1
|
Sécher T, Cortes M, Boisseau C, Barba Goudiaby MT, Pitiot A, Parent C, Thomas M, Heuzé-Vourc’h N. Synergy between Lactobacillus murinus and anti-PcrV antibody delivered in the airways to boost protection against Pseudomonas aeruginosa. Mol Ther Methods Clin Dev 2024; 32:101330. [PMID: 39314638 PMCID: PMC11418128 DOI: 10.1016/j.omtm.2024.101330] [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: 12/14/2023] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Therapeutic antibodies (Ab) have revolutionized the management of multiple illnesses including respiratory tract infections (RTIs). However, anti-infectious Ab displayed several limitations including antigen restrictiveness, narrowed therapeutic windows, and limited dose in the vicinity of the target when delivered by parenteral routes. Strategies enhancing further Ab-dependent containment of infection are currently needed. Here we showed that a combination of inhaled anti-infectious Ab and probiotics is an efficient formulation to protect against lung infection. Using a mouse model of Pseudomonas aeruginosa-induced pneumonia, we demonstrated a synergistic effect reducing both bacterial burden and pro-inflammatory response affording protection against primary and secondary infections. This is the first study showing that the local combination in the airways of anti-infective Ab and probiotics subverts suboptimal potency of Ab monotherapy and provides protection against respiratory pathogen.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Mélanie Cortes
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Chloé Boisseau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Marie-Thérèse Barba Goudiaby
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, UMR1319 Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Aubin Pitiot
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| | - Muriel Thomas
- Institut Micalis, INRA, AgroParisTech, Université Paris-Saclay, UMR1319 Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Nathalie Heuzé-Vourc’h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
- Université de Tours, Centre d'Etude des Pathologies Respiratoires, U1100 Tours, France
| |
Collapse
|
2
|
Byun AS, Vitetta L, Chan HK, Kwok PCL. Respiratory Delivery of Lacticaseibacillus rhamnosus GG by Vibrating-Mesh and Jet Nebulisation. Pharmaceutics 2024; 16:1326. [PMID: 39458655 PMCID: PMC11510752 DOI: 10.3390/pharmaceutics16101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The use of probiotic bacteria to improve lung health has been gaining interest. Although the oral delivery of probiotics and their effects are well documented, there is currently limited knowledge on the respiratory delivery of probiotics. OBJECTIVES This study aimed to investigate whether nebulisation is suitable for delivering Lacticaseibacillus rhamnosus GG (LGG) into the lungs for the potential treatment of bacterial pulmonary infections. METHODS It compared the dose output and aerosol performance of a vibrating-mesh nebuliser (VMN) and a jet nebuliser (JN) in nebulising LGG suspended in de Man Rogosa Sharpe (MRS) broth, phosphate-buffered saline (PBS), or normal saline (0.9% w/v sodium chloride in water). RESULTS The VMN consistently produced a higher output than the JN for all liquid media, indicating that VMN was more efficient. The fine-particle fractions of both nebulisers were comparable for a given medium. The highest fine-particle fraction was achieved with LGG suspended in MRS broth for both nebulisers (20.5 ± 2.8% for VMN; 18.7 ± 3.4% for JN). This suggests that the aerosol performance of nebulised probiotics may depend on the medium in which the probiotic bacteria were suspended. CONCLUSIONS Therefore, this study demonstrated that the nebulisation efficiency of LGG depended on the nebuliser type and liquid medium of the probiotic suspension.
Collapse
Affiliation(s)
| | | | | | - Philip Chi Lip Kwok
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia; (A.S.B.); (L.V.); (H.-K.C.)
| |
Collapse
|
3
|
Seo H, Yoon Y, Kim S, Ghorbanian F, Tajdozian H, Jo S, Barman I, Lee S, Lee Y, Rahim MA, Hossain MS, Lee S, Song HY. Anti-tuberculosis effect of microbiome therapeutic PMC205 in extensively drug-resistant pulmonary tuberculosis in vivo. Int J Antimicrob Agents 2024; 64:107274. [PMID: 39002701 DOI: 10.1016/j.ijantimicag.2024.107274] [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: 06/15/2023] [Revised: 06/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis, and the increase in antibiotic resistance threatens humankind. Therefore, there is an urgent need to develop new anti-tuberculosis drugs that can overcome the limitations of existing drugs. Here, we report the anti-tuberculosis effect of microbiome therapeutic PMC205, a strain of Bacillus subtilis. METHODS The anti-tuberculosis activity of probiotics was evaluated in mouse models of lethal and latent pulmonary tuberculosis induced by high or low-dose infection of the extensively drug-resistant strain. Probiotics were administered by inhalation, and the burden of M. tuberculosis in the lungs, along with mortality and clinical observations, were monitored for 12 weeks and 8 months, respectively. For an in-depth understanding, analysis of the microbiome and inflammatory profile of the lung microenvironment and induction of autophagy in vitro were explored. RESULTS After inhalation administration of PMC205 for 3 months, the survival rate was 100%, unlike all deaths in the saline-treated group, and the burden of M. tuberculosis in the lungs was reduced by log 1.3 in the 8-month latent tuberculosis model. Moreover, PMC205 induced recovery of disrupted lung microflora, increased butyric acid, and suppressed excessive inflammation. It also promoted autophagy. CONCLUSIONS These results confirm PMC205's anti-tuberculosis effect, suggesting that it can be developed as an adjuvant to current antibiotic therapy to solve the drug-resistant tuberculosis problem.
Collapse
Affiliation(s)
- Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Hanieh Tajdozian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sujin Jo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Soyeon Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Saebim Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea.
| |
Collapse
|
4
|
Zemanick ET, Rosas-Salazar C. The Role of the Microbiome in Pediatric Respiratory Diseases. Clin Chest Med 2024; 45:587-597. [PMID: 39069323 DOI: 10.1016/j.ccm.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Numerous studies have examined the role of the microbiome and microbiome-based therapeutics in many childhood airway and lung diseases. In this narrative review, the authors first give a brief overview of the current methods used in microbiome research. The authors then review the literature linking the microbiome with (1) early-life acute respiratory infections due to respiratory syncytial virus, (2) childhood asthma onset, (3) cystic fibrosis, and (4) bronchopulmonary dysplasia, focusing on recent studies that have used culture-independent methods to characterize the respiratory or gut microbiome in the pediatric population.
Collapse
Affiliation(s)
- Edith T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, 13123 East 16th Avenue, B-395, Aurora, CO 80045, USA
| | - Christian Rosas-Salazar
- Department of Pediatrics, Vanderbilt University Medical Center and Monroe Carell Jr. Children's Hospital at Vanderbilt, 2200 Children's Way, Doctors' Office Tower, Suite 11215, Nashville, TN 37232, USA.
| |
Collapse
|
5
|
Lyu J, Gao M, Zhao S, Liu X, Zhao X, Zou Y, Zhong Y, Ge L, Zhang H, Huang L, Fan S, Xiao L, Zhang X. From whole genomes to probiotic candidates: A study of potential lactobacilli strains selection for vaginitis treatment. Heliyon 2024; 10:e30495. [PMID: 38765070 PMCID: PMC11098787 DOI: 10.1016/j.heliyon.2024.e30495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024] Open
Abstract
Vaginitis, characterized by pathogenic invasion and a deficiency in beneficial lactobacilli, has recognized lactobacilli supplementation as a novel therapeutic strategy. However, due to individual differences in vaginal microbiota, identifying universally effective Lactobacillus strains is challenging. Traditional methodologies for probiotic selection, which heavily depend on extensive in vitro experiments, are both time-intensive and laborious. The aim of this study was to pinpoint possible vaginal probiotic candidates based on whole-genome screening. We sequenced the genomes of 98 previously isolated Lactobacillus strains, annotating their genes involved in probiotic metabolite biosynthesis, adherence, acid/bile tolerance, and antibiotic resistance. A scoring system was used to assess the strains based on their genomic profiles. The highest-scoring strains underwent further in vitro evaluation. Consequently, two strains, Lactobacillus crispatus LG55-27 and Lactobacillus gasseri TM13-16, displayed an outstanding ability to produce d-lactate and adhere to human vaginal epithelial cells. They also showed higher antimicrobial activity against Gardnerella vaginalis, Escherichia coli, Candida albicans, Staphylococcus aureus, and Pseudomonas aeruginosa compared to reference Lactobacillus strains. Their resilience to acid and bile environments highlights the potential for oral supplementation. Oral and vaginal administration of these two strains were tested in a bacterial vaginosis (BV) rat model at various doses. Results indicated that combined vaginal administration of these strains at 1 × 106 CFU/day significantly mitigated BV in rats. This research offers a probiotic dosage guideline for vaginitis therapy, underscoring an efficient screening process for probiotics using genome sequencing, in vitro testing, and in vivo BV model experimentation.
