1
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
2
|
Fallah A, Sedighian H, Kachuei R, Fooladi AAI. Human microbiome in post-acute COVID-19 syndrome (PACS). CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100324. [PMID: 39717208 PMCID: PMC11665312 DOI: 10.1016/j.crmicr.2024.100324] [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] [Indexed: 12/25/2024] Open
Abstract
The global COVID-19 pandemic, which began in 2019, is still ongoing. SARS-CoV-2, also known as the severe acute respiratory syndrome coronavirus 2, is the causative agent. Diarrhea, nausea, and vomiting are common GI symptoms observed in a significant number of COVID-19 patients. Additionally, the respiratory and GI tracts express high level of transmembrane protease serine 2 (TMPRSS2) and angiotensin-converting enzyme-2 (ACE2), making them primary sites for human microbiota and targets for SARS-CoV-2 infection. A growing body of research indicates that individuals with COVID-19 and post-acute COVID-19 syndrome (PACS) exhibit considerable alterations in their microbiome. In various human disorders, including diabetes, obesity, cancer, ulcerative colitis, Crohn's disease, and several viral infections, the microbiota play a significant immunomodulatory role. In this review, we investigate the potential therapeutic implications of the interactions between host microbiota and COVID-19. Microbiota-derived metabolites and components serve as primary mediators of microbiota-host interactions, influencing host immunity. We discuss the various mechanisms through which these metabolites or components produced by the microbiota impact the host's immune response to SARS-CoV-2 infection. Additionally, we address confounding factors in microbiome studies. Finally, we examine and discuss about a range of potential microbiota-based prophylactic measures and treatments for COVID-19 and PACS, as well as their effects on clinical outcomes and disease severity.
Collapse
Affiliation(s)
- Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Villanueva BHA, Huang HY, Tyan YC, Lin PJ, Li CW, Minh H, Tayo LL, Chuang KP. Immune mRNA Expression and Fecal Microbiome Composition Change Induced by Djulis ( Chenopodium formosanum Koidz.) Supplementation in Aged Mice: A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1545. [PMID: 39336586 PMCID: PMC11434560 DOI: 10.3390/medicina60091545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: The aging process has always been associated with a higher susceptibility to chronic inflammatory lung diseases. Several studies have demonstrated the gut microbiome's influence on the lungs through cross-talk or the gut-lungs axis maintaining nutrient-rich microenvironments. Taiwan djulis (Chenopodium formosanum Koidz.) provides antioxidant and anti-inflammatory characteristics that could modulate the gut microbiome. This could induce the gut-lung axis through microbial cross-talk, thus favoring the modulation of lung inflammation. Materials and Methods: Here, we investigate the immune mRNA expression in the spleen, fecal microbiome composition, and hyperplasia of the bronchial epithelium in aged 2-year-old BALB/c mice after 60 days of supplementation of djulis. Results: The pro-inflammatory cytokines IFN-γ, TNF-α, and IL-1β, T; cells CD4 and CD8; and TLRs TLR3, TLR4, TLR5, TLR7, TLR8, and TLR9 were reduced in their mRNA expression levels, while the anti-inflammatory cytokines IL-2, IL-4, and IL-10 were highly expressed in the C. formosanum-treated group. Interestingly, the fecal microbiome composition analysis indicated higher diversity in the C. formosanum-treated group and the presence of butyrate-producing bacteria that are beneficial in the gut microbiome. The histopathology showed reduced hyperplasia of the bronchial epithelium based on the degree of lesions. Conclusions: Our findings suggest that Taiwan djulis can modulate the gut microbiome, leading to microbial cross-talk; reducing the mRNA expression of pro-inflammatory cytokines, T cells, and TLRs; and increasing anti-inflammatory cytokines in the spleen, as cytokines migrate in the lungs, preventing lung inflammation damage in aged mice or the gut-lung axis. Thus, Taiwan djulis could be considered a beneficial dietary component for the older adult population. The major limitation includes a lack of protein validation of cytokines and TLRs and quantification of the T cell population in the spleen as a marker of the gut-lung axis.
Collapse
Affiliation(s)
- Brian Harvey Avanceña Villanueva
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Huai-Ying Huang
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Demin Veterinary Hospital, Kaohsiung 811, Taiwan
- Department of Pet Care and Grooming, Ta Jen University, Pingtung 912, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Tropical Medicine and Infectious Disease Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Pei-Ju Lin
- Livestock Disease Control Center of Chiayi County, Chiayi 612, Taiwan
- Department of Veterinary Medicine, National Chiayi University, Chiayi 600, Taiwan
| | | | - Hoang Minh
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi 100000, Vietnam
| | - Lemmuel L Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines
- Department of Biology, School of Medicine and Health Sciences, Mapúa University, Makati City 1200, Philippines
| | - Kuo-Pin Chuang
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Companion Animal Research Center, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| |
Collapse
|
4
|
Seo H, Yoon Y, Kim S, Ghorbanian F, Tajdozian H, Jo S, Barman I, Lee S, Lee Y, Rahim MA, Hossain MS, Lee S, Song HY. Anti-tuberculosis effect of microbiome therapeutic PMC205 in extensively drug-resistant pulmonary tuberculosis in vivo. Int J Antimicrob Agents 2024; 64:107274. [PMID: 39002701 DOI: 10.1016/j.ijantimicag.2024.107274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 06/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis, and the increase in antibiotic resistance threatens humankind. Therefore, there is an urgent need to develop new anti-tuberculosis drugs that can overcome the limitations of existing drugs. Here, we report the anti-tuberculosis effect of microbiome therapeutic PMC205, a strain of Bacillus subtilis. METHODS The anti-tuberculosis activity of probiotics was evaluated in mouse models of lethal and latent pulmonary tuberculosis induced by high or low-dose infection of the extensively drug-resistant strain. Probiotics were administered by inhalation, and the burden of M. tuberculosis in the lungs, along with mortality and clinical observations, were monitored for 12 weeks and 8 months, respectively. For an in-depth understanding, analysis of the microbiome and inflammatory profile of the lung microenvironment and induction of autophagy in vitro were explored. RESULTS After inhalation administration of PMC205 for 3 months, the survival rate was 100%, unlike all deaths in the saline-treated group, and the burden of M. tuberculosis in the lungs was reduced by log 1.3 in the 8-month latent tuberculosis model. Moreover, PMC205 induced recovery of disrupted lung microflora, increased butyric acid, and suppressed excessive inflammation. It also promoted autophagy. CONCLUSIONS These results confirm PMC205's anti-tuberculosis effect, suggesting that it can be developed as an adjuvant to current antibiotic therapy to solve the drug-resistant tuberculosis problem.
