1
|
Zhang W, Jia Q, Han M, Zhang X, Guo L, Sun S, Yin W, Bo C, Han R, Sai L. Bifidobacteria in disease: from head to toe. Folia Microbiol (Praha) 2024; 69:1-15. [PMID: 37644256 DOI: 10.1007/s12223-023-01087-3] [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] [Indexed: 08/31/2023]
Abstract
Bifidobacteria as a strictly anaerobic gram-positive bacteria, is widely distributed in the intestine, vagina and oral cavity, and is one of the first gut flora to colonize the early stages of life. Intestinal flora is closely related to health, and dysbiosis of intestinal flora, especially Bifidobacteria, has been found in a variety of diseases. Numerous studies have shown that in addition to maintaining intestinal homeostasis, Bifidobacteria may be involved in diseases covering all parts of the body, including the nervous system, respiratory system, genitourinary system and so on. This review collects evidence for the variation of Bifidobacteria in typical diseases among various systems, provides mild and effective therapeutic options for those diseases that are difficult to cure, and moves Bifidobacteria from basic research to further clinical applications.
Collapse
Affiliation(s)
- Weiliang Zhang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingming Han
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xin Zhang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Limin Guo
- Rongcheng Municipal Hospital of Traditional Chinese Medicine, Rongcheng, Shandong, China
| | - Shichao Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Shandong University of Traditional Chinese Medicine Doctoral candidate Class of 2022, Jinan, Shandong, China
| | - Wenhui Yin
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ru Han
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
2
|
Shin HJ, Yang WK, Lee YC, Kim S, Moon SO, Kwon YJ, Noh HJ, Kim KH, Kim BK, Shin CH, Chae MY, Yun SH, Kim SH. Protective effect of the mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt extract on respiratory disorders. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115856. [PMID: 38134637 DOI: 10.1016/j.ecoenv.2023.115856] [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: 10/11/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Air pollutants, such as particulate matter (PM) and diesel exhaust particles (DEP), are associated with respiratory diseases. Therefore, preventive and therapeutic strategies against PM-and DEP (PM10D)-induced respiratory diseases are needed. Herein, we evaluate the protective effects of a mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt (LJH) extract against airway inflammation associated with exposure to PM10D. To determine the anti-inflammatory effects of the LJH extract, reactive oxygen species (ROS) production and the expression of inflammatory pathways were determined in PM10-induced MH-S cells. For the respiratory protective effects, BALB/c mice were exposed to PM10D via intranasal injection, and a mixture of L. plantarum KC3 and LJH extract was administered orally for 12 days. LJH extract inhibited ROS production and the phosphorylation of downstream factors of NF-κB in PM10-stimulated MH-S cells. The mixture of L. plantarum KC3 and LJH repressed the infiltration of neutrophils, reduced the immune cells number, and suppressed the proinflammatory mediators and cyclooxygenase (COX)-2 expressions in PM10D-induced airway inflammation with reduced phosphorylation of downstream factors of NF-κB. In addition, these effects were not observed in an alveolar macrophage depleted PM10D-induced mouse model using clodronate liposomes. The extract mixture also regulated gut microbiota in feces and upregulated the mRNA expression of Foxp3, transforming growth factor (TGF)-β1, and interleukin (IL)-10 in the colon. The L. plantarum KC3 and LJH extract mixture may inhibit alveolar macrophage- and neutrophil-mediated inflammatory responses and regulate gut microbiota and immune response in PM10D-induced airway inflammation, suggesting it is a potential remedy to prevent and cure airway inflammation and respiratory disorders.
Collapse
Affiliation(s)
- Han Jae Shin
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Won-Kyung Yang
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Young Chul Lee
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Soeun Kim
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Sung Ok Moon
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Yoo Jin Kwon
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Hye-Ji Noh
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Kyung Hwan Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Byoung Kook Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Chang Hun Shin
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Min-Young Chae
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Su-Hyeon Yun
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea.
| |
Collapse
|
3
|
Gupta N, Abd EL-Gawaad N, Osman Abdallah SA, Al-Dossari M. Possible modulating functions of probiotic Lactiplantibacillus plantarum in particulate matter-associated pulmonary inflammation. Front Cell Infect Microbiol 2024; 13:1290914. [PMID: 38264731 PMCID: PMC10803600 DOI: 10.3389/fcimb.2023.1290914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Pulmonary disease represents a substantial global health burden. Increased air pollution, especially fine particulate matter (PM2.5) is the most concerned proportion of air pollutants to respiratory health. PM2.5 may carry or combine with other toxic allergens and heavy metals, resulting in serious respiratory allergies and anaphylactic reactions in the host. Available treatment options such as antihistamines, steroids, and avoiding allergens/dust/pollutants could be limited due to certain side effects and immense exposure to air pollutants, especially in most polluted countries. In this mini-review, we summarized how PM2.5 triggers respiratory hyperresponsiveness and inflammation, and the probiotic Lactiplantibacillus plantarum supplementation could minimize the risk of the same. L. plantarum may confer beneficial effects in PM2.5-associated pulmonary inflammation due to significant antioxidant potential. We discussed L. plantarum's effect on PM2.5-induced reactive oxygen species (ROS), inflammatory cytokines, lipid peroxidation, and DNA damage. Available preclinical evidence shows L. plantarum induces gut-lung axis, SCFA, GABA, and other neurotransmitter signaling via gut microbiota modulation. SCFA signals are important in maintaining lung homeostasis and regulating intracellular defense mechanisms in alveolar cells. However, significant research is needed in this direction to contemplate L. plantarum's therapeutic potential in pulmonary allergies.
Collapse
Affiliation(s)
- Nishant Gupta
- Medical Research and Development, River Engineering, Greater Noida, India
| | - N.S. Abd EL-Gawaad
- Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - M. Al-Dossari
- Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
4
|
Du B, Fu Y, Han Y, Sun Q, Xu J, Yang Y, Rong R. The lung-gut crosstalk in respiratory and inflammatory bowel disease. Front Cell Infect Microbiol 2023; 13:1218565. [PMID: 37680747 PMCID: PMC10482113 DOI: 10.3389/fcimb.2023.1218565] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023] Open
Abstract
Both lung and gut belong to the common mucosal immune system (CMIS), with huge surface areas exposed to the external environment. They are the main defense organs against the invasion of pathogens and play a key role in innate and adaptive immunity. Recently, more and more evidence showed that stimulation of one organ can affect the other, as exemplified by intestinal complications during respiratory disease and vice versa, which is called lung-gut crosstalk. Intestinal microbiota plays an important role in respiratory and intestinal diseases. It is known that intestinal microbial imbalance is related to inflammatory bowel disease (IBD), this imbalance could impact the integrity of the intestinal epithelial barrier and leads to the persistence of inflammation, however, gut microbial disturbances have also been observed in respiratory diseases such as asthma, allergy, chronic obstructive pulmonary disease (COPD), and respiratory infection. It is not fully clarified how these disorders happened. In this review, we summarized the latest examples and possible mechanisms of lung-gut crosstalk in respiratory disease and IBD and discussed the strategy of shaping intestinal flora to treat respiratory diseases.
Collapse
Affiliation(s)
- Baoxiang Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Fu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuxiu Han
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qihui Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinke Xu
- Shandong Center for Disease Control and Prevention, Jinan, China
| | - Yong Yang
- Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rong Rong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Zhang R, Chen S, Wang Z, Ye L, Jiang Y, Li M, Jiang X, Peng H, Guo Z, Chen L, Zhang R, Niu Y, Aschner M, Li D, Chen W. Assessing the Effects of Nicotinamide Mononucleotide Supplementation on Pulmonary Inflammation in Male Mice Subchronically Exposed to Ambient Particulate Matter. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:77006. [PMID: 37458712 PMCID: PMC10351503 DOI: 10.1289/ehp12259] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 03/27/2023] [Accepted: 06/16/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Chronic lung injury and dysregulated cellular homeostasis in response to particulate matter (PM) exposure are closely associated with adverse health effects. However, an effective intervention for preventing the adverse health effects has not been developed. OBJECTIVES This study aimed to evaluate the protective effects of nicotinamide mononucleotide (NMN) supplementation on lung injury and elucidate the mechanism by which NMN improved immune function following subchronic PM exposure. METHODS Six-week-old male C57BL/6J mice were placed in a real-ambient PM exposