1
|
Lai Y, Qiu R, Zhou J, Ren L, Qu Y, Zhang G. Fecal Microbiota Transplantation Alleviates Airway Inflammation in Asthmatic Rats by Increasing the Level of Short-Chain Fatty Acids in the Intestine. Inflammation 2025:10.1007/s10753-024-02233-w. [PMID: 39775370 DOI: 10.1007/s10753-024-02233-w] [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: 10/12/2024] [Revised: 11/22/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
Asthma is a prevalent chronic inflammatory disorder of the respiratory tract that not only manifests with respiratory symptoms but also often involves intestinal flora disorders and gastrointestinal dysfunction. Recent studies have confirmed the close relationship between the gut and lungs, known as the "gut-lung axis" theory. Fecal microbiota transplantation (FMT), a method for restoring normal intestinal flora, has shown promise in treating common gastrointestinal diseases. The "gut-lung axis" theory suggests that FMT may have significant therapeutic potential for asthma. In this study, we established an Ovalbumin (OVA)-induced rat model of asthma to investigate the protective effect of FMT on airway inflammation and the restoration of intestinal short-chain fatty acids (SCFAs), aiming to explore its underlying mechanism. Rats in the Control group underwent fecal treatment via gavage (Control-FMT, C-FMT group), while rats in the Asthma group underwent fecal treatment via gavage after asthma induction (Asthma-FMT, A-FMT group). Following a two-week period of continuous intragastric administration, various measurements were conducted to assess pulmonary function, peripheral blood neutrophil, lymphocyte, and eosinophil content, lung tissue pathology, and collagen fiber deposition in the lungs. Additionally, neutrophil and eosinophil content in bronchoalveolar lavage fluid (BALF), expression levels of Interleukin-4 (IL-4), IL-5, IL-13, IL-17, IL-33, leukotrienes (LT), thymic stromal lymphopoietin (TSLP), prostaglandin D2 (PGD2) protein and mRNA in lung tissue, and SCFAs content in stool were evaluated. In the C-FMT group, lung function significantly improved, inflammatory cell content in peripheral blood and BALF decreased, lung tissue pathology and collagen fiber deposition significantly improved, the protein and mRNA levels of lung inflammatory factors IL-4, IL-5, IL-13, IL-17, IL-33, LT, TSLP, PGD2 were significantly decreased, and SCFAs such as acetate (C2), propionate (C3), butyrate (C4), isobutyric acid (I-C4), valeric acid (C5), and isovaleric acid (I-C5) content in stool significantly increased. However, the indexes in the A-FMT group did not show significant recovery, and the treatment effect on asthma symptoms in rats was inferior to that in the C-FMT group. Asthma induced intestinal flora disorders in rats, and FMT treatment improved the inflammatory response in asthmatic rat models and corrected their intestinal SCFAs disorders. Encouraging the recovery of intestinal SCFAs may play a significant role, and beneficial bacteria present in feces may improve asthma symptoms by promoting the remodeling of intestinal flora. This experiment provides further scientific evidence supporting the "gut-lung axis" theory.
Collapse
Affiliation(s)
- Yitian Lai
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ranran Qiu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Jingying Zhou
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ling Ren
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yizhuo Qu
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Guoshan Zhang
- College of Acupuncture-Moxibustion-Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
2
|
Liu S, Yang Y, Han L, He C, Li M, Tian X, Meng J, Wang L, Zhang F. Gastrointestinal lesions of eosinophilic granulomatosis with polyangiitis: a prediction model and clinical patterns. Arthritis Res Ther 2025; 27:3. [PMID: 39755665 DOI: 10.1186/s13075-024-03467-7] [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: 08/12/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025] Open
Abstract
OBJECTIVE Severe gastrointestinal lesions are associated with a poor prognosis in eosinophilic granulomatosis with polyangiitis (EGPA). The goal of this study was to develop an effective predictive model for gastrointestinal lesions and to examine clinical patterns, associated factors, treatment, and outcomes of gastrointestinal lesions in EGPA. METHODS We retrospectively enrolled 165 EGPA patients. The independent associated factors were analyzed using multivariate logistic regression. A nomogram was conducted to quantify the predictive factors. The correlation between different organ lesions was calculated to explore the clinical patterns. RESULTS A total of 52 patients had gastrointestinal lesions, and 22 developed severe disorders. Common manifestations included abdominal pain (78%), diarrhea (40.4%), and nausea and/or vomiting (32.7%). Severe gastrointestinal lesions included hemorrhage (26.9%), ulcers (17.3%), obstruction (9.6%), and pancreatitis (5.8%). Eosinophilic tissue infiltration, weight loss, and myalgia were independently associated with gastrointestinal involvement. Patients with severe gastrointestinal lesions had a shorter duration from initial symptoms to EGPA diagnosis, less frequent asthma, and ear-nose-throat involvement, and were more likely to receive methylprednisolone pulse. Weight loss, central nervous system involvement, myalgia, and eosinophilic tissue infiltration were retained in the nomogram. An eosinophil ratio of over 19.2% identified gastrointestinal lesions. Significantly more patients with gastrointestinal involvement had a Five Factor Score ≥ 2. Five well-defined clinical models were identified, including the brain-gut pattern. CONCLUSIONS Severe gastrointestinal lesions are common in EGPA and early detection is critical. Eosinophils are an important factor associated with gastrointestinal involvement of EGPA. We developed a model to predict the risk of gastrointestinal lesions. The brain-gut pattern might deserve further investigation in EGPA.
Collapse
Affiliation(s)
- Suying Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
- Department of Rheumatology and Clinical Immunology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yunjiao Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Linna Han
- Department of Rheumatology and Clinical Immunology, the Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengmei He
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Juan Meng
- Department of Rheumatology and Clinical Immunology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, the Ministry of Education Key Laboratory, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| |
Collapse
|
3
|
Ding J, Xu J, Wu H, Li M, Xiao Y, Fu J, Zhu X, Wu N, Sun Q, Liu Y. The cross-talk between the metabolome and microbiome in a double-hit neonatal rat model of bronchopulmonary dysplasia. Genomics 2025; 117:110969. [PMID: 39615804 DOI: 10.1016/j.ygeno.2024.110969] [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: 04/09/2024] [Revised: 10/15/2024] [Accepted: 11/24/2024] [Indexed: 01/13/2025]
Abstract
Bronchopulmonary dysplasia (BPD), a chronic lung disease in preterm infants, is associated with inflammation and high oxygen exposure. However, the effects of antenatal inflammation and postnatal extended hyperoxia on the metabolome and microbiome remain unclear. In this study, pregnant rats received lipopolysaccharide or saline injections on gestational day 20 and were exposed to either 21 % or 80 % oxygen for 4 weeks post-birth. Analysis revealed an increase in Firmicutes, Proteobacteria, and Actinobacteria, with a decrease in Bacteroidetes in BPD rats. Metabolomic analysis identified 78 altered metabolites, primarily lipids, enriched in pathways including arginine biosynthesis, sphingolipid metabolism, and primary bile acid biosynthesis in BPD rats. Integration analysis revealed strong correlations between intestinal microbiota and metabolites in BPD rats. These findings underscored the impact of antenatal inflammation and prolonged postnatal hyperoxia on gut microbiota and serum metabolome, suggesting their role in BPD pathogenesis.
Collapse
Affiliation(s)
- Jing Ding
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, PR China
| | - Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, PR China
| | - Hongkun Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, PR China
| | - Mei Li
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, PR China
| | - Yihan Xiao
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, PR China
| | - Jie Fu
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, PR China
| | - Xiangyu Zhu
- Department of Laboratory Animal Unit, Peking University People's Hospital, Beijing 100044, PR China
| | - Na Wu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, PR China
| | - Qiang Sun
- Center for RNA Medicine, the Fourth Affilliated Hospital of School of Medicine, and International school of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, Zhejiang, PR China.
| | - Yaran Liu
- Institute of Medical Artificial Intelligence, Binzhou Medical College, Yantai 264003, Shandong, PR China.
| |
Collapse
|
4
|
Xia Z, Zhao X, Wang L, Huang L, Yang Y, Yin X, He L, Aga Y, Kahaer A, Yang S, Hao L, Chen C. Amelioration of Inflammation in Rats with Experimentally Induced Asthma by Spenceria ramalana Trimen Polyphenols via the PI3K/Akt Signaling Pathway. Int J Mol Sci 2024; 26:165. [PMID: 39796021 PMCID: PMC11720363 DOI: 10.3390/ijms26010165] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/13/2025] Open
Abstract
Asthma is a chronic inflammatory respiratory disease that affects millions globally and poses a serious public health challenge. Current therapeutic strategies, including corticosteroids, are constrained by variable patient responses and adverse effects. In this study, a polyphenolic extract derived from the Tibetan medicinal plant Spenceria ramalana Trimen (SRT) was employed and shown to improve experimentally (ovalbumin + cigarette smoke, OVA + CS) induced asthma in rats. Initially, the potential therapeutic mechanism of the polyphenolic components in SRT on OVA + CS-induced asthma was predicated by network pharmacology analysis. Subsequently, in vivo experiments identified that SRT polyphenols exhibit significant anti-asthmatic activities, primarily mediated by lowering inflammatory cell counts such as the WBC (white blood cell), eosinophils, and neutrophils, decreasing the expression of inflammatory cytokines (IL-4, IL-5, IL-13, and TNF-α), alleviating lung histological damage (reduced inflammation, collagen deposition, and mucus secretion), and enhancing the epithelial barrier integrity (upregulation of ZO-1, occludin, and claudin-1). Additionally, SRT polyphenols downregulated the PI3K/Akt (Phosphoinositide 3-kinase/protein kinase B) signaling pathway, improved gut microbiota disruption, and regulated fecal metabolites (glucose-6-glutamate, PS (16:0/0:0), 8-aminocaprylic acid, galactonic acid, Ascr#10, 2,3,4,5,6,7-hexahydroxyheptanoic acid, phosphodimethylethanolamine, muramic acid, 9-oxohexadeca-10e-enoic acid, and sedoheptulose) in asthmatic rats. In conclusion, SRT polyphenols exerted multifaceted protective effects against OVA + CS-induced asthma in rats, highlighting their potential value in preventing asthma via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zhaobin Xia
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Xing Zhao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Lu Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Lin Huang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Yanwen Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Xiangyu Yin
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Luyu He
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Yuebumo Aga
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Ankaer Kahaer
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Shiyu Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
| | - Lili Hao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| | - Chaoxi Chen
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China; (Z.X.); (X.Z.); (L.W.); (L.H.); (Y.Y.); (X.Y.); (L.H.); (Y.A.); (A.K.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
5
|
Dai C, Liu D, Qin C, Fang J, Cheng G, Xu C, Wang Q, Lu T, Guo Z, Wang J, Zhong T, Guo Q. Guben Kechuan granule attenuates bronchial asthma by inhibiting NF-κB/STAT3 signaling pathway-mediated apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 340:119124. [PMID: 39694430 DOI: 10.1016/j.jep.2024.119124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/20/2024] [Accepted: 11/15/2024] [Indexed: 12/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic asthma caused by allergies is a lung illness marked by airway remodeling and hyperresponsiveness. Guben Kechuan (GK) granule is a clinically proven formula for treating lung disease. It relieves cough and helps to clear phlegm, but the mechanisms underlying its treatment for asthma are not clear. AIM OF THE STUDY We aimed to elucidate the efficacy and potential mechanisms by which GK ameliorates allergic asthma. MATERIALS AND METHODS Ultra-performance liquid chromatography (UHPLC-LTQ-Orbitrap-MS) identified the main chemical components of GK. The efficacy of GK was studied in an ovalbumin/alum (OVA)/AL(OH)3-sensitized rat model of bronchial asthma by measuring cytokine concentrations in serum and alveolar lavage samples, examining tissue pathology, and performing leukocyte counts. The mechanisms underlying its effectiveness in asthma were investigated by both transcriptomic and proteomic analyses. RESULTS GK relieved asthma-induced airway inflammation and remodeling, reduced inflammatory cell infiltration, and decreased the levels of the inflammatory cytokines TNF-α, IL-4, IL-5, IL-6, and IL-10. Analysis of the transcriptomic and proteomic results found that asthma activated the transcription factors STAT3 and NF-κB and induced oxidative-stress damage and apoptosis. GK was found to reduce Bax and caspase-3 expression, increase Bcl-2 expression, and inhibit asthma-induced apoptosis. GK downregulated the expression of the transcription factors STAT3 and NF-kB, which decreased the inflammatory response. Decreases in CAT, SOD, and GSH reduced asthma-induced oxidative-stress damage. CONCLUSIONS Our findings provide evidence that GK alleviates bronchial asthma by inhibiting apoptosis and oxidative stress damage mediated by the NF-κB/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Chuanhao Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Dewen Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Cuiying Qin
- Development Center of Medical Science & Technology National Health Commission of the People's Republic of China, Beijing, 100044, China
| | - Jingya Fang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chunhong Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qixin Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Tianming Lu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zuchang Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; Department of Critical Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
| | - Tianyu Zhong
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
6
|
Gou Y, Lin F, Dan L, Zhang D. Exposure to toluene diisocyanate induces dysbiosis of gut-lung homeostasis: Involvement of gut microbiota. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125119. [PMID: 39414067 DOI: 10.1016/j.envpol.2024.125119] [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: 05/19/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Toluene diisocyanate (TDI) is a major industrial compound that induces occupational asthma with steroid-resistant properties. Recent studies suggest that the gastrointestinal tract may be an effective target for the treatment of respiratory diseases. However, the alterations of the gut-lung axis in TDI-induced asthma remain unexplored. Therefore, in this study, a model of stable occupational asthma caused by TDI exposure was established to detect the alteration of the gut-lung axis. Exposure to TDI resulted in dysbiosis of the gut microbiome, with significant decreases in Barnesiella_intestinihominis, Faecalicoccus_pleomorphus, Lactobacillus_apodemi, and Lactobacillus_intestinalis, but increases in Alistipes_shahii and Odoribacter_laneus. The largest change in abundance was in Barnesiella_intestinihominis, which decreased from 12.14 per cent to 6.18 per cent. The histopathological abnormalities, including shorter length of intestinal villi, thinner thickness of muscularis, reduced number of goblet cells and inflammatory cell infiltration, were found in TDI-treated mice compared to control mice. In addition, increased permeability (evidenced by significantly reduced levels of ZO-1, Occludin and Claudin-1) and activation of TLR4/NF-κB signaling were observed in the intestine of these TDI-exposed mice. Concurrently, exposure to TDI resulted in airway hyperresponsiveness, overt cytokine production (e.g., IL-4, IL-5, IL-13, IL-25, and IL-33), and elevated IgE level within the respiratory tract. The expression of tight junction proteins is reduced and TLR4/NF-κB signaling is activated in the lung following TDI treatment. In addition, correlation analyses showed that changes in the gut microbiota were correlated with TDI exposure-induced airway inflammation. In conclusion, the present study suggests that the immune gut-lung axis may be involved in the development of TDI-induced asthma, which may have implications for potential interventions against steroid-resistant asthma.
Collapse
Affiliation(s)
- Yuxuan Gou
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China.
| | - Fu Lin
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Li Dan
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Dianyu Zhang
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| |
Collapse
|
7
|
Ren M, Ma J, Qu M. Network pharmacology integrated with molecular docking and molecular dynamics simulations to explore the mechanism of Shaoyao Gancao Tang in the treatment of asthma and irritable bowel syndrome. Medicine (Baltimore) 2024; 103:e40929. [PMID: 39686413 DOI: 10.1097/md.0000000000040929] [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] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Numerous studies have demonstrated a correlation between asthma and irritable bowel syndrome (IBS). The Chinese herbal compound Shaoyao Gancao Tang (SYGCT) has been found to have therapeutic effects on both asthma and IBS, but the underlying mechanisms are not yet fully understood. This study aims to explore the key components, key targets, and potential mechanisms of SYGCT in treating asthma with IBS by using network pharmacology, molecular docking techniques and molecular dynamics simulation. METHODS The major chemical components and potential target genes of SYGCT were screened by bioinformatics. The key targets of Asthma-IBS comorbidity were identified based on network modules. The intersection of the drug targets and disease targets was identified as the potential targets of SYGCT in treating asthma-IBS. Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed to identify the biological processes and signaling pathways involved in these potential targets. A protein-protein interaction network was constructed to identify hub targets, while a drug-compound-target topological network was built to screen key compounds. Molecular docking was used to verify the affinity between the hub targets and key compounds. Molecular dynamics analysis was utilized to assess the binding stability of these interactions. RESULTS Network pharmacology analysis revealed that the therapeutic effect of SYGCT on asthma-IBS involved multiple biological processes and signaling pathways. It may exert therapeutic effects primarily through signaling pathways such as IL-17, TNF, and Th17 cell differentiation. The possible targets of SYGCT in the treatment of asthma-IBS could be IL6, TNF, JUN, PTGS2, STAT3, IL1B, CASP3, NFKBIA, IL10, and PPARG. Molecular docking verification showed that the predicted targets had good binding affinity with the compounds, among which PTGS2, CASP3, and PPARG had higher binding energy. Molecular dynamics simulation revealed that PTGS2, CASP3, and PPARG proteins had good stability and high binding strength with the compounds 2-[(3R)-8,8-dimethyl-3,4-dihydro-2H-pyrano[6,5-f]chromen-3-yl]-5-methoxyphenol and shinpterocarpin. CONCLUSION SYGCT plays a therapeutic role in asthma and IBS through multiple targets and pathways, providing a theoretical basis for explaining the mechanism and clinical application of SYGCT in treating different diseases with the same treatment in asthma and IBS.
Collapse
Affiliation(s)
- Mengjiao Ren
- Department of Warm Disease, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | | |
Collapse
|
8
|
Burrows K, Ngai L, Chiaranunt P, Watt J, Popple S, Forde B, Denha S, Olyntho VM, Tai SL, Cao EY, Tejeda-Garibay S, Koenig JFE, Mayer-Barber KD, Streutker CJ, Hoyer KK, Osborne LC, Liu J, O'Mahony L, Mortha A. A gut commensal protozoan determines respiratory disease outcomes by shaping pulmonary immunity. Cell 2024:S0092-8674(24)01336-9. [PMID: 39706191 DOI: 10.1016/j.cell.2024.11.020] [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: 04/27/2022] [Revised: 05/07/2024] [Accepted: 11/13/2024] [Indexed: 12/23/2024]
Abstract
The underlying mechanisms used by the intestinal microbiota to shape disease outcomes of the host are poorly understood. Here, we show that the gut commensal protozoan, Tritrichomonas musculis (T.mu), remotely shapes the lung immune landscape to facilitate perivascular shielding of the airways by eosinophils. Lung-specific eosinophilia requires a tripartite immune network between gut-derived inflammatory group 2 innate lymphoid cells and lung-resident T cells and B cells. This network exacerbates the severity of allergic airway inflammation while hindering the systemic dissemination of pulmonary Mycobacterium tuberculosis. The identification of protozoan DNA sequences in the sputum of patients with severe allergic asthma further emphasizes the relevance of commensal protozoa in human disease. Collectively, these findings demonstrate that a commensal protozoan tunes pulmonary immunity via a gut-operated lung immune network, promoting both beneficial and detrimental disease outcomes in response to environmental airway allergens and pulmonary infections.
Collapse
Affiliation(s)
- Kyle Burrows
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Pailin Chiaranunt
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jacqueline Watt
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sarah Popple
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Brian Forde
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Saven Denha
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Vitoria M Olyntho
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Eric Yixiao Cao
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Susana Tejeda-Garibay
- Health Sciences Research Institute, University of California Merced, Merced, CA, USA
| | - Joshua F E Koenig
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Catherine J Streutker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Katrina K Hoyer
- Health Sciences Research Institute, University of California Merced, Merced, CA, USA
| | - Lisa C Osborne
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Jun Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Liam O'Mahony
- Department of Medicine, University College Cork, Cork, Ireland
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Utembe W, Kamng'ona AW. Inhalation exposure to chemicals, microbiota dysbiosis and adverse effects on humans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176938. [PMID: 39414049 DOI: 10.1016/j.scitotenv.2024.176938] [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: 06/05/2024] [Revised: 09/21/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
As revealed by culture-independent methodologies, disruption of the normal lung microbiota (LM) configuration (LM dysbiosis) is a potential mediator of adverse effects from inhaled chemicals. LM, which consists of microbiota in the upper and lower respiratory tract, is influenced by various factors, including inter alia environmental exposures. LM dysbiosis has been associated with multiple respiratory pathologies such as asthma, lung cancer, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF). Chemically-induced LM dysbiosis appears to play significant roles in human respiratory diseases, as has been shown for some air pollutants, cigarette smoke and some inhalable chemical antibiotics. Lung microbiota are also linked with the central nervous system (CNS) in the so-called lung-brain axis. Inhaled chemicals that undergo mucociliary clearance may be linked to respiratory conditions through gut microbiota (GM) dysbiosis in the so-called Gut-Lung axis. However, current linkages of various disease states to LM appears to be associative, with causal linkages requiring further studies using more robust approaches, methods and techniques that are different from those applied in studies involving (GM). Most importantly, the sampling techniques determine the level of risk of cross contamination. Furthermore, the development of continuous or semi-continuous systems designed to replicate the lung microbiome will go a long way to further LM dysbiosis studies. These challenges notwithstanding, the preponderance of evidence points to the significant role of LM-mediated chemical toxicity in human disease and conditions.
Collapse
Affiliation(s)
- W Utembe
- Toxicology and Biochemistry Department, National Institute for Occupational Health, National Health Laboratory Services, Johannesburg 2000, South Africa; Environmental Health Division, School of Public Health and Family Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - A W Kamng'ona
- School of Life Sciences and Allied Health Professions, Kamuzu University of Health Sciences, Blantyre Campus, Mahatma Gandhi Road, Blantyre 312224, Malawi
| |
Collapse
|
10
|
Kortekaas Krohn I, Callewaert C, Belasri H, De Pessemier B, Diez Lopez C, Mortz CG, O'Mahony L, Pérez-Gordo M, Sokolowska M, Unger Z, Untersmayr E, Homey B, Gomez-Casado C. The influence of lifestyle and environmental factors on host resilience through a homeostatic skin microbiota: An EAACI Task Force Report. Allergy 2024; 79:3269-3284. [PMID: 39485000 DOI: 10.1111/all.16378] [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: 01/28/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Human skin is colonized with skin microbiota that includes commensal bacteria, fungi, arthropods, archaea and viruses. The composition of the microbiota varies at different anatomical locations according to changes in body temperature, pH, humidity/hydration or sebum content. A homeostatic skin microbiota is crucial to maintain epithelial barrier functions, to protect from invading pathogens and to interact with the immune system. Therefore, maintaining homeostasis holds promise to be an achievable goal for microbiome-directed treatment strategies as well as a prophylactic strategy to prevent the development of skin diseases, as dysbiosis or disruption of homeostatic skin microbiota is associated with skin inflammation. A healthy skin microbiome is likely modulated by genetic as well as environmental and lifestyle factors. In this review, we aim to provide a complete overview of the lifestyle and environmental factors that can contribute to maintaining the skin microbiome healthy. Awareness of these factors could be the basis for a prophylactic strategy to prevent the development of skin diseases or to be used as a therapeutic approach.
Collapse
Affiliation(s)
- Inge Kortekaas Krohn
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Department of Dermatology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Chris Callewaert
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Hafsa Belasri
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Department of Dermatology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Britta De Pessemier
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Celia Diez Lopez
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Charlotte G Mortz
- Department of Dermatology and Allergy Centre, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Liam O'Mahony
- APC Microbiome Ireland, School of Microbiology, and Department of medicine, University College Cork, Cork, Ireland
| | - Marina Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Zsofia Unger
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Centre of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Homey
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| | - Cristina Gomez-Casado
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| |
Collapse
|
11
|
Yang X, Jiao W, Zeng X, Yu J, Xiao J, Jiang T, Tang H, Bi J, Chen Y, Li X, Chen W, Chen Y, Shen A, Sun L. Assessment of lower respiratory tract microbiota associated with pulmonary tuberculosis in children. Pediatr Pulmonol 2024; 59:3550-3559. [PMID: 39282716 DOI: 10.1002/ppul.27253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND The respiratory microbiota plays a crucial role in the development of tuberculosis (TB). While existing research has underscored imbalances in the respiratory microbiota of adult patients with TB, information regarding the lower respiratory tract (LRT) microbiota in pediatric patients with TB remains scarce. METHODS We employed 16S rRNA gene sequencing technology to investigate the LRT microbial communities of 85 children of different ages with active TB of different severities, 33 children with infectious diseases other than TB, and 48 sex- and age-matched healthy children. RESULTS A marked imbalance in the respiratory microbiota was observed in children with TB, highlighted by reduced alpha diversity and a distinct microbial community structure. Comparative analysis indicated that patients with severe TB exhibited lower Neisseria levels than those with non-severe TB (1.01% vs. 3.93%, respectively; p = .02). Streptococcus and Gemella levels were lower in bacteriologically confirmed TB cases compared with clinically diagnosed cases, and higher in healthy children younger than 10 years old than in the older group. Spearman correlation analysis demonstrated significant associations between the microbiota of the LRT and cytokine concentrations in the sputum of children with TB (e.g., an inverse correlation between Veillonella and interleukin-17A). CONCLUSIONS TB induced significant dysbiosis in the LRT microbiota of children that was associated with disease severity and the immunological response in the respiratory tract. Our findings may offer a deeper understanding of the role of the respiratory microbiome in TB pathogenesis and progression.
Collapse
Affiliation(s)
- Xuemei Yang
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Weiwei Jiao
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xi Zeng
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Yu
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jing Xiao
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tingting Jiang
- Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, China
| | - He Tang
- Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, China
| | - Jing Bi
- Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, China
| | - Yiyi Chen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoxue Li
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wanning Chen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yu Chen
- Shenyang Tenth People's Hospital (Shenyang Chest Hospital), Shenyang, Liaoning, China
| | - Adong Shen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Lin Sun
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Respiratory Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
12
|
Umakanthan S, Katwaroo AR, Bukelo M, Bg S, Boralingaiah P, Ranade AV, Rangan P, Shashidhar S, Kini JR, Kini G. Post-Acute Sequelae of Covid-19: A System-wise Approach on the Effects of Long-Covid-19. AMERICAN JOURNAL OF MEDICINE OPEN 2024; 12:100071. [PMID: 39268246 PMCID: PMC11387218 DOI: 10.1016/j.ajmo.2024.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/01/2024] [Indexed: 09/15/2024]
Abstract
The SARS-CoV-2 virus responsible for the COVID-19 pandemic has profoundly impacted global health, economics, and society. This review seeks to encompass an overview of current knowledge on COVID-19, including its transmission, pathogenesis, and clinical presentation related to various systems within the human body. COVID-19 is a highly contagious illness that has rapidly spread worldwide. As of August 4, 2023, the WHO reported over 570 million confirmed cases of COVID-19 and over 6.3 million deaths. Although the virus is most common in adults, children can also be infected. Respiratory droplets that are produced when an infected person coughs or sneezes are the primary transmission mode for COVID-19. Additionally, the virus can be disseminated via contact with contaminated surfaces or objects, as it can remain viable for several hours or days. SARS-CoV-2 is a respiratory virus that enters cells by bonding with the angiotensin-converting enzyme 2 (ACE2) receptor. Once inside the cell, the virus replicates and produces new particles that can infect other cells. Interestingly, the effects of post-acute sequelae of SARS-CoV-2 infection (PASC) encompass more than just respiratory system. The findings presented in the data suggest that PASC significantly impacts multiple organs and their respective physiological processes. In light of these observations, we aim to provide a detailed discussion of the relevant findings in this paper. Through our review, we hope to provide healthcare professionals with a deeper understanding of the effects of PASC on the human body, which could ultimately lead to improved patient outcomes and treatment strategies.
Collapse
Affiliation(s)
- Srikanth Umakanthan
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Arun Rabindra Katwaroo
- Trinidad Institute of Medical Technology, Department of Medicine, St. Augustine, Trinidad and Tobago
| | - Maryann Bukelo
- Department of Anatomical Pathology, Laboratory Services, North Central Regional Health Authority, Champ Fleurs, Trinidad and Tobago
| | - Shashidhar Bg
- Department of Critical care Medicine, Manipal Hospital, Bengaluru, India
| | - Prashanth Boralingaiah
- Early Psychosis Prevention and Intervention Center (EPPIC), Orygen Youth Health, Sunshine, Australia
| | - Anu V Ranade
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | | | - Jyoti Ramanath Kini
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Karnataka, India
| | - Gayathri Kini
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Karnataka, India
| |
Collapse
|
13
|
Yang Q, Wang Z, Liu M, Gan L. Causal Relationship Between Gut Microbiota and Leukemia: Future Perspectives. Oncol Ther 2024; 12:663-683. [PMID: 39217582 PMCID: PMC11573970 DOI: 10.1007/s40487-024-00300-8] [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: 05/25/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota plays a crucial role in maintaining homeostasis in the human gastrointestinal tract. Numerous studies have shown a strong association between the gut microbiota and the emergence and progression of various diseases. Leukemia is one of the most common hematologic malignancies. Although standardized protocols and expert consensus have been developed for routine diagnosis and treatment, limitations remain due to individual differences. Nevertheless, a large number of studies have established a link between the gut microbiota and leukemia, with disturbances in the gut microbiota directly or indirectly affecting the development of leukemia. However, the causal relationship between the two remains unclear, and studying and exploring the causal relationship may open up entirely new avenues and protocols for use in the prevention and/or treatment of leukemia, offering new insights into diagnosis and treatment. In this review, the intricate relationship between the gut microbiota and leukemia is explored in depth, including causal associations, metabolite effects, therapeutic applications, and complications. Based on the characteristics of the gut microbiota, the future applications and prospects of gut microbiota are discussed to provide useful information for clinical treatment of leukemia.
Collapse
Affiliation(s)
- Qiang Yang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Zexin Wang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China.
| | - Miao Liu
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Lingling Gan
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| |
Collapse
|
14
|
Ito K, Kanemitsu Y, Ueda T, Kamiya T, Kubota E, Mori Y, Fukumitsu K, Tajiri T, Fukuda S, Uemura T, Ohkubo H, Ito Y, Shibata Y, Kumamoto N, Ugawa S, Niimi A. Comorbid functional dyspepsia reflects IL-33-mediated airway neuronal dysfunction in asthma. J Allergy Clin Immunol 2024; 154:1422-1433. [PMID: 38909633 DOI: 10.1016/j.jaci.2024.06.008] [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: 12/18/2023] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Neuronal dysfunction is implicated in the pathophysiology of asthma and functional dyspepsia (FD). However, the relationship between these diseases remains unclear. OBJECTIVE This study aimed to clarify the clinical implications of comorbid FD in asthma and to explore the unified pathway between asthma and FD by focusing on airway neuronal dysfunction. METHODS Clinical indices and biomarkers, including capsaicin cough sensitivity (C-CS), were compared between patients with asthma with and without FD. C-CS was determined on the basis of capsaicin concentration that induced at least 2 coughs (C2) or 5 coughs (C5). Additionally, the associations of airway inflammation with airway innervation and gastrointestinal motility were evaluated in mouse models of type 2 airway inflammation. RESULTS Patients with asthma with FD had worse asthma control and cough severity and lower C2 and C5 thresholds than those without FD. The severity of FD symptoms was negatively correlated with C2 and C5 thresholds. FD and poor asthma control were predictors of heightened C-CS (defined as C5 ≤ 2.44 μmol) in asthma. A mouse model of papain-induced airway inflammation developed airway hyperinnervation and gastrointestinal dysmotility, and both pathologies were ameliorated by an anti-IL-33 antibody. Moreover, papain-induced gastrointestinal dysmotility was mitigated by silencing the airway sensory neurons using QX-314, a sodium channel blocker. Furthermore, sputum IL-33 levels were significantly elevated in patients with asthma with FD or heightened C-CS compared to their counterparts. CONCLUSION FD is significantly associated with airway neuronal dysfunction in asthma. IL-33-mediated airway neuronal dysfunction may contribute to the interaction between asthma and FD.
Collapse
Affiliation(s)
- Keima Ito
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshihiro Kanemitsu
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Takashi Ueda
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Takeshi Kamiya
- Department of Medical Innovation, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Eiji Kubota
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuta Mori
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kensuke Fukumitsu
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tomoko Tajiri
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Fukuda
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirotsugu Ohkubo
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Ito
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuhiro Shibata
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Natsuko Kumamoto
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinya Ugawa
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
15
|
Quan J, Xie D, Li Z, Yu X, Liang Z, Chen Y, Wu L, Huang D, Lin L, Fan L. Luteolin alleviates airway remodeling in asthma by inhibiting the epithelial-mesenchymal transition via β-catenin regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156090. [PMID: 39393303 DOI: 10.1016/j.phymed.2024.156090] [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: 04/19/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Asthma is a prevalent long-term inflammatory condition that causes airway inflammation and remodeling. Increasing evidence indicates that epithelial-mesenchymal transition (EMT) holds a prominent implication in airway reconstruction in patients with asthma. Flavonoids obtained from Chinese Materia Medica (CMM), such as Luteolin (Lut), exhibit various beneficial effects in various asthma models. Lut has been shown to mitigate various asthma symptoms, including airway inflammation, hyperresponsiveness, bronchoconstriction, excessive mucus production, pulmonary autophagy, and neutrophilic asthma. However, whether flavonoids can suppress EMT-associated airway remodeling in asthma and the fundamental mechanisms involved remain unclear, with no studies specifically addressing Lut in this context. PURPOSE To evaluate the inhibition of airway remodeling in asthma by Lut and its potential mechanisms, while examining the significance of β-catenin in this process through cellular and animal studies. METHODS A BEAS-2B cell model stimulated by lipopolysaccharide (LPS) was established in vitro. Wound closure and Transwell assays were utilized to assess the cellular migratory ability. EMT- and fibrosis-related markers in LPS-stimulated cells were evaluated using RT-qPCR and western blotting. The status of the β-catenin/E-cadherin and β-catenin destruction complexes was evaluated using western blotting, immunofluorescence (IF) staining, and co-immunoprecipitation (Co-IP) analysis. The regulatory function of Lut in β-catenin-dependent EMT was further validated by β-catenin overexpression with adenovirus transduction and siRNA-mediated knockdown of β-catenin. Moreover, the counts of different types of bronchoalveolar lavage fluid (BALF) inflammatory cells from mice with asthma induced by ovalbumin (OVA) were evaluated in vivo using Congo red staining. Hematoxylin and eosin (H&E), Masson's trichrome, and periodic acid-Schiff (PAS) staining were used to evaluate collagen deposition, mucus production, and inflammation in murine lung tissues. Western blotting and immunohistochemistry (IHC) assays were used to assess EMT- and fibrosis-related markers in the lung tissues in vivo. RESULT Six naturally derived flavonoids, including Lut, attenuated cell migration and prevented EMT in LPS-treated BEAS-2B cells. Moreover, Lut suppressed TGF-β1, MMP-9, fibronectin (FN), and α-smooth muscle actin (α-SMA) levels in LPS-stimulated BEAS-2B cells. Additionally, Lut downregulated the levels of β-catenin by modulating the β-catenin/E-cadherin and β-catenin destruction complexes, highlighting the pivotal role of β-catenin in EMT inhibition by Lut in LPS-stimulated BEAS-2B cells. Furthermore, Lut suppressed airway inflammation and attenuated EMT-associated airway remodeling through β-catenin blockade in OVA-induced asthmatic mice. The bronchial wall thickness notably reduced from 37.24 ± 4.00 μm in the asthmatic model group to 30.06 ± 4.40 μm in the Lut low-dose group and 24.69 ± 2.87 μm in the Lut high-dose group. CONCLUSION According to our current understanding, this research is the first to reveal that Lut diminishes airway remodeling in asthma by inhibiting EMT via β-catenin regulation, thereby filling a research gap concerning Lut and flavonoids. These results provide a theoretical basis for treating asthma with anti-asthmatic CMM, as well as a candidate and complementary therapeutic approach to treat asthma.
Collapse
Affiliation(s)
- Jingyu Quan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Zihong Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xuhua Yu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Ziyao Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Yuanbin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lei Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Donghui Huang
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, China.
| | - Lin Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| | - Long Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| |
Collapse
|
16
|
Qian X, Liu Y, Wei X, Chen X, Rong G, Hu X. Unique Gut Microbiome and Metabolic Profiles in Chinese Workers Exposed to Dust: Insights From a Case-Control Study. J Occup Environ Med 2024; 66:1072-1082. [PMID: 39393924 DOI: 10.1097/jom.0000000000003243] [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: 10/13/2024]
Abstract
OBJECTIVES This study aimed to identify distinct gut microbiome and serum metabolic features in workers exposed to dust compared to healthy controls. METHODS A case-control study was conducted with dust-exposed workers without silicosis and age-matched healthy controls. Gut microbiome composition was analyzed using 16S rRNA sequencing, and serum and fecal metabolomic profiles were assessed by LC-MS. RESULTS Dust-exposed workers showed higher levels of Blautia and Trichoderma and lower levels of Anaplasma , Aspergillus , Plasmodiophoromycetes, and Escherichia coli-Shigella . Metabolites such as indole-3-acetate and gentamicin C1a were downregulated, while adenine, 2-phenylacetamide, and 4-pyridoxic acid were upregulated. CONCLUSIONS Blautia spp. were linked to altered metabolites in dust-exposed workers, suggesting microbiome-metabolite interactions that may affect silicosis progression. However, the small sample size and cross-sectional design limit generalizability, and further longitudinal studies are needed.
Collapse
Affiliation(s)
- Xiaojun Qian
- From the Department of Respiratory and Critical Care Medicine, The Third People's Hospital of Hefei, Hefei, Anhui, China (X.Q., X.W., X.C., G.R.); Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China (X.Q., Y.L.); Department of Respiratory and Critical Care Medicine, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China (X.Q.); and Department of Science and Education, The Third People's Hospital of Hefei, Hefei Third Clinical College of Anhui Medical University, Hefei, Anhui, China (X.H.)
| | | | | | | | | | | |
Collapse
|
17
|
Rana S, Singh P, Bhardwaj T, Somvanshi P. A Comprehensive Metagenome Study Identifies Distinct Biological Pathways in Asthma Patients: An In-Silico Approach. Biochem Genet 2024; 62:4264-4279. [PMID: 38285123 DOI: 10.1007/s10528-023-10635-y] [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: 04/18/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
Asthma is a multifactorial disease with phenotypes and several clinical and pathophysiological characteristics. Besides innate and adaptive immune responses, the gut microbiome generates Treg cells, mediating the allergic response to environmental factors and exposure to allergens. Because of the complexity of asthma, microbiome analysis and other precision medicine methods are now widely regarded as essential elements of efficient disease therapy. An in-silico pipeline enables the comparative taxonomic profiling of 16S rRNA metagenomic profiles of 20 asthmatic patients and 15 healthy controls utilizing QIIME2. Further, PICRUSt supports downstream gene enrichment and pathway analysis, inferring the enriched pathways in a diseased state. A significant abundance of the phylum Proteobacteria, Sutterella, and Megamonas is identified in asthma patients and a diminished genus Akkermansia. Nasal samples reveal a high relative abundance of Mycoplasma in the nasal samples. Further, differential functional profiling identifies the metabolic pathways related to cofactors and amino acids, secondary metabolism, and signaling pathways. These findings support that a combination of bacterial communities is involved in mediating the responses involved in chronic respiratory conditions like asthma by exerting their influence on various metabolic pathways.
Collapse
Affiliation(s)
- Samiksha Rana
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, JNU Campus, New Delhi, 110067, India
| | - Pooja Singh
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, JNU Campus, New Delhi, 110067, India
| | - Tulika Bhardwaj
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Pallavi Somvanshi
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, JNU Campus, New Delhi, 110067, India.
- Special Centre of Systems Medicine (SCSM), Jawaharlal Nehru University, JNU Campus, New Delhi, 110067, India.
| |
Collapse
|
18
|
Ma H, Mueed A, Ma Y, Ibrahim M, Su L, Wang Q. Fecal Microbiota Transplantation Activity of Floccularia luteovirens Polysaccharides and Their Protective Effect on Cyclophosphamide-Induced Immunosuppression and Intestinal Injury in Mice. Foods 2024; 13:3881. [PMID: 39682952 DOI: 10.3390/foods13233881] [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: 10/18/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Floccularia luteovirens polysaccharides (FLP1s) have potential biological activities. Our previous study showed that FLP1s positively regulated gut immunity and microbiota. However, it is still unclear whether FLP1s mediate gut microbiota in immunosuppressed mice. This research aims to explore the relationship between FLP1-mediated gut microbes and intestinal immunity in immunosuppressed mice through fecal microbiota transplantation (FMT). The results demonstrated that FLP1s exhibited prebiotic and anti-immunosuppressive effects on CTX-induced immunosuppressed mice. FFLP1 treatment (microbiota transplantation from the fecal sample) remarkably elevated the production of sIgA and secretion of the anti-inflammatory cytokines IL-4, TNF-α, and IFN-γ in the intestine of CTX-treated mice, inducing activation of the MAPK pathway. Moreover, FFLP1s mitigated oxidative stress by activating the Nrf2/Keap1 signaling pathway and strengthened the intestinal barrier function by upregulating the expression level of tight junction proteins (occludin, claudin-1, MUC-2, and ZO-1). Furthermore, FFPL1s restored gut dysbiosis in CTX-treated immunosuppressed mice by increasing the abundance of Alloprevotella, Lachnospiraceae, and Bacteroides. They also modified the composition of fecal metabolites, leading to enhanced regulation of lipolysis in adipocytes, the cGMP-PKG pathway, the Rap1 signaling pathway, and ovarian steroidogenesis, as indicated by KEGG pathway analysis. These findings indicate that FLP1s could modulate the response of the intestinal immune system through regulation of the gut microbiota, thus promoting immune activation in CTX-treated immunosuppressed mice. FLP1s can serve as a natural protective agent against CTX-induced immune injury.
Collapse
Affiliation(s)
- He Ma
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Abdul Mueed
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Yanxu Ma
- Jilin Sericulture Science Research Institute, Changchun 130012, China
| | - Muhammad Ibrahim
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Ling Su
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130012, China
| | - Qi Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| |
Collapse
|
19
|
Yang B, Guo X, Shi C, Liu G, Qin X, Chen S, Gan L, Liang D, Shao K, Xu R, Zhong J, Mo Y, Li H, Luo D. Alterations in purine and pyrimidine metabolism associated with latent tuberculosis infection: insights from gut microbiome and metabolomics analyses. mSystems 2024; 9:e0081224. [PMID: 39436103 PMCID: PMC11575419 DOI: 10.1128/msystems.00812-24] [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: 06/25/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Individuals with latent tuberculosis infection (LTBI) account for almost 30% of the population worldwide and have the potential to develop active tuberculosis (ATB). Despite this, the current understanding of the pathogenesis of LTBI is limited. The gut microbiome can be altered in tuberculosis patients, and an understanding of the changes associated with the progression from good health to LTBI to ATB can provide novel perspectives for understanding the pathogenesis of LTBI by identifying microbial and molecular biomarkers associated therewith. In this study, fecal samples from healthy controls (HC), individuals with LTBI and ATB patients were collected for gut microbiome and metabolomics analyses. Compared to HC and LTBI subjects, participants with ATB showed a significant decrease in gut bacterial α-diversity. Additionally, there were significant differences in gut microbial communities and metabolism among the HC, LTBI, and ATB groups. PICRUSt2 analysis revealed that microbiota metabolic pathways involving the degradation of purine and pyrimidine metabolites were upregulated in LTBI and ATB individuals relative to HCs. Metabolomic profiling similarly revealed that purine and pyrimidine metabolite levels were decreased in LTBI and ATB samples relative to those from HCs. Further correlation analyses revealed that the levels of purine and pyrimidine metabolites were negatively correlated with those of gut microbial genera represented by Ruminococcus_gnavus_group (R. gnavus), and the levels of R. gnavus were also positively correlated with adenosine nucleotide degradation II, which is a purine degradation pathway. Moreover, a combined signature including hypoxanthine and xanthine was found to effectively distinguish between LTBI and HC samples (area under the curve [AUC] of training set = 0.796; AUC of testing set = 0.924). Therefore, through gut microbiome and metabolomic analyses, these findings provide valuable clues regarding how alterations in gut purine and pyrimidine metabolism are linked to the pathogenesis of LTBI.IMPORTANCEThis study provides valuable insight into alterations in the gut microbiome and metabolomic profiles in a cohort of adults with LTBI and ATB. Perturbed gut purine and pyrimidine metabolism in LTBI was associated with the compositional alterations of gut microbiota, which may be an impetus for developing novel diagnostic strategies and interventions targeting LTBI.
Collapse
Affiliation(s)
- Boyi Yang
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- The First Clinical College, Guangxi Medical University, Nanning, China
| | - Xiaojing Guo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Chongyu Shi
- Molecular Biology Laboratory of Respiratory Disease, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Gang Liu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaoling Qin
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Shiyi Chen
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Li Gan
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Dongxu Liang
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Kai Shao
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Ruolan Xu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Jieqing Zhong
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Yujie Mo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Hai Li
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, China
| | - Dan Luo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, China
| |
Collapse
|
20
|
Chen L, Yan H, Di S, Guo C, Zhang H, Zhang S, Gold A, Wang Y, Hu M, Wu D, Johnson CH, Wang X, Zhu J. Mapping Pesticide-Induced Metabolic Alterations in Human Gut Bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623895. [PMID: 39605636 PMCID: PMC11601348 DOI: 10.1101/2024.11.15.623895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pesticides can modulate gut microbiota (GM) composition, but their specific effects on GM remain largely elusive. Our study demonstrated that pesticides inhibit or promote growth in various GM species, even at low concentrations, and can accumulate in GM to prolong their presence in the host. Meanwhile, the pesticide induced changes in GM composition are associated with significant alterations in gut bacterial metabolism that reflected by the changes of hundreds of metabolites. We generated a pesticide-GM-metabolites (PMM) network that not only reveals pesticide-sensitive gut bacteria species but also report specific metabolic changes in 306 pesticide-GM pairs (PGPs). Using an in vivo mice model, we further demonstrated a PGP's interactions and verified the inflammation-inducing effects of pesticides on the host through dysregulated lipid metabolism of microbes. Taken together, our findings generate a PMM interactions atlas, and shed light on the molecular level of how pesticides impact host health by modulating GM metabolism.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Shanshan Di
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Chao Guo
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Huan Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Shiqi Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Gold
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Yu Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Ming Hu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Dayong Wu
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Rodrigues e-Lacerda R, Barra NG, Fang H, Anhê GF, Schertzer JD. NOD2 protects against allergic lung inflammation in obese female mice. iScience 2024; 27:111130. [PMID: 39507249 PMCID: PMC11539594 DOI: 10.1016/j.isci.2024.111130] [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: 09/25/2023] [Revised: 08/07/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Obesity is associated with compartmentalized changes in immune responses that can be protective or pathogenic. It has been proposed that obesity-related changes in the microbiota influence allergic lung inflammation. We hypothesized that sensors of the bacterial cell wall influenced allergenic lung inflammation during obesity. Ovalbumin (OVA)-induced lung inflammation was similar in female Nod1-/- and wild-type mice during high-fat-diet-induced obesity, but allergic lung inflammation was higher in obese, high-fat-diet-fed female Nod2-/- mice. Obese Nod2-/- mice had higher inflammatory cell infiltration in the bronchial alveolar lavage (BAL) and lungs, pulmonary fibrosis, mucus levels, hypertrophy and hyperplasia of goblet cells, M2 alveolar macrophage infiltration, interleukin-4 (IL-4), IL-5, IL-6, and lower CXCL1 and IL-22. Therefore, Nod2 protects against excessive lung inflammation and is a bacterial sensor that relays protective responses to allergenic lung inflammation in obese female mice.
Collapse
Affiliation(s)
- Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Nicole G. Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Gabriel Forato Anhê
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
22
|
Wang H, Zhang L, Shang Y. DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1. Biol Direct 2024; 19:114. [PMID: 39533404 PMCID: PMC11556204 DOI: 10.1186/s13062-024-00557-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Endoplasmic reticulum (ER) stress has been shown to play a pivotal role in the pathogenesis of asthma. DEPTOR (DEP Domain Containing MTOR Interacting Protein) is an endogenous mTOR inhibitor that participates in various physiological processes such as cell growth, apoptosis, autophagy, and ER homeostasis. However, the role of DEPTOR in the pathogenesis of asthma is still unknown. In this study, an ovalbumin (OVA)-induced mice model and IL-13 induced 16HBE cells were used to evaluate the effect of DEPTOR on asthma. A decreased DEPTOR expression was shown in the lung tissues of OVA-mice and IL-13 induced 16HBE cells. Upregulation of DEPTOR attenuated airway goblet cell hyperplasia, inhibited mucus hypersecretion, decreased the expression of mucin MUC5AC, and suppressed the level of inflammatory factors IL-4 and IL-5, which were all induced by OVA treatment. The increased protein expression of ER stress markers GRP78, CHOP, unfolded protein response (UPR) related proteins, and apoptosis markers in OVA mice were also inhibited by DEPTOR overexpression. In IL-13 induced 16HBE cells, overexpression of DEPTOR decreased IL-4, IL-5, and MUC5AC levels, preventing ER stress response and UPR process. Furthermore, from the proteomics results, we identified that SOD1 (Cu/Zn Superoxide Dismutase 1) may be the downstream factor of DEPTOR. Similar to DEPTOR, upregulation of SOD1 alleviated asthma progression. Rescue experiments showed that SOD1 inhibition abrogates the remission effect of DEPTOR on ER stress in vitro. In conclusion, these data suggested that DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Lei Zhang
- Department of General Surgery, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Yunxiao Shang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
23
|
Li L, Xu Z, Ni H, Meng Y, Xu Y, Xu H, Zheng Y, Zhang Y, Xue G, Shang Y. Hydrogen-rich water alleviates asthma airway inflammation by modulating tryptophan metabolism and activating aryl hydrocarbon receptor via gut microbiota regulation. Free Radic Biol Med 2024; 224:50-61. [PMID: 39147072 DOI: 10.1016/j.freeradbiomed.2024.08.009] [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: 05/22/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Hydrogen-rich water (HRW) is a beverage containing a high concentration of hydrogen that has been researched for its antioxidant, anti-apoptotic, and anti-inflammatory properties in asthma. This study investigates the potential therapeutic impact of HRW on the gut-lung axis. Using 16S rRNA and serum metabolomics, we examined changes in gut microbiota and serum metabolites in asthmatic mice after HRW intervention, followed by validation experiments. The findings revealed that HRW influenced gut microbiota by increasing Ligilactobacillus and Bifidobacterium abundance and enhancing the presence of indole-3-acetic acid (IAA), a microbially derived serum metabolite. Both in vivo and in vitro experiments showed that HRW's protective effects against airway inflammation in asthmatic mice may be linked to the gut microbiota, with IAA potentially playing a role in reducing asthmatic airway inflammation through the aryl hydrocarbon receptors (AhR) signaling pathway. In summary, HRW can modify gut microbiota, increase Bifidobacterium abundance, elevate microbial-derived IAA levels, and activate AhR, which could potentially alleviate inflammation in asthma.
Collapse
Affiliation(s)
- Li Li
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China.
| | - Ziqian Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Haoran Ni
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yesong Meng
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yongzhuang Xu
- Department of General Practice, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Hao Xu
- Department of General Practice, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yuyang Zheng
- Department of General Practice, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Yi Zhang
- Department of General Practice, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China.
| | - Geng Xue
- Department of Medical Genetics, College of Basic Medical Sciences, Naval Military Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Yan Shang
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China; Department of General Practice, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Military Medical University (Second Military Medical University), Shanghai, 200433, China.
| |
Collapse
|
24
|
Lin X, Hu X, Zhang J, Luo J, Qin G, Jiang L. Gut microbiota, allergic rhinitis, vasomotor rhinitis, Mendelian randomization, causal association. Braz J Otorhinolaryngol 2024; 90:101491. [PMID: 39243698 PMCID: PMC11409179 DOI: 10.1016/j.bjorl.2024.101491] [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: 04/23/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024] Open
Abstract
OBJECTIVE Continuous research on the structure and function of intestinal microecology has confirmed the association between gut microbiota and the occurrence, development, and outcome of allergic diseases. Here, we explored the genetic causality between gut microbiota and rhinitis. METHODS We conducted a two-sample Mendelian Randomization (MR) study to investigate the genetic causal relationship between gut microbiota and allergic rhinitis and vasomotor rhinitis. Genetic variations in the human gut microbiota were obtained from the summary statistics of the MiBioGen study. Genome-wide summary statistics of rhinitis were obtained from the FinnGen consortium. The causal effect between gut microbiota and rhinitis was assessed using the inverse variance weighted, MR-Egger regression, and weighted median methods. In addition, sensitivity analyses were conducted using different methods, including maximum likelihood, simple mode, and weighted model methods. RESULTS The IVW approach revealed a causal association of the genus Ruminococcus gauvreauii group with an increased risk of allergic rhinitis (IVW Odds Ratio [OR = 1.26] [1.04, 1.53], p-value = 0.01645). In addition, the genus Fusicatenibacter (IVW OR = 1.20 [1.02, 1.41], p-value = 0.02868) was causally associated with an increased risk of vasomotor rhinitis. CONCLUSION Gut microbiota belonging to different genera exert different effects on allergic rhinitis and vasomotor rhinitis, including reducing the risk of rhinitis, and increasing the risk of rhinitis. New insights into the mechanisms of underlying gut microbiota-associated rhinitis are provided. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Xitan Lin
- Affiliated Hospital of Southwest Medical University, Department of Otolaryngology Head and Neck Surgery, Sichuan, China
| | - Xiaoyan Hu
- School of Basic Medicine, Department of Pathogen Biology, Southwest Medical University, Public Center of Experimental Technology of Pathogen Biology Technology Platform, Sichuan, China
| | - Jing Zhang
- Affiliated Hospital of Southwest Medical University, Department of Otolaryngology Head and Neck Surgery, Sichuan, China
| | - Jing Luo
- Affiliated Hospital of Southwest Medical University, Department of Otolaryngology Head and Neck Surgery, Sichuan, China
| | - Gang Qin
- Affiliated Hospital of Southwest Medical University, Department of Otolaryngology Head and Neck Surgery, Sichuan, China
| | - Liang Jiang
- Affiliated Hospital of Southwest Medical University, Department of Otolaryngology Head and Neck Surgery, Sichuan, China.
| |
Collapse
|
25
|
Kabil A, Nayyar N, Brassard J, Li Y, Chopra S, Hughes MR, McNagny KM. Microbial intestinal dysbiosis drives long-term allergic susceptibility by sculpting an ILC2-B1 cell-innate IgE axis. J Allergy Clin Immunol 2024; 154:1260-1276.e9. [PMID: 39134158 DOI: 10.1016/j.jaci.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND The abundance and diversity of intestinal commensal bacteria influence systemic immunity with impact on disease susceptibility and severity. For example, loss of short chain fatty acid (SCFA)-fermenting bacteria in early life (humans and mice) is associated with enhanced type 2 immune responses in peripheral tissues including the lung. OBJECTIVE Our goal was to reveal the microbiome-dependent cellular and molecular mechanisms driving enhanced susceptibility to type 2 allergic lung disease. METHODS We used low-dose vancomycin to selectively deplete SCFA-fermenting bacteria in wild-type mice. We then examined the frequency and activation status of innate and adaptive immune cell lineages with and without SCFA supplementation. Finally, we used ILC2-deficient and signal transducer and activator of transcription 6 (STAT6)-deficient transgenic mouse strains to delineate the cellular and cytokine pathways leading to enhanced allergic disease susceptibility. RESULTS Mice with vancomycin-induced dysbiosis exhibited a 2-fold increase in lung ILC2 primed to produce elevated levels of IL-2, -5, and -13. In addition, upon IL-33 inhalation, mouse lung ILC2 displayed a novel ability to produce high levels of IL-4. These expanded and primed ILC2s drove B1 cell expansion and IL-4-dependent production of IgE that in turn led to exacerbated allergic inflammation. Importantly, these enhanced lung inflammatory phenotypes in mice with vancomycin-induced dysbiosis were reversed by administration of dietary SCFA (specifically butyrate). CONCLUSION SCFAs regulate an ILC2-B1 cell-IgE axis. Early-life administration of vancomycin, an antibiotic known to deplete SCFA-fermenting gut bacteria, primes and amplifies this axis and leads to lifelong enhanced susceptibility to type 2 allergic lung disease.
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Natalia Nayyar
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yicong Li
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sameeksha Chopra
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael R Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Kelly M McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada; Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
26
|
Zhou Z, Yang J, Liu Q, Gao J, Ji W. Patho-immunological mechanisms of atopic dermatitis: The role of the three major human microbiomes. Scand J Immunol 2024; 100:e13403. [PMID: 39267301 DOI: 10.1111/sji.13403] [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: 11/29/2023] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/17/2024]
Abstract
Atopic dermatitis (AD) is a genetically predisposed allergic inflammatory dermatosis with chronic, pruritic, and recurrent features. Patients with AD have dry and itchy skin, often accompanied by chronic eczematous lesions, allergic rhinitis, or asthma, which has a considerable impact on their daily lives. With advances in genome sequencing technology, it has been demonstrated that microorganisms are involved in this disease, and the microorganisms associated with AD are attracting considerable research attention. An increasing number of studies conducted in recent years have demonstrated that an imbalanced microbiome in AD patients has substantial impact on disease prognosis, and the causes are closely tied to various immune mechanisms. However, the involvement of microorganisms in the pathogenesis of AD remains poorly understood. In this paper, we review the advances in research on the immunological mechanisms of the skin microbiome, intestinal microbiome, and lung microbiome that are related to AD prognosis and immunotherapy protocols. It is hoped that this approach will lay the foundation for exploring the pathogenesis of and emerging treatments for AD.
Collapse
Affiliation(s)
- Zhaosen Zhou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qin Liu
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jing Gao
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenting Ji
- Department of Nursing in Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Callanan S, Talaei M, Delahunt A, Shaheen SO, McAuliffe FM. Low glycaemic index diet in pregnancy and child asthma: follow-up of the ROLO trial. Br J Nutr 2024:1-11. [PMID: 39466114 DOI: 10.1017/s0007114524001612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Epidemiological evidence suggests that a higher intake of sugar during pregnancy is associated with a higher risk of childhood asthma and atopy. However, randomised trial evidence supporting such a link is lacking. This study aimed to examine whether a low glycaemic index (GI) dietary intervention during pregnancy decreases the risk of childhood asthma and eczema. This is a secondary analysis of 514 children from the ROLO trial. Healthy women were randomised to receive an intervention of low GI dietary advice or routine care from early pregnancy. Mothers reported current doctor-diagnosed eczema in their children at 2 years (n 271) and current doctor-diagnosed asthma and eczema in their children at 5 (n 357) and 9-11 years (n 391) of age. Multivariable logistic regression models were used test the effect of the intervention on child outcomes overall and stratified by maternal education. There was a suggestion of a reduction in asthma at 5 years of age in children whose mothers received the low GI dietary intervention during pregnancy compared with usual care (adjusted OR 0·46 (95 % CI 0·19, 1·09); P = 0·08). In stratified adjusted analyses, the intervention was associated with a reduced risk of asthma at 5 years of age in children born to mothers with incomplete tertiary level education but not in those with complete tertiary level education (OR 0·14 (95 % CI 0·02, 0·69); P = 0·010 and OR 1·03 (95 % CI 0·34, 3·13); P = 0·94, respectively). A low GI diet in pregnancy may reduce the risk of developing asthma in childhood, particularly amongst children born to mothers with lower educational attainment.
Collapse
Affiliation(s)
- Sophie Callanan
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
| | - Mohammad Talaei
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anna Delahunt
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
| | - Seif O Shaheen
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Republic of Ireland
| |
Collapse
|
28
|
Donald K, Finlay BB. Experimental models of antibiotic exposure and atopic disease. FRONTIERS IN ALLERGY 2024; 5:1455438. [PMID: 39525399 PMCID: PMC11543581 DOI: 10.3389/falgy.2024.1455438] [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: 06/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
In addition to numerous clinical studies, research using experimental models have contributed extensive evidence to the link between antibiotic exposure and atopic disease. A number of mouse models of allergy have been developed and used to uncover the specific effects of various microbiota members and perturbations on allergy development. Studies in mice that lack microbes entirely have also demonstrated the various components of the immune system that require microbial exposure. The importance of the early-life period and the mechanisms by which atopy "protective" species identified in human cohorts promote immune development have been elucidated in mice. Finally, non-animal models involving human-derived cells shed light on specific effects of bacteria on human epithelial and immune responses. When considered alongside clinical cohort studies, experimental model systems have provided crucial evidence for the link between the neonatal gut microbiota and allergic disease, immensely supporting the stewardship of antibiotic administration in infants. The following review aims to describe the range of experimental models used for studying factors that affect the relationship between the gut microbiota and allergic disease and summarize key findings that have come from research in animal and in vitro models.