Collapse
Affiliation(s)
- Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Fatemeh Ghorbanian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Hanieh Tajdozian
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Sujin Jo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Soyeon Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea
| | - Saebim Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, Chungnam, Republic of Korea; Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan-si, Chungnam, Republic of Korea.
| |
Collapse
|
5
|
Giancola ML, Fontana A, Panebianco C, Mazzarelli A, Beccacece A, De Marco P, Cocomazzi G, De Giuli C, Grassi G, Fontana C, Baldini GM, Contu V, Copetti M, Perri F, Nicastri E, Pazienza V. Efficacy of a Multistrain Synbiotic Treatment in Acute and Post-Acute COVID-19 Patients: A Double-Blind, Placebo-Controlled Randomized Trial. Microorganisms 2024; 12:1443. [PMID: 39065211 PMCID: PMC11279369 DOI: 10.3390/microorganisms12071443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIMS Several studies reported the effect of COVID-19 on inducing gut dysbiosis, which is also correlated with disease severity. This study aims to investigate the effect of a nutraceutical formula on the shift of microbiota profiles and, secondly, on the clinical-pathological parameters of acute and post-acute COVID-19 patients. METHODS In this randomised, double-blind, placebo-controlled trial conducted at National Institute for Infectious diseases (INMI) Lazzaro Spallanzani (Italy), 52 patients were randomly assigned (1:1) to receive a multistrain synbiotic formula (Kebirah®) or placebo orally for 35 days at COVID-19 diagnosis. Health professionals, investigators, and patients were masked to group assignments. The V3-V4 hypervariable region of 16S rRNA gene sequencing was employed to study the gut microbiota composition in the two groups. RESULTS Supplementation with Kebirah® prevented the decrease in the Shannon diversity index of gut microbiota, which was instead observed in patients receiving the placebo. In addition, decreases in lymphocyte count and haemoglobin levels were observed only in the placebo group and not in the treated group, which was also characterised by an amelioration of the gut microbial profile, with an enrichment in beneficial bacteria and a preservation of species diversity. CONCLUSIONS Our data suggest that modulating the gut microbiota in acute disease through administration of a specific symbiotic formula could be a useful strategy in the frame of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Maria Letizia Giancola
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Andrea Fontana
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Concetta Panebianco
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Antonio Mazzarelli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Alessia Beccacece
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Patrizia De Marco
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giovanna Cocomazzi
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Chiara De Giuli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Germana Grassi
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Carla Fontana
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giorgio Maria Baldini
- AO Consorziale Policlinico di Bari, Università Aldo Moro di Bari, 70121 Bari, BA, Italy;
| | - Viviana Contu
- Integrative Medicine Unit, Humanitas Gradenigo, Corso Regina Margherita 8/10, 10153 Torino, TO, Italy;
| | - Massimiliano Copetti
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Francesco Perri
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Emanuele Nicastri
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Valerio Pazienza
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| |
Collapse
|
6
|
Shen HT, Fang YT, Tsai WH, Chou CH, Huang MS, Yeh YT, Wu JT, Huang CH, Wang BY, Chang WW. A Lactobacillus Combination Ameliorates Lung Inflammation in an Elastase/LPS-induced Mouse Model of Chronic Obstructive Pulmonary Disease. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10300-9. [PMID: 38865030 DOI: 10.1007/s12602-024-10300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is the world's leading lung disease and lacks effective and specific clinical strategies. Probiotics are increasingly used to support the improvement of the course of inflammatory diseases. In this study, we evaluated the potential of a lactic acid bacteria (LAB) combination containing Limosilactobacillus reuteri GMNL-89 and Lacticaseibacillus paracasei GMNL-133 to decrease lung inflammation and emphysema in a COPD mouse model. This model was induced by intranasal stimulation with elastase and LPS for 4 weeks, followed by 2 weeks of oral LAB administration. The results showed that the LAB combination decreased lung emphysema and reduced inflammatory cytokines (IL-1β, IL-6, TNF-α) in the lung tissue of COPD mice. Microbiome analysis revealed that Bifidobacterium and Akkermansia muciniphila, reduced in the gut of COPD mice, could be restored after LAB treatment. Microbial α-diversity in the lungs decreased in COPD mice but was reversed after LAB administration, which also increased the relative abundance of Candidatus arthromitus in the gut and decreased Burkholderia in the lungs. Furthermore, LAB-treated COPD mice exhibited increased levels of short-chain fatty acids, specifically acetic acid and propionic acid, in the cecum. Additionally, pulmonary emphysema and inflammation negatively correlated with C. arthromitus and Adlercreutzia levels. In conclusion, the combination of L. reuteri GMNL-89 and L. paracasei GMNL-133 demonstrates beneficial effects on pulmonary emphysema and inflammation in experimental COPD mice, correlating with changes in gut and lung microbiota, and providing a potential strategy for future adjuvant therapy.