Collapse
Affiliation(s)
- Jinli Lyu
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Mengyu Gao
- BGI-Shenzhen, Shenzhen, 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, 518083, China
| | - Shaowei Zhao
- BGI-Shenzhen, Shenzhen, 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, 518083, China
| | - Xinyang Liu
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Xinlong Zhao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanqiang Zou
- BGI-Shenzhen, Shenzhen, 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yiyi Zhong
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Lan Ge
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Hiafeng Zhang
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Liting Huang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Shangrong Fan
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Liang Xiao
- BGI-Shenzhen, Shenzhen, 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, 518083, China
| | - Xiaowei Zhang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- BGI-Shenzhen, Shenzhen, 518083, China
| |
Collapse
|
6
|
Sarnelli G, Del Re A, Palenca I, Franzin SB, Lu J, Seguella L, Zilli A, Pesce M, Rurgo S, Esposito G, Sanseverino W, Esposito G. Intranasal administration of Escherichia coli Nissle expressing the spike protein of SARS-CoV-2 induces long-term immunization and prevents spike protein-mediated lung injury in mice. Biomed Pharmacother 2024; 174:116441. [PMID: 38518597 DOI: 10.1016/j.biopha.2024.116441] [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: 02/23/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
While current anti-Spike protein (SP) vaccines have been pivotal in managing the pandemic, their limitations in delivery, storage, and the inability to provide mucosal immunization (preventing infections) highlight the ongoing necessity for research and innovation. To tackle these constraints, our research group developed a bacterial-based vaccine using a non-pathogenic E. coli Nissle 1917 (EcN) strain genetically modified to express the SARS-CoV-2 spike protein on its surface (EcN-pAIDA1-SP). We intranasally delivered the EcN-pAIDA1-SP in two doses and checked specific IgG/IgA production as well as the key immune mediators involved in the process. Moreover, following the initial and booster vaccine doses, we exposed both immunized and non-immunized mice to intranasal delivery of SARS-CoV-2 SP to assess the effectiveness of EcN-pAIDA1-SP in protecting lung tissue from the inflammation damage. We observed detectable levels of anti-SARS-CoV-2 spike IgG in serum samples and IgA in bronchoalveolar lavage fluid two weeks after the initial treatment, with peak concentrations in the respective samples on the 35th day. Moreover, immunoglobulins displayed a progressively enhanced avidity index, suggesting a selective binding to the spike protein. Finally, the pre-immunized group displayed a decrease in proinflammatory markers (TLR4, NLRP3, ILs) following SP challenge, compared to the non-immunized groups, along with better preservation of tissue morphology. Our probiotic-based technology provides an effective immunobiotic tool to protect individuals against disease and control infection spread.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy; Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Alessandro Del Re
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Jie Lu
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Anatomy and Cell Biology, China Medical University, Shenyang 110122, China.
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, Section of Gastroenterology, University Federico II, Naples 80138, Italy.
| | - Giovanni Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Molecular Medicine and Medical Biotechnologies, Centro Ingegneria Genetica-Biotecnologie Avanzate s.c.a rl, Naples 80131, Italy.
| | - Walter Sanseverino
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy.
| | - Giuseppe Esposito
- Nextbiomics S.R.L. (Società a Responsabilità Limitata), Naples 80100, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
7
|
Taufer CR, da Silva J, Rampelotto PH. The Influence of Probiotic Lactobacilli on COVID-19 and the Microbiota. Nutrients 2024; 16:1350. [PMID: 38732597 PMCID: PMC11085918 DOI: 10.3390/nu16091350] [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: 03/07/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 05/13/2024] Open
Abstract
This comprehensive review explores the potential of using lactobacilli as a probiotic in the management of COVID-19. Our findings suggest that lactobacilli show promise in reducing the risk of death, gastrointestinal and overall symptoms, and respiratory failure, as well as in lowering cytokines and inflammatory markers associated with the disease. The molecular mechanisms by which lactobacilli protect against COVID-19 and other viral infections may be related to the reduction in inflammation, modulation of the immune response, and direct interaction with viruses to produce antiviral substances. However, the selected studies demonstrate the presence of mixed findings for various clinical, biochemical, hematological, and immunological parameters, which may be attributed to methodological differences among studies. We highlight the importance of clearly describing randomization processes to minimize bias and caution against small sample sizes and inappropriate statistical tests that could lead to errors. This review offers valuable insights into the therapeutic potential of lactobacilli in the context of COVID-19 and identifies avenues for further research and applications. These findings hold promise for the development of novel approaches to managing COVID-19 and warrant further investigation into the potential benefits of lactobacilli in combating the disease.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Juliana da Silva
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
- Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
8
|
Cao Z, Pang Y, Pu J, Liu J. Bacteria-based drug delivery for treating non-oncological diseases. J Control Release 2024; 366:668-683. [PMID: 38219912 DOI: 10.1016/j.jconrel.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
Bacteria inhabit all over the human body, especially the skin, gastrointestinal tract, respiratory tract, urogenital tract, as well as specific lesion sites, such as wound and tumor. By leveraging their distinctive attributes including rapid proliferation, inherent abilities to colonize various biointerfaces in vivo and produce diverse biomolecules, and the flexibility to be functionalized via genetic engineering or surface modification, bacteria have been widely developed as living therapeutic agents, showing promising potential to make a great impact on the exploration of advanced drug delivery systems. In this review, we present an overview of bacteria-based drug delivery and its applications in treating non-oncological diseases. We systematically summarize the physiological positions where living bacterial therapeutic agents can be delivered to, including the skin, gastrointestinal tract, respiratory tract, and female genital tract. We discuss the success of using bacteria-based drug delivery systems in the treatment of diseases that occur in specific locations, such as skin wound healing/infection, inflammatory bowel disease, respiratory diseases, and vaginitis. We also discuss the advantages as well as the limitations of these living therapeutics and bacteria-based drug delivery, highlighting the key points that need to be considered for further translation. This review article may provide unique insights for designing next-generation bacteria-based therapeutics and developing advanced drug delivery systems.
Collapse
Affiliation(s)
- Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
9
|
Yarlagadda T, Zhu Y, Snape N, Carey A, Bryan E, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. Lactobacillus rhamnosus dampens cytokine and chemokine secretion from primary human nasal epithelial cells infected with rhinovirus. J Appl Microbiol 2024; 135:lxae018. [PMID: 38268489 DOI: 10.1093/jambio/lxae018] [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: 12/03/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
AIMS To investigate the effect of Lactobacillus rhamnosus on viral replication and cellular response to human rhinovirus (HRV) infection, including the secretion of antiviral and inflammatory mediators from well-differentiated nasal epithelial cells (WD-NECs). METHODS AND RESULTS The WD-NECs from healthy adult donors (N = 6) were cultured in vitro, exposed to different strains of L. rhamnosus (D3189, D3160, or LB21), and infected with HRV (RV-A16) after 24 h. Survival and adherence capacity of L. rhamnosus in a NEC environment were confirmed using CFSE-labelled isolates, immunofluorescent staining, and confocal microscopy. Shed virus and viral replication were quantified using TCID50 assays and RT-qPCR, respectively. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity. Pro-inflammatory mediators were measured by multiplex immunoassay, and interferon (IFN)-λ1/3 was measured using a standard ELISA kit. Lactobacillus rhamnosus was able to adhere to and colonize WD-NECs prior to the RV-A16 infection. Lactobacillus rhamnosus did not affect shed RV-A16, viral replication, RV-A16-induced IFN-λ1/3 production, or LDH release. Pre-exposure to L. rhamnosus, particularly D3189, reduced the secretion of RV-A16-induced pro-inflammatory mediators by WD-NECs. CONCLUSIONS These findings demonstrate that L. rhamnosus differentially modulates RV-A16-induced innate inflammatory immune responses in primary NECs from healthy adults.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Yanshan Zhu
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Natale Snape
- University of Queensland Frazer Institute, Woolloongabba 4102, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| |
Collapse
|
10
|
Taufer CR, Rampelotto PH. Lactobacilli in COVID-19: A Systematic Review Based on Next-Generation Sequencing Studies. Microorganisms 2024; 12:284. [PMID: 38399688 PMCID: PMC10891515 DOI: 10.3390/microorganisms12020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The global pandemic was caused by the SARS-CoV-2 virus, known as COVID-19, which primarily affects the respiratory and intestinal systems and impacts the microbial communities of patients. This systematic review involved a comprehensive search across the major literature databases to explore the relationship between lactobacilli and COVID-19. Our emphasis was on investigations employing NGS technologies to explore this connection. Our analysis of nine selected studies revealed that lactobacilli have a reduced abundance in the disease and an association with disease severity. The protective mechanisms of lactobacilli in COVID-19 and other viral infections are likely to be multifaceted, involving complex interactions between the microbiota, the host immune system, and the virus itself. Moreover, upon closely examining the NGS methodologies and associated statistical analyses in each research study, we have noted concerns regarding the approach used to delineate the varying abundance of lactobacilli, which involves potential biases and the exclusion of pertinent data elements. These findings provide new insight into the relationship between COVID-19 and lactobacilli, highlighting the potential for microbiota modulation in COVID-19 treatment.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
11
|
Li R, Li J, Zhou X. Lung microbiome: new insights into the pathogenesis of respiratory diseases. Signal Transduct Target Ther 2024; 9:19. [PMID: 38228603 PMCID: PMC10791971 DOI: 10.1038/s41392-023-01722-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
The lungs were long thought to be sterile until technical advances uncovered the presence of the lung microbial community. The microbiome of healthy lungs is mainly derived from the upper respiratory tract (URT) microbiome but also has its own characteristic flora. The selection mechanisms in the lung, including clearance by coughing, pulmonary macrophages, the oscillation of respiratory cilia, and bacterial inhibition by alveolar surfactant, keep the microbiome transient and mobile, which is different from the microbiome in other organs. The pulmonary bacteriome has been intensively studied recently, but relatively little research has focused on the mycobiome and virome. This up-to-date review retrospectively summarizes the lung microbiome's history, composition, and function. We focus on the interaction of the lung microbiome with the oropharynx and gut microbiome and emphasize the role it plays in the innate and adaptive immune responses. More importantly, we focus on multiple respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), fibrosis, bronchiectasis, and pneumonia. The impact of the lung microbiome on coronavirus disease 2019 (COVID-19) and lung cancer has also been comprehensively studied. Furthermore, by summarizing the therapeutic potential of the lung microbiome in lung diseases and examining the shortcomings of the field, we propose an outlook of the direction of lung microbiome research.
Collapse
Affiliation(s)
- Ruomeng Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Calzada-Fraile D, Sánchez-Madrid F. Reprogramming dendritic cells through the immunological synapse: A two-way street. Eur J Immunol 2023; 53:e2350393. [PMID: 37598303 DOI: 10.1002/eji.202350393] [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: 06/16/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/21/2023]
Abstract
Dendritic cells (DCs) bridge innate and adaptive immunity. Their main function is to present antigens to prime T cells and initiate and shape adaptive responses. Antigen presentation takes place through intimate contacts between the two cells, termed immune synapses (IS). During the formation of IS, information travels towards the T-cell side to induce and tune its activation; but it also travels in reverse via engagement of membrane receptors and within extracellular vesicles transferred to the DC. Such reverse information transfer and its consequences on DC fate have been largely neglected. Here, we review the events and effects of IS-mediated antigen presentation on DCs. In addition, we discuss novel technological advancements that enable monitoring DCs interactions with T lymphocytes, the main effects of DCs undergoing productive IS (postsynaptic DCs, or psDCs), and how reverse information transfer could be harnessed to modulate immune responses for therapeutic intervention.
Collapse
Affiliation(s)
- Diego Calzada-Fraile
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Immunology Department, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
13
|
Chen CM, Yang YCSH, Chou HC, Lin S. Intranasal administration of Lactobacillus johnsonii attenuates hyperoxia-induced lung injury by modulating gut microbiota in neonatal mice. J Biomed Sci 2023; 30:57. [PMID: 37517995 PMCID: PMC10388480 DOI: 10.1186/s12929-023-00958-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Supplemental oxygen impairs lung development in newborn infants with respiratory distress. Lactobacillus johnsonii supplementation attenuates respiratory viral infection in mice and exhibits anti-inflammatory effects. This study investigated the protective effects of intranasal administration of L. johnsonii on lung development in hyperoxia-exposed neonatal mice. METHODS Neonatal C57BL/6N mice were reared in either room air (RA) or hyperoxia condition (85% O2). From postnatal days 0 to 6, they were administered intranasal 10 μL L. johnsonii at a dose of 1 × 105 colony-forming units. Control mice received an equal volume of normal saline (NS). We evaluated the following four study groups: RA + NS, RA + probiotic, O2 + NS, and O2 + probiotic. On postnatal day 7, lung and intestinal microbiota were sampled from the left lung and lower gastrointestinal tract, respectively. The right lung of each mouse was harvested for Western blot, cytokine, and histology analyses. RESULTS The O2 + NS group exhibited significantly lower body weight and vascular density and significantly higher mean linear intercept (MLI) and lung cytokine levels compared with the RA + NS and RA + probiotic groups. At the genus level of the gut microbiota, the O2 + NS group exhibited significantly higher Staphylococcus and Enterobacter abundance and significantly lower Lactobacillus abundance compared with the RA + NS and RA + probiotic groups. Intranasal L. johnsonii treatment increased the vascular density, decreased the MLI and cytokine levels, and restored the gut microbiota in hyperoxia-exposed neonatal mice. CONCLUSIONS Intranasal administration of L. johnsonii protects against hyperoxia-induced lung injury and modulates the gut microbiota.
Collapse
Affiliation(s)
- Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shan Lin
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan, Taiwan
| |
Collapse
|
14
|
Tomkinson S, Triscott C, Schenk E, Foey A. The Potential of Probiotics as Ingestible Adjuvants and Immune Modulators for Antiviral Immunity and Management of SARS-CoV-2 Infection and COVID-19. Pathogens 2023; 12:928. [PMID: 37513775 PMCID: PMC10384479 DOI: 10.3390/pathogens12070928] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Probiotic bacteria are able to modulate general antiviral responsiveness, including barrier functionality and innate and adaptive immune responses. The COVID-19 pandemic, resulting from SARS-CoV-2 infection, has created a need to control and treat this viral infection and its ensuing immunopathology with a variety of approaches; one such approach may involve the administration of probiotic bacteria. As with most viral infections, its pathological responses are not fully driven by the virus, but are significantly contributed to by the host's immune response to viral infection. The potential adoption of probiotics in the treatment of COVID-19 will have to appreciate the fine line between inducing antiviral immunity without over-provoking immune inflammatory responses resulting in host-derived immunopathological tissue damage. Additionally, the effect exerted on the immune system by SARS-CoV-2 evasion strategies will also have to be considered when developing a robust response to this virus. This review will introduce the immunopathology of COVID-19 and the immunomodulatory effects of probiotic strains, and through their effects on a range of respiratory pathogens (IAV, SARS-CoV, RSV), as well as SARS-CoV-2, will culminate in a focus on how these bacteria can potentially manipulate both infectivity and immune responsiveness via barrier functionality and both innate and adaptive immunity. In conclusion, the harnessing of induction and augmentation of antiviral immunity via probiotics may not only act as an ingestible adjuvant, boosting immune responsiveness to SARS-CoV-2 infection at the level of barrier integrity and innate and adaptive immunity, but also act prophylactically to prevent infection and enhance protection afforded by current vaccine regimens.
Collapse
Affiliation(s)
- Sophie Tomkinson
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Cloe Triscott
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Emily Schenk
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
- Peninsula Medical School, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - Andrew Foey
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| |
Collapse
|
15
|
Rasai D, Hosseinian SA, Asasi K, Shekarforosh SS, Tafti K. The beneficial effects of spraying of probiotic Bacillus and Lactobacillus bacteria on broiler chickens experimentally infected with avian influenza virus H9N2. Poult Sci 2023; 102:102669. [PMID: 37146538 DOI: 10.1016/j.psj.2023.102669] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
This study investigated the clinical, antiviral, and immunological effects of spraying Bacillus spp. and Lactobacillus spp. as a single or mixture probiotic compound on experimentally infected broiler chickens with AIV H9N2. Two hundred and forty 1-day-old broilers were randomly assigned to 6 groups as follows: Ctrl- (no challenge AIV; no spray probiotic), Ctrl+ (AIV challenged; no spray probiotic), AI+B (AIV challenged; daily spraying of probiotic Bacillus spp.), AI+L group (AIV challenged; daily spraying of probiotic Lactobacillus spp.), AIV+BL (AIV challenged; daily spraying of probiotic Bacillus spp. and Lactobacillus spp.), and G-DW (daily spraying of normal saline; no AIV challenged). The birds were reared for 35 d. On the 22nd day of age, broiler chickens were challenged by AIV H9N2. The probiotics were sprayed at 9×109 CFU/m2 daily for 35 d. Growth performance, clinical signs, virus shedding, macroscopic and microscopic lesions were evaluated at various days in all groups. Spraying with probiotics improved the body weight gain and food conversion ratio in the AI+B, AI+L, and AI+BL groups compared to the Ctrl+. The severity of clinical signs, gross lesions, pathological lesions and viral shedding in the probiotic treatment groups was lower than in the Ctrl+ group. The findings of this study suggest the daily spraying of Lactobacillus and Bacillus probiotics alone or in combination during the rearing period reduce clinical and nonclinical aspects of H9N2 virus infection; so, it can be effective as a preventive protocol for controlling the severity of AIV H9N2 infection in broilers.
Collapse
|
16
|
Kennedy K, Khaddour K, Ramnath N, Weinberg F. The Lung Microbiome in Carcinogenesis and Immunotherapy Treatment. Cancer J 2023; 29:61-69. [PMID: 36957975 DOI: 10.1097/ppo.0000000000000644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Lung cancer is the leading cause of cancer-related deaths. Over the past 10 years, significant advances in treatment modalities, including immune checkpoint inhibitor (ICI) blockade, have led to improved outcomes. Elucidating predicative biomarkers in responders and nonresponders to ICI will lead to development of therapeutic targets that could enhance ICI efficacy. Recently, the gut microbiome was identified as a predictive biomarker for ICI in patients with multiple cancer types. However, it is unclear how other host microbiomes influence tumorigenesis and response to ICI. Other groups have explored the lung microbiome as it relates to carcinogenesis and immunotherapy efficacy. In this review, we explore the role of the lung microbiome in health and disease. We also review the current state of lung microbiome research as it relates to tumorigenesis and treatments and provide potential insights into how the lung microbiome could improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Kathleen Kennedy
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Karam Khaddour
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| | | | - Frank Weinberg
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| |
Collapse
|
17
|
Harnessing Innate Immunity to Treat Mycobacterium tuberculosis Infections: Heat-Killed Caulobacter crescentus as a Novel Biotherapeutic. Cells 2023; 12:cells12040560. [PMID: 36831226 PMCID: PMC9954702 DOI: 10.3390/cells12040560] [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: 11/24/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is a serious and devastating infectious disease worldwide. Approximately a quarter of the world population harbors latent Mtb infection without pathological consequences. Exposure of immunocompetent healthy individuals with Mtb does not result in active disease in more than 90% individuals, suggesting a defining role of host immunity to prevent and/or clear early infection. However, innate immune stimulation strategies have been relatively underexplored for the treatment of tuberculosis. In this study, we used cell culture and mouse models to examine the role of a heat-killed form of a non-pathogenic microbe, Caulobacter crescentus (HKCC), in inducing innate immunity and limiting Mtb infection. We also examined the added benefits of a distinct chemo-immunotherapeutic strategy that incorporates concurrent treatments with low doses of a first-line drug isoniazid and HKCC. This therapeutic approach resulted in highly significant reductions in disseminated Mtb in the lungs, liver, and spleen of mice compared to either agent alone. Our studies demonstrate the potential of a novel innate immunotherapeutic strategy with or without antimycobacterial drugs in controlling Mtb infection in mice and open new avenues for the treatment of tuberculosis in humans.
Collapse
|
18
|
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: 5.0] [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
|
19
|
De Boeck I, Cauwenberghs E, Spacova I, Gehrmann T, Eilers T, Delanghe L, Wittouck S, Bron PA, Henkens T, Gamgami I, Simons A, Claes I, Mariën J, Ariën KK, Bakokimi D, Loens K, Jacobs K, Ieven M, Bruijning-Verhagen P, Delputte P, Coenen S, Verhoeven V, Lebeer S. Randomized, Double-Blind, Placebo-Controlled Trial of a Throat Spray with Selected Lactobacilli in COVID-19 Outpatients. Microbiol Spectr 2022; 10:e0168222. [PMID: 36154666 PMCID: PMC9604152 DOI: 10.1128/spectrum.01682-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/09/2022] [Indexed: 12/31/2022] Open
Abstract
Primary care urgently needs treatments for coronavirus disease 2019 (COVID-19) patients because current options are limited, while these patients who do not require hospitalization encompass more than 90% of the people infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we evaluated a throat spray containing three Lactobacillaceae strains with broad antiviral properties in a randomized, double-blind, placebo-controlled trial. Before the availability of vaccines, 78 eligible COVID-19 patients were randomized to verum (n = 41) and placebo (n = 37) within 96 h of a positive PCR-based SARS-CoV-2 diagnosis, and a per-protocol analysis was performed. Symptoms and severity were reported daily via an online diary. Combined nose-throat swabs and dried blood spots were collected at regular time points in the study for microbiome, viral load, and antibody analyses. The daily reported symptoms were highly variable, with no added benefit for symptom resolution in the verum group. However, based on 16S V4 amplicon sequencing, the acute symptom score (fever, diarrhea, chills, and muscle pain) was significantly negatively associated with the relative abundance of amplicon sequence variants (ASVs) that included the applied lactobacilli (P < 0.05). Furthermore, specific monitoring of these applied lactobacilli strains showed that they were detectable via quantitative PCR (qPCR) analysis in 82% of the patients in the verum group. At the end of the trial, a trend toward lower test positivity for SARS-CoV-2 was observed for the verum group (2/30; 6.7% positive) than for the placebo group (7/27; 26% positive) (P = 0.07). These data indicate that the throat spray with selected antiviral lactobacilli could have the potential to reduce nasopharyngeal viral loads and acute symptoms but should be applied earlier in the viral infection process and substantiated in larger trials. IMPORTANCE Viral respiratory tract infections result in significant health and economic burdens, as highlighted by the COVID-19 pandemic. Primary care patients represent 90% of those infected with SARS-CoV-2, yet their treatment options are limited to analgesics and antiphlogistics, and few broadly acting antiviral strategies are available. Microbiome or probiotic therapy is a promising emerging treatment option because it is based on the multifactorial action of beneficial bacteria against respiratory viral disease. In this study, an innovative topical throat spray with select beneficial lactobacilli was administered to primary COVID-19 patients. A remote study setup (reducing the burden on hospitals and general practitioners) was successfully implemented using online questionnaires and longitudinal self-sampling. Our results point toward the potential mechanisms of action associated with spray administration at the levels of viral loads and microbiome modulation in the upper respiratory tract and pave the way for future clinical applications of beneficial bacteria against viral diseases.
Collapse
Affiliation(s)
- Ilke De Boeck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Eline Cauwenberghs
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Thies Gehrmann
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Tom Eilers
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Lize Delanghe
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Stijn Wittouck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Peter A. Bron
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | - Joachim Mariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Evolutionary Ecology Group, Department of Biology, University of Antwerp, Antwerp, Belgium
| | - Kevin K. Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Diana Bakokimi
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Katherine Loens
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
- Department of Microbiology, University Hospital Antwerp, Edegem, Belgium
| | - Kevin Jacobs
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Margareta Ieven
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
- Department of Microbiology, University Hospital Antwerp, Edegem, Belgium
| | - Patricia Bruijning-Verhagen
- Julius Centre for Health Sciences and Primary Care, Department of Epidemiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Samuel Coenen
- Family Medicine and Population Health (FAMPOP), University of Antwerp, Antwerp, Belgium
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Veronique Verhoeven
- Family Medicine and Population Health (FAMPOP), University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
20
|
Anti-Influenza Virus Potential of Probiotic Strain Lactoplantibacillus plantarum YML015 Isolated from Korean Fermented Vegetable. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Lactic acid bacteria are one of the potential natural remedies used worldwide, commonly known as probiotics. Here, the aim of this research investigation was to isolate a probiotic Lactobacilli strain, YLM015, from the popular Korean fermented vegetable “Kimchi” and to evaluate its anti-viral potential against influenza virus A (IFVA) H1N1 using the MDCK cell line in vitro, and in embryonated eggs in ovo. The YML015 strain was selected from among the 1200 Lactobacilli isolates for further studies based on its potent anti-viral efficacy. YML015 was identified and characterized as Lactoplantibacillus plantarum YML015 based on the 16S rRNA gene sequencing and biochemically with an API 50 CHL Kit. In ovo assay experienced with embryonated eggs and the hemagglutination inhibition method, as well as cytopathogenic reduction assay, was performed individually to observe anti-influenza viral activity of YML015 against influenza virus A H1N1. Additionally, YML015 was classified for its non-resistance nature as safe for humans and animals as confirmed by the antibiotic susceptibility (MIC) test, cell viability, and hemolysis assay. The heat stability test was also experienced by using different heat-treated cell-free supernatant (CFS) samples of YML015. As a result, YML015 showed highly potent anti-viral activity against influenza virus A H1N1 in vitro in the MDCK cell line. Overall findings suggest that anti-influenza viral activity of L. plantarum YML015 makes it a potential candidate of choice for use as an influential probiotic in pharmacological preparations to protect humans and animals from flu and viral infection.
Collapse
|
21
|
Zeinali T, Faraji N, Joukar F, Khan Mirzaei M, Kafshdar Jalali H, Shenagari M, Mansour-Ghanaei F. Gut bacteria, bacteriophages, and probiotics: Tripartite mutualism to quench the SARS-CoV2 storm. Microb Pathog 2022; 170:105704. [PMID: 35948266 PMCID: PMC9357283 DOI: 10.1016/j.micpath.2022.105704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
Patients with SARS-CoV-2 infection, exhibit various clinical manifestations and severity including respiratory and enteric involvements. One of the main reasons for death among covid-19 patients is excessive immune responses directed toward cytokine storm with a low chance of recovery. Since the balanced gut microbiota could prepare health benefits by protecting against pathogens and regulating immune homeostasis, dysbiosis or disruption of gut microbiota could promote severe complications including autoimmune disorders; we surveyed the association between the imbalanced gut bacteria and the development of cytokine storm among COVID-19 patients, also the impact of probiotics and bacteriophages on the gut bacteria community to alleviate cytokine storm in COVID-19 patients. In present review, we will scrutinize the mechanism of immunological signaling pathways which may trigger a cytokine storm in SARS-CoV2 infections. Moreover, we are explaining in detail the possible immunological signaling pathway-directing by the gut bacterial community. Consequently, the specific manipulation of gut bacteria by using probiotics and bacteriophages for alleviation of the cytokine storm will be investigated. The tripartite mutualistic cooperation of gut bacteria, probiotics, and phages as a candidate prophylactic or therapeutic approach in SARS-CoV-2 cytokine storm episodes will be discussed at last.
Collapse
Affiliation(s)
- Tahereh Zeinali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764, Neuherberg, Germany
| | - Hossnieh Kafshdar Jalali
- Department of Microbiology, Faculty of Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Caspian Digestive Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
22
|
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: 37] [Impact Index Per Article: 18.5] [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
|
23
|
Rather IA, Kamli MR, Sabir JSM, Paray BA. Potential Antiviral Activity of Lactiplantibacillus plantarum KAU007 against Influenza Virus H1N1. Vaccines (Basel) 2022; 10:vaccines10030456. [PMID: 35335088 PMCID: PMC8954428 DOI: 10.3390/vaccines10030456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
The development of antiviral resistance has exacerbated a growing threat to public health. As a result, there is increasing demand for unconventional antivirals that can effectively replace the presently in-use drugs. Lactic acid-producing bacteria (LAB) are among the most common bacteria used in the food industry. These bacteria play an essential role in the fermentation of many foods and feed. Additionally, these bacteria are considered more economical, efficient, and safe “nutraceuticals” in the health care arsenal. Therefore, we carried out the screening and molecular characterization of raw camel milk LAB isolates and tested their inhibitory activity against influenza virus H1N1. The strain that exhibited the highest antiviral activity against the H1N1 virus, confirmed by hemagglutination assay, was identified as Lactiplantibacillus plantarum KAU007. The study also confirmed the non-cytotoxic behavior of CFCS isolated from KAU007 against MDCK cells, approving its safety concern against the mammalian cells. Besides, CFCS at 5 and 10 mg/mL significantly decreased the level of IFN-γ (p < 0.001 and p < 0.001) and IL-6 (p < 0.001 and p < 0.005) in a dose-dependent manner, respectively. This is a preliminary report about the anti-influenza activity of KAU007 isolated from camel milk. This study reinforces that camel milk contains beneficial LAB isolates with antagonistic properties against the H1N1 influenza virus.
Collapse
Affiliation(s)
- Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (M.R.K.); (J.S.M.S.)
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Correspondence:
| | - Majid Rasool Kamli
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (M.R.K.); (J.S.M.S.)
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Jamal S. M. Sabir
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (M.R.K.); (J.S.M.S.)
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Bilal Ahmad Paray
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
24
|
Shi HY, Zhu X, Li WL, Mak JWY, Wong SH, Zhu ST, Guo SL, Chan FKL, Zhang ST, Ng SC. Modulation of gut microbiota protects against viral respiratory tract infections: a systematic review of animal and clinical studies. Eur J Nutr 2021; 60:4151-4174. [PMID: 33852069 PMCID: PMC8044287 DOI: 10.1007/s00394-021-02519-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Earlier studies suggest that probiotics have protective effects in the prevention of respiratory tract infections (RTIs). Whether such benefits apply to RTIs of viral origin and mechanisms supporting the effect remain unclear. AIM To determine the role of gut microbiota modulation on clinical and laboratory outcomes of viral RTIs. METHODS We conducted a systematic review of articles published in Embase and MEDLINE through 20 April 2020 to identify studies reporting the effect of gut microbiota modulation on viral RTIs in clinical studies and animal models. The incidence of viral RTIs, clinical manifestations, viral load and immunological outcomes was evaluated. RESULTS We included 58 studies (9 randomized controlled trials; 49 animal studies). Six of eight clinical trials consisting of 726 patients showed that probiotics administration was associated with a reduced risk of viral RTIs. Most commonly used probiotics were Lactobacillus followed by Bifidobacterium and Lactococcus. In animal models, treatment with probiotics before viral challenge had beneficial effects against influenza virus infection by improving infection-induced survival (20/22 studies), mitigating symptoms (21/21 studies) and decreasing viral load (23/25 studies). Probiotics and commensal gut microbiota exerted their beneficial effects through strengthening host immunity. CONCLUSION Modulation of gut microbiota represents a promising approach against viral RTIs via host innate and adaptive immunity regulation. Further research should focus on next generation probiotics specific to viral types in prevention and treatment of emerging viral RTIs.
Collapse
Affiliation(s)
- Hai Yun Shi
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, China
| | - Xi Zhu
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Lin Li
- Department of Medicine, Division of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joyce W Y Mak
- Department of Medicine and Therapeutics, State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H Wong
- Department of Medicine and Therapeutics, State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheng Tao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, China
| | - Shui Long Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, China
| | - Francis K L Chan
- Department of Medicine and Therapeutics, State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Microbiota I-Center (MagIC) Limited, The Chinese University of Hong Kong, Hong Kong, China
| | - Shu Tian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, China
| | - Siew C Ng
- Department of Medicine and Therapeutics, State Key Laboratory for Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Microbiota I-Center (MagIC) Limited, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
25
|
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: 20] [Impact Index Per Article: 6.7] [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
|
26
|
Salva S, Kolling Y, Ivir M, Gutiérrez F, Alvarez S. The Role of Immunobiotics and Postbiotics in the Recovery of Immune Cell Populations From Respiratory Mucosa of Malnourished Hosts: Effect on the Resistance Against Respiratory Infections. Front Nutr 2021; 8:704868. [PMID: 34458307 PMCID: PMC8387655 DOI: 10.3389/fnut.2021.704868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Malnutrition is associated with a state of secondary immunodeficiency, which is characterized by a worsening of the immune response against infectious agents. Despite important advances in vaccines and antibiotic therapies, the respiratory infections are among the leading causes of increased morbidity and mortality, especially in immunosuppressed hosts. In this review, we examine the interactions between immunobiotics-postbiotics and the immune cell populations of the respiratory mucosa. In addition, we discuss how this cross talk affects the maintenance of a normal generation of immune cells, that is crucial for the establishment of protective innate and adaptive immune responses. Particular attention will be given to the alterations in the development of phagocytic cells, T and B lymphocytes in bone marrow, spleen and thymus in immunosuppression state by protein deprivation. Furthermore, we describe our research that demonstrated that the effectiveness of immunobiotics nasal administration in accelerating the recovery of the respiratory immune response in malnourished hosts. Finally, we propose the peptidoglycan from the immunobiotic Lactobacillus rhamnosus CRL1505 as the key cellular component for the effects on mucosal immunity, which are unique and cannot be extrapolated to other L. rhamnosus or probiotic strains. In this way, we provide the scientific bases for its application as a mucosal adjuvant in health plans, mainly aimed to improve the immune response of immunocompromised hosts. The search for safe vaccine adjuvants that increase their effectiveness at the mucosal level is a problem of great scientific relevance today.
Collapse
Affiliation(s)
- Susana Salva
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucuman, Argentina
| | - Yanina Kolling
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucuman, Argentina
| | - Maximiliano Ivir
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucuman, Argentina
| | - Florencia Gutiérrez
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucuman, Argentina
| | - Susana Alvarez
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucuman, Argentina.,Clinical Biochemistry I, Institute of Applied Biochemistry, National University of Tucuman, San Miguel de Tucuman, Argentina
| |
Collapse
|
27
|
Effects of Heat-Killed Levilactobacillus brevis KB290 in Combination with β-Carotene on Influenza Virus Infection in Healthy Adults: A Randomized Controlled Trial. Nutrients 2021; 13:nu13093039. [PMID: 34578917 PMCID: PMC8467669 DOI: 10.3390/nu13093039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 01/05/2023] Open
Abstract
Influenza, a seasonal acute respiratory disease caused primarily by the influenza virus A or B, manifests with severe symptoms leading to considerable morbidity and mortality and is a major concern worldwide. Therefore, effective preventive measures against it are required. The aim of this trial was to evaluate the preventive effects of heat-killed Levilactobacillus brevis KB290 (KB290) in combination with β-carotene (βC) on influenza virus infections in healthy Japanese subjects aged between 20 and 59 y throughout the winter season. We performed a randomized, double-blind, placebo-controlled, parallel-group trial from 16 December 2019 to 8 March 2020, comparing KB290 + βC beverage with placebo beverage. The primary endpoint was the incidence of influenza based on a doctor’s certificate. The incidence of influenza was not significantly different between the two groups. However, the subgroup analysis showed a significant difference between the two groups (influenza incidence: the KB290 + βC group 1.9%, and the placebo group 3.9%) in the subgroup of subjects aged ˂40 y, but not in the subgroup of subjects aged ≥40 y. The results of this trial suggest that the combination of KB290 and βC might be a possible candidate supplement for protection against the seasonal influenza virus infection in humans aged <40 y, although further clinical studies are needed to confirm the concrete preventive effect of this combination on influenza.
Collapse
|
28
|
Physical properties of lactic acid bacteria influence the level of protection against influenza infection in mice. PLoS One 2021; 16:e0251784. [PMID: 34003877 PMCID: PMC8130949 DOI: 10.1371/journal.pone.0251784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/03/2021] [Indexed: 11/20/2022] Open
Abstract
We evaluated whether the water dispersibility of lactic acid bacteria (Enterococcus faecalis KH2) affects their efficacy. When cultured lactic acid bacteria are washed, heat-killed, and powdered, adhesion occurs between results in aggregation (non-treated lactic acid bacteria, n-LAB). However, dispersed lactic acid bacteria (d-LAB) with a lower number of aggregates can be prepared by treating them with a high-pressure homogenizer and adding an excipient during powdering. Mice were administered n-LAB or d-LAB Peyer’s patches in the small intestine were observed. Following n-LAB administration, a high amount of aggregated bacteria drifting in the intestinal mucosa was observed; meanwhile, d-LAB reached the Peyer’s patches and was absorbed into them. Evaluation in a mouse influenza virus infection model showed that d-LAB was more effective than n-LAB in the influenza yield of bronchoalveolar lavage fluids on day 3 post-infection and neutralizing antibody titers of sera and influenza virus-specific immunoglobulin A in the feces on day 14 post-infection. Therefore, the physical properties of lactic acid bacteria affect their efficacy; controlling their water dispersibility can improve their effectiveness.
Collapse
|
29
|
Spacova I, De Boeck I, Bron PA, Delputte P, Lebeer S. Topical Microbial Therapeutics against Respiratory Viral Infections. Trends Mol Med 2021; 27:538-553. [PMID: 33879402 DOI: 10.1016/j.molmed.2021.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/08/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Emerging evidence suggests that microbial therapeutics can prevent and treat respiratory viral diseases, especially when applied directly to the airways. This review presents established beneficial effects of locally administered microbial therapeutics against respiratory viral diseases and the inferred related molecular mechanisms. Several mechanisms established in the intestinal probiotics field as well as novel, niche-specific insights are relevant in the airways. Studies at cellular and organism levels highlight biologically plausible but strain-specific and host and virus context-dependent mechanisms, underlying the potential of beneficial bacteria. Large-scale clinical studies can now be rationally designed to provide a bench-to-bedside translation of the multifactorial bacterial mechanisms within the host respiratory tract, to diminish the incidence and severity of viral infections and the concomitant complications.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Ilke De Boeck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Peter A Bron
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, B-2610, Belgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium. @uantwerpen.be
| |
Collapse
|
30
|
A meta-analysis reveals the effectiveness of probiotics and prebiotics against respiratory viral infection. Biosci Rep 2021; 41:227885. [PMID: 33604601 PMCID: PMC7955103 DOI: 10.1042/bsr20203638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/28/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Experimental experience suggests that microbial agents including probiotics and prebiotics (representative microbial agents) play a critical role in defending against respiratory virus infection. We aim to systematically examine these agents' effect on respiratory viral infection and encourage research into clinical applications. An electronic literature search was conducted from published data with a combination of a microbial agents search component containing synonyms for microbial agents-related terms and a customized search component for respiratory virus infection. Hazard ratio (HR), risk ratio (RR) and standard deviation (SD) were employed as effect estimates. In 45 preclinical studies, the mortality rates decreased in the respiratory viral infection models that included prebiotics or prebiotics as interventions (HR: 0.70; 95% confidence interval (CI): 0.56-0.87; P=0.002). There was a significant decrease in viral load due to improved gut microbiota (SD: -1.22; 95% CI: -1.50 to -0.94; P<0.001). Concentrations of interferon (IFN)-α (SD: 1.05; 95% CI: 0.33-1.77; P=0.004), IFN-γ (SD: 0.83; 95% CI: 0.01-1.65; P=0.05) and interleukin (IL)-12 (SD: 2.42; 95% CI: 0.32-4.52; P=0.02), IL-1β (SD: 0.01; 95% CI: -0.37 to 0.40; P=0.94) increased, whereas those of TNF-α (SD: -0.58; 95% CI: -1.59 to 0.43; P=0.26) and IL-6 (SD: -0.59; 95% CI: -1.24 to 0.07; P=0.08) decreased. Six clinical studies had lower symptom scores (SD: -0.09; 95% CI: -0.44 to 0.26; P=0.61) and less incidence of infection (RR: 0.80; 95% CI: 0.64-1.01; P=0.06). Our research indicates that probiotics and prebiotics pose a defensive possibility on respiratory viral infection and may encourage the clinical application.
Collapse
|
31
|
Singh K, Rao A. Probiotics: A potential immunomodulator in COVID-19 infection management. Nutr Res 2021; 87:1-12. [PMID: 33592454 PMCID: PMC7881295 DOI: 10.1016/j.nutres.2020.12.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/02/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 caused by SARS-CoV-2 is an ongoing global pandemic. SARS-CoV-2 affects the human respiratory tract's epithelial cells, leading to a proinflammatory cytokine storm and chronic lung inflammation. With numerous patients dying daily, a vaccine and specific antiviral drug regimens are being explored. Probiotics are live microorganisms with proven beneficial effects on human health. While probiotics as nutritional supplements are long practiced in different cuisines across various countries, the emerging scientific evidence supports the antiviral and general immune-strengthening health effects of the probiotics. Here, we present an overview of the experimental studies published in the last 10 years that provide a scientific basis for unexplored probiotics as a preventive approach to respiratory viral infections. Based on collated insights from these experimental data, we identify promising microbial strains that may serve as lead prophylactic and immune-boosting probiotics in COVID-19 management.
Collapse
Affiliation(s)
- Kuljit Singh
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh 160036 India
| | - Alka Rao
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh 160036 India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India.
| |
Collapse
|
32
|
Watanabe T, Hayashi K, Kan T, Ohwaki M, Kawahara T. Anti-Influenza virus effects of Enterococcus faecalis KH2 and Lactobacillus plantarum SNK12 RNA. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2021; 40:43-49. [PMID: 33520568 PMCID: PMC7817512 DOI: 10.12938/bmfh.2020-019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/28/2020] [Indexed: 01/22/2023]
Abstract
Bacterial RNA has recently emerged as an immune-stimulating factor during viral
infection. The immune response in an organism is directly related to the progression of
virus infections. Lactic acid bacteria in particular have anticancer, bioprotective, and
antiallergic effects by modulating immunity. Here, we aimed to demonstrate the effect of
bacterial RNA on in vitro production of IL-12, a proinflammatory
cytokine, and on in vivo activity against influenza A virus (IFV)
infection. Oral administration of heat-killed Enterococcus faecalis KH2
(KH2) or Lactobacillus plantarum SNK12 (SNK) in IFV-infected mice
suppressed viral replication and stimulated production of virus-specific antibodies.
However, ribonuclease-treated KH2 or SNK abrogated the effect, reducing IL-12 production
in vitro and anti-IFV effects in vivo. Taken together,
KH2 or SNK showed antiviral effects in vivo when administered orally, and
the RNAs of KH2 and SNK play a part in these effects, despite the phylogenetic differences
between the bacteria.
Collapse
Affiliation(s)
- Takumi Watanabe
- Graduate School of Engineering, Chubu University, 1200 Matsumoto, Kasugai, Aichi 487-8501, Japan.,Bio-Lab Co., Ltd., 2-1-3 Komagawa, Hidaka, Saitama 350-1249, Japan
| | - Kyoko Hayashi
- Graduate School of Engineering, Chubu University, 1200 Matsumoto, Kasugai, Aichi 487-8501, Japan
| | - Tatsuhiko Kan
- Bio-Lab Co., Ltd., 2-1-3 Komagawa, Hidaka, Saitama 350-1249, Japan
| | - Makoto Ohwaki
- Non-Profit Organisation, The Japanese Association of Clinical Research on Supplements, 1-9-24 Shihogi, Hidaka, Saitama 350-1248, Japan
| | - Toshio Kawahara
- College of Life and Health Sciences, Chubu University, 1200 Matsumoto, Kasugai, Aichi 487-8501, Japan
| |
Collapse
|
33
|
Li L, Fang Z, Liu Z, Zhao J, Zhang H, Wang S, He J, Lu W, Chen W. Lactobacillus reuteri CCFM1072 and CCFM1040 with the role of Treg cells regulation alleviate airway inflammation through modulating gut microbiota in allergic asthma mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
34
|
Hsue VB, Itamura K, Wu AW, Illing EA, Sokoloski KJ, Weaver BA, Anthony BP, Hughes N, Ting JY, Higgins TS. Topical Oral and Intranasal Antiviral Agents for Coronavirus Disease 2019 (COVID-19). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1327:169-189. [PMID: 34279838 DOI: 10.1007/978-3-030-71697-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
With the largest viral loads in both symptomatic and asymptomatic patients with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) present in the oral and nasal cavities, agents that act on these two areas have the potential for large therapeutic and prophylactic benefit. A literature review was conducted to elucidate the possible agents useful in treatment of SARS-CoV-2. These agents were evaluated for their current applications, adverse reactions, their current state of study, and any future considerations in their management of coronavirus disease 2019 (COVID-2019). Our review has found that, while there are many promising agents with proven efficacy in their in-vitro efficacy against SARS-CoV-2, more clinical trials and in-vivo studies, as well as safety trials, must be conducted before these agents can be effectively implemented.
Collapse
Affiliation(s)
- Victor B Hsue
- Department of Otolaryngology-Head and Neck Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Kyohei Itamura
- Department of Otolaryngology-Head and Neck Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Arthur W Wu
- Department of Otolaryngology-Head and Neck Surgery, Cedars Sinai, Los Angeles, CA, USA
| | - Elisa A Illing
- Department of Otolaryngology-Head and Neck Surgery, Indiana University, Indianapolis, IN, USA
| | - Kevin J Sokoloski
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Bree A Weaver
- Division of Infectious Diseases, Departments of Internal Medicine and Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benjamin P Anthony
- Department of Otolaryngology-Head and Neck Surgery, Indiana University, Indianapolis, IN, USA
| | - Nathan Hughes
- Pharmacy Operations, Kindred Healthcare Support Center, Louisville, KY, USA
| | - Jonathan Y Ting
- Department of Otolaryngology-Head and Neck Surgery, Indiana University, Indianapolis, IN, USA
| | - Thomas S Higgins
- Department of Otolaryngology-Head and Neck Surgery and Communicative Disorders, University of Louisville, Louisville, KY, USA. .,Rhinology, Sinus & Skull Base, Kentuckiana Ear, Nose, and Throat, Louisville, KY, USA.
| |
Collapse
|
35
|
The Lung Microbiome: A Central Mediator of Host Inflammation and Metabolism in Lung Cancer Patients? Cancers (Basel) 2020; 13:cancers13010013. [PMID: 33375062 PMCID: PMC7792810 DOI: 10.3390/cancers13010013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Lung cancer is the major cause of cancer related deaths in the world. New therapies have improved outcomes. Unfortunately, overall 5 year survival is ~20%. Therefore, better understanding of tumor biology and the microenvironment may lead to new therapeutic targets. The lung microbiome has recently emerged as a major mediator of host inflammation and pathogenesis. Understanding how the lung microbiota exerts its effects on lung cancer and the tumor microenvironment will allow for novel development of therapies. Abstract Lung cancer is the leading cause of cancer-related death. Over the past 5–10 years lung cancer outcomes have significantly improved in part due to better treatment options including immunotherapy and molecularly targeted agents. Unfortunately, the majority of lung cancer patients do not enjoy durable responses to these new treatments. Seminal research demonstrated the importance of the gut microbiome in dictating responses to immunotherapy in melanoma patients. However, little is known regarding how other sites of microbiota in the human body affect tumorigenesis and treatment responses. The lungs were traditionally thought to be a sterile environment; however, recent research demonstrated that the lung contains its own dynamic microbiota that can influence disease and pathophysiology. Few studies have explored the role of the lung microbiome in lung cancer biology. In this review article, we discuss the links between the lung microbiota and cancer, with particular focus on immune responses, metabolism and strategies to target the lung microbiome for cancer prevention.
Collapse
|
36
|
Probiotics in Treatment of Viral Respiratory Infections and Neuroinflammatory Disorders. Molecules 2020; 25:molecules25214891. [PMID: 33105830 PMCID: PMC7660077 DOI: 10.3390/molecules25214891] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a biological response to the activation of the immune system by various infectious or non-infectious agents, which may lead to tissue damage and various diseases. Gut commensal bacteria maintain a symbiotic relationship with the host and display a critical function in the homeostasis of the host immune system. Disturbance to the gut microbiota leads to immune dysfunction both locally and at distant sites, which causes inflammatory conditions not only in the intestine but also in the other organs such as lungs and brain, and may induce a disease state. Probiotics are well known to reinforce immunity and counteract inflammation by restoring symbiosis within the gut microbiota. As a result, probiotics protect against various diseases, including respiratory infections and neuroinflammatory disorders. A growing body of research supports the beneficial role of probiotics in lung and mental health through modulating the gut-lung and gut-brain axes. In the current paper, we discuss the potential role of probiotics in the treatment of viral respiratory infections, including the COVID-19 disease, as major public health crisis in 2020, and influenza virus infection, as well as treatment of neurological disorders like multiple sclerosis and other mental illnesses.
Collapse
|
37
|
The potential application of probiotics and prebiotics for the prevention and treatment of COVID-19. NPJ Sci Food 2020; 4:17. [PMID: 33083549 PMCID: PMC7536434 DOI: 10.1038/s41538-020-00078-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
COVID-19 is a pandemic disease caused by the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This new viral infection was first identified in China in December 2019, and it has subsequently spread globally. The lack of a vaccine or curative treatment for COVID-19 necessitates a focus on other strategies to prevent and treat the infection. Probiotics consist of single or mixed cultures of live microorganisms that can beneficially affect the host by maintaining the intestinal or lung microbiota that play a major role in human health. At present, good scientific evidence exists to support the ability of probiotics to boost human immunity, thereby preventing colonization by pathogens and reducing the incidence and severity of infections. Herein, we present clinical studies of the use of probiotic supplementation to prevent or treat respiratory tract infections. These data lead to promising benefits of probiotics in reducing the risk of COVID-19. Further studies should be conducted to assess the ability of probiotics to combat COVID-19.
Collapse
|
38
|
Satomi S, Khanum S, Miller P, Suzuki S, Suganuma H, Heiser A, Gupta SK. Short Communication: Oral Administration of Heat-killed Lactobacillus brevis KB290 in Combination with Retinoic Acid Provides Protection against Influenza Virus Infection in Mice. Nutrients 2020; 12:nu12102925. [PMID: 32987850 PMCID: PMC7600661 DOI: 10.3390/nu12102925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
Influenza virus type A (IAV) is a seasonal acute respiratory disease virus with severe symptoms, and an effective preventive measure is required. Despite many reports describing the potentially protective effects of lactic acid bacteria, few studies have investigated the effects of nutritional supplement combinations. This study reports the effect of the combined intake of heat-killed Lactobacillus brevis KB290 (KB290) and vitamin A (VA) on mice challenged with a sublethal dose of IAV. For 2 weeks, five groups of mice were fed either placebo, KB290, VA, or a combination of KB290 and VA (KB290+VA). After subsequent IAV challenge, bodyweight and general health were monitored for up to 2 weeks. Viral titres were determined in the lungs of animal subgroups euthanised at days 3, 7, and 14 after IAV challenge. A significant loss was observed in the bodyweights of IAV-infected animals from day 1 post-IAV challenge, whereas the mice fed KB290+VA did not lose any weight after IAV infection, indicating successful protection from the infection. Additionally, mice in the KB290+VA group showed the highest reduction in lung viral titres. In conclusion, the combination of KB290 and VA could be a useful food supplement relevant for protection against seasonal influenza virus infection in humans.
Collapse
Affiliation(s)
- Shohei Satomi
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
- Correspondence: (S.S.); (S.K.G.); Tel.: +81-80-8132-3813 (S.S.); +64-06351-8697 (S.K.G.)
| | - Sofia Khanum
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Poppy Miller
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Shigenori Suzuki
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
| | - Hiroyuki Suganuma
- Department of Nature & Wellness Research, Innovation Division, KAGOME CO., LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (S.S.); (H.S.)
| | - Axel Heiser
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
| | - Sandeep K Gupta
- AgResearch Ltd., Hopkirk Research Institute, Grasslands Research Centre, Private Bag 11008, Palmerston North 4442, New Zealand; (S.K.); (P.M.); (A.H.)
- Correspondence: (S.S.); (S.K.G.); Tel.: +81-80-8132-3813 (S.S.); +64-06351-8697 (S.K.G.)
| |
Collapse
|
39
|
Lira-Lucio JA, Falfán-Valencia R, Ramírez-Venegas A, Buendía-Roldán I, Rojas-Serrano J, Mejía M, Pérez-Rubio G. Lung Microbiome Participation in Local Immune Response Regulation in Respiratory Diseases. Microorganisms 2020; 8:E1059. [PMID: 32708647 PMCID: PMC7409050 DOI: 10.3390/microorganisms8071059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
The lung microbiome composition has critical implications in the regulation of innate and adaptive immune responses. Next-generation sequencing techniques have revolutionized the understanding of pulmonary physiology and pathology. Currently, it is clear that the lung is not a sterile place; therefore, the investigation of the participation of the pulmonary microbiome in the presentation, severity, and prognosis of multiple pathologies, such as asthma, chronic obstructive pulmonary disease, and interstitial lung diseases, contributes to a better understanding of the pathophysiology. Dysregulation of microbiota components in the microbiome-host interaction is associated with multiple lung pathologies, severity, and prognosis, making microbiome study a useful tool for the identification of potential therapeutic strategies. This review integrates the findings regarding the activation and regulation of the innate and adaptive immune response pathways according to the microbiome, including microbial patterns that could be characteristic of certain diseases. Further studies are required to verify whether the microbial profile and its metabolites can be used as biomarkers of disease progression or poor prognosis and to identify new therapeutic targets that restore lung dysbiosis safely and effectively.
Collapse
Affiliation(s)
- Juan Alberto Lira-Lucio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| | - Alejandra Ramírez-Venegas
- Tobacco Smoking and COPD Research Department, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Jorge Rojas-Serrano
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-S.); (M.M.)
| | - Mayra Mejía
- Interstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.R.-S.); (M.M.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (J.A.L.-L.); (R.F.-V.)
| |
Collapse
|
40
|
Stachowska E, Folwarski M, Jamioł-Milc D, Maciejewska D, Skonieczna-Żydecka K. Nutritional Support in Coronavirus 2019 Disease. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E289. [PMID: 32545556 PMCID: PMC7353890 DOI: 10.3390/medicina56060289] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
The epidemic that broke out in Chinese Wuhan at the beginning of 2020 presented how important the rapid diagnosis of malnutrition (elevating during intensive care unit stay) and the immediate implementation of caloric and protein-balanced nutrition care are. According to specialists from the Chinese Medical Association for Parenteral and Enteral Nutrition (CSPEN), these activities are crucial for both the therapy success and reduction of mortality rates. The Chinese have published their recommendations including principles for the diagnosis of nutritional status along with the optimal method for nutrition supply including guidelines when to introduce education approach, oral nutritional supplement, tube feeding, and parenteral nutrition. They also calculated energy demand and gave their opinion on proper monitoring and supplementation of immuno-nutrients, fluids and macronutrients intake. The present review summarizes Chinese observations and compares these with the latest European Society for Clinical Nutrition and Metabolism guidelines. Nutritional approach should be an inseparable element of therapy in patients with COVID-19.
Collapse
Affiliation(s)
- Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (D.J.-M.); (D.M.); (K.S.-Ż.)
| | - Marcin Folwarski
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-210 Gdańsk, Poland;
- Home Enteral and Parenteral Nutrition Unit, Nicolaus Copernicus Hospital, 80-803 Gdańsk, Poland
| | - Dominika Jamioł-Milc
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (D.J.-M.); (D.M.); (K.S.-Ż.)
| | - Dominika Maciejewska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (D.J.-M.); (D.M.); (K.S.-Ż.)
| | - Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (D.J.-M.); (D.M.); (K.S.-Ż.)
| |
Collapse
|
41
|
Ren C, Faas MM, de Vos P. Disease managing capacities and mechanisms of host effects of lactic acid bacteria. Crit Rev Food Sci Nutr 2020; 61:1365-1393. [PMID: 32366110 DOI: 10.1080/10408398.2020.1758625] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Consumption of lactic acid bacteria (LAB) has been suggested to confer health-promoting effects on the host. However, effects of LABs have been reported to be species- and strain-specific and the mechanisms involved are subjects of discussion. Here, the possible mechanisms by which LABs induce antipathogenic, gut barrier enhancing and immune modulating effects in consumers are reviewed. Specific strains for which it has been proven that health is improved by these mechanisms are discussed. However, most strains probably act via several or combinations of mechanisms depending on which effector molecules they express. Current insight is that these effector molecules are either present on the cell wall of LAB or are excreted. These molecules are reviewed as well as the ligand binding receptors in the host. Also postbiotics are discussed. Finally, we provide an overview of the efficacy of LABs in combating infections caused by Helicobacter pylori, Salmonella, Escherichia coli, Streptococcus pneumoniae, and influenza virus, in controlling gut inflammatory diseases, in managing allergic disorders, and in alleviating cancer.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
42
|
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: 25] [Impact Index Per Article: 6.3] [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
|
43
|
Le Noci V, Guglielmetti S, Arioli S, Camisaschi C, Bianchi F, Sommariva M, Storti C, Triulzi T, Castelli C, Balsari A, Tagliabue E, Sfondrini L. Modulation of Pulmonary Microbiota by Antibiotic or Probiotic Aerosol Therapy: A Strategy to Promote Immunosurveillance against Lung Metastases. Cell Rep 2019; 24:3528-3538. [PMID: 30257213 DOI: 10.1016/j.celrep.2018.08.090] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/04/2018] [Accepted: 08/29/2018] [Indexed: 12/15/2022] Open
Abstract
Pulmonary immunological tolerance to inhaled particulates might create a permissive milieu for lung metastasis. Lung microbiota contribute to pulmonary tolerance; here, we explored whether its manipulation via antibiotic or probiotic aerosolization favors immune response against melanoma metastasis. In lungs of vancomycin/neomycin-aerosolized mice, a decrease in bacterial load was associated with reduced regulatory T cells and enhanced T cell and NK cell activation that paralleled a significant reduction of melanoma B16 lung metastases. Reduction of metastases also occurred in lungs transplanted with bacterial isolates from antibiotic-treated lungs. Aerosolized Lactobacillus rhamnosus strongly promoted immunity against B16 lung metastases as well. Furthermore, probiotics or antibiotics improved chemotherapy activity against advanced B16 metastases. Thus, we identify a role for lung microbiota in metastasis and show that its targeting via aerosolization is a therapy that can prevent metastases and enhance responses to chemotherapy.
Collapse
Affiliation(s)
- Valentino Le Noci
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Simone Guglielmetti
- Dipartimento di Scienze degli Alimenti, Nutrizione e Ambiente (DeFENS), Università degli Studi di Milano, Milan 20133, Italy
| | - Stefania Arioli
- Dipartimento di Scienze degli Alimenti, Nutrizione e Ambiente (DeFENS), Università degli Studi di Milano, Milan 20133, Italy
| | - Chiara Camisaschi
- Immunotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Francesca Bianchi
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy; Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| | - Chiara Storti
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Chiara Castelli
- Immunotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Andrea Balsari
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy; Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy.
| | - Elda Tagliabue
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|
44
|
Yang D, Xing Y, Song X, Qian Y. The impact of lung microbiota dysbiosis on inflammation. Immunology 2019; 159:156-166. [PMID: 31631335 DOI: 10.1111/imm.13139] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Host-microbiota interaction plays fundamental roles in the homeostasis of mucosal immunity. Dysbiosis of intestinal microbiota has been demonstrated to participate in various immune responses and many multifactorial diseases. Study of intestinal microbiota has moved beyond the consequences of dysbiosis to the causal microbiota associated with diseases. However, studies of pulmonary microbiota and its dysbiosis are still in their infancy. Improvement of culture-dependent and -independent techniques has facilitated our understanding of lung microbiota that not only exists in healthy lung tissue but also exerts great impact on immune responses under both physiological and pathological conditions. In this review, we summarize recent progresses of lung microbiota dysbiosis and its impact on the local immune system that determines the balance of tolerance and inflammation. We discuss the causal roles of pulmonary dysbiosis under disease settings, and propose that the interaction between lung microbiota and host is critical for establishing the immune homeostasis in lung.
Collapse
Affiliation(s)
- Daping Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yingying Xing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xinyang Song
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Youcun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
45
|
Dhakal S, Klein SL. Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019; 93:e00797-19. [PMID: 31391269 PMCID: PMC6803252 DOI: 10.1128/jvi.00797-19] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
Affiliation(s)
- Santosh Dhakal
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019. [PMID: 31391269 DOI: 10.1128/jvi.00797‐19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
|
47
|
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: 5.6] [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
|
48
|
Prevention of respiratory syncytial virus infection with probiotic lactic acid bacterium Lactobacillus gasseri SBT2055. Sci Rep 2019; 9:4812. [PMID: 30886158 PMCID: PMC6423325 DOI: 10.1038/s41598-019-39602-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/19/2018] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus gasseri SBT2055 (LG2055) is a probiotic lactic acid bacterium with multifunctional effects, including the prevention of influenza A virus infection in mice, reduction of adipocyte size in mice, and increased lifespan in C. elegans. We investigated whether LG2055 exhibits antiviral activity against respiratory syncytial virus (RSV), a global pathogen for which a preventive strategy is required. Following oral administration of LG2055 in mice, the RSV titre in the lung was significantly decreased, while body weight was not decreased after virus infection. Additionally, the elevated expression of pro-inflammatory cytokines in the lung upon RSV infection decreased after LG2055 administration. Moreover, interferon and interferon stimulated genes were upregulated by LG2055 treatment. Comparative cellular proteomic analysis revealed that SWI2/SNF2-related CREB-binding protein activator protein (SRCAP) was a candidate for the antiviral activity of LG2055 against RSV. There was a positive correlation between the inhibition of RSV replication and the suppression of SRCAP expression and RSV replication was suppressed by SRCAP silencing. Since SRCAP is a scaffold protein to which viral non-structural proteins bind, the downregulation of SRCAP induced by LG2055 could provide new insights about the inhibition of RSV replication. In summary, our study demonstrated that LG2055 has prophylactic potential against RSV infection.
Collapse
|
49
|
Kara SS, Volkan B, Erten I. Lactobacillus rhamnosus GG can protect malnourished children. Benef Microbes 2019; 10:237-244. [PMID: 30638398 DOI: 10.3920/bm2018.0071] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Malnutrition affects virtually all organ systems, and malnourished children are more prone to infections. These children have dysbiosis, but probiotics can restore the disrupted gut microbiome. We investigated the protective effects of Lactobacillus rhamnosus GG in malnourished children in terms of incidence of infection, and anthropometric and metabolic parameters. 50 intervention and 50 control patients, aged 6 months to 5 years, with body weight and height below -2 SD, were randomly and prospectively recruited. The controls received a calorie and protein-appropriate diet for 3 months, while the study group additionally received approximately 109 L. rhamnosus GG for 3 months. Infection episodes and nutritional status were compared between the groups. 38 intervention, 33 control patients completed the study and the two groups were similar at baseline. The study group had fewer upper respiratory tract infections and gastroenteritis episodes at each month and at the end of the study. Children in the study group experienced fewer total upper respiratory infections and urinary tract infections. Hospitalisation was more frequent in the control group during the third month and at the end of the study. Total infection numbers were higher in the control group at each month and at the end of the study (P<0.001 for each). Increments in body mass index (BMI) and BMI Z-scores were more pronounced in the study group (P=0.008 and P=0.02, respectively). Daily prophylactic use of L. rhamnosus GG at 109 bacteria in malnourished children prevents most infections and improves nutritional status when used together with appropriate diet.
Collapse
Affiliation(s)
- S S Kara
- 1 Department of Pediatric Infectious Diseases, Regional Training and Research Hospital, 25280 Erzurum, Turkey
| | - B Volkan
- 2 Department of Pediatric Gastroenterology, Regional Training and Research Hospital, 25280 Erzurum, Turkey
| | - I Erten
- 3 Department of Pediatrics, Regional Training and Research Hospital, 25280 Erzurum, Turkey
| |
Collapse
|
50
|
Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O. The role of the lung microbiota and the gut-lung axis in respiratory infectious diseases. Cell Microbiol 2018; 20:e12966. [PMID: 30329198 DOI: 10.1111/cmi.12966] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/05/2018] [Accepted: 10/14/2018] [Indexed: 12/13/2022]
Abstract
The pulmonary microbial community, described only a few years ago, forms a discreet part of the human host microbiota. The airway microbiota has been found to be substantially altered in the context of numerous respiratory disorders; nonetheless, its role in health and disease is as yet only poorly understood. Another important parameter to consider is the gut-lung axis, where distal (gut) immune modulation during respiratory disease is mediated by the gut microbiota. The use of specific microbiota strains, termed "probiotics," with beneficial effects on the host immunity and/or against pathogens, has proven successful in the treatment of intestinal disorders and is also showing promise in the context of airway diseases. In this review, we highlight the beneficial role of the body's commensal bacteria during airway infectious diseases, including recent evidence highlighting their local (lung) or distal (gut) contribution in this process.
Collapse
Affiliation(s)
- Alexia Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lucie Bernard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yannick Poquet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|