Collapse
Affiliation(s)
- Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Han M, Wang X, Su L, Pan S, Liu N, Li D, Liu L, Cui J, Zhao H, Yang F. Intestinal microbiome dysbiosis increases Mycobacteria pulmonary colonization in mice by regulating the Nos2-associated pathways. eLife 2024; 13:RP99282. [PMID: 39412514 PMCID: PMC11483126 DOI: 10.7554/elife.99282] [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] [Indexed: 10/19/2024] Open
Abstract
Increasing researches reveal gut microbiota was associated with the development of tuberculosis (TB). How to prevent or reduce Mycobacterium tuberculosis colonization in the lungs is a key measure to prevent TB. However, the data on gut microbiota preventing Mycobacterium colonization in the lungs were scarce. Here, we established the clindamycin-inducing intestinal microbiome dysbiosis and fecal microbial transplantation models in mice to identify gut microbiota's effect on Mycobacterium's colonization in the mouse lungs and explore its potential mechanisms. The results showed that clindamycin treatment altered the diversity and composition of the intestinal bacterial and fungal microbiome, weakened the trans-kingdom network interactions between bacteria and fungi, and induced gut microbiome dysbiosis in the mice. Gut microbiota dysbiosis increases intestinal permeability and enhances the susceptibility of Mycobacterium colonization in the lungs of mice. The potential mechanisms were gut microbiota dysbiosis altered the lung transcriptome and increased Nos2 expression through the 'gut-lung axis'. Nos2 high expression disrupts the intracellular antimicrobial and anti-inflammatory environment by increasing the concentration of nitric oxide, decreasing the levels of reactive oxygen species and Defb1 in the cells, and promoting Mycobacteria colonization in the lungs of mice. The present study raises a potential strategy for reducing the risks of Mycobacteria infections and transmission by regulating the gut microbiome balance.
Collapse
Affiliation(s)
- MeiQing Han
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Xia Wang
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
| | - Lin Su
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Shiqi Pan
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Ningning Liu
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Duan Li
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Liang Liu
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - JunWei Cui
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
| | - Huajie Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Fan Yang
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| |
Collapse
|
30
|
Pan Y, Zhang H, Zhu L, Tan J, Wang B, Li M. The role of gut microbiota in MP/NP-induced toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124742. [PMID: 39153541 DOI: 10.1016/j.envpol.2024.124742] [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: 04/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Microplastics (MPs) and nanoplastics (NPs) are globally recognized as emerging environmental pollutants in various environmental media, posing potential threats to ecosystems and human health. MPs/NPs are unavoidably ingested by humans, mainly through contaminated food and drinks, impairing the gastrointestinal ecology and seriously impacting the human body. The specific role of gut microbiota in the gastrointestinal tract upon MP/NP exposure remains unknown. Given the importance of gut microbiota in metabolism, immunity, and homeostasis, this review aims to enhance our current understanding of the role of gut microbiota in MP/NP-induced toxicity. First, it discusses human exposure to MPs/NPs through the diet and MP/NP-induced adverse effects on the respiratory, digestive, neural, urinary, reproductive, and immune systems. Second, it elucidates the complex interactions between the gut microbiota and MPs/NPs. MPs/NPs can disrupt gut microbiota homeostasis, while the gut microbiota can degrade MPs/NPs. Third, it reveals the role of the gut microbiota in MP/NP-mediated systematic toxicity. MPs/NPs cause direct intestinal toxicity and indirect toxicity in other organs via regulating the gut-brain, gut-liver, and gut-lung axes. Finally, novel approaches such as dietary interventions, prebiotics, probiotics, polyphenols, engineered bacteria, microalgae, and micro/nanorobots are recommended to reduce MP/NP toxicity in humans. Overall, this review provides a theoretical basis for targeting the gut microbiota to study MP/NP toxicity and develop novel strategies for its mitigation.
Collapse
Affiliation(s)
- Yinping Pan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, PR China
| | - Haojie Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, PR China
| | - Liancai Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, PR China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological & Chemical engineering, Chongqing University of Education, Chongqing, 400067, PR China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, PR China
| | - Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, PR China; Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.
| |
Collapse
|
31
|
Panpan Z, Jinli H, Qiuhong L, Bo D, Juan Z, Hui S, Xin S. Changes in respiratory tract and gut microbiota in AR mice and their relationship with Th1/Th2/Treg. Microb Pathog 2024; 195:106881. [PMID: 39197690 DOI: 10.1016/j.micpath.2024.106881] [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: 01/29/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND The etiology of allergic rhinitis (AR) is not fully understood. Studies have shown that the maturation of children's immune systems is closely related to microecology. However, few studies have focused simultaneously on changes in respiratory and gut microbiota in AR and their correlation between microecological changes and Th1/Th2/Treg. OBJECTIVE The aim is to investigate the pathogenesis of AR based on respiratory microecology, gut microecology, and Th1/Th2/Treg levels by applying microbiome techniques and correlation analysis. METHODS Standardized OVA-induced AR mice were established. Serum OVA-sIgE, IL-4, IFN-γ, IL-10 were measured by ELISA, Tregs in lymph nodes were determined by flow cytometry, and the histological characteristics of nasal tissues were evaluated by Hematoxylin & Eosin (H&E). Nasal symptoms were observed to determine the reliability of the AR mouse model. Nasal lavage fluid (NALF) and fecal samples were collected after the last OVA challenge. The composition of respiratory microbiota in NALF and gut microbial in feces samples via 16S rRNA gene sequencing between the two groups, further explored the relationship between microbiota and Th1/Th2/Treg levels. RESULTS In the AR group, the incidence of nose rubbing and sneezing in each mouse was significantly increased compared with the control group (all P < 0.001) and the inflammatory cell infiltration of NALF shows a significant increase in eosinophilic and neutrophilic infiltrates upon the AR group; H&E showed that the nasal mucosa of AR mice infiltration of massive eosinophils cells and neutrophils cells. OVA-sIgE and IL-4 in the AR group were increased (P < 0.01, P < 0.05) and IFN-γ, IL-10 were significantly decreased (P < 0.01, P < 0.05). Tregs showed a downward trend in the AR group, but there was no statistical difference. Compared with the control group, the respiratory microbiota of AR mice did not change significantly, while the gut microbiota changed significantly. In gut microbiota, compared to the control group, Shannon index in the AR group revealed a significant decrease at the genus level (P < 0.01), and Simpson index was significantly increased at all levels (all P < 0.05). PCoA also showed significant differences in beta diversity between the two groups (all P < 0.05). Compared to the control group, Deferribacteres at phylum level, Roseburia, Ruminiclostridium, Anaerotruncus at genus level were significantly decreased in the AR group (all P < 0.05). Spearman's rank correlation showed that OVA-sIgE was positively correlated with Bacteroidetes, Muribaculaceae and Erysipelotrichaceae (all P < 0.05); IL-4 was significantly negatively correlated with Epsilonbacteraeota and Deferribacteres (all P < 0.05). Treg was significantly positively correlated with Patescibacteria, Lachnospiraceae, and Saccharimonadaceae in gut microecology. CONCLUSION Our results showed that the respiratory microbiota of AR mice was not significantly altered, but the gut microbiota varied significantly and there was a correlation between gut microbiota and Th1/Th2/Treg.
Collapse
Affiliation(s)
- Zhang Panpan
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Huang Jinli
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Li Qiuhong
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Dong Bo
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhang Juan
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Su Hui
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China.
| | - Sun Xin
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
32
|
Ma H, Dong Z, Zhang X, Liu C, Liu Z, Zhou X, He J, Zhang S. Airway bacterial microbiome signatures correlate with occupational pneumoconiosis progression. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116875. [PMID: 39142114 DOI: 10.1016/j.ecoenv.2024.116875] [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: 05/06/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Recent evidence has pinpointed a key role of the microbiome in human respiratory health and disease. However, significant knowledge gaps still exist regarding the connection between bacterial communities and adverse effects caused by particulate matters (PMs). Here, we characterized the bacterial microbiome along different airway sites in occupational pneumoconiosis (OP) patients. The sequencing data revealed that OP patients exhibited distinct dysbiosis in the composition and function of the respiratory microbiota. To different extents, there was an overall increase in the colonization of microbiota, such as Streptococcus, implying a possible intrusion pathway provided by exogenous PMs. Compared to those of healthy subjects, unhealthy living habits (i.e., smoking) had a greater impact on microbiome changes in OP patients. Importantly, the associations between the bacterial community and disease indicators indicated that specific bacterial species, including Prevotella, Actinobacillus, and Leptotrichia, might be surrogate markers of OP disease progression. Collectively, our results highlighted the potential participation of the bacterial microbiota in the pathogenesis of respiratory diseases and helped in the discovery of microbiome-based diagnostics for PM-induced disorders.
Collapse
Affiliation(s)
- Huimin Ma
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Department of Stomatology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Zheng Dong
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Xu Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Conghe Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhihao Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xi Zhou
- Occupational Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250062, China; Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250062, China
| | - Jin He
- Occupational Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250062, China; Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong 250062, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| |
Collapse
|
33
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [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: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
34
|
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
|
35
|
Zheng X, Chen M, Zhuang Y, Zhao L, Qian Y, Shi C. Unveiling genetic links between gut microbiota and asthma: a Mendelian randomization. Front Microbiol 2024; 15:1448629. [PMID: 39372270 PMCID: PMC11449699 DOI: 10.3389/fmicb.2024.1448629] [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: 06/13/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
Background Multiple studies suggest a potential connection between the gut microbiome and asthma. Our objective is to use advanced genetic and metagenomic techniques to elucidate the causal relationships and underlying mechanisms between gut microbiota and asthma. Methods The study utilized comprehensive Linkage Disequilibrium Score Regression (LDSC) and Mendelian randomization (MR) analyses to examine the relationship between 119 gut microbiota genera and asthma, using publicly accessible genome-wide association studies (GWAS). The meta-analysis synthesized summary effect estimates obtained from LDSC, forward MR, and reverse MR. The MiBioGen collaboration, involving 18,340 individuals, identified genetic variations associated with gut bacteria. Asthma data were collected from the UK Biobank, FinnGen, and GERA, encompassing a total of 82,060 cases and 641,049 controls. Results LDSC analysis revealed significant negative genetic correlations between asthma and RuminococcaceaeUCG004 (Rg = -0.55, p = 7.66 × 10-5) and Subdoligranulum (Rg = -0.35, p = 3.61 × 10-4). Forward MR analysis suggested associations between Butyricicoccus (OR = 0.92, p = 0.01), Turicibacter (OR = 0.95, p = 0.025), Butyrivibrio (OR = 0.98, p = 0.047), and reduced asthma risk. Conversely, Coprococcus2 (OR = 1.10, p = 0.035) and Roseburia (OR = 1.07, p = 0.039) were associated with increased risk. Reverse MR analysis indicated significant associations between genetically predicted asthma and Eubacteriumxylanophilumgroup (Beta = -0.08, p = 9.25 × 10-7), LachnospiraceaeNK4A136group (Beta = -0.05, p = 1.26 × 10-4), and Eisenbergiella (Beta = 0.06, p = 0.015, Rg_P = 0.043). Conclusion The findings underscore significant genetic correlations and causal relationships between specific gut microbiota and asthma. These insights highlight the potential of gut microbiota as both markers and modulators of asthma risk, offering new avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - ChengCheng Shi
- Emergency Department, Wujin People’s Hospital Affiliated with Jiangsu University, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
36
|
Wang Z, Lloyd D, Zhao S, Motsinger-Reif A. Taxanorm: a novel taxa-specific normalization approach for microbiome data. BMC Bioinformatics 2024; 25:304. [PMID: 39285319 PMCID: PMC11406911 DOI: 10.1186/s12859-024-05918-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: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND In high-throughput sequencing studies, sequencing depth, which quantifies the total number of reads, varies across samples. Unequal sequencing depth can obscure true biological signals of interest and prevent direct comparisons between samples. To remove variability due to differential sequencing depth, taxa counts are usually normalized before downstream analysis. However, most existing normalization methods scale counts using size factors that are sample specific but not taxa specific, which can result in over- or under-correction for some taxa. RESULTS We developed TaxaNorm, a novel normalization method based on a zero-inflated negative binomial model. This method assumes the effects of sequencing depth on mean and dispersion vary across taxa. Incorporating the zero-inflation part can better capture the nature of microbiome data. We also propose two corresponding diagnosis tests on the varying sequencing depth effect for validation. We find that TaxaNorm achieves comparable performance to existing methods in most simulation scenarios in downstream analysis and reaches a higher power for some cases. Specifically, it balances power and false discovery control well. When applying the method in a real dataset, TaxaNorm has improved performance when correcting technical bias. CONCLUSION TaxaNorm both sample- and taxon- specific bias by introducing an appropriate regression framework in the microbiome data, which aids in data interpretation and visualization. The 'TaxaNorm' R package is freely available through the CRAN repository https://CRAN.R-project.org/package=TaxaNorm and the source code can be downloaded at https://github.com/wangziyue57/TaxaNorm .
Collapse
Affiliation(s)
- Ziyue Wang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, 27709, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Dillon Lloyd
- Department of Biological Sciences and Statistics, North Carolina State University, Raleigh, NC, 27695, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA
| | - Shanshan Zhao
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, 27709, USA
| | - Alison Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, 27709, USA.
| |
Collapse
|
37
|
Fu H, Gao Y. Correlation analysis of serum Rac1 level with asthma control, airway inflammatory response and lung function in asthmatic children. BMC Pulm Med 2024; 24:455. [PMID: 39285415 PMCID: PMC11406832 DOI: 10.1186/s12890-024-03266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE To investigate the correlation between serum Rac1 enzyme (Rac1) level with asthma control, airway inflammatory response and lung function in asthmatic children. METHODS A retrospective analysis was performed on 79 children with asthma who were diagnosed and treated in our hospital from June 2020 to January 2023. According to the severity of the disease, the children were divided into mild group (25 cases), moderate group (30 cases) and severe group (24 cases). 36 healthy children who underwent physical examination at the same period in our hospital were selected as the control group. The state of an illness, control level, serum mRNA Rac1, inflammatory factors, and lung function of the children in two groups were compared between the control group and the observation group. RESULTS The Rac1 mRNA levels, forced vital capacity (FVC), forced expiratory volume in one second/FVC (FEV1/FVC), peak expiratory flow (PEF), and maximum mid-expiratory flow (MMEF) in the observation group were significantly lower than these in the control group (P < 0.05). The tumor necrosis factor-alpha (TNF-α), interleukin-5 (IL-5), IL-6, and IL-33 in the observation group were markedly higher than these in the control group (P < 0.05). As the state of an illness worsened, the Rac1 mRNA levels, FVC, FEV1/FVC, PEF, and MMEF gradually reduced (P < 0.05), while the levels of TNF-α, IL-5, IL-6, and IL-33 increased (P < 0.05). As the degree of disease control improved, the Rac1 mRNA levels, FVC, FEV1/FVC, PEF, and MMEF gradually elevated (P < 0.05), and the levels of TNF- α, IL-5, IL-6, and IL-33 showed the opposite trend (P < 0.05). Rac1 was negatively related to the levels of TNF-α, IL-5, IL-6 and IL-33 (P < 0.05), and positively to the levels of FVC, FEV1/FVC, PEF and MMEF (P < 0.001). Rac1 mRNA levels, FVC, FEV1/FVC, PEF and MMEF were protective factors, while TNF-α, IL-5, IL-6 and IL-33 were risk factors for the prognosis of children with asthma (P < 0.05). CONCLUSION Children with asthma have obviously lower serum Rac1 mRNA levels, higher inflammatory factor levels and lower lung function. Serum Rac1 mRNA level may be associated with better asthma control, lower airway inflammatory response, better lung function and lower disease severity. It has important reference value for the evaluation of the state of an illness, efficacy and prognosis of children with bronchial asthma.
Collapse
Affiliation(s)
- Hui Fu
- Department of Pediatrics, The Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, 213000, China
| | - Yun Gao
- Department of Rehabilitation, Wujin Affiliated Hospital, Nanjing University of Chinese Medicine, 699 Renmin Middle Road, Hutang Town, Wujin District, Changzhou City, 213161, Jiangsu Province, China.
| |
Collapse
|
38
|
Dély S, Gerber V, Peters LM, Sage SE. Association between equine asthma and fungal elements in the tracheal wash: An environment-matched case-control study. PLoS One 2024; 19:e0309835. [PMID: 39240830 PMCID: PMC11379288 DOI: 10.1371/journal.pone.0309835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/19/2024] [Indexed: 09/08/2024] Open
Abstract
The presence of fungi in tracheal wash (TW) of horses was recently linked to mild-moderate equine asthma, indicating a possible causal role; however, increased numbers of fungi may also stem from asthma-related alteration of tracheal mucus clearance or from environmental exposure. Our objective was to elucidate the association between the presence of fungi in TW and asthma status while controlling for relevant confounders. We conducted a retrospective case-control study involving 73 horses, including 34 controls and 39 asthmatic cases. Each asthmatic horse was matched with a control from the same barn to account for the influence of environmental exposure. All horses underwent respiratory clinical scoring, endoscopy, TW, and bronchoalveolar lavage (BAL). The association between asthma status and presence of TW fungi was tested with multivariable logistic regression modelling, accounting for selected management factors, tracheal mucus accumulation, and selected TW and BAL cytological characteristics, including multinucleated giant cells (MGCs) in the TW. Given the variability in MGC definitions in the literature, particularly concerning their morphology and number of nuclei, we constructed two distinct models for each outcome (asthma status or presence of fungi in TW): one considering MGCs as cells with ≥ 3 nuclei, and another using a criterion of ≥ 10 nuclei. Horses with a tracheal mucus score ≥ 2 exhibited 3.6 to 4.3 higher odds of being asthmatic, depending on the MGC definition. None of the other variables examined were associated with either asthma status or TW fungi detection. Notably, the presence of fungal elements in the TW was not associated with equine asthma.
Collapse
Affiliation(s)
- Sarah Dély
- Vetsuisse Faculty, Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine (ISME), University of Bern, Bern, Switzerland
| | - Vinzenz Gerber
- Vetsuisse Faculty, Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine (ISME), University of Bern, Bern, Switzerland
| | - Laureen M Peters
- Vetsuisse Faculty, Department of Clinical Veterinary Medicine, Clinical Diagnostic Laboratory, University of Bern, Bern, Switzerland
| | - Sophie E Sage
- Vetsuisse Faculty, Department of Clinical Veterinary Medicine, Swiss Institute of Equine Medicine (ISME), University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol 2024; 15:1434804. [PMID: 39301033 PMCID: PMC11410641 DOI: 10.3389/fimmu.2024.1434804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
This comprehensive review undertakes a multidisciplinary exploration of the gut-lung axis, from the foundational aspects of anatomy, embryology, and histology, through the functional dynamics of pathophysiology, to implications for clinical science. The gut-lung axis, a bidirectional communication pathway, is central to understanding the interconnectedness of the gastrointestinal- and respiratory systems, both of which share embryological origins and engage in a continuous immunological crosstalk to maintain homeostasis and defend against external noxa. An essential component of this axis is the mucosa-associated lymphoid tissue system (MALT), which orchestrates immune responses across these distant sites. The review delves into the role of the gut microbiome in modulating these interactions, highlighting how microbial dysbiosis and increased gut permeability ("leaky gut") can precipitate systemic inflammation and exacerbate respiratory conditions. Moreover, we thoroughly present the implication of the axis in oncological practice, particularly in lung cancer development and response to cancer immunotherapies. Our work seeks not only to synthesize current knowledge across the spectrum of science related to the gut-lung axis but also to inspire future interdisciplinary research that bridges gaps between basic science and clinical application. Our ultimate goal was to underscore the importance of a holistic understanding of the gut-lung axis, advocating for an integrated approach to unravel its complexities in human health and disease.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Viktória Halasy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csenge Somodi
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Mógor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
40
|
Zhou J, Hou W, Zhong H, Liu D. Lung microbiota: implications and interactions in chronic pulmonary diseases. Front Cell Infect Microbiol 2024; 14:1401448. [PMID: 39233908 PMCID: PMC11372588 DOI: 10.3389/fcimb.2024.1401448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
The lungs, as vital organs in the human body, continuously engage in gas exchange with the external environment. The lung microbiota, a critical component in maintaining internal homeostasis, significantly influences the onset and progression of diseases. Beneficial interactions between the host and its microbial community are essential for preserving the host's health, whereas disease development is often linked to dysbiosis or alterations in the microbial community. Evidence has demonstrated that changes in lung microbiota contribute to the development of major chronic lung diseases, including chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), asthma, and lung cancer. However, in-depth mechanistic studies are constrained by the small scale of the lung microbiota and its susceptibility to environmental pollutants and other factors, leaving many questions unanswered. This review examines recent research on the lung microbiota and lung diseases, as well as methodological advancements in studying lung microbiota, summarizing the ways in which lung microbiota impacts lung diseases and introducing research methods for investigating lung microbiota.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang Hou
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huilin Zhong
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Passos FC, de Oliveira LMG, Jesus FR, Zanette DL, Neto OLL, Neves MCLC, Lemos ACM, Baccan GC. Beneficial Bacteria in the Gut Microbiota May Lead to Improved Metabolic and Immunological Status in Chronic Obstructive Pulmonary Disease. Med Sci (Basel) 2024; 12:41. [PMID: 39189204 PMCID: PMC11348168 DOI: 10.3390/medsci12030041] [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: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024] Open
Abstract
The progression of chronic obstructive pulmonary disease (COPD) is characterized by functional changes in the airways. The lung-gut axis and gut microbiota (GM) have been linked to the pathophysiology of airway diseases. Regarding COPD, studies have shown that GM alterations could be related the stages of this disease. However, the relationship between GM and clinical, biochemical and immunological parameters in patients with COPD are not well understood. The aim of this study was to compare the relative abundance of specific groups of beneficial gut bacteria between COPD patients and healthy controls (CTLs) in order to evaluate relationships with metabolic and inflammatory markers in COPD. METHODS We included 16 stable COPD patients and 16 healthy volunteer CTLs. The relative abundances of Bifidobacterium spp. (Bf) and Akkermansia muciniphila (Akk) bacteria and the Bacteroidetes and Firmicutes phyla were assessed by qPCR. Pulmonary function was evaluated by spirometry, biochemical parameters by colorimetric methods and plasma cytokine levels by cytometric bead array analysis. RESULTS The Firmicutes/Bacteroides ratio was related to emergency hospital visits and six-minute walk test (6MWT) results. Furthermore, the relative abundance of Bf was associated with plasma concentrations of glucose, triglycerides, HDL-C and IL-10. In addition, Firmicutes levels and the Firmicutes/Bacteroidetes ratio were associated with the IL-12/IL-10 ratio, while Akk abundance was linked to IL-12 levels. CONCLUSIONS The present findings suggest that the abundance of beneficial bacteria in the GM could influence clinical presentation and immunoregulation in COPD.
Collapse
Affiliation(s)
- Fabine Correia Passos
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Lucas Matheus Gonçalves de Oliveira
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Fabíola Ramos Jesus
- Maternidade Climério de Oliveira (MCO/EBSERH), Universidade Federal da Bahia, Salvador 40055-150, Bahia, Brazil;
| | | | - Odilon Lobão Leal Neto
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| | - Margarida Célia Lima Costa Neves
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil; (M.C.L.C.N.); (A.C.M.L.)
| | - Antônio Carlos Moreira Lemos
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil; (M.C.L.C.N.); (A.C.M.L.)
| | - Gyselle Chrystina Baccan
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40170-110, Bahia, Brazil; (F.C.P.); (L.M.G.d.O.); (O.L.L.N.)
| |
Collapse
|
42
|
Liu X, Li B, Liu S, Zong J, Zheng X. To investigate the function of age-related genes in different subtypes of asthma based on bioinformatics analysis. Heliyon 2024; 10:e34766. [PMID: 39144919 PMCID: PMC11320208 DOI: 10.1016/j.heliyon.2024.e34766] [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: 04/10/2024] [Revised: 07/01/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
Asthma is a heterogeneous airway inflammatory disease that can be classified according to the inflammatory phenotype. The pathogenesis, clinical features, response to hormone therapy, and prognosis of different inflammatory phenotypes differ significantly. This condition also refers to age-related chronic ailments. Here, we intend to identify the function of aging-related genes in different inflammatory phenotypes of asthma using bioinformatic analyses. Initially, the research adopted the GSEA analysis to understand the fundamental mechanisms that govern different inflammatory phenotypes of asthma pathogenesis and use the CIBERSORT algorithm to assess the immune cell composition. The differentially expressed genes (DEGs) of eosinophilic asthma (EA), neutrophilic asthma (NA), and paucigranulocytic asthma (PGA) were identified through the limma R package. Aging-related genes, screened from multiple databases, were intersected with DEGs of asthma to obtain the asthma-aging-related DEGs. Then, the GO and KEGG pathway enrichment analyses showed that the NA- and EA-aging-related DEGs are involved in the various cytokine-mediated signaling pathways. PPI network and correlation analysis were performed to identify and evaluate the correlation of the hub genes. Further, the clinical characteristics of asthma-aging-related DEGs were explored through ROC analysis. 3 and 12 aging-related DEGs in EA and NA patients show high diagnostic accuracy, respectively (AUC >0.7). This study provided valuable insights into aging-related gene therapy for phenotype-specific asthma. Moreover, the study suggests that effective interventions against asthma may operate by disrupting the detrimental cycle of "aging induces metabolic diseases, which exacerbate aging".
Collapse
Affiliation(s)
- Xinning Liu
- Central Laboratory, Clinical Laboratory and Qingdao Key Laboratory of Immunodiagnosis, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266034, China
| | - Bing Li
- Department of Neurology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266034, China
| | - Shuya Liu
- Department of Clinical Pharmacy, Qingdao Women and Children's Hospital, Qingdao, 266034, China
| | - Jinbao Zong
- Central Laboratory, Clinical Laboratory and Qingdao Key Laboratory of Immunodiagnosis, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266034, China
| | - Xin Zheng
- Department of Respiratory Medicine, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266034, China
| |
Collapse
|
43
|
Li N, Tan G, Xie Z, Chen W, Yang Z, Wang Z, Liu S, He M. Distinct enterotypes and dysbiosis: unraveling gut microbiota in pulmonary and critical care medicine inpatients. Respir Res 2024; 25:304. [PMID: 39127664 DOI: 10.1186/s12931-024-02943-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The gut-lung axis, pivotal for respiratory health, is inadequately explored in pulmonary and critical care medicine (PCCM) inpatients. METHODS Examining PCCM inpatients from three medical university-affiliated hospitals, we conducted 16S ribosomal RNA sequencing on stool samples (inpatients, n = 374; healthy controls, n = 105). We conducted statistical analyses to examine the gut microbiota composition in PCCM inpatients, comparing it to that of healthy controls. Additionally, we explored the associations between gut microbiota composition and various clinical factors, including age, white blood cell count, neutrophil count, platelet count, albumin level, hemoglobin level, length of hospital stay, and medical costs. RESULTS PCCM inpatients exhibited lower gut microbiota diversity than healthy controls. Principal Coordinates Analysis revealed marked overall microbiota structure differences. Four enterotypes, including the exclusive Enterococcaceae enterotype in inpatients, were identified. Although no distinctions were found at the phylum level, 15 bacterial families exhibited varying abundances. Specifically, the inpatient population from PCCM showed a significantly higher abundance of Enterococcaceae, Lactobacillaceae, Erysipelatoclostridiaceae, Clostridiaceae, and Tannerellaceae. Using random forest analyses, we calculated the areas under the receiver operating characteristic curves (AUCs) to be 0.75 (95% CIs 0.69-0.80) for distinguishing healthy individuals from inpatients. The four most abundant genera retained in the classifier were Blautia, Subdoligranulum, Enterococcus, and Klebsiella. CONCLUSIONS Evidence of gut microbiota dysbiosis in PCCM inpatients underscores the gut-lung axis's significance, promising further avenues in respiratory health research.
Collapse
Affiliation(s)
- Naijian Li
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guiyan Tan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zhiling Xie
- Department of Pulmonary and Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, People's Republic of China
| | - Weixin Chen
- Department of Chinese and Western Medicine in Clinical Medicine, The Clinical School of Chinese and Western Medicine, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhaowei Yang
- Department of Allergy and Clinical Immunology, State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhang Wang
- Biomedical Research Center, Institute of Ecological Sciences, School of Life Sciences, State Key Laboratory of Respiratory Disease, South China Normal University, Guangzhou, People's Republic of China
| | - Sha Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People's Republic of China.
| | - Mengzhang He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
44
|
Jaswal K, Todd OA, Flores Audelo RC, Santus W, Paul S, Singh M, Miao J, Underhill DM, Peters BM, Behnsen J. Commensal Yeast Promotes Salmonella Typhimurium Virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606421. [PMID: 39211098 PMCID: PMC11360897 DOI: 10.1101/2024.08.08.606421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Enteric pathogens engage in complex interactions with the host and the resident microbiota to establish gut colonization. Although mechanistic interactions between enteric pathogens and bacterial commensals have been extensively studied, whether and how commensal fungi affect pathogenesis of enteric infections remains largely unknown. Here we show that colonization with the common human gut commensal fungus Candida albicans worsened infections with the enteric pathogen Salmonella enterica serovar Typhimurium. Presence of C. albicans in the mouse gut increased Salmonella cecum colonization and systemic dissemination. We investigated the underlying mechanism and found that Salmonella binds to C. albicans via Type 1 fimbriae and uses its Type 3 Secretion System (T3SS) to deliver effector proteins into C. albicans . A specific effector, SopB, was sufficient to manipulate C. albicans metabolism, triggering increased arginine biosynthesis in C. albicans and the release of millimolar amounts of arginine into the extracellular environment. The released arginine, in turn, induced T3SS expression in Salmonella , increasing its invasion of epithelial cells. C. albicans deficient in arginine production was unable to increase Salmonella virulence in vitro or in vivo . In addition to modulating pathogen invasion, arginine also directly influenced the host response to infection. Arginine-producing C. albicans dampened the inflammatory response during Salmonella infection, whereas C. albicans deficient in arginine production did not. Arginine supplementation in the absence of C. albicans increased the systemic spread of Salmonella and decreased the inflammatory response, phenocopying the presence of C. albicans . In summary, we identified C. albicans colonization as a susceptibility factor for disseminated Salmonella infection, and arginine as a central metabolite in the cross-kingdom interaction between fungi, bacteria, and host.
Collapse
|
45
|
Radhouani M, Starkl P. Adjuvant-independent airway sensitization and infection mouse models leading to allergic asthma. FRONTIERS IN ALLERGY 2024; 5:1423938. [PMID: 39157265 PMCID: PMC11327155 DOI: 10.3389/falgy.2024.1423938] [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: 04/26/2024] [Accepted: 07/05/2024] [Indexed: 08/20/2024] Open
Abstract
Asthma is a chronic respiratory disease of global importance. Mouse models of allergic asthma have been instrumental in advancing research and novel therapeutic strategies for patients. The application of relevant allergens and physiological routes of exposure in such models has led to valuable insights into the complexities of asthma onset and development as well as key disease mechanisms. Furthermore, environmental microbial exposures and infections have been shown to play a fundamental part in asthma pathogenesis and alter disease outcome. In this review, we delve into physiological mouse models of allergic asthma and explore literature reports on most significant interplays between microbial infections and asthma development with relevance to human disease.
Collapse
Affiliation(s)
- Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
46
|
Vijay A, Valdes AM. Retraction Note: Role of the gut microbiome in chronic diseases: a narrative review. Eur J Clin Nutr 2024; 78:736. [PMID: 38926608 PMCID: PMC11300294 DOI: 10.1038/s41430-024-01467-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Amrita Vijay
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK.
| | - Ana M Valdes
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, The University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| |
Collapse
|
47
|
Kleniewska P, Pawliczak R. Can probiotics be used in the prevention and treatment of bronchial asthma? Pharmacol Rep 2024; 76:740-753. [PMID: 38951480 PMCID: PMC11294272 DOI: 10.1007/s43440-024-00618-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Asthma is a lifelong condition with varying degrees of severity and susceptibility to symptom control. Recent studies have examined the effects of individual genus, species, and strains of probiotic microorganisms on the course of asthma. The present review aims to provide an overview of current knowledge on the use of probiotic microorganisms, mainly bacteria of the genus Lactobacillus and Bifidobacterium, in asthma prevention and treatment. Recent data from clinical trials and mouse models of allergic asthma indicate that probiotics have therapeutic potential in this condition. Animal studies indicate that probiotic microorganisms demonstrate anti-inflammatory activity, attenuate airway hyperresponsiveness (AHR), and reduce airway mucus secretion. A randomized, double-blind, placebo-controlled human trials found that combining multi-strain probiotics with prebiotics yielded promising outcomes in the treatment of clinical manifestations of asthma. It appears that probiotic supplementation is safe and significantly reduces the frequency of asthma exacerbations, as well as improved forced expiratory volume and peak expiratory flow parameters, and greater attenuation of inflammation. Due to the small number of available clinical trials, and the use of a wide range of probiotic microorganisms and assessment methods, it is not possible to draw clear conclusions regarding the use of probiotics as asthma treatments.
Collapse
Affiliation(s)
- Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9, Łódź, 90-752, Poland.
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9, Łódź, 90-752, Poland
| |
Collapse
|
48
|
Guérin M, Lepeltier E. Nanomedicines via the pulmonary route: a promising strategy to reach the target? Drug Deliv Transl Res 2024; 14:2276-2297. [PMID: 38587757 DOI: 10.1007/s13346-024-01590-1] [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] [Accepted: 03/22/2024] [Indexed: 04/09/2024]
Abstract
Over the past decades, research on nanomedicines as innovative tools in combating complex pathologies has increased tenfold, spanning fields from infectiology and ophthalmology to oncology. This process has further accelerated since the introduction of SARS-CoV-2 vaccines. When it comes to human health, nano-objects are designed to protect, transport, and improve the solubility of compounds to allow the delivery of active ingredients on their targets. Nanomedicines can be administered by different routes, such as intravenous, oral, intramuscular, or pulmonary routes. In the latter route, nanomedicines can be aerosolized or nebulized to reach the deep lung. This review summarizes existing nanomedicines proposed for inhalation administration, from their synthesis to their potential clinical use. It also outlines the respiratory organs, their structure, and particularities, with a specific emphasis on how these factors impact the administration of nanomedicines. Furthermore, the review addresses the organs accessible through pulmonary administration, along with various pathologies such as infections, genetic diseases, or cancer that can be addressed through inhaled nanotherapeutics. Finally, it examines the existing devices suitable for the aerosolization of nanomedicines and the range of nanomedicines in clinical development.
Collapse
Affiliation(s)
- Mélina Guérin
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, 49000, Angers, France
| | - Elise Lepeltier
- Univ Angers, INSERM, CNRS, MINT, SFR ICAT, 49000, Angers, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
49
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
50
|
Deng X, Yang H, Tian L, Ling J, Ruan H, Ge A, Liu L, Fan H. Bibliometric analysis of global research trends between gut microbiota and breast cancer: from 2013 to 2023. Front Microbiol 2024; 15:1393422. [PMID: 39144230 PMCID: PMC11322113 DOI: 10.3389/fmicb.2024.1393422] [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: 02/29/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Background Breast cancer is the most prevalent cancer globally and is associated with significant mortality. Recent research has provided crucial insights into the role of gut microbiota in the onset and progression of breast cancer, confirming its impact on the disease's management. Despite numerous studies exploring this relationship, there is a lack of comprehensive bibliometric analyses to outline the field's current state and emerging trends. This study aims to fill that gap by analyzing key research directions and identifying emerging hotspots. Method Publications from 2013 to 2023 were retrieved from the Web of Science Core Collection database. The VOSviewer, R language and SCImago Graphica software were utilized to analyze and visualize the volume of publications, countries/regions, institutions, authors, and keywords in this field. Results A total of 515 publications were included in this study. The journal Cancers was identified as the most prolific, contributing 21 papers. The United States and China were the leading contributors to this field. The University of Alabama at Birmingham was the most productive institution. Peter Bai published the most papers, while James J. Goedert was the most cited author. Analysis of highly cited literature and keyword clustering confirmed a close relationship between gut microbiota and breast cancer. Keywords such as "metabolomics" and "probiotics" have been prominently highlighted in the keyword analysis, indicating future research hotspots in exploring the interaction between metabolites in the breast cancer microenvironment and gut microbiota. Additionally, these keywords suggest significant interest in the therapeutic potential of probiotics for breast cancer treatment. Conclusion Research on the relationship between gut microbiota and breast cancer is expanding. Attention should be focused on understanding the mechanisms of their interaction, particularly the metabolite-microbiota-breast cancer crosstalk. These insights have the potential to advance prevention, diagnosis, and treatment strategies for breast cancer. This bibliometric study provides a comprehensive assessment of the current state and future trends of research in this field, offering valuable perspectives for future studies on gut microbiota and breast cancer.
Collapse
Affiliation(s)
- Xianguang Deng
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hua Yang
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lingjia Tian
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Ling
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Ruan
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Anqi Ge
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hongqiao Fan
- Department of Cosmetic and Plastic Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|