1
|
Ye F, Li L, Wang J, Yang H. Advances in gut-lung axis research: clinical perspectives on pneumonia prevention and treatment. Front Immunol 2025; 16:1576141. [PMID: 40330490 PMCID: PMC12052896 DOI: 10.3389/fimmu.2025.1576141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
In recent years, the study of the interaction between gut microbiota and distant organs such as the heart, lungs, brain, and liver has become a hot topic in the field of gut microbiology. With a deeper understanding of its immune regulation and mechanisms of action, these findings have increasingly highlighted their guiding value in clinical practice. The gut is not only the largest digestive organ in the human body but also the habitat for most microorganisms. Imbalances in gut microbial communities have been associated with various lung diseases, such as allergic asthma and cystic fibrosis. Furthermore, gut microbial communities have significant impacts on metabolic function and immune responses. Their metabolites not only regulate gastrointestinal immune systems but may also affect distant organs such as the lungs and brain. As one of the most common types of respiratory system diseases worldwide, pulmonary infections have high morbidity and mortality rates. Pulmonary infections caused by immune dysfunction can lead to gastrointestinal problems like diarrhea, further resulting in imbalances within complex interactions that are associated with abnormal manifestations under disequilibrium conditions. Meanwhile, clinical interventions can significantly modulate the composition of gut microbiota, and alteration in gut microbiota may subsequently indicate susceptibility to pulmonary infections and even contribute to the prevention or regulation of their progression. This review delves into the interaction between gut microbiota and pulmonary infections, elucidating the latest advancements in gut-lung axis research and providing a fresh perspective for the treatment and prevention of pneumonia.
Collapse
Affiliation(s)
| | | | | | - Hongfeng Yang
- Department of Critical Care Medicine, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
2
|
Zhong H, Yu Y, Abdullah, Zhang H, Du J, Sun J, Chen L, Feng F, Guan R. Lactiplantibacillus plantarum N1 derived lipoteichoic acid alleviates insulin resistance in association with modulation of the gut microbiota and amino acid metabolism. Food Funct 2025; 16:1371-1388. [PMID: 39877991 DOI: 10.1039/d4fo06100d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
This study aimed to investigate the effects of heat-killed Lactiplantibacillus plantarum N1 (HK-N1) and lipoteichoic acid (LTA) derived from it on alleviating insulin resistance by modulating the gut microbiota and amino acid metabolism. High-fat diet (HFD)-fed mice were administered live bacteria or HK-N1, and the results demonstrated that HK-N1 significantly reduced epididymal adipocyte size and serum low density lipoprotein-cholesterol, and improved insulin resistance by increasing the YY peptide and glucagon-like peptide levels. HK-N1 also modulated the gut microbiome composition, enhancing microbiota uniformity and reducing the abundance of Ruminococcus, Oscillospira and norank_f_Mogibacteriaceae. Three main active substances obtained from HK-N1 (membrane protein, peptidoglycan, and lipoteichoic acid) were also used to investigate their potential effects in hyperglycemic zebrafish. Only LTA reduced blood sugar and altered the gut microbiome, particularly reducing Aeromonas, which is positively related to hyperglycemia. Untargeted metabolomics revealed that LTA improved vitamin and amino acid metabolism, thereby alleviating metabolic disorders in zebrafish. Collectively, our findings indicate that HK-N1, primarily through LTA, modulated insulin sensitivity by regulating the gut microbiota and amino acid metabolism, offering a potential therapeutic strategy for insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Yufen Yu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Haoxuan Zhang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Juan Du
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
- Hangzhou Kangyuan Food Science & Technology Co., Ltd, Hangzhou 310012, China
| | - Jiangwei Sun
- Sanya Branch of Hainan Academy of Inspection and Testing, Shanghai 201700, China
| | - Ling Chen
- Sanya Branch of Hainan Food and Drug Inspection Institute, San Ya, 572011, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
3
|
Bano Y, Shrivastava A, Shukla P, Chaudhary AA, Khan SUD, Khan S. The implication of microbiome in lungs cancer: mechanisms and strategies of cancer growth, diagnosis and therapy. Crit Rev Microbiol 2025; 51:128-152. [PMID: 38556797 DOI: 10.1080/1040841x.2024.2324864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 04/02/2024]
Abstract
Available evidence illustrates that microbiome is a promising target for the study of growth, diagnosis and therapy of various types of cancer. Lung cancer is a leading cause of cancer death worldwide. The relationship of microbiota and their products with diverse pathologic conditions has been getting large attention. The novel research suggests that the microbiome plays an important role in the growth and progression of lung cancer. The lung microbiome plays a crucial role in maintaining mucosal immunity and synchronizing the stability between tolerance and inflammation. Alteration in microbiome is identified as a critical player in the progression of lung cancer and negatively impacts the patient. Studies suggest that healthy microbiome is essential for effective therapy. Various clinical trials and research are focusing on enhancing the treatment efficacy by altering the microbiome. The regulation of microbiota will provide innovative and promising treatment strategies for the maintenance of host homeostasis and the prevention of lung cancer in lung cancer patients. In the current review article, we presented the latest progress about the involvement of microbiome in the growth and diagnosis of lung cancer. Furthermore, we also assessed the therapeutic status of the microbiome for the management and treatment of lung cancer.
Collapse
Affiliation(s)
- Yasmin Bano
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
| | - Abhinav Shrivastava
- Department of Biotechnology, College of Life Sciences, Cancer Hospital and research Institute, Gwalior, India
| | - Piyush Shukla
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, India
- Laboratory of Natural Products, Department of Rural Technology and Social Development, Guru Ghasidas University, Bilaspur, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health Technology (IIHT), Deoband, Saharanpur, UP, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, Australia
| |
Collapse
|
4
|
Zhang M, Zhan M, Song X. Echinacoside attenuates Klebsiella pneumoniae-induced pneumonia via inhibition of the TLR4/NF-κB signaling. APMIS 2025; 133:e13507. [PMID: 39757690 DOI: 10.1111/apm.13507] [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: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025]
Abstract
The Gram-negative bacterium Klebsiella pneumoniae (K. pneumoniae) is one major causative agent of community- and hospital-acquired pneumonia. Echinacoside (ECH) is a phenylethanoid glycoside isolated from Cistanche deserticola that possesses anti-inflammatory activity. Our research aimed to confirm whether ECH alleviates K. pneumoniae-induced pneumonia and explore the underlying regulatory mechanisms. BEAS-2B cells and BALB/c mice were infected by K. pneumoniae to establish the cellular and animal models, respectively, followed by ECH treatment. Inflammatory cytokine levels were detected by RT-qPCR and ELISA. The lung wet/dry (W/D) weight ratio and the myeloperoxidase (MPO) activity in lung tissues were examined. The pulmonary histopathologic changes were observed through hematoxylin and eosin (H&E) staining. The levels of TLR4/NF-κB pathway-associated molecules were estimated through western blotting, immunohistochemical, and immunohistochemical staining. K. pneumoniae infection caused lung histopathologic damage, enhanced MPO activity, elevated lung W/D weight ratio, and upregulated inflammatory cytokine levels in mice and promoted inflammatory cytokine expression in BEAS-2B cells, which were reversed by ECH treatment. K. pneumoniae infection-induced upregulation in TLR4, phosphorylated (p)-p65, and p-IκBα levels, and downregulation in IκBα levels in BEAS-2B cells and pneumonia mice were overturned by ECH treatment. ECH ameliorates K. pneumoniae-induced pneumonia through suppressing the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| | - Ming Zhan
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| | - Xinyu Song
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
5
|
He G, Long H, He J, Zhu C. The Immunomodulatory Effects and Applications of Probiotic Lactiplantibacillus plantarum in Vaccine Development. Probiotics Antimicrob Proteins 2024; 16:2229-2250. [PMID: 39101975 DOI: 10.1007/s12602-024-10338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Lactiplantibacillus plantarum (previously known as Lactobacillus plantarum) is a lactic acid bacterium that exists in various niches. L. plantarum is a food-grade microorganism that is commonly considered a safe and beneficial microorganism. It is widely used in food fermentation, agricultural enhancement, and environmental protection. L. plantarum is also part of the normal flora that can regulate the intestinal microflora and promote intestinal health. Some strains of L. plantarum are powerful probiotics that induce and modulate the innate and adaptive immune responses. Due to its outstanding immunoregulatory capacities, an increasing number of studies have examined the use of probiotic L. plantarum strains as natural immune adjuvants or alternative live vaccine carriers. The present review summarizes the main immunomodulatory characteristics of L. plantarum and discusses the preliminary immunological effects of L. plantarum as a vaccine adjuvant and delivery carrier. Different methods for improving the immune capacities of recombinant vector vaccines are also discussed.
Collapse
Affiliation(s)
- Guiting He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Huanbing Long
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Jiarong He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China
| | - Cuiming Zhu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
Rühle J, Schwarz J, Dietz S, Rückle X, Schoppmeier U, Lajqi T, Poets CF, Gille C, Köstlin-Gille N. Impact of perinatal administration of probiotics on immune cell composition in neonatal mice. Pediatr Res 2024; 96:1645-1654. [PMID: 38278847 PMCID: PMC11772233 DOI: 10.1038/s41390-024-03029-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Newborns and especially preterm infants are much more susceptible to infections than adults. The pathogens causing infections in newborns are often detectable in the intestinal flora of affected children even before disease onset. Therefore, it seems reasonable to prevent dysbiosis in newborns and preterm infants. An approach followed in many neonatal intensive care units (NICUs) is to prevent infections in preterm infants with probiotics however their mechanisms of action of probiotics are incompletely understood. Here, we investigated the effect of perinatal probiotic exposure on immune cells in newborn mice. METHODS Pregnant mice were orally treated with a combination of Lactobacillus acidophilus and Bifidobacterium bifidum (Infloran®) from mid-pregnancy until the offspring were harvested. Immune cell composition in organs of the offspring were analyzed by flow cytometry. RESULTS Perinatal probiotic exposure had profound effects on immune cell composition in the intestine, liver and lungs of newborn mice with reduction of myeloid and B cells and induction of T cells in the probiotic treated animals' organs at weaning. Furthermore, probiotic exposure had an effect on T cell development in the thymus. CONCLUSION Our results contribute to a better understanding of the interaction of probiotics with the developing immune system. IMPACT probiotics have profound effects on immune cell composition in intestines, livers and lungs of newborn mice. probiotics modulate T cell development in thymus of newborn mice. effects of probiotics on neonatal immune cells are particularly relevant in transition phases of the microbiome. our results contribute to a better understanding of the mechanisms of action of probiotics in newborns.
Collapse
Affiliation(s)
- Jessica Rühle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Xenia Rückle
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Ulrich Schoppmeier
- Institute for Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tuebingen University Children's Hospital, Tuebingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| |
Collapse
|
7
|
Zhang N, Zheng Y, Wei Y, Wang L, Chen X, Li J. Yak DEFB123 alleviates lung injury caused by Klebsiella pneumoniae through MAPKs signaling pathway. Vet Microbiol 2024; 298:110248. [PMID: 39265281 DOI: 10.1016/j.vetmic.2024.110248] [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: 04/01/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Beta-defensins, such as β-defensin 123 (DEFB123), are vital components of the immune system's defense against infections due to their strong antimicrobial properties and capacity for modulating the body's immunological responses. In this study, we successfully cloned and analyzed the yak DEFB123 gene sequence. Subsequently, we obtained recombinant protein DEFB123 (rDEFB123) through prokaryotic expression. Our results demonstrate that rDEFB123 effectively inhibits the growth of Escherichia coli, Klebsiella pneumoniae, and Staphylococcus aureus. Furthermore, rDEFB123 enhances the phagocytic activity of macrophages by regulating specific factors. In a mouse model infected with Klebsiella pneumoniae, the administration of rDEFB123 showed significantly lower levels of serum ALT and AST compared to the control group. Moreover, IFN-γ and IgG were significantly increased in the rDEFB123-treated groups, indicating an enhanced immune response. In the MAPKs signaling pathway of the infected mouse lungs, the expressions of JNK, TRAF2, TRAF6, MIF, and IL-1β genes were downregulated in the rDEFB123-treated groups. Moreover, the levels of p-JNK protein were significantly decreased in these groups as well. Klebsiella pneumoniae caused systemic infection with organ damage in mice. However, the administration of rDEFB123 suppressed the expressions of inflammatory factors, thereby mitigating organ injury and regulating the activity of apoptosis-related factors to enhance immunity. These findings provide valuable theoretical data for future exploration of the functionality and potential applications of DEFB123 in yak.
Collapse
Affiliation(s)
- Nanchi Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu 610041, China
| | - Yao Zheng
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu 610041, China
| | - Yong Wei
- Animal Genetics and Breeding Key Laboratory of Sichuan Province, Sichuan Animal Sciences Academy, Chengdu 610041, China
| | - Li Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu 610041, China.
| | - Xiwen Chen
- Animal Disease Prevention and Control and Healthy Breeding Engineering Technology Research Center, Mianyang Normal University, Mianyang 621000, China.
| | - Juan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
8
|
Long H, He G, He J, Du TF, Feng P, Zhu C. The protective effect and immunomodulatory ability of orally administrated Lacticaseibacillus rhamnosus GG against Mycoplasma pneumoniae infection in BALB/c mice. PLoS One 2024; 19:e0312318. [PMID: 39453930 PMCID: PMC11508164 DOI: 10.1371/journal.pone.0312318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/27/2024] [Indexed: 10/27/2024] Open
Abstract
Mycoplasma pneumoniae represents one of the significant etiologies of community-acquired pneumonia in pediatric patients. However, clinical treatment of M. pneumoniae infection in children has encountered challenges due to the escalating resistance to quinolones. Numerous studies have highlighted the potential of probiotic lactobacillus administration in boosting immune responses to bacterial and viral respiratory infections. In this study, the protective efficacy of pre-oral administration of Lacticaseibacillus rhamnosus GG (LGG), Limosilactobacillus reuteri F275, Lactiplantibacillus plantarum NCIMB 8826, L. plantarum S1 or L. plantarum S2 was evaluated in the BALB/c mice model; it was observed that among these five strains of lactobacillus, the supplementation of LGG exhibited the most significant protective effect against M. pneumoniae infection. Moreover, when administered orally, both live LGG and heat-inactivated LGG have demonstrated efficacy in reducing the burden of M. pneumoniae in the lungs and alleviating pulmonary inflammation. Oral supplementation with LGG resulted in the inhibition of neutrophil recruitment into the lungs and increased recruitment of alveolar macrophages in M. pneumoniae-infected mice. Additionally, LGG supplementation led to increased production of IL-10 and secretory IgA (sIgA), while suppressing the levels of IL-1β, IL-6, IL-17A, and TNF-α in the lungs of mice infected with M. pneumoniae. The data suggests that supplementation with LGG can modulate immune responses, decrease pathogen load, and alleviate inflammatory injury in the lungs of M. pneumoniae-infected mice.
Collapse
Affiliation(s)
- Huanbing Long
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Guiting He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jiarong He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Ting feng Du
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Pengxiao Feng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Cuiming Zhu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
9
|
Raboso B, Pou C, Abril R, Erro M, Sánchez C, Manzano C, Zamarrón E, Suarez-Cuartin G, González J. Bronchiectasis. OPEN RESPIRATORY ARCHIVES 2024; 6:100339. [PMID: 39026515 PMCID: PMC11255363 DOI: 10.1016/j.opresp.2024.100339] [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: 02/27/2024] [Accepted: 05/02/2024] [Indexed: 07/20/2024] Open
Abstract
Non-cystic fibrosis bronchiectasis, a condition that remains relatively underrecognized, has garnered increasing research focus in recent years. This scientific interest has catalyzed advancements in diagnostic methodologies, enabling comprehensive clinical and molecular profiling. Such progress facilitates the development of personalized treatment strategies, marking a significant step toward precision medicine for these patients. Bronchiectasis poses significant diagnostic challenges in both clinical settings and research studies. While computed tomography (CT) remains the gold standard for diagnosis, novel alternatives are emerging. These include artificial intelligence-powered algorithms, ultra-low dose chest CT, and magnetic resonance imaging (MRI) techniques, all of which are becoming recognized as feasible diagnostic tools. The precision medicine paradigm calls for refined characterization of bronchiectasis patients by analyzing their inflammatory and molecular profiles. Research into the underlying mechanisms of inflammation and the evaluation of biomarkers such as neutrophil elastase, mucins, and antimicrobial peptides have led to the identification of distinct patient endotypes. These endotypes present variable clinical outcomes, necessitating tailored therapeutic interventions. Among these, eosinophilic bronchiectasis is notable for its prevalence and specific prognostic factors, calling for careful consideration of treatable traits. A deeper understanding of the microbiome's influence on the pathogenesis and progression of bronchiectasis has inspired a holistic approach, which considers the multibiome as an interconnected microbial network rather than treating pathogens as solitary entities. Interactome analysis therefore becomes a vital tool for pinpointing alterations during both stable phases and exacerbations. This array of innovative approaches has revolutionized the personalization of treatments, incorporating therapies such as inhaled mannitol or ARINA-1, brensocatib for anti-inflammatory purposes, and inhaled corticosteroids specifically for patients with eosinophilic bronchiectasis.
Collapse
Affiliation(s)
| | | | - Rosa Abril
- University Hospital Complex Insular-Materno Infantil (CHUIMI) of Gran Canaria, Gran Canaria, Spain
| | - Marta Erro
- Puerta del Hierro University Hospital, Madrid, Spain
| | | | - Carlos Manzano
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
| | | | - Guillermo Suarez-Cuartin
- Hospital Universitari Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Jessica González
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Kim H, Oh S, Song S. Lactobacillus Persisters Formation and Resuscitation. J Microbiol Biotechnol 2024; 34:854-862. [PMID: 38326923 DOI: 10.4014/jmb.2312.12035] [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: 12/27/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/09/2024]
Abstract
Lactobacillus is a commonly used probiotic, and many researchers have focused on its stress response to improve its functionality and survival. However, studies on persister cells, dormant cells that aid bacteria in surviving general stress, have focused on pathogenic bacteria that cause infection, not Lactobacillus. Thus, understanding Lactobacillus persister cells will provide essential clues for understanding how Lactobacillus survives and maintains its function under various environmental conditions. We treated Lactobacillus strains with various antibiotics to determine the conditions required for persister formation using kill curves and transmission electron microscopy. In addition, we observed the resuscitation patterns of persister cells using single-cell analysis. Our results show that Lactobacillus creates a small population of persister cells (0.0001-1% of the bacterial population) in response to beta-lactam antibiotics such as ampicillin and amoxicillin. Moreover, only around 0.5-1% of persister cells are heterogeneously resuscitated by adding fresh media; the characteristics are typical of persister cells. This study provides a method for forming and verifying the persistence of Lactobacillus and demonstrates that antibiotic-induced Lactobacillus persister cells show characteristics of dormancy, sensitivity of antibiotics, same as exponential cells, multi-drug tolerance, and resuscitation, which are characteristics of general persister cells. This study suggests that the mechanisms of formation and resuscitation may vary depending on the characteristics, such as the membrane structure of the bacterial species.
Collapse
Affiliation(s)
- Hyein Kim
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Sejong Oh
- Division of Animal Science, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| | - Sooyeon Song
- Department of Animal Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
11
|
Luo L, Tang J, Du X, Li N. Chronic obstructive pulmonary disease and the airway microbiome: A review for clinicians. Respir Med 2024; 225:107586. [PMID: 38460708 DOI: 10.1016/j.rmed.2024.107586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/30/2023] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex heterogeneous disease characterized by progressive airflow limitation and chronic inflammation. The progressive development and long-term repeated acute exacerbation of COPD make many patients still unable to control the deterioration of the disease after active treatment, and even eventually lead to death. An increasing number of studies have shown that the occurrence and development of COPD are closely related to the composition and changes of airway microbiome. This article reviews the interaction between COPD and airway microbiome, the potential mechanisms of interaction, and the treatment methods related to microbiome. We elaborated the internal correlation between airway microbiome and different stages of COPD, inflammatory endotypes, glucocorticoid and antibiotic treatment, analyze the pathophysiological mechanisms such as the "vicious cycle" hypothesis, abnormal inflammation-immune response of the host and the "natural selection" of COPD to airway microbiome, introduce the treatment of COPD related to microbiome and emphasize the predictive value of airway microbiome for the progression, exacerbation and prognosis of COPD, as well as the guiding role for clinical management of patients, in order to provide a new perspective for exploring the pathogenesis of COPD, and also provide clues and guidance for finding new treatment targets.
Collapse
Affiliation(s)
- Lingxin Luo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Junli Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xianzhi Du
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Na Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
12
|
Shen X, Yang Z, Wang Q, Chen X, Zhu Q, Liu Z, Patel N, Liu X, Mo X. Lactobacillus plantarum L168 improves hyperoxia-induced pulmonary inflammation and hypoalveolarization in a rat model of bronchopulmonary dysplasia. NPJ Biofilms Microbiomes 2024; 10:32. [PMID: 38553470 PMCID: PMC10980738 DOI: 10.1038/s41522-024-00504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
Alteration of gut microbiota can affect chronic lung diseases, such as asthma and chronic obstructive pulmonary disease, through abnormal immune and inflammatory responses. Previous studies have shown a feasible connection between gut microbiota and bronchopulmonary dysplasia (BPD) in preterm infants. However, whether BPD can be ameliorated by restoring the gut microbiota remains unclear. In preterm infants with BPD, we found variance in the diversity and structure of gut microbiota. Similarly, BPD rats showed gut dysbiosis, characterized by a deficiency of Lactobacillus, which was abundant in normal rats. We therefore explored the effect and potential mechanism of action of a probiotic strain, Lactobacillus plantarum L168, in improving BPD. The BPD rats were treated with L. plantarum L168 by gavage for 2 weeks, and the effect was evaluated by lung histopathology, lung function, and serum inflammatory markers. Subsequently, we observed reduced lung injury and improved lung development in BPD rats exposed to L. plantarum L168. Further evaluation revealed that L. plantarum L168 improved intestinal permeability in BPD rats. Serum metabolomics showed altered inflammation-associated metabolites following L. plantarum L168 intervention, notably a marked increase in anti-inflammatory metabolites. In agreement with the metabolites analysis, RNA-seq analysis of the intestine and lung showed that inflammation and immune-related genes were down-regulated. Based on the information from RNA-seq, we validated that L. plantarum L168 might improve BPD relating to down-regulation of TLR4 /NF-κB /CCL4 pathway. Together, our findings suggest the potential of L. plantarum L168 to provide probiotic-based therapeutic strategies for BPD.
Collapse
Affiliation(s)
- Xian Shen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qihui Zhu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Zhi Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China.
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Mina SA, Zhu G, Fanian M, Chen S, Yang G. Exploring reduced macrophage cell toxicity of hypervirulent Klebsiella pneumoniae compared to classical Klebsiella pneumoniae. Microbiol Res 2024; 278:127515. [PMID: 37837829 DOI: 10.1016/j.micres.2023.127515] [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: 06/17/2023] [Revised: 09/21/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
Bacterial pneumoniae caused by Klebsiella pneumoniae (Kp) is a major concern due to the prevalence of multiple antibiotic-resistant strains, which limit treatment options and increase mortality rates. Patients with Kp infections often experience an uncontrolled immune response in the lungs, leading to excessive inflammation and elevated levels of proinflammatory cytokines. This study aimed to investigate the cytotoxicity, the inflammatory cytokine response, and the longevity of intracellular bacterial load in RAW 264.7 macrophages, infected with two different Kp strains - cKP (HKU1: Classical Kp) and HvKP (17ZR101: Hypervirulent Kp). This study found that after infecting macrophages with cKP and HvKP, the internalization rate was faster and the intracellular cKP load was higher than that of HvKP. Additionally, the number of intracellular Kp was correlated with the presence of M1 macrophage polarization marker CD86 and expressions of proinflammatory cytokines. Interestingly, the expression of these proinflammatory cytokines was significantly higher in cKP-infected macrophages than in HvKP-infected macrophages. Thus, a higher intracellular cKP load is suggested to play a significant role in causing more proinflammatory cytokines and killing macrophages compared to HvKP infection. This finding highlights the importance of understanding the mechanisms behind Kp infections and developing effective treatment strategies.
Collapse
Affiliation(s)
- Sohana Akter Mina
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Gaochen Zhu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Maryam Fanian
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hom Hung, Kowloon, Hong Kong Special Administrative Region of China.
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China.
| |
Collapse
|
14
|
Agbemavor WSK, Buys EM. Dynamic Interactions between Diarrhoeagenic Enteroaggregative Escherichia coli and Presumptive Probiotic Bacteria: Implications for Gastrointestinal Health. Microorganisms 2023; 11:2942. [PMID: 38138086 PMCID: PMC10745617 DOI: 10.3390/microorganisms11122942] [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: 10/29/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
This study delves into the temporal dynamics of bacterial interactions in the gastrointestinal tract, focusing on how probiotic strains and pathogenic bacteria influence each other and human health. This research explores adhesion, competitive exclusion, displacement, and inhibition of selected diarrhoeagenic Escherichia coli (D-EAEC) and potential probiotic strains under various conditions. Key findings reveal that adhesion is time-dependent, with both D-EAEC K2 and probiotic L. plantarum FS2 showing increased adhesion over time. Surprisingly, L. plantarum FS2 outperformed D-EAEC K2 in adhesion and exhibited competitive exclusion and displacement, with inhibition of adhesion surpassing competitive exclusion. This highlights probiotics' potential to slow pathogen attachment when not in competition. Pre-infecting with L. plantarum FS2 before pathogenic infection effectively inhibited adhesion, indicating probiotics' ability to prevent pathogen attachment. Additionally, adhesion correlated strongly with interleukin-8 (IL-8) secretion, linking it to the host's inflammatory response. Conversely, IL-8 secretion negatively correlated with trans-epithelial electrical resistance (TEER), suggesting a connection between tight junction disruption and increased inflammation. These insights offer valuable knowledge about the temporal dynamics of gut bacteria interactions and highlight probiotics' potential in competitive exclusion and inhibiting pathogenic bacteria, contributing to strategies for maintaining gastrointestinal health and preventing infections.
Collapse
Affiliation(s)
- Wisdom Selorm Kofi Agbemavor
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Pretoria 0028, South Africa
- Radiation Technology Centre, Biotechnology and Nuclear Agriculture Research Institute, Ghana Atomic Energy Commission, Legon, Accra P.O. Box LG 80, Ghana
| | - Elna Maria Buys
- Department of Consumer and Food Sciences, University of Pretoria, Private Bag X20, Pretoria 0028, South Africa
| |
Collapse
|
15
|
Khataybeh B, Jaradat Z, Ababneh Q. Anti-bacterial, anti-biofilm and anti-quorum sensing activities of honey: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116830. [PMID: 37400003 DOI: 10.1016/j.jep.2023.116830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Man has used honey to treat diseases since ancient times, perhaps even before the history of medicine itself. Several civilizations have utilized natural honey as a functional and therapeutic food to ward off infections. Recently, researchers worldwide have been focusing on the antibacterial effects of natural honey against antibiotic-resistant bacteria. AIM OF THE STUDY This review aims to summarize research on the use of honey properties and constituents with their anti-bacterial, anti-biofilm, and anti-quorum sensing mechanisms of action. Further, honey's bacterial products, including probiotic organisms and antibacterial agents which are produced to curb the growth of other competitor microorganisms is addressed. MATERIALS AND METHODS In this review, we have provided a comprehensive overview of the antibacterial, anti-biofilm, and anti-quorum sensing activities of honey and their mechanisms of action. Furthermore, the review addressed the effects of antibacterial agents of honey from bacterial origin. Relevant information on the antibacterial activity of honey was obtained from scientific online databases such as Web of Science, Google Scholar, ScienceDirect, and PubMed. RESULTS Honey's antibacterial, anti-biofilm, and anti-quorum sensing activities are mostly attributed to four key components: hydrogen peroxide, methylglyoxal, bee defensin-1, and phenolic compounds. The performance of bacteria can be altered by honey components, which impact their cell cycle and cell morphology. To the best of our knowledge, this is the first review that specifically summarizes every phenolic compound identified in honey along with their potential antibacterial mechanisms of action. Furthermore, certain strains of beneficial lactic acid bacteria such as Bifidobacterium, Fructobacillus, and Lactobacillaceae, as well as Bacillus species can survive and even grow in honey, making it a potential delivery system for these agents. CONCLUSION Honey could be regarded as one of the best complementary and alternative medicines. The data presented in this review will enhance our knowledge of some of honey's therapeutic properties as well as its antibacterial activities.
Collapse
Affiliation(s)
- Batool Khataybeh
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ziad Jaradat
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Qutaiba Ababneh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
16
|
Chen D, Zeng Q, Liu L, Zhou Z, Qi W, Yu S, Zhao L. Global Research Trends on the Link Between the Microbiome and COPD: A Bibliometric Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:765-783. [PMID: 37180751 PMCID: PMC10167978 DOI: 10.2147/copd.s405310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
Background The pathogenesis of chronic obstructive pulmonary disease (COPD) has been studied in relation to the microbiome, providing space for more targeted interventions and new treatments. Numerous papers on the COPD microbiome have been reported in the last 10 years, yet few publications have used bibliometric methods to evaluate this area. Methods We searched the Web of Science Core Collection for all original research articles in the field of COPD microbiome from January 2011 to August 2022 and used CiteSpace for visual analysis. Results A total of 505 relevant publications were obtained, and the number of global publications in this field is steadily increasing every year, with China and the USA occupying the first two spots in international publications. Imperial College London and the University of Leicester produced the most publications. Brightling C from the UK was the most prolific writer, while Huang Y and Sze M from the USA were first and second among the authors cited. The American Journal of Respiratory and Critical Care Medicine had the highest frequency of citations. The top 10 institutions, cited authors and journals are mostly from the UK and the US. In the ranking of citations, the first article was a paper published by Sze M on changes in the lung tissue's microbiota in COPD patients. The keywords "exacerbation", "gut microbiota", "lung microbiome", "airway microbiome", "bacterial colonization", and "inflammation" were identified as cutting-edge research projects for 2011-2022. Conclusion Based on the visualization results, in the future, we can use the gut-lung axis as the starting point to explore the immunoinflammatory mechanism of COPD, and study how to predict the effects of different treatments of COPD by identifying the microbiota, and how to achieve the optimal enrichment of beneficial bacteria and the optimal consumption of harmful bacteria to improve COPD.
Collapse
Affiliation(s)
- Daohong Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qian Zeng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Lu Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ziyang Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenchuan Qi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Shuguang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
17
|
Dentice Maidana S, Imamura Y, Elean M, Albarracín L, Nishiyama K, Suda Y, Kurata S, Jure MÁ, Kitazawa H, Villena J. Oral Administration of Lacticaseibacillus rhamnosus CRL1505 Modulates Lung Innate Immune Response against Klebsiella pneumoniae ST25. Microorganisms 2023; 11:1148. [PMID: 37317122 DOI: 10.3390/microorganisms11051148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
Orally administered Lacticaseibacillus rhamnosus CRL1505 enhances respiratory immunity, providing protection against respiratory viruses and Streptococcus pneumoniae. However, the capacity of the CRL1505 strain to improve respiratory immunity against Gram-negative bacterial infections has not been evaluated before. The aim of this work was to evaluate whether the Lcb. rhamnosus CRL1505 was able to beneficially regulate the respiratory innate immune response and enhance the resistance to hypermucoviscous KPC-2-producing Klebsiella pneumoniae of the sequence type 25 (ST25). BALB/c mice were treated with the CRL1505 strain via the oral route and then nasally challenged with K. pneumoniae ST25 strains LABACER 01 or LABACER 27. Bacterial cell counts, lung injuries and the respiratory and systemic innate immune responses were evaluated after the bacterial infection. The results showed that K. pneumoniae ST25 strains increased the levels of TNF-α, IL-1β, IL-6, IFN-γ, IL-17, KC and MPC-1 in the respiratory tract and blood, as well as the numbers of BAL neutrophils and macrophages. Mice treated with Lcb. rhamnosus CRL1505 had significantly lower K. pneumoniae counts in their lungs, as well as reduced levels of inflammatory cells, cytokines and chemokines in the respiratory tract and blood when compared to infected controls. Furthermore, higher levels of the regulatory cytokines IL-10 and IL-27 were found in the respiratory tract and blood of CRL1505-treated mice than controls. These results suggest that the ability of Lcb. rhamnosus CRL1505 to help with the control of detrimental inflammation in lungs during K. pneumoniae infection would be a key feature to improve the resistance to this pathogen. Although further mechanistic studies are necessary, Lcb. rhamnosus CRL1505 can be proposed as a candidate to improve patients' protection against hypermucoviscous KPC-2-producing strains belonging to the ST25, which is endemic in the hospitals of our region.
Collapse
Affiliation(s)
- Stefania Dentice Maidana
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
- Laboratory of Antimicrobials, Institute of Microbiology "Luis C. Verna", Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, Tucuman 4000, Argentina
| | - Yoshiya Imamura
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
| | - Leonardo Albarracín
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
| | - Keita Nishiyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan
| | - Shoichiro Kurata
- Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - María Ángela Jure
- Laboratory of Antimicrobials, Institute of Microbiology "Luis C. Verna", Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, Tucuman 4000, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| |
Collapse
|
18
|
Narayana JK, Aliberti S, Mac Aogáin M, Jaggi TK, Ali NABM, Ivan FX, Cheng HS, Yip YS, Vos MIG, Low ZS, Lee JXT, Amati F, Gramegna A, Wong SH, Sung JJY, Tan NS, Tsaneva-Atanasova K, Blasi F, Chotirmall SH. Microbial Dysregulation of the Gut-Lung Axis in Bronchiectasis. Am J Respir Crit Care Med 2023; 207:908-920. [PMID: 36288294 PMCID: PMC10111978 DOI: 10.1164/rccm.202205-0893oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Emerging data support the existence of a microbial "gut-lung" axis that remains unexplored in bronchiectasis. Methods: Prospective and concurrent sampling of gut (stool) and lung (sputum) was performed in a cohort of n = 57 individuals with bronchiectasis and subjected to bacteriome (16S rRNA) and mycobiome (18S Internal Transcribed Spacer) sequencing (total, 228 microbiomes). Shotgun metagenomics was performed in a subset (n = 15; 30 microbiomes). Data from gut and lung compartments were integrated by weighted similarity network fusion, clustered, and subjected to co-occurrence analysis to evaluate gut-lung networks. Murine experiments were undertaken to validate specific Pseudomonas-driven gut-lung interactions. Results: Microbial communities in stable bronchiectasis demonstrate a significant gut-lung interaction. Multibiome integration followed by unsupervised clustering reveals two patient clusters, differing by gut-lung interactions and with contrasting clinical phenotypes. A high gut-lung interaction cluster, characterized by lung Pseudomonas, gut Bacteroides, and gut Saccharomyces, is associated with increased exacerbations and greater radiological and overall bronchiectasis severity, whereas the low gut-lung interaction cluster demonstrates an overrepresentation of lung commensals, including Prevotella, Fusobacterium, and Porphyromonas with gut Candida. The lung Pseudomonas-gut Bacteroides relationship, observed in the high gut-lung interaction bronchiectasis cluster, was validated in a murine model of lung Pseudomonas aeruginosa infection. This interaction was abrogated after antibiotic (imipenem) pretreatment in mice confirming the relevance and therapeutic potential of targeting the gut microbiome to influence the gut-lung axis. Metagenomics in a subset of individuals with bronchiectasis corroborated our findings from targeted analyses. Conclusions: A dysregulated gut-lung axis, driven by lung Pseudomonas, associates with poorer clinical outcomes in bronchiectasis.
Collapse
Affiliation(s)
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Micheál Mac Aogáin
- Biochemical Genetics Laboratory, Department of Biochemistry, St. James’s Hospital, Dublin, Ireland
- Clinical Biochemistry Unit, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | | | | | - Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andrea Gramegna
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sunny H. Wong
- Lee Kong Chian School of Medicine and
- Department of Gastroenterology and
| | - Joseph J. Y. Sung
- Lee Kong Chian School of Medicine and
- Department of Gastroenterology and
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine and
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics and
- Living Systems Institute, University of Exeter, Exeter, United Kingdom
| | - Francesco Blasi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine and
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore; and
| |
Collapse
|
19
|
Han B, Zhang X, Wang L, Yuan W. Dysbiosis of Gut Microbiota Contributes to Uremic Cardiomyopathy via Induction of IFNγ-Producing CD4 + T Cells Expansion. Microbiol Spectr 2023; 11:e0310122. [PMID: 36788674 PMCID: PMC9927280 DOI: 10.1128/spectrum.03101-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Uremic cardiomyopathy (UCM) correlates with chronic kidney disease (CKD)-induced morbidity and mortality. Gut microbiota has been involved in the pathogenesis of certain cardiovascular disease, but the role of gut microbiota in the pathogenesis of UCM remains unknown. Here, we performed a case-control study to compare the gut microbiota of patients with CKD and healthy controls by 16S rRNA (rRNA) gene sequencing. To test the causative relationship between gut microbiota and UCM, we performed fecal microbiota transplantation (FMT) in 5/6th nephrectomy model of CKD. We found that opportunistic pathogens, particularly Klebsiella pneumoniae (K. pneumoniae), are markedly enriched in patients with CKD. FMT from CKD patients aggravated diastolic dysfunction in the mouse model. The diastolic dysfunction was associated with microbiome-dependent increases in heart-infiltrating IFNγ+ CD4+ T cells. Monocolonization with K. pneumoniae increased cardiac IFNγ+ CD4+ T cells infiltration and promoted UCM development of the mouse model. A probiotic Bifidobacterium animalis decreased the relative abundance of K. pneumoniae, reduced levels of cardiac IFNγ+ CD4+ T cells and ameliorated the severity of diastolic dysfunction in the mice. Thus, the aberrant gut microbiota in CKD patients, especially K. pneumoniae, contributed to UCM pathogenesis through the induction of heart-infiltrating IFNγ+ CD4+ T cells expansion, proposing that a Gut Microbiota-Gut-Kidney-Heart axis could play a critical role in elucidating the etiology of UCM, and suggesting that modulation of the gut bacteria may serve as a promising target for the amelioration of UCM. IMPORTANCE Uremic cardiomyopathy (UCM) correlates tightly with increased mortality in patients with chronic kidney disease (CKD), yet the pathogenesis of UCM remains incompletely understood, limiting therapeutic approaches. Our study proposed that a Gut Microbiota-Gut-Kidney-Heart axis could play a critical role in understanding etiology of UCM. There is a major need in future clinical trials of patients with CKD to explore if modulation of gut microbiota by fecal microbiota transplantation (FMT), probiotics or antibiotics can alleviate cardiac dysfunction, reduce mortality, and improve life quality.
Collapse
Affiliation(s)
- Bin Han
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoqian Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
MiR-181a-5p Delivered by Adipose-Derived Mesenchymal Stem Cell Exosomes Alleviates Klebsiella pneumonia Infection-Induced Lung Injury by Targeting STAT3 Signaling. Mediators Inflamm 2022; 2022:5188895. [PMID: 36570020 PMCID: PMC9771653 DOI: 10.1155/2022/5188895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/16/2022] [Accepted: 01/27/2022] [Indexed: 12/23/2022] Open
Abstract
Background Klebsiella pneumoniae (K. pneu) is a leading cause of gram-negative pneumonia, which requires effective treatment. Adipose-derived mesenchymal stem cell- (ADSC-) derived exosomal microRNAs (miRNAs) have presented the inhibitory effect of multiple diseases. However, the function of ADSC-derived exosomal miRNAs in K. pneu remains unclear. Aim In this study, we aimed to explore the effect of ADSC-derived exosomal miR-181-5p on K. pneu infection-induced lung injury. Methods C57BL/6 mouse model was established by infection of K. pneu. ADSCs and exosomes were extracted and characterized in vitro. The translocation of ADSC-derived exosomes to bone marrow-derived macrophages (BMDMs) was detected. The level of miR-181a-5p was detected by real-time PCR. The secretion of inflammatory factors was determined by ELISA. The interaction between miR-181a-5p with STAT3 was identified. Results We successfully isolated the ADSCs that express positive markers CD90 and CD105 rather than CD31 and CD45. The exosomal miR-181a-5p secreted by ADSCs were internalized by BMDM and K. pneu infection stimulated the miR-181a-5p level in bronchoalveolar lavage fluid (BALF) and BMDM. ADSC-derived exosomal miR-181a-5p repressed pulmonary outgrowth and dissemination of K. pneu infection in mice, repressed cellular infiltration in lung tissue, and attenuated the inflammasome activity and the levels of IL-1β and IL-18 in the lung. Mechanically, miR-181a-5p was able to inhibit STAT3 expression at posttranscriptional levels and repressed Nlrp3 and Asc expression in BMDM. Conclusion Consequently, we concluded that ADSC-derived exosomal miR-181a-5p alleviated Klebsiella pneumonia infection-induced lung injury by targeting STAT3 signaling. ADSC-derived exosomal miR-181a-5p may serve as a potential candidate for the treatment of Klebsiella pneumonia infection-induced lung injury.
Collapse
|
21
|
Adapen C, Réot L, Menu E. Role of the human vaginal microbiota in the regulation of inflammation and sexually transmitted infection acquisition: Contribution of the non-human primate model to a better understanding? FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:992176. [PMID: 36560972 PMCID: PMC9763629 DOI: 10.3389/frph.2022.992176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
The human vaginal microbiota has a central role in the regulation of the female reproductive tract (FRT) inflammation. Indeed, on one hand an optimal environment leading to a protection against sexually transmitted infections (STI) is associated with a high proportion of Lactobacillus spp. (eubiosis). On the other hand, a more diverse microbiota with a high amount of non-Lactobacillus spp. (dysbiosis) is linked to a higher local inflammation and an increased STI susceptibility. The composition of the vaginal microbiota is influenced by numerous factors that may lead to a dysbiotic environment. In this review, we first discuss how the vaginal microbiota composition affects the local inflammation with a focus on the cytokine profiles, the immune cell recruitment/phenotype and a large part devoted on the interactions between the vaginal microbiota and the neutrophils. Secondly, we analyze the interplay between STI and the vaginal microbiota and describe several mechanisms of action of the vaginal microbiota. Finally, the input of the NHP model in research focusing on the FRT health including vaginal microbiota or STI acquisition/control and treatment is discussed.
Collapse
Affiliation(s)
- Cindy Adapen
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Louis Réot
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Université Paris-Saclay, Inserm, Commissariat à l'énergie Atomique et aux énergies Alternatives (CEA), Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB)/Department of Infectious Disease Models and Innovative Therapies (IDMIT), Fontenay-aux-Roses, France
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Paris, France
| |
Collapse
|
22
|
Lacticaseibacillus rhamnosus attenuates acute lung inflammation in a murine model of acute respiratory distress syndrome: Relevance to cytokines associated to STAT4/T-bet and STAT3/RORɣt”. Microb Pathog 2022; 173:105831. [DOI: 10.1016/j.micpath.2022.105831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
|
23
|
Saha UB, Saroj SD. Lactic acid bacteria: prominent player in the fight against human pathogens. Expert Rev Anti Infect Ther 2022; 20:1435-1453. [PMID: 36154442 DOI: 10.1080/14787210.2022.2128765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The human microbiome is a unique repository of diverse bacteria. Over 1000 microbial species reside in the human gut, which predominantly influences the host's internal environment and plays a significant role in host health. Lactic acid bacteria have long been employed for multiple purposes, ranging from food to medicines. Lactobacilli, which are often used in commercial food fermentation, have improved to the point that they might be helpful in medical applications. AREAS COVERED This review summarises various clinical and experimental evidence on efficacy of lactobacilli in treating a wide range of infections. Both laboratory based and clinical studies have been discussed. EXPERT OPINION Lactobacilli are widely accepted as safe biological treatments and host immune modulators (GRAS- Generally regarded as safe) by the US Food and Drug Administration and Qualified Presumption of Safety. Understanding the molecular mechanisms of lactobacilli in the treatment and pathogenicity of bacterial infections can help with the prediction and development of innovative therapeutics aimed at pathogens which have gained resistance to antimicrobials. To formulate effective lactobacilli based therapy significant research on the effectiveness of different lactobacilli strains and its association with demographic distribution is required. Also, the side effects of such therapy needs to be evaluated.
Collapse
Affiliation(s)
- Ujjayni B Saha
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, India
| | - Sunil D Saroj
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Symbiosis Knowledge Village, Lavale, Pune, India
| |
Collapse
|
24
|
Rastogi S, Singh A. Gut microbiome and human health: Exploring how the probiotic genus Lactobacillus modulate immune responses. Front Pharmacol 2022; 13:1042189. [PMID: 36353491 PMCID: PMC9638459 DOI: 10.3389/fphar.2022.1042189] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
The highest density of microbes resides in human gastrointestinal tract, known as “Gut microbiome”. Of note, the members of the genus Lactobacillus that belong to phyla Firmicutes are the most important probiotic bacteria of the gut microbiome. These gut-residing Lactobacillus species not only communicate with each other but also with the gut epithelial lining to balance the gut barrier integrity, mucosal barrier defence and ameliorate the host immune responses. The human body suffers from several inflammatory diseases affecting the gut, lungs, heart, bone or neural tissues. Mounting evidence supports the significant role of Lactobacillus spp. and their components (such as metabolites, peptidoglycans, and/or surface proteins) in modulatingimmune responses, primarily through exchange of immunological signals between gastrointestinal tract and distant organs. This bidirectional crosstalk which is mediated by Lactobacillus spp. promotes anti-inflammatory response, thereby supporting the improvement of symptoms pertaining to asthma, chronic obstructive pulmonary disease (COPD), neuroinflammatory diseases (such as multiple sclerosis, alzheimer’s disease, parkinson’s disease), cardiovascular diseases, inflammatory bowel disease (IBD) and chronic infections in patients. The metabolic disorders, obesity and diabetes are characterized by a low-grade inflammation. Genus Lactobacillus alleviates metabolic disorders by regulating the oxidative stress response and inflammatory pathways. Osteoporosis is also associated with bone inflammation and resorption. The Lactobacillus spp. and their metabolites act as powerful immune cell controllers and exhibit a regulatory role in bone resorption and formation, supporting bone health. Thus, this review demonstrated the mechanisms and summarized the evidence of the benefit of Lactobacillus spp. in alleviating inflammatory diseases pertaining to different organs from animal and clinical trials. The present narrative review explores in detail the complex interactions between the gut-dwelling Lactobacillus spp. and the immune components in distant organs to promote host’s health.
Collapse
|
25
|
Dentice Maidana S, Ortiz Moyano R, Vargas JM, Fukuyama K, Kurata S, Melnikov V, Jure MÁ, Kitazawa H, Villena J. Respiratory Commensal Bacteria Increase Protection against Hypermucoviscous Carbapenem-Resistant Klebsiella pneumoniae ST25 Infection. Pathogens 2022; 11:pathogens11091063. [PMID: 36145495 PMCID: PMC9501321 DOI: 10.3390/pathogens11091063] [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: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 12/18/2022] Open
Abstract
In a previous work, we demonstrated that nasally administered Corynebacterium pseudodiphtheriticum 090104 beneficially modulated the respiratory innate immune response and improved the protection against Respiratory Syncytial Virus and Streptococcus pneumoniae in mice. In this work, we aimed to evaluate whether the immunomodulatory 090104 strain was able to enhance the resistance against the respiratory infection induced by hypermucoviscous carbapenemase-producing (KPC-2) Klebsiella pneumoniae strains belonging to the sequence type (ST) 25. The nasal treatment of mice with C. pseudodiphtheriticum 090104 before the challenge with multiresistant K. pneumoniae ST25 strains significantly reduced lung bacterial cell counts and lung tissue damage. The protective effect of the 090104 strain was related to its ability to regulate the respiratory innate immune response triggered by K. pneumoniae challenge. C. pseudifteriticum 090104 differentially modulated the recruitment of leukocytes into the lung and the production of TNF-α, IFN-γ and IL-10 levels in the respiratory tract and serum. Our results make an advance in the positioning of C. pseudodiphtheriticum 090104 as a next-generation probiotic for the respiratory tract and encourage further research of this bacterium as a promising alternative to develop non-antibiotic therapeutical approaches to enhance the prevention of infections produced by microorganisms with multiple resistance to antimicrobials such as KPC-2-producing hypermucoviscous K. pneumoniae strains belonging to ST25.
Collapse
Affiliation(s)
- Stefania Dentice Maidana
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina
- Laboratory of Antimicrobials, Institute of Microbiology “Luis C. Verna”, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, San Miguel de Tucumán 4000, Argentina
| | - Ramiro Ortiz Moyano
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina
| | - Juan Martin Vargas
- Laboratory of Antimicrobials, Institute of Microbiology “Luis C. Verna”, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, San Miguel de Tucumán 4000, Argentina
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Shoichiro Kurata
- Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Vyacheslav Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | - María Ángela Jure
- Laboratory of Antimicrobials, Institute of Microbiology “Luis C. Verna”, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucuman, San Miguel de Tucumán 4000, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: (H.K.); (J.V.)
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: (H.K.); (J.V.)
| |
Collapse
|
26
|
Russo C, Colaianni V, Ielo G, Valle MS, Spicuzza L, Malaguarnera L. Impact of Lung Microbiota on COPD. Biomedicines 2022; 10:biomedicines10061337. [PMID: 35740358 PMCID: PMC9219765 DOI: 10.3390/biomedicines10061337] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
There is a fine balance in maintaining healthy microbiota composition, and its alterations due to genetic, lifestyle, and environmental factors can lead to the onset of respiratory dysfunctions such as chronic obstructive pulmonary disease (COPD). The relationship between lung microbiota and COPD is currently under study. Little is known about the role of the microbiota in patients with stable or exacerbated COPD. Inflammation in COPD disorders appears to be characterised by dysbiosis, reduced lung activity, and an imbalance between the innate and adaptive immune systems. Lung microbiota intervention could ameliorate these disorders. The microbiota’s anti-inflammatory action could be decisive in the onset of pathologies. In this review, we highlight the feedback loop between microbiota dysfunction, immune response, inflammation, and lung damage in relation to COPD status in order to encourage the development of innovative therapeutic goals for the prevention and management of this disease.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.R.); (V.C.)
| | - Valeria Colaianni
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.R.); (V.C.)
| | - Giuseppe Ielo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (G.I.); (L.S.)
| | - Maria Stella Valle
- Laboratory of Neuro-Biomechanics, Section of Physiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Correspondence: (M.S.V.); (L.M.)
| | - Lucia Spicuzza
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (G.I.); (L.S.)
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.R.); (V.C.)
- Correspondence: (M.S.V.); (L.M.)
| |
Collapse
|
27
|
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: 67] [Impact Index Per Article: 22.3] [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
|
28
|
Hamouda RA, Hamza HA, Salem ML, Kamal S, Alhasani RH, Alsharif I, Mahrous H, Abdella A. Synergistic Hypolipidemic and Immunomodulatory Activity of Lactobacillus and Spirulina platensis. FERMENTATION-BASEL 2022; 8:220. [DOI: 10.3390/fermentation8050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Hyperlipidemia is a significant risk factor for atherosclerosis and coronary heart disease (CHD). The aim of this study was to investigate hypolipidemic effects of Lactobacillus, Spirulina and their combination on Swiss albino mice fed a regular or high-cholesterol diet. Rosuvastatin was used as a reference drug The highest body weight, total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol LDL-C and the lowest high-density lipoprotein cholesterol were recorded in a positive control group (G5). Treatment with Lactobacillus or Spirulina or by their combination resulted in a significant decrease in body weight, TC, TG, LDL-C and significant increase in HDL-C (p < 0.05) in both mice fed a regular diet or high-cholesterol diet. The treatments induced a significant increase in Hb, MCHC and HCT levels in mice fed a regular diet (p < 0.05). They did not induce a significant effect on these parameters in mice fed a high-cholesterol diet, while treatment with standard rosuvastatin induced a significant decrease in these parameters (p < 0.05). The treatments induced a significant increase in the platelet count and WBC number in mice fed a regular diet p < 0.05), while they induced significant decrease in these parameters in mice fed a high-cholesterol diet p < 0.05. They also stimulated the innate immunity represented by both monocyte and neutrophil cells in mice fed a regular diet, while this immunity was reduced in mice fed a high-cholesterol diet. It also caused a marked reduction in inflammation and an improvement in the congestion of cardiac tissues, the aorta, and the spleen. The treatment of hyperlipidemic mice with combination of Lactobacillus and Spirulina gave similar results to those obtained with treatment by rosuvastatin.
Collapse
Affiliation(s)
- Ragaa A. Hamouda
- Biology Department, College of Science and Arts at Khulis, University of Jeddah, Jeddah 21921, Saudi Arabia
- Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 32897, Egypt
| | - Hanafy A. Hamza
- Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 32897, Egypt
| | - Mohammed L. Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta 31111, Egypt
| | - Shymaa Kamal
- National Organization for Drug Control and Research, Cairo 12561, Egypt
| | - Reem Hasaballah Alhasani
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah 21961, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Hoda Mahrous
- Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 32897, Egypt
| | - Asmaa Abdella
- Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat City 32897, Egypt
| |
Collapse
|
29
|
Human microbiota: a crucial gatekeeper in lung cancer initiation, progression, and treatment. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
30
|
Gu X, Zhang S, Ma W, Wang Q, Li Y, Xia C, Xu Y, Zhang T, Yang L, Zhou M. The Impact of Instant Coffee and Decaffeinated Coffee on the Gut Microbiota and Depression-Like Behaviors of Sleep-Deprived Rats. Front Microbiol 2022; 13:778512. [PMID: 35283829 PMCID: PMC8914519 DOI: 10.3389/fmicb.2022.778512] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/04/2022] [Indexed: 12/21/2022] Open
Abstract
Objective Based on our previous research, chronic paradoxical sleep deprivation (PSD) can cause depression-like behaviors and microbial changes in gut microbiota. Coffee, as the world’s most popular drink for the lack of sleep, is beneficial to health and attention and can eliminate the cognitive sequelae caused by poor sleep. The purpose of this study is to investigate the effects of coffee and decaffeinated coffee on PSD rats. Research Design and Methods A total of 32 rats were divided into four groups: control group, PSD model group, conventional coffee group, and decaffeinated coffee group. Behavioral tests, including sucrose preference test, open field test, forced swimming test, and tail suspension test, as well as biochemical detection for inflammatory and antioxidant indexes were performed. The effects of coffee and decaffeinated coffee on the gut microbiota of PSD rats were investigated by 16S rRNA gene sequencing. Results Coffee and decaffeinated coffee significantly improved the depression-like behaviors. Moreover, the serum levels of interleukin-6 and tumor necrosis factor alpha were decreased in both coffee and decaffeinated coffee groups, as well as the levels of superoxide dismutase and GSH-Px were increased. Gut microbiota analysis revealed that the abundance of S24-7, Lachnospiraceae, Oscillospira, and Parabacteroides were significantly increased in PSD rats, while the abundance of Akkermansia and Klebsiella were significantly decreased. After the treatment of coffee and decaffeinated coffee, the abundance of the above gut microbiota was all restored in different degrees. Coffee had relatively more significant effects on PSD-induced depressive-like behaviors, while the difference between coffee and decaffeinated coffee was not obvious in correcting the disorder of gut microbiota. Conclusions These findings have shown that both coffee and decaffeinated coffee are effective for sleep deprivation-induced depression-like behaviors and the dysbiosis of gut microbiota and indicated that caffeine may be not the only key substance of coffee for regulating gut microbiota.
Collapse
Affiliation(s)
- Xinyi Gu
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuyi Zhang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weini Ma
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qixue Wang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Li
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Xia
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Zhang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Yang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mingmei Zhou,
| |
Collapse
|
31
|
Gao J, Wang R, Liu J, Wang W, Chen Y, Cai W. Effects of novel microecologics combined with traditional Chinese medicine and probiotics on growth performance and health of broilers. Poult Sci 2022; 101:101412. [PMID: 34920387 PMCID: PMC8683594 DOI: 10.1016/j.psj.2021.101412] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/10/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
In this study, we prepared a kind of novel microecologics, namely Chinese medicine-probiotic compound microecological preparation (CPCMP), which is composed of 5 traditional Chinese medicine herbs (Galla Chinensis, Andrographis paniculata, Arctii Fructus, Glycyrrhizae Radix, and Schizonepeta tenuifolia) fermented by Aspergillus niger and a kind of compound probiotics (Lactobacillus plantarum A37 and L. plantarum MIII). The effects of the CPCMP in broilers on growth performance, serum parameters, immune function, and intestinal health were investigated. A total of 450 one-day-old male Arbor Acres broilers were randomly divided into 6 treatment groups with 5 replicates, 15 birds per replicate. Treatments consisted of: blank control, CPCMP, positive control, commercial CPCMP, traditional Chinese medicine, and probiotics groups, which were birds fed with basal diet supplemented with no extra additives, 0.2% CPCMP, 0.0035% chlortetracycline, 0.2% commercially available CPCMP, 0.2% fermented traditional Chinese medicines, and 0.2% compound probiotics, respectively. CPCMP obviously increased the average body weight and average daily gain (P < 0.05, compared with any other group) and decreased the feed:gain ratio of broilers (P < 0.05, compared with the blank control, commercial CPCMP, traditional Chinese medicine, or probiotics group). Moreover, it significantly increased glutathione peroxidase and secretory immunoglobulin A levels and spleen/bursa indices (P < 0.05 for all, compared with the blank control, commercial CPCMP, traditional Chinese medicine, or probiotics group). Villus heights in duodenum, jejunum, and ileum were also elevated by CPCMP treatment (P < 0.05, compared with any other group). Furthermore, CPCMP substantially increased jejunal mRNA levels of occludin and zonula occludens-1 (P < 0.05, compared with the blank control, positive control, or probiotics group) and facilitated the growth and colonization of beneficial cecal bacteria, such as Olsenella, Barnesiella, and Lactobacillus. Overall results show that the CPCMP prepared in our work contributes to improving growth performance, serum parameters, immune function, and intestinal health of broilers and exerts synergistic effects of traditional Chinese medicines and probiotics to some extent. Our findings suggest that CPCMP is a promising antibiotic substitute in the livestock and poultry industry in the future.
Collapse
Affiliation(s)
- Jin Gao
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Rui Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Jingxuan Liu
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Wenling Wang
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Yong Chen
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Wentao Cai
- Hubei Province Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China.
| |
Collapse
|
32
|
Salvador PBU, Dalmacio LMM, Kim SH, Kang DK, Balolong MP. Immunomodulatory potential of four candidate probiotic Lactobacillus strains from plant and animal origin using comparative genomic analysis. Access Microbiol 2022; 3:000299. [PMID: 35024559 PMCID: PMC8749136 DOI: 10.1099/acmi.0.000299] [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: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 12/11/2022] Open
Abstract
Probiotic strains from different origins have shown promise in recent decades for their health benefits, for example in promoting and regulating the immune system. The immunomodulatory potential of four Lactobacillus strains from animal and plant origins was evaluated in this paper based on their genomic information. Comparative genomic analysis was performed through genome alignment, average nucleotide identity (ANI) analysis and gene mining for putative immunomodulatory genes. The genomes of the four Lactobacillus strains show relative similarities in multiple regions, as observed in the genome alignment. However, ANI analysis showed that L. mucosae LM1 and L. fermentum SK152 are the most similar when considering their nucleotide sequences alone. Gene mining of putative immunomodulatory genes studied from L. plantarum WCFS1 yielded multiple results in the four potential probiotic strains, with L. plantarum SK151 showing the largest number of genes at around 74 hits, followed by L. johnsonii PF01 at 41 genes when adjusted for matches with at least 30 % identity. Looking at the immunomodulatory genes in each strain, L. plantarum SK151 and L. johnsonii PF01 may have wider activity, covering both immune activation and immune suppression, as compared to L. mucosae LM1 and L. fermentum SK152, which could be more effective in activating immune cells and the pro-inflammatory cascade rather than suppressing it. The similarities and differences between the four Lactobacillus species showed that there is no definitive trend based on the origin of isolation alone. Moreover, higher percentage identities between genomes do not directly correlate with higher similarities in potential activity, such as in immunomodulation. The immunomodulatory function of each of the four Lactobacillus strains should be observed and verified experimentally in the future, since some the activity of some genes may be strain-specific, which would not be identified through comparative genomics alone.
Collapse
Affiliation(s)
- Paul Benedic U Salvador
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| | - Leslie Michelle M Dalmacio
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| | - Sang Hoon Kim
- Department of Animal Resources Science, College of Biotechnology and Bioengineering, Dankook University, Republic of Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, College of Biotechnology and Bioengineering, Dankook University, Republic of Korea
| | - Marilen P Balolong
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| |
Collapse
|
33
|
Kim YJ, Lee JY, Lee JJ, Jeon SM, Silwal P, Kim IS, Kim HJ, Park CR, Chung C, Han JE, Choi JW, Tak EJ, Yoo JH, Jeong SW, Kim DY, Ketphan W, Kim SY, Jhun BW, Whang J, Kim JM, Eoh H, Bae JW, Jo EK. Arginine-mediated gut microbiome remodeling promotes host pulmonary immune defense against nontuberculous mycobacterial infection. Gut Microbes 2022; 14:2073132. [PMID: 35579969 PMCID: PMC9116420 DOI: 10.1080/19490976.2022.2073132] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Nontuberculous mycobacterial pulmonary diseases (NTM-PDs) are emerging as global health threats with issues of antibiotic resistance. Accumulating evidence suggests that the gut-lung axis may provide novel candidates for host-directed therapeutics against various infectious diseases. However, little is known about the gut-lung axis in the context of host protective immunity to identify new therapeutics for NTM-PDs. This study was performed to identify gut microbes and metabolites capable of conferring pulmonary immunity to NTM-PDs. Using metabolomics analysis of sera from NTM-PD patients and mouse models, we showed that the levels of l-arginine were decreased in sera from NTM-PD patients and NTM-infected mice. Oral administration of l-arginine significantly enhanced pulmonary antimicrobial activities with the expansion of IFN-γ-producing effector T cells and a shift to microbicidal (M1) macrophages in the lungs of NTM-PD model mice. Mice that received fecal microbiota transplants from l-arginine-treated mice showed increased protective host defense in the lungs against NTM-PD, whereas l-arginine-induced pulmonary host defense was attenuated in mice treated with antibiotics. Using 16S rRNA sequencing, we further showed that l-arginine administration resulted in enrichment of the gut microbiota composition with Bifidobacterium species. Notably, oral treatment with either Bifidobacterium pseudolongum or inosine enhanced antimicrobial pulmonary immune defense against NTM infection, even with multidrug-resistant clinical NTM strains. Our findings indicate that l-arginine-induced gut microbiota remodeling with enrichment of B. pseudolongum boosts pulmonary immune defense against NTM infection by driving the protective gut-lung axis in vivo.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - June-Young Lee
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Jae Jin Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
| | - In Soo Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Cho Rong Park
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| | - Chaeuk Chung
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Chungnam National University School of MedicineDaejeon, Korea
| | - Jeong Eun Han
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Jee-Won Choi
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Euon Jung Tak
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Ji-Ho Yoo
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Su-Won Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Do-Yeon Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
| | - Warisa Ketphan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, South Korea
| | - Byung Woo Jhun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoul, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC) & Basic Research Section, The Korean Institute of Tuberculosis (KIT), Cheongju, Korea
| | - Jin-Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyungjin Eoh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern CaliforniaCA, USA
| | - Jin-Woo Bae
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Seoul, Korea
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of MedicineDaejeon, Korea
- Infection Control Convergence Research Center, Chungnam National University School of MedicineDaejeon, Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon, Korea
| |
Collapse
|
34
|
Li QR, Tan SR, Yang L, He W, Chen L, Shen FX, Wang Z, Wang HF. Mechanism of chlorogenic acid in alveolar macrophage polarization in Klebsiella pneumoniae-induced pneumonia. J Leukoc Biol 2021; 112:9-21. [PMID: 34585429 DOI: 10.1002/jlb.3hi0721-368r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chlorogenic acid (CA) has been discovered to regulate macrophage polarization in pneumonia. This study aims to analyze the functional mechanism of CA in alveolar macrophage (AM) polarization and provide a theoretical basis for treatment of Klebsiella pneumoniae (Kp)-induced pneumonia. Mice were infected with Kp, and treated with CA and silent information regulator 1 (SIRT1) inhibitor (Selisistat). Mouse survival rate was recorded and bacterial burden was detected. AM polarization and pathologic change of lung tissues were evaluated. Expressions of SIRT1 and HMGB1 and cytokine levels were detected. MH-S cells were infected with Kp to establish the pneumonia cell model, followed by transfection of si-SIRT1 and HMGB1 overexpression vector. The HMGB1 expression in the nucleus and cytoplasm was detected. HMGB1 subcellular localization and HMGB1 acetylation level were detected. Kp led to high death rates, SIRT down-regulation and increases in inflammatory factor level and bacterial burden, and promoted M1 polarization. CA treatment improved mouse survival rate and promoted M2 polarization and SIRT1 expression. SIRT1 decreased HMGB1 acetylation level to inhibit nuclear to the cytoplasm translocation. Silencing SIRT1 or HMGB1 overexpression reversed the effect of CA on Kp-induced pneumonia. Overall, CA activated SIRT1 to inhibit HMGB1 acetylation level and nuclear translocation, thereby promoting M2 polarization in AMs and alleviating Kp-induced pneumonia.
Collapse
Affiliation(s)
- Qing Rong Li
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shi Rui Tan
- School of Agriculture, Chenggong Campus, Yunnan University, Kunming, China
| | - Lu Yang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei He
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Chen
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fen Xiu Shen
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhuo Wang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hai Feng Wang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
35
|
Liu X, Cheng Y, Zang D, Zhang M, Li X, Liu D, Gao B, Zhou H, Sun J, Han X, Lin M, Chen J. The Role of Gut Microbiota in Lung Cancer: From Carcinogenesis to Immunotherapy. Front Oncol 2021; 11:720842. [PMID: 34490119 PMCID: PMC8417127 DOI: 10.3389/fonc.2021.720842] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
The influence of microbiota on host health and disease has attracted adequate attention, and gut microbiota components and microbiota-derived metabolites affect host immune homeostasis locally and systematically. Some studies have found that gut dysbiosis, disturbance of the structure and function of the gut microbiome, disrupts pulmonary immune homeostasis, thus leading to increased disease susceptibility; the gut-lung axis is the primary cross-talk for this communication. Gut dysbiosis is involved in carcinogenesis and the progression of lung cancer through genotoxicity, systemic inflammation, and defective immunosurveillance. In addition, the gut microbiome harbors the potential to be a novel biomarker for predicting sensitivity and adverse reactions to immunotherapy in patients with lung cancer. Probiotics and fecal microbiota transplantation (FMT) can enhance the efficacy and depress the toxicity of immune checkpoint inhibitors by regulating the gut microbiota. Although current studies have found that gut microbiota closely participates in the development and immunotherapy of lung cancer, the mechanisms require further investigation. Therefore, this review aims to discuss the underlying mechanisms of gut microbiota influencing carcinogenesis and immunotherapy in lung cancer and to provide new strategies for governing gut microbiota to enhance the prevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Xiangjun Liu
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Ye Cheng
- Department of Oncology, The Third Hospital of Dalian Medical University, Dalian, China
| | - Dan Zang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Min Zhang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xiuhua Li
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dan Liu
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Bing Gao
- Department of Oncology, The Third Hospital of Dalian Medical University, Dalian, China
| | - Huan Zhou
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jinzhe Sun
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xu Han
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Lin
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
36
|
Zhang L, Zhang XT, Jin P, Zhao H, Liu X, Sheng Q. Effects of oral administration of Spirulina platensis and probiotics on serum immunity indexes, colonic immune factors, fecal odor, and fecal flora in mice. Anim Sci J 2021; 92:e13593. [PMID: 34289202 DOI: 10.1111/asj.13593] [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: 12/11/2020] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/01/2022]
Abstract
To evaluate the effects of Spirulina platensis and probiotics on growth, immunity indexes, fecal flora, and fecal odor in mice, 40 mice were randomly allotted to four groups, and each was administrated with nothing, S. platensis, probiotics, or both for 28 days, respectively. Then, many indexes were measured. The results showed that S. platensis was more effective (P < 0.001) than probiotics in improving mice's feed conversion ration (FCR). In immunity, probiotics administration increased (P < 0.042) serum IgE, IgM, IFN-γ, colonic AHR, TLR4, and NF-κB protein expression and decreased (P < 0.039) serum IL-1α, IL-21, IL-22, and colonic ARNT gene expression. However, the S. platensis showed weaker effect, which increased (P < 0.025) the serum IgE, IgM, TNF-α, and the colonic AHR and NF-κB protein expression, and decreased (P < 0.01) serum IL-21. Probiotics consumption decreased the fecal odor by decreasing (P < 0.02) fecal Escherichia coli, indole-3-acetic acid (IAA), and skatole contents, and the S. platensis decreased (P = 0.04) the IAA. These results indicated that oral administration of probiotics, S. platensis, or both of them in mice probably benefited body's immunity and reduced fecal odor. However, their mechanisms were still unclear and need further study.
Collapse
Affiliation(s)
- Lingyan Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Xing Tao Zhang
- The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, China
| | - Pingting Jin
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Hongbo Zhao
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China
| | - Xue Liu
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingkai Sheng
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Jinan, China.,College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
37
|
From Klebsiella pneumoniae Colonization to Dissemination: An Overview of Studies Implementing Murine Models. Microorganisms 2021; 9:microorganisms9061282. [PMID: 34204632 PMCID: PMC8231111 DOI: 10.3390/microorganisms9061282] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/31/2022] Open
Abstract
Klebsiella pneumoniae is a Gram-negative pathogen responsible for community-acquired and nosocomial infections. The strains of this species belong to the opportunistic group, which is comprised of the multidrug-resistant strains, or the hypervirulent group, depending on their accessory genome, which determines bacterial pathogenicity and the host immune response. The aim of this survey is to present an overview of the murine models mimicking K. pneumoniae infectious processes (i.e., gastrointestinal colonization, urinary, pulmonary, and systemic infections), and the bacterial functions deployed to colonize and disseminate into the host. These in vivo approaches are pivotal to develop new therapeutics to limit K. pneumoniae infections via a modulation of the immune responses and/or microbiota.
Collapse
|
38
|
Zhao L, Mao Y, Yu H, Liu H, Wang C, Liu J, Han Y, Bi Y, Zhang D. The Preventive Effects of Lactobacillus casei on Acute Lung Injury Induced by Lipopolysaccharide. Indian J Microbiol 2021; 61:370-382. [PMID: 34092818 PMCID: PMC8169435 DOI: 10.1007/s12088-021-00949-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/24/2021] [Indexed: 12/29/2022] Open
Abstract
Lactobacillus has been reported to inhibit acute lung injury (ALI). However, the molecular mechanism of Lactobacillus casei (L. casei) in preventing ALI has not been identified, so we investigated whether L. casei pretreatment could inhibit the activation of TLR4/MyD88/NF-κB signaling pathway following ALI. ALI model was established by intraperitoneal injection of 2 mg/kg lipopolysaccharide (LPS) to female BALB/c mice. In L. casei LC2W group, mice were intragastrically administrated L. casei LC2W for a week, before the ALI modeling. The serum of normal BALB/c mice after intragastric administration of L. casei LC2W was used for in vitro cell assays. The serum was pre-incubated with mouse macrophage cell line (RAW264.7) and human lung cell line (HLF-A), then LPS was added to co-incubate. Compared with ALI model group, L. casei LC2W pretreatment significantly reduced lung pathological damage, the number of neutrophils and total cells in bronchoalveolar lavage fluid. Besides, L. casei LC2W pretreatment could significantly reverse the abnormal expression of ICAM-1, IL-6, TNF-α and IL-10 in lung tissue and serum, plus, L. casei LC2W significantly reduced the phosphorylation levels of IRAK-1 and NF-κB p65. In vitro, the serum decreased the up-regulation of IL-6 and TNF-α in cell lines induced by LPS. In conclusion, L. casei LC2W intragastric administration pretreatment could significantly improve LPS-induced ALI in mice, probably through circulation to reach the lungs so as to inhibit the inflammatory response induced by activation of TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lihui Zhao
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Ying Mao
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Haiming Yu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - He Liu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Chao Wang
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Jianwei Liu
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Yutong Han
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Yang Bi
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| | - Donghai Zhang
- the Second Affiliated Hospital of Qiqihar Medical University, No. 37 Zhonghua West Road, Jianhua DistrictHeilongjiang Province, Qiqihar City, 161000 China
| |
Collapse
|
39
|
Balderrama-Gutierrez G, Milovic A, Cook VJ, Islam MN, Zhang Y, Kiaris H, Belisle JT, Mortazavi A, Barbour AG. An Infection-Tolerant Mammalian Reservoir for Several Zoonotic Agents Broadly Counters the Inflammatory Effects of Endotoxin. mBio 2021; 12:e00588-21. [PMID: 33849979 PMCID: PMC8092257 DOI: 10.1128/mbio.00588-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Animals that are competent reservoirs of zoonotic pathogens commonly suffer little morbidity from the infections. To investigate mechanisms of this tolerance of infection, we used single-dose lipopolysaccharide (LPS) as an experimental model of inflammation and compared the responses of two rodents: Peromyscus leucopus, the white-footed deermouse and reservoir for the agents of Lyme disease and other zoonoses, and the house mouse Mus musculus Four hours after injection with LPS or saline, blood, spleen, and liver samples were collected and subjected to transcriptome sequencing (RNA-seq), metabolomics, and specific reverse transcriptase quantitative PCR (RT-qPCR). Differential expression analysis was at the gene, pathway, and network levels. LPS-treated deermice showed signs of sickness similar to those of exposed mice and had similar increases in corticosterone levels and expression of interleukin 6 (IL-6), tumor necrosis factor, IL-1β, and C-reactive protein. By network analysis, the M. musculus response to LPS was characterized as cytokine associated, while the P. leucopus response was dominated by neutrophil activity terms. In addition, dichotomies in the expression levels of arginase 1 and nitric oxide synthase 2 and of IL-10 and IL-12 were consistent with type M1 macrophage responses in mice and type M2 responses in deermice. Analysis of metabolites in plasma and RNA in organs revealed species differences in tryptophan metabolism. Two genes in particular signified the different phenotypes of deermice and mice: the Slpi and Ibsp genes. Key RNA-seq findings for P. leucopus were replicated in older animals, in a systemic bacterial infection, and with cultivated fibroblasts. The findings indicate that P. leucopus possesses several adaptive traits to moderate inflammation in its balancing of infection resistance and tolerance.IMPORTANCE Animals that are natural carriers of pathogens that cause human diseases commonly manifest little or no sickness as a consequence of infection. Examples include the deermouse, Peromyscus leucopus, which is a reservoir for Lyme disease and several other disease agents in North America, and some types of bats, which are carriers of viruses with pathogenicity for humans. Mechanisms of this phenomenon of infection tolerance and entailed trade-off costs are poorly understood. Using a single injection of lipopolysaccharide (LPS) endotoxin as a proxy for infection, we found that deermice differed from the mouse (Mus musculus) in responses to LPS in several diverse pathways, including innate immunity, oxidative stress, and metabolism. Features distinguishing the deermice cumulatively would moderate downstream ill effects of LPS. Insights gained from the P. leucopus model in the laboratory have implications for studying infection tolerance in other important reservoir species, including bats and other types of wildlife.
Collapse
Affiliation(s)
- Gabriela Balderrama-Gutierrez
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Ana Milovic
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, California, USA
| | - Vanessa J Cook
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, California, USA
| | - M Nurul Islam
- Department of Microbiology, Immunology, & Pathology, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Youwen Zhang
- Department of Drug Discovery & Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Hippokratis Kiaris
- Peromyscus Genetic Stock Center, University of South Carolina, Columbia, South Carolina, USA
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, California, USA
| | - John T Belisle
- Department of Microbiology, Immunology, & Pathology, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Alan G Barbour
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, California, USA
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, California, USA
- Department of Ecology & Evolutionary Biology, School of Biological Sciences, University of California Irvine, Irvine, California, USA
| |
Collapse
|
40
|
Layús BI, Gomez MA, Cazorla SI, Rodriguez AV. Drops of Lactiplantibacillus plantarum CRL 759 culture supernatant attenuates eyes inflammation induced by lipopolysaccharide. Benef Microbes 2021; 12:163-174. [PMID: 33769229 DOI: 10.3920/bm2020.0101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anti-inflammatory effect of soluble secreted compounds of probiotic bacteria was widely demonstrated as therapy for different inflammatory diseases, but was not investigated in inflammatory eye disorders. The aim of this study was to determine whether Lactiplantibacillus plantarum CRL759 cell-free supernatant reduced inflammatory parameters and clinical signs in ocular inflammations. First, we evaluated the effect of L. plantarum CRL759 supernatant in vitro on human retinal cell line, ARPE-19 cells, stimulated with lipopolysaccharide (LPS). Then, we investigated in vivo its capacity to decrease inflammation by local administration on the eyes of mice with endotoxin induced inflammation. In vitro assays demonstrated that L. plantarum CRL759 supernatant reduced the production of interleukin (IL)-6, IL-8, nitric oxide and thiobarbituric acid reactive substances in LPS-stimulated ARPE-19 cells. Our in vivo data proved that L. plantarum supernatant significantly reduced the clinical score of endotoxin treated mice and diminished levels of tumour necrosis factor alpha, interferon gamma and protein concentration in aqueous humour. Histological examination showed reduction of infiltrating inflammatory cells in the posterior segment of the eyes. As far as we know, this is the first report showing that Lactobacillus spp. supernatant administered as drops reduces some parameters of ocular inflammation. This promising strategy is safe and could alleviate symptoms and signs of ocular inflammation in people that are refractories to the conventional therapies.
Collapse
Affiliation(s)
- B I Layús
- CONICET, CERELA, Batalla de Chacabuco 145, 4000 San Miguel de Tucumán, Argentina
| | - M A Gomez
- Hospital Ángel C. Padilla, Juan Bautista Alberdi 550, 4000 San Miguel de Tucumán, Argentina
| | - S I Cazorla
- CONICET, CERELA, Batalla de Chacabuco 145, 4000 San Miguel de Tucumán, Argentina
| | - A V Rodriguez
- CONICET, Cell Signaling Laboratory, Batalla de Chacabuco 145, 4000 San Miguel de Tucumán, Argentina
| |
Collapse
|
41
|
Zhao W, Peng C, Sakandar HA, Kwok LY, Zhang W. Meta-Analysis: Randomized Trials of Lactobacillus plantarum on Immune Regulation Over the Last Decades. Front Immunol 2021; 12:643420. [PMID: 33828554 PMCID: PMC8019694 DOI: 10.3389/fimmu.2021.643420] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/24/2021] [Indexed: 12/18/2022] Open
Abstract
Lactobacillus (L.) plantarum strains, belong to lactic acid bacteria group, are considered indispensable probiotics. Here, we performed meta-analysis to evaluate the regulatory effects of L. plantarum on the immunity during clinical trials. This meta-analysis was conducted by searching across four most common literature databases, namely, Cochrane Central Register of Controlled Trials, Web of Science, Embase, and PubMed. Clinical trial articles that met the inclusion and exclusion criteria were analyzed by Review Manager (version 5.3). p-value < 0.05 of the total effect was considered statistically significant. Finally, total of 677 references were retrieved, among which six references and 18 randomized controlled trials were included in the meta-analysis. The mean differences observed at 95% confidence interval: interleukin (IL)-4, -0.48 pg/mL (-0.79 to -0.17; p < 0.05); IL-10, 9.88 pg/mL (6.52 to 13.2; p < 0.05); tumor necrosis factor (TNF)-α, -2.34 pg/mL (-3.5 to -1.19; p < 0.05); interferon (IFN)-γ, -0.99 pg/mL (-1.56 to -0.41; p < 0.05). Therefore, meta-analysis results suggested that L. plantarum could promote host immunity by regulating pro-inflammatory and anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Wei Zhao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Chuantao Peng
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China.,Qingdao Special Food Research Institute, Qingdao Agricultural University, Qingdao, China
| | - Hafiz Arbab Sakandar
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
42
|
Saint-Criq V, Lugo-Villarino G, Thomas M. Dysbiosis, malnutrition and enhanced gut-lung axis contribute to age-related respiratory diseases. Ageing Res Rev 2021; 66:101235. [PMID: 33321253 DOI: 10.1016/j.arr.2020.101235] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Older people are at an increased risk of developing respiratory diseases such as chronic obstructive pulmonary diseases, asthma, idiopathic pulmonary fibrosis or lung infections. Susceptibility to these diseases is partly due to the intrinsic ageing process, characterized by genomic, cellular and metabolic hallmarks and immunosenescence, and is associated with changes in the intestinal microbiota. Importantly, in the lungs, ageing is also associated with a dysbiosis and loss of resilience of the resident microbiota and alterations of the gut-lung axis. Notably, as malnutrition is often observed in the elderly, nutrition is one of the most accessible modifiable factors affecting both senescence and microbiota. This article reviews the changes affecting the lung and its resident microbiota during ageing, as well as the interconnections between malnutrition, senescence, microbiota, gut-lung axis and respiratory health. As the communication along the gut-lung axis becomes more permissive with ageing, this review also explores the evidence that the gut and lung microbiota are key players in the maintenance of healthy lungs, and as such, are potential targets for nutrition-based preventive strategies against lung disease in elderly populations.
Collapse
|
43
|
Spangler JR, Caruana JC, Medintz IL, Walper SA. Harnessing the potential of Lactobacillus species for therapeutic delivery at the lumenal-mucosal interface. Future Sci OA 2021; 7:FSO671. [PMID: 33815818 PMCID: PMC8015674 DOI: 10.2144/fsoa-2020-0153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus species have been studied for over 30 years in their role as commensal organisms in the human gut. Recently there has been a surge of interest in their abilities to natively and recombinantly stimulate immune activities, and studies have identified strains and novel molecules that convey particular advantages for applications as both immune adjuvants and immunomodulators. In this review, we discuss the recent advances in Lactobacillus-related activity at the gut/microbiota interface, the efforts to probe the boundaries of the direct and indirect therapeutic potential of these bacteria, and highlight the continued interest in harnessing the native capacity for the production of biogenic compounds shown to influence nervous system activity. Taken together, these aspects underscore Lactobacillus species as versatile therapeutic delivery vehicles capable of effector production at the lumenal-mucosal interface, and further establish a foundation of efficacy upon which future engineered strains can expand.
Collapse
Affiliation(s)
- Joseph R Spangler
- National Research Council Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Julie C Caruana
- American Society for Engineering Education Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Igor L Medintz
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Scott A Walper
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| |
Collapse
|
44
|
Shi Y, Zhao W, Liu G, Ali T, Chen P, Liu Y, Kastelic JP, Han B, Gao J. Bacteriophages isolated from dairy farm mitigated Klebsiella pneumoniae-induced inflammation in bovine mammary epithelial cells cultured in vitro. BMC Vet Res 2021; 17:37. [PMID: 33468111 PMCID: PMC7814619 DOI: 10.1186/s12917-020-02738-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/29/2020] [Indexed: 12/02/2022] Open
Abstract
Background Klebsiella pneumoniae, an environmental pathogen causing mastitis in dairy cattle, is often resistant to antibiotics. K. pneumoniae was used as the host bacteria to support bacteriophage replication; 2 bacteriophages, CM8-1 and SJT-2 were isolated and considered to have therapeutic potential. In the present study, we determined the ability of these 2 bacteriophages to mitigate cytotoxicity, pathomorphological changes, inflammatory responses and apoptosis induced by K. pneumoniae (bacteriophage to K. pneumoniae MOI 1:10) in bovine mammary epithelial cells (bMECs) cultured in vitro. Results Bacteriophages reduced bacterial adhesion and invasion and cytotoxicity (lactate dehydrogenase release). Morphological changes in bMECs, including swelling, shrinkage, necrosis and hematoxylin and eosin staining of cytoplasm, were apparent 4 to 8 h after infection with K. pneumoniae, but each bacteriophage significantly suppressed damage and decreased TNF-α and IL-1β concentrations. K. pneumoniae enhanced mRNA expression of TLR4, NF-κB, TNF-α, IL-1β, IL-6, IL-8, caspase-3, caspase-9 and cyt-c in bMECs and increased apoptosis of bMECs, although these effects were mitigated by treatment with either bacteriophage for 8 h. Conclusions Bacteriophages CM8-1 and SJT-2 mitigated K. pneumoniae-induced inflammation in bMECs cultured in vitro. Therefore, the potential of these bacteriophages for treating mastitis in cows should be determined in clinical trials.
Collapse
Affiliation(s)
- Yuxiang Shi
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China.,College of Life Sciences and Food Engineering, Hebei University of Engineering, 056038, Handan, Hebei, P.R. China
| | - Wenpeng Zhao
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China
| | - Gang Liu
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China
| | - Tariq Ali
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China.,Center of Microbiology & Biotechnology, Veterinary Research Institute, Peshawar, Pakistan
| | - Peng Chen
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, 271018, Taìan, China
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, T2N 4N1, Calgary, AB, Canada
| | - Bo Han
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China.
| | - Jian Gao
- College of Veterinary Medicine, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, 100193, Beijing, P. R. China.
| |
Collapse
|
45
|
Sun T, Yu H, Fu J. Respiratory Tract Microecology and Bronchopulmonary Dysplasia in Preterm Infants. Front Pediatr 2021; 9:762545. [PMID: 34966701 PMCID: PMC8711720 DOI: 10.3389/fped.2021.762545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe respiratory complication in preterm infants. Although the etiology and pathogenesis of BPD are complex and remain to be clarified, recent studies have reported a certain correlation between the microecological environment of the respiratory tract and BPD. Changes in respiratory tract microecology, such as abnormal microbial diversity and altered evolutional patterns, are observed prior to the development of BPD in premature infants. Therefore, research on the colonization and evolution of neonatal respiratory tract microecology and its relationship with BPD is expected to provide new ideas for its prevention and treatment. In this paper, we review microecological changes in the respiratory tract and the mechanisms by which they can lead to BPD in preterm infants.
Collapse
Affiliation(s)
- Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
46
|
De Rudder C, Garcia-Tímermans C, De Boeck I, Lebeer S, Van de Wiele T, Calatayud Arroyo M. Lacticaseibacillus casei AMBR2 modulates the epithelial barrier function and immune response in a donor-derived nasal microbiota manner. Sci Rep 2020; 10:16939. [PMID: 33037304 PMCID: PMC7547715 DOI: 10.1038/s41598-020-73857-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023] Open
Abstract
Live biotherapeutic products (LBP) are emerging as alternative treatment strategies for chronic rhinosinusitis. The selection of interesting candidate LBPs often involves model systems that do not include the polymicrobial background (i.e. the host microbiota) in which they will be introduced. Here, we performed a screening in a simplified model system of upper respiratory epithelium to assess the effect of nasal microbiota composition on the ability to attach and grow of a potential LBP, Lacticaseibacillus casei AMBR2, in this polymicrobial background. After selecting the most permissive and least permissive donor, L. casei AMBR2 colonisation in their respective polymicrobial backgrounds was assessed in more physiologically relevant model systems. We examined cytotoxicity, epithelial barrier function, and cytokine secretion, as well as bacterial cell density and phenotypic diversity in differentiated airway epithelium based models, with or without macrophage-like cells. L. casei AMBR2 could colonize in the presence of both selected donor microbiota and increased epithelial barrier resistance in presence of donor-derived nasal bacteria, as well as anti-inflammatory cytokine secretion in the presence of macrophage-like cells. This study highlights the potential of L. casei AMBR2 as LBP and the necessity to employ physiologically relevant model systems to investigate host–microbe interaction in LBP research.
Collapse
Affiliation(s)
- Charlotte De Rudder
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium
| | - Cristina Garcia-Tímermans
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium
| | - Ilke De Boeck
- Research Group of Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Research Group of Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium.
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Coupure Links 653, Ghent University, 9000, Ghent, Belgium.,Group of Lactic Bacteria and Probiotics, Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA), Spanish Research Council (CSIC), Valencia, Spain
| |
Collapse
|
47
|
The Gut Microbiota and Respiratory Diseases: New Evidence. J Immunol Res 2020; 2020:2340670. [PMID: 32802893 PMCID: PMC7415116 DOI: 10.1155/2020/2340670] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
Abstract
Human body surfaces, such as the skin, intestines, and respiratory and urogenital tracts, are colonized by a large number of microorganisms, including bacteria, fungi, and viruses, with the gut being the most densely and extensively colonized organ. The microbiome plays an essential role in immune system development and tissue homeostasis. Gut microbiota dysbiosis not only modulates the immune responses of the gastrointestinal (GI) tract but also impacts the immunity of distal organs, such as the lung, further affecting lung health and respiratory diseases. Here, we review the recent evidence of the correlations and underlying mechanisms of the relationship between the gut microbiota and common respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), lung cancer, and respiratory infection, and probiotic development as a therapeutic intervention for these diseases.
Collapse
|
48
|
Yang K, Dong W. Perspectives on Probiotics and Bronchopulmonary Dysplasia. Front Pediatr 2020; 8:570247. [PMID: 33194897 PMCID: PMC7649774 DOI: 10.3389/fped.2020.570247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease of preterm infants, associated with high morbidity and hospitalization expenses. With the revolutionary advances in microbiological analysis technology, increasing evidence indicates that children with BPD are affected by lung microbiota dysbiosis, which may be related to the illness occurrence and progression. However, dysbiosis treatment in BPD patients has not been fully investigated. Probiotics are living microorganisms known to improve human health for their anti-inflammatory and anti-tumor effects, and particularly by balancing gut microbiota composition, which promotes gut-lung axis recovery. The aim of the present review is to examine current evidence of lung microbiota dysbiosis and explore potential applications of probiotics in BPD, which may provide new insights into treatment strategies of this disease.
Collapse
Affiliation(s)
- Kun Yang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|