28 completed the study. While no effect was observed on nasal congestion (primary outcome), treatment with NCC2461 significantly decreased nasal pruritus (determined by VAS), and leukocytes in nasal fluid samples, enhanced IL-5, IL-13 and IL-10 production by peripheral blood mononuclear cells in an allergen specific manner and tended to decrease IL-5 secretion in nasal fluid, in contrast to treatment with the blend of L. acidophilus and B. lactis. Conclusions Despite short-term consumption, NCC2461 was able to reduce subjective nasal pruritus while not affecting nasal congestion in adults suffering from grass pollen allergic rhinitis. The associated decrease in nasal fluid leukocytes and IL-5 secretion, and the enhanced IL-10 secretion in an allergen specific manner may partly explain the decrease in nasal pruritus. However, somewhat unexpected systemic immune changes were also noted. These data support the study of NCC2461 consumption in a seasonal clinical trial to further demonstrate its potentially beneficial effect.
Collapse
Affiliation(s)
- Yannick Perrin
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Régine Audran
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Rodrigo Bibiloni
- Nestlé Research Centre, Lausanne, Switzerland.,Current address: AgResearch Ltd, Food and Bio-based Products, Hamilton, New Zealand
| | - Jacqueline Wassenberg
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Nathalie Barbier
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Vincent Aubert
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | | | | | - François Spertini
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
49
|
Probiotic strategies for treatment of respiratory diseases. Trends Microbiol 2013; 21:485-92. [DOI: 10.1016/j.tim.2013.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/12/2013] [Accepted: 04/23/2013] [Indexed: 02/03/2023]
|
50
|
Howarth GS, Wang H. Role of endogenous microbiota, probiotics and their biological products in human health. Nutrients 2013; 5:58-81. [PMID: 23306189 PMCID: PMC3571638 DOI: 10.3390/nu5010058] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 12/05/2012] [Accepted: 12/14/2012] [Indexed: 02/06/2023] Open
Abstract
Although gut diseases such as inflammatory bowel disease, mucositis and the alimentary cancers share similar pathogenetic features, further investigation is required into new treatment modalities. An imbalance in the gut microbiota, breached gut integrity, bacterial invasion, increased cell apoptosis to proliferation ratio, inflammation and impaired immunity may all contribute to their pathogenesis. Probiotics are defined as live bacteria, which when administered in sufficient amounts, exert beneficial effects to the gastrointestinal tract. More recently, probiotic-derived factors including proteins and other molecules released from living probiotics, have also been shown to exert beneficial properties. In this review we address the potential for probiotics, with an emphasis on probiotic-derived factors, to reduce the severity of digestive diseases and further discuss the known mechanisms by which probiotics and probiotic-derived factors exert their physiological effects.
Collapse
Affiliation(s)
- Gordon S. Howarth
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus, South Australia 5371, Australia; E-Mail:
- Centre for Paediatric and Adolescent Gastroenterology, Children, Youth and Women’s Health Service, North Adelaide, South Australia 5006, Australia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +61-8-8313-7885; Fax: +61-8-8313-7972
| | - Hanru Wang
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus, South Australia 5371, Australia; E-Mail:
| |
Collapse
|