Collapse
Affiliation(s)
- Huan-Ting Shen
- Department of Pulmonary Medicine, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 88, Sec. 1, Fengxing Rd., Tanzi Dist., Taichung City, 427003, Taiwan
| | - Yi-Ting Fang
- Research and Development Department, GenMont Biotech Incorporation, No.8, Nanke 7th Rd., Shanhua Dist., Tainan City, 741014, Taiwan
| | - Wan-Hua Tsai
- Research and Development Department, GenMont Biotech Incorporation, No.8, Nanke 7th Rd., Shanhua Dist., Tainan City, 741014, Taiwan
| | - Chia-Hsuan Chou
- Research and Development Department, GenMont Biotech Incorporation, No.8, Nanke 7th Rd., Shanhua Dist., Tainan City, 741014, Taiwan
| | - Ming-Shyan Huang
- Division of Respiratory and Chest Medicine, Department of Internal Medicine, E-Da Cancer Hospital, No. 1, Yida Rd, Yanchao Dist, Kaohsiung City, 824005, Taiwan
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, No. 151, Jinxue Rd., Daliao Dist., Kaohsiung City, 831301, Taiwan
| | - Jiun-Ting Wu
- Division of Respiratory and Chest Medicine, Department of Internal Medicine, E-Da Cancer Hospital, No. 1, Yida Rd, Yanchao Dist, Kaohsiung City, 824005, Taiwan
| | - Cheng-Hsieh Huang
- Aging and Disease Prevention Research Center, Fooyin University, No. 151, Jinxue Rd., Daliao Dist., Kaohsiung City, 831301, Taiwan
| | - Bing-Yen Wang
- Division of Thoracic Surgery, Department of Surgery, Changhua Christian Hospital, No. 135, Nanhsiao Street, Changhua County, 500209, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, No. 145, Xingda Rd., South Dist., Taichung City, 402202, Taiwan.
| | - Wen-Wei Chang
- Department of Biomedical Sciences, Chung Shan Medical University, No.110, Sec.1, Jianguo N.Rd, Taichung City, 402306, Taiwan.
- Department of Medical Research, Chung Shan Medical University Hospital, No.110, Sec.1, Jianguo N.Rd, Taichung City, 402306, Taiwan.
| |
Collapse
|
7
|
Cheng ZX, Zhang J. Exploring the Role of Gut-Lung Interactions in COPD Pathogenesis: A Comprehensive Review on Microbiota Characteristics and Inflammation Modulation. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2024; 11:311-325. [PMID: 38563747 PMCID: PMC11216226 DOI: 10.15326/jcopdf.2023.0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a paramount contributor to global morbidity and mortality. Over the past decade, the concept of the "gut-lung axis" has emerged, offering a lens through which to examine the intricate interplay between the host, microbiome, and respiratory diseases, including COPD. An expanding body of evidence underscores that the composition of both the gastrointestinal and respiratory microbiome deviates in COPD patients compared to healthy individuals, leading to distinct host immune responses and clinical manifestations. The objective of this review is to provide a concise overview of the role both gut and respiratory microbiome play in the development of COPD. This was accomplished by compiling current literature on the microbiome profile in stable and exacerbated cases of COPD, as well as exploring the biological mechanisms through a discussion of relevant experiments conducted on murine models. Hallmark characteristics of the microbial profile in COPD encompass reduced Prevotella species in the respiratory microbiome, culminating in a loss of anti-inflammatory protection, and diminished Bacteroidetes in the gut microbiome, leading to a decrease in protective short-chain fatty acids. The proliferation of Proteobacteria, particularly the Haemophilus species, Moraxellaspecies, and Pseudomonas species contribute to COPD pathologies via recognition of proinflammatory lipopolysaccharide via Toll-like receptors. As a consequence, deteriorated pulmonary function, enhanced severity, increased onset of exacerbations, and elevated mortality were observed.
Collapse
Affiliation(s)
- Zi-Xuan Cheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai, China
- *PhD candidate
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai, China
| |
Collapse
|
8
|
Chen Z, Wang Z, Ma H, Bao H, Jiang T, Yang T, Ma S. Immune cells mediated the causal relationship between the gut microbiota and lung cancer: a Mendelian randomization study. Front Microbiol 2024; 15:1390722. [PMID: 38765682 PMCID: PMC11099228 DOI: 10.3389/fmicb.2024.1390722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction The gut microbiota (GM) influences the occurrence and progression of lung cancer (LC), with potential involvement of immune cells (IC). We aimed to investigate the causal impact of GM on LC and identify potential immune cell mediators. Methods The utilized data for the Genome-Wide Association Studies (GWAS) were summarized as follows: gut microbiota data from the Dutch Microbiome Project (DMP) (N = 7,738), lung cancer data from the Transdisciplinary Research in Cancer of the Lung (TRICL) and International Lung Cancer Consortium (ILCCO) (Ncase = 29,266, Ncontrol = 56,450) included four types of cancer: NSCLC, LUAD, LUSC, and SCLC, and immune cell data from European populations (N = 3,757). We employed bi-directional two-sample univariable Mendelian randomization (UVMR), multivariable Mendelian randomization (MVMR), and mediation analysis to assess the causal relationship between GM and LC and potential immune cell mediators. Results Bi-directional UVMR analysis revealed that 24 gut microbiota species can affect LC, while LC can affect the abundance of 17 gut microbiota species. Mediation analysis demonstrated that six immune cells mediated the causal relationships of seven gut microbiota species on LC: "CCR7 on naive CD8+ T cell" mediated the causal relationship between s_Alistipes_putredinis and LUAD, with a mediation proportion of 9.5% and P = 0.018; "IgD- CD27- B cell %lymphocyte" mediated the causal relationships between g_Gordonibacter and s_Gordonibacter_pamelaeae with LUSC, with mediation proportions of 11.8% and 11.9%, respectively and P = 0.029; "CD20- CD38- B cell %lymphocyte" mediated the causal relationship between s_Bacteroides_clarus and SCLC, with a mediation proportion of 13.8% and P = 0.005; "CD20 on IgD+ CD38- unswitched memory B cell" mediated the causal relationship between s_Streptococcus_thermophilus and SCLC, with a mediation proportion of 14.1% and P = 0.023; "HLA DR on CD14- CD16+ monocyte" mediated the causal relationship between s_Bifidobacterium_bifidum and SCLC, with a mediation proportion of 8.7% and P = 0.012; "CD45 on Granulocytic Myeloid-Derived Suppressor Cells" mediated the causal relationship between f_Lactobacillaceae and SCLC, with a mediation proportion of 4.0% and P = 0.021. Conclusion This Mendelian randomization study identified several specific gut microbiotas that exhibit causal relationships with lung cancer and potentially mediate immune cells.
Collapse
Affiliation(s)
- Zhiting Chen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Ma
- Department of Obstetrics and Gynecology, Shijing People’s Hospital, Guangzhou, China
| | - Hejing Bao
- Department of Oncology, Panyu Central Hospital, Guangzhou, China
| | - Ting Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shudong Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Yousef M, Rob M, Varghese S, Rao S, Zamir F, Paul P, Chaari A. The effect of microbiome therapy on COVID-19-induced gut dysbiosis: A narrative and systematic review. Life Sci 2024; 342:122535. [PMID: 38408636 DOI: 10.1016/j.lfs.2024.122535] [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: 12/15/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
AIMS Emerging evidence highlights the role of COVID-19 in instigating gut dysbiosis, with repercussions on disease severity and bidirectional gut-organ communication involving the lung, heart, brain, and liver. This study aims to evaluate the efficacy of probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT) in addressing gut dysbiosis associated with COVID-19, as well as their impact on related disease severity and clinical outcomes. MATERIALS AND METHODS We systematically review 27 studies exploring the efficacy of different microbiome-modulating therapies: probiotics, prebiotics, synbiotics, and fecal microbiota transplantation as potential interventions for COVID-19. KEY FINDINGS The probiotics and synbiotics investigated encompassed a spectrum of eight bacterial and fungal genera, namely Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, Pediococcus, Bacillus, Saccharomyces, and Kluyveromyces. Noteworthy prebiotics employed in these studies included chestnut tannin, galactooligosaccharides, fructooligosaccharides, xylooligosaccharide, and resistant dextrin. The majority of the investigated biotics exhibited positive effects on COVID-19 patients, manifesting in symptom alleviation, inflammation reduction, and notable decreases in mortality rates. Five studies reported death rates, showing an average mortality ranging from 0 % to 11 % in the intervention groups, as compared to 3 % to 30 % in the control groups. Specifically, probiotics, prebiotics, and synbiotics demonstrated efficacy in diminishing the duration and severity of symptoms while significantly accelerating viral and symptomatic remission. FMT emerged as a particularly effective strategy, successfully restoring gut microbiota and ameliorating gastrointestinal disorders. SIGNIFICANCE The insights gleaned from this review significantly contribute to our broader comprehension of the therapeutic potential of biotics in addressing COVID-19-related gut dysbiosis and mitigating secondary multi-organ complications.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Mlaak Rob
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Sanish Varghese
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Shrinidhi Rao
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Fahad Zamir
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar.
| |
Collapse
|
10
|
Qu X, Song Y, Li Q, Xu Q, Li Y, Zhang H, Cheng X, Mackay CR, Wang Q, Liu W. Indole-3-acetic acid ameliorates dextran sulfate sodium-induced colitis via the ERK signaling pathway. Arch Pharm Res 2024; 47:288-299. [PMID: 38489148 DOI: 10.1007/s12272-024-01488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Microbiota-derived catabolism of nutrients is closely related to ulcerative colitis (UC). The level of indole-3-acetic acid (IAA), a microbiota-dependent metabolite of tryptophan, was decreased significantly in the feces of UC patients. Thus supplementation with IAA could be a potential therapeutic method for ameliorating colitis. In this work, the protective effect of supplementation with IAA on dextran sulfate sodium (DSS)-induced colitis was evaluated, and the underlying mechanism was elucidated. The results indicated that the administration of IAA significantly relieved DSS-induced weight loss, reduced the disease activity index (DAI), restored colon length, alleviated intestinal injury, and improved the intestinal tight junction barrier. Furthermore, IAA inhibited intestinal inflammation by reducing the expression of proinflammatory cytokines and promoting the production of IL-10 and TGF-β1. In addition, the ERK signaling pathway is an important mediator of various physiological processes including inflammatory responses and is closely associated with the expression of IL-10. Notably, IAA treatment induced the activation of extracellular signal-regulated kinase (ERK), which is involved in the progression of colitis, while the ERK inhibitor U0126 attenuated the beneficial effects of IAA. In summary, IAA could attenuate the clinical symptoms of colitis, and the ERK signaling pathway was involved in the underlying mechanism. Supplementation with IAA could be a potential option for preventing or ameliorating UC.
Collapse
Affiliation(s)
- Xinyan Qu
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Yingying Song
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Qingjun Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qi Xu
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Yanru Li
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Huimin Zhang
- Shandong Academy of Chinese Medicine, Jinan, China
| | - Xuemei Cheng
- Shandong Center for Disease Control and Prevention, Jinan, China
| | - Charles R Mackay
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Quanbo Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.
| | - Wei Liu
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China.
| |
Collapse
|
11
|
Kim HS, Kim B, Holzapfel WH, Kang H. Lactiplantibacillusplantarum APsulloc331261 (GTB1 ™) promotes butyrate production to suppress mucin hypersecretion in a murine allergic airway inflammation model. Front Microbiol 2024; 14:1292266. [PMID: 38449878 PMCID: PMC10915089 DOI: 10.3389/fmicb.2023.1292266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/15/2023] [Indexed: 03/08/2024] Open
Abstract
Introduction Allergic airway diseases are one of the serious health problems in worldwide and allergic airway inflammation is a prerequisite led to the exacerbated situation such as mucus hypersecretion, epithelial barrier damage and microbiota dysbiosis. Because of side effects and low efficiencies of current therapeutics, the need for novel alternatives has been urged. Probiotics in which have diverse and beneficial modulatory effects have been applied to the airway inflammation model and the underlying mechanism needs to be investigated. Methods We aimed to evaluate whether our target strain, Lactiplantibacillus plantarum APsulloc331261 (GTB1TM) isolated from green tea, can ameliorate allergic airway inflammation in mice and to figure out the mechanism. We induced allergic airway inflammation to mice by ovalbumin (OVA) and administered GTB1 orally and the immune and epithelial barrier markers were assessed. The gut metabolite and microbiota were also analysed, and the in vitro cell-line experiment was introduced to confirm the hypothesis of the study. Results GTB1 ameliorated type 2 inflammation and suppressed mucin hypersecretion with the inhibition of MUC5AC in inflamed mice. Moreover, GTB1 increased the butyrate production and the relative abundance of butyrate producer, Clostridium cluster IV. We assumed that butyrate may have a potential role and investigated the effect of butyrate in mucin regulation via human airway epithelial cell line, A549. Butyrate significantly reduced the gene expression of MUC5AC in A549 cells suggesting its regulatory role in mucus production. Conclusion Therefore, our study demonstrates that the oral administration of GTB1 can ameliorate allergic airway inflammation and mucin hypersecretion by butyrate production.
Collapse
Affiliation(s)
- Hye-Shin Kim
- Department of Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- HEM Pharma, Pohang, Republic of Korea
| | - Bobae Kim
- HEM Pharma, Pohang, Republic of Korea
| | - Wilhelm H. Holzapfel
- Department of Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- HEM Pharma, Pohang, Republic of Korea
| | | |
Collapse
|
12
|
Scheithauer TPM, Montijn RC, Mieremet A. Gut microbe-host interactions in post-COVID syndrome: a debilitating or restorative partnership? Gut Microbes 2024; 16:2402544. [PMID: 39287023 PMCID: PMC11409505 DOI: 10.1080/19490976.2024.2402544] [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: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Post-COVID syndrome (PCS) patients have reported a wide range of symptoms, including fatigue, shortness of breath, and diarrhea. Particularly, the presence of gastrointestinal symptoms has led to the hypothesis that the gut microbiome is involved in the development and severity of PCS. The objective of this review is to provide an overview of the role of the gut microbiome in PCS by describing the microbial composition and microbial metabolites in COVID-19 and PCS. Moreover, host-microbe interactions via the microbiota-gut-brain (MGB) and the microbiota-gut-lung (MGL) axes are described. Furthermore, we explore the potential of therapeutically targeting the gut microbiome to support the recovery of PCS by reviewing preclinical model systems and clinical studies. Overall, current studies provide evidence that the gut microbiota is affected in PCS; however, diversity in symptoms and highly individual microbiota compositions suggest the need for personalized medicine. Gut-targeted therapies, including treatments with pre- and probiotics, have the potential to improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Torsten P M Scheithauer
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Roy C Montijn
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Arnout Mieremet
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
13
|
Sapra L, Saini C, Das S, Mishra PK, Singh A, Mridha AR, Yadav PK, Srivastava RK. Lactobacillus rhamnosus (LR) ameliorates pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) via targeting neutrophils. Clin Immunol 2024; 258:109872. [PMID: 38113963 DOI: 10.1016/j.clim.2023.109872] [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: 08/11/2023] [Revised: 11/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Pulmonary and extrapulmonary acute respiratory distress syndrome (ARDS) is a life-threatening respiratory failure associated with high mortality. Despite progress in our understanding of the pathological mechanism causing the crippling illness, there are currently no targeted pharmaceutical treatments available for it. Recent discoveries have emphasized the existence of a potential nexus between gut and lung health fueling novel approaches including probiotics for the treatment of ARDS. We thus investigated the prophylactic-potential of Lactobacillus rhamnosus-(LR) in lipopolysaccharide (LPS)-induced pulmonary and cecal ligation puncture (CLP) induced extrapulmonary ARDS mice. Our in-vivo findings revealed that pretreatment with LR significantly ameliorated vascular-permeability (edema) of the lungs via modulating the neutrophils along with significantly reducing the expression of inflammatory-cytokines in the BALF, lungs and serum in both pulmonary and extrapulmonary mice-models. Interestingly, our ex-vivo immunofluorescence and flow cytometric data suggested that mechanistically LR via short chain fatty acids (butyrate being the most potent and efficient in ameliorating the pathophysiology of both pulmonary and extra-pulmonary ARDS) targets the phagocytic and neutrophils extracellular traps (NETs) releasing potential of neutrophils. Moreover, our in-vivo data further corroborated our ex-vivo findings and suggested that butyrate exhibits enhanced potential in ameliorating the pathophysiology of ARDS via reducing the infiltration of neutrophils into the lungs. Altogether, our study establishes the prophylactic role of LR and its associated metabolites in the prevention and management of both pulmonary and extrapulmonary ARDS via targeting neutrophils.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sneha Das
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Anurag Singh
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Asit R Mridha
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Pardeep K Yadav
- Central Animal Facility, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
14
|
Lim EY, Song EJ, Shin HS. Gut Microbiome as a Possible Cause of Occurrence and Therapeutic Target in Chronic Obstructive Pulmonary Disease. J Microbiol Biotechnol 2023; 33:1111-1118. [PMID: 37164760 PMCID: PMC10580882 DOI: 10.4014/jmb.2301.01033] [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: 01/26/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/12/2023]
Abstract
As a long-term condition that affects the airways and lungs, chronic obstructive pulmonary disease (COPD) is characterized by inflammation, emphysema, breathlessness, chronic cough, and sputum production. Currently, the bronchodilators and anti-inflammatory drugs prescribed for COPD are mostly off-target, warranting new disease management strategies. Accumulating research has revealed the gut-lung axis to be a bidirectional communication system. Cigarette smoke, a major exacerbating factor in COPD and lung inflammation, affects gut microbiota composition and diversity, causing gut microbiota dysbiosis, a condition that has recently been described in COPD patients and animal models. For this review, we focused on the gut-lung axis, which is influenced by gut microbial metabolites, bacterial translocation, and immune cell modulation. Further, we have summarized the findings of preclinical and clinical studies on the association between gut microbiota and COPD to provide a basis for using gut microbiota in therapeutic strategies against COPD. Our review also proposes that further research on probiotics, prebiotics, short-chain fatty acids, and fecal microbiota transplantation could assist therapeutic approaches targeting the gut microbiota to alleviate COPD.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Eun-Ji Song
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Hee Soon Shin
- Food Functionality Research Division, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
15
|
Li Y, Wang K, Zhang Y, Yang J, Wu Y, Zhao M. Revealing a causal relationship between gut microbiota and lung cancer: a Mendelian randomization study. Front Cell Infect Microbiol 2023; 13:1200299. [PMID: 37829610 PMCID: PMC10565354 DOI: 10.3389/fcimb.2023.1200299] [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: 04/04/2023] [Accepted: 08/18/2023] [Indexed: 10/14/2023] Open
Abstract
Background The gut microbiota has been found to be associated with the risk of lung cancer. However, its causal relationship with various types of lung cancer remains unclear. Methods We conducted a Mendelian randomization (MR) study using the largest genome-wide association analysis of gut microbiota data to date from the MiBioGen consortium, with pooled statistics for various types of lung cancer from the Transdisciplinary Research in Cancer of the Lung, the International Lung Cancer Consortium, and FinnGen Consortium R7 release data. Inverse variance weighted, weighted model, MR-Egger regression, and weighted median were adapted to assess the causal relationship between gut microbiota and various types of lung cancer. Sensitivity analysis was used to test for the presence of pleiotropy and heterogeneity in instrumental variables. A reverse MR analysis was performed on these bacteria to determine their potential role in causing lung cancer. A reverse MR analysis was performed on these bacteria to determine their potential role in causing lung cancer. Multivariable Mendelian randomization (MVMR) was conducted to assess the direct causal impact of gut microbiota on the risk of various types of lung cancer. Results Using IVW as the primary analytical method, we identified a total of 40 groups of gut microbiota with potential causal associations with various subtypes of lung cancer, of which 10 were associated with lung cancer, 10 with lung adenocarcinoma, 9 with squamous cell lung cancer, and 11 groups of bacteria associated with small cell lung cancer. After performing FDR correction, we further found that there was still a significant causal relationship between Peptococcaceae and lung adenocarcinoma. Sensitivity analyses demonstrated the robustness of these results, with no heterogeneity or pleiotropy found. Conclusions Our results confirm a causal relationship between specific gut microbiota and lung cancer, providing new insights into the role of gut microbiota in mediating the development of lung cancer.
Collapse
Affiliation(s)
- Yingchen Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuchong Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jitao Yang
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Wu
- Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang, China
| | - Mingfang Zhao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Yu H, Yuan L, Yan Z, Zhou M, Ye J, Wu K, Chen W, Chen R, Xia N, Guan Y, Zhu H. Butyrate Protects against SARS-CoV-2-Induced Tissue Damage in Golden Hamsters. Int J Mol Sci 2023; 24:14191. [PMID: 37762492 PMCID: PMC10532055 DOI: 10.3390/ijms241814191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Butyrate, produced by gut microbe during dietary fiber fermentation, has anti-inflammatory and antioxidant effects on chronic inflammation diseases, yet it remains to be explored whether butyrate has protective effects against viral infections. Here, we demonstrated that butyrate alleviated tissue injury in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected golden hamsters supplemented with butyrate before and during the infection. Butyrate-treated hamsters showed augmentation of type I interferon (IFN) response and activation of endothelial cells without exaggerated inflammation. In addition, butyrate regulated redox homeostasis by enhancing the activity of superoxide dismutase (SOD) to inhibit excessive apoptotic cell death. Therefore, butyrate exhibited effective prevention against SARS-CoV-2 by upregulating antiviral immune responses and promoting cell survival.
Collapse
Affiliation(s)
- Huan Yu
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhigang Yan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Ming Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jianghui Ye
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kun Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wenjia Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Rirong Chen
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yi Guan
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- EKIH (Gewuzhikang) Advanced Pathogen Research Institute, Futian District, Shenzhen 518045, China
| | - Huachen Zhu
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases (Key Laboratory of Ministry of Education), Joint Institute of Virology (Shantou University/The University of Hong Kong), Shantou University Medical College, Shantou 515063, China
- State Key Laboratory of Emerging Infectious Diseases (SKLEID), School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- EKIH (Gewuzhikang) Advanced Pathogen Research Institute, Futian District, Shenzhen 518045, China
| |
Collapse
|
17
|
Martín R, Rios-Covian D, Huillet E, Auger S, Khazaal S, Bermúdez-Humarán LG, Sokol H, Chatel JM, Langella P. Faecalibacterium: a bacterial genus with promising human health applications. FEMS Microbiol Rev 2023; 47:fuad039. [PMID: 37451743 PMCID: PMC10410495 DOI: 10.1093/femsre/fuad039] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/08/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
In humans, many diseases are associated with alterations in gut microbiota, namely increases or decreases in the abundance of specific bacterial groups. One example is the genus Faecalibacterium. Numerous studies have underscored that low levels of Faecalibacterium are correlated with inflammatory conditions, with inflammatory bowel disease (IBD) in the forefront. Its representation is also diminished in the case of several diseases, including colorectal cancer (CRC), dermatitis, and depression. Additionally, the relative presence of this genus is considered to reflect, at least in part, intestinal health status because Faecalibacterium is frequently present at reduced levels in individuals with gastrointestinal diseases or disorders. In this review, we first thoroughly describe updates to the taxonomy of Faecalibacterium, which has transformed a single-species taxon to a multispecies taxon over the last decade. We then explore the links discovered between Faecalibacterium abundance and various diseases since the first IBD-focused studies were published. Next, we examine current available strategies for modulating Faecalibacterium levels in the gut. Finally, we summarize the mechanisms underlying the beneficial effects that have been attributed to this genus. Together, epidemiological and experimental data strongly support the use of Faecalibacterium as a next-generation probiotic (NGP) or live biotherapeutic product (LBP).
Collapse
Affiliation(s)
- Rebeca Martín
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - David Rios-Covian
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Eugénie Huillet
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Sandrine Auger
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Sarah Khazaal
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Harry Sokol
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012 Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, F-75012, Paris, France
| | - Jean-Marc Chatel
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Philippe Langella
- Paris-Saclay University, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| |
Collapse
|
18
|
Lo Russo G, Prelaj A, Dolezal J, Beninato T, Agnelli L, Triulzi T, Fabbri A, Lorenzini D, Ferrara R, Brambilla M, Occhipinti M, Mazzeo L, Provenzano L, Spagnoletti A, Viscardi G, Sgambelluri F, Brich S, Miskovic V, Pedrocchi ALG, Trovo' F, Manglaviti S, Giani C, Ambrosini P, Leporati R, Franza A, McCulloch J, Torelli T, Anichini A, Mortarini R, Trinchieri G, Pruneri G, Torri V, De Braud F, Proto C, Ganzinelli M, Garassino MC. PEOPLE (NTC03447678), a phase II trial to test pembrolizumab as first-line treatment in patients with advanced NSCLC with PD-L1 <50%: a multiomics analysis. J Immunother Cancer 2023; 11:e006833. [PMID: 37286305 PMCID: PMC10254948 DOI: 10.1136/jitc-2023-006833] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Chemoimmunotherapy represents the standard of care for patients with advanced non-small cell lung cancer (NSCLC) and programmed death-ligand 1 (PD-L1) <50%. Although single-agent pembrolizumab has also demonstrated some activity in this setting, no reliable biomarkers yet exist for selecting patients likely to respond to single-agent immunotherapy. The main purpose of the study was to identify potential new biomarkers associated with progression-free-survival (PFS) within a multiomics analysis. METHODS PEOPLE (NTC03447678) was a prospective phase II trial evaluating first-line pembrolizumab in patients with advanced EGFR and ALK wild type treatment-naïve NSCLC with PD-L1 <50%. Circulating immune profiling was performed by determination of absolute cell counts with multiparametric flow cytometry on freshly isolated whole blood samples at baseline and at first radiological evaluation. Gene expression profiling was performed using nCounter PanCancer IO 360 Panel (NanoString) on baseline tissue. Gut bacterial taxonomic abundance was obtained by shotgun metagenomic sequencing of stool samples at baseline. Omics data were analyzed with sequential univariate Cox proportional hazards regression predicting PFS, with Benjamini-Hochberg multiple comparisons correction. Biological features significant with univariate analysis were analyzed with multivariate least absolute shrinkage and selection operator (LASSO). RESULTS From May 2018 to October 2020, 65 patients were enrolled. Median follow-up and PFS were 26.4 and 2.9 months, respectively. LASSO integration analysis, with an optimal lambda of 0.28, showed that peripheral blood natural killer cells/CD56dimCD16+ (HR 0.56, 0.41-0.76, p=0.006) abundance at baseline and non-classical CD14dimCD16+monocytes (HR 0.52, 0.36-0.75, p=0.004), eosinophils (CD15+CD16-) (HR 0.62, 0.44-0.89, p=0.03) and lymphocytes (HR 0.32, 0.19-0.56, p=0.001) after first radiologic evaluation correlated with favorable PFS as well as high baseline expression levels of CD244 (HR 0.74, 0.62-0.87, p=0.05) protein tyrosine phosphatase receptor type C (HR 0.55, 0.38-0.81, p=0.098) and killer cell lectin like receptor B1 (HR 0.76, 0.66-0.89, p=0.05). Interferon-responsive factor 9 and cartilage oligomeric matrix protein genes correlated with unfavorable PFS (HR 3.03, 1.52-6.02, p 0.08 and HR 1.22, 1.08-1.37, p=0.06, corrected). No microbiome features were selected. CONCLUSIONS This multiomics approach was able to identify immune cell subsets and expression levels of genes associated to PFS in patients with PD-L1 <50% NSCLC treated with first-line pembrolizumab. These preliminary data will be confirmed in the larger multicentric international I3LUNG trial (NCT05537922). TRIAL REGISTRATION NUMBER 2017-002841-31.
Collapse
Affiliation(s)
- Giuseppe Lo Russo
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Arsela Prelaj
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Lombardia, Italy
| | - James Dolezal
- Thoracic Oncology Program, Section of Hematology/Oncology, University of Chicago Department of Medicine, Chicago, Illinois, USA
| | - Teresa Beninato
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Luca Agnelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Lombardia, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Alessandra Fabbri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniele Lorenzini
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Ferrara
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
- Medical Oncology, Università Vita Salute San Raffaele, Milano, Lombardia, Italy
| | - Marta Brambilla
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Mario Occhipinti
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Laura Mazzeo
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Leonardo Provenzano
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Andrea Spagnoletti
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Giuseppe Viscardi
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Caserta, Campania, Italy
| | - Francesco Sgambelluri
- Department of Research, Human Tumors Immunobiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Silvia Brich
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vanja Miskovic
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Lombardia, Italy
| | | | - Francesco Trovo'
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Lombardia, Italy
| | - Sara Manglaviti
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Claudia Giani
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Paolo Ambrosini
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Rita Leporati
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Andrea Franza
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - John McCulloch
- Genetics and Microbiome Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NCI, Bethesda, Maryland, USA
| | - Tommaso Torelli
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Anichini
- Department of Research, Human Tumors Immunobiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Roberta Mortarini
- Department of Research, Human Tumors Immunobiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NIH, Bethesda, Maryland, USA
| | - Giancarlo Pruneri
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Lombardia, Italy
| | - Valter Torri
- Oncology department, Mario Negri Institute for Pharmacological Research (IRCCS), Milano, Lombardia, Italy
| | - Filippo De Braud
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Lombardia, Italy
| | - Claudia Proto
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Monica Ganzinelli
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Marina Chiara Garassino
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
- Thoracic Oncology Program, Section of Hematology/Oncology, University of Chicago Department of Medicine, Chicago, Illinois, USA
| |
Collapse
|
19
|
Wang X, Hou L, Cui M, Liu J, Wang M, Xie J. The traditional Chinese medicine and non-small cell lung cancer: from a gut microbiome perspective. Front Cell Infect Microbiol 2023; 13:1151557. [PMID: 37180438 PMCID: PMC10167031 DOI: 10.3389/fcimb.2023.1151557] [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: 01/26/2023] [Accepted: 03/29/2023] [Indexed: 05/16/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most serious diseases affecting human health today, and current research is focusing on gut flora. There is a correlation between intestinal flora imbalance and lung cancer, but the specific mechanism is not clear. Based on the "lung and large intestine being interior-exteriorly related" and the "lung-intestinal axis" theory. Here, based on the theoretical comparisons of Chinese and western medicine, we summarized the regulation of intestinal flora in NSCLC by active ingredients of traditional Chinese medicine and Chinese herbal compounds and their intervention effects, which is conducive to providing new strategies and ideas for clinical prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Xuelin Wang
- School of Food Science and Engineering (School of Biological and Pharmaceutical Sciences), Shaanxi University of Science & Technology, Xi an, China
| | - Liming Hou
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi an, China
| | - Meng Cui
- School of Food Science and Engineering (School of Biological and Pharmaceutical Sciences), Shaanxi University of Science & Technology, Xi an, China
| | - Junnan Liu
- School of Food Science and Engineering (School of Biological and Pharmaceutical Sciences), Shaanxi University of Science & Technology, Xi an, China
| | - Mengzhou Wang
- School of Food Science and Engineering (School of Biological and Pharmaceutical Sciences), Shaanxi University of Science & Technology, Xi an, China
| | - Jianwu Xie
- School of Food Science and Engineering (School of Biological and Pharmaceutical Sciences), Shaanxi University of Science & Technology, Xi an, China
| |
Collapse
|
20
|
Jin Q, Ren F, Dai D, Sun N, Qian Y, Song P. The causality between intestinal flora and allergic diseases: Insights from a bi-directional two-sample Mendelian randomization analysis. Front Immunol 2023; 14:1121273. [PMID: 36969260 PMCID: PMC10033526 DOI: 10.3389/fimmu.2023.1121273] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
Background Growing evidence shows a significant association between intestinal flora and allergic diseases, specifically atopic dermatitis (AD), allergic rhinitis (AR), and allergic asthma (AA). However, the causality has not yet been clarified. Objective We conducted a bidirectional two-sample Mendelian randomization (TSMR) analysis to study the causal relationships between intestinal flora classification and AD, AR, or AA. Materials and methods We obtained summary data of intestinal flora, AD, AR, and AA from a genome-wide association research. The inverse-variance weighted method is the primary method for analyzing causality in the TSMR analysis. Several sensitivity analyses were conducted to examine the stability of TSMR results. Reverse TSMR analysis was also performed to assess whether there was a reverse causality. Results A total of 7 bacterial taxa associated with AD, AR, and AA were identified by the current TSMR analysis. Specifically, the genus Dialister(P=0.034)and genus Prevotella(P=0.047)were associated with a higher risk of AD, whereas class Coriobacteriia (P=0.034) and its child taxon, order Coriobacteriales (P=0.034) and family Coriobacteriaceae (P=0.034), all had a protective effect on AR. In addition, the family Victivallaceae (P=0.019) was identified as a risk factor for AR. We also noticed a positive association between the genus Holdemanella (P=0.046) and AA. The reverse TSMR analysis didn't suggest any evidence of reverse causality from allergic diseases to the intestinal flora. Conclusion We confirmed the causal relationship between intestinal flora and allergic diseases and provided an innovative perspective for research on allergic diseases: targeted regulation of dysregulation of specific bacterial taxa to prevent and treat AD, AR, and AA.
Collapse
Affiliation(s)
- Qiubai Jin
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feihong Ren
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Dai
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiyun Qian
- Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Ping Song
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Ping Song,
| |
Collapse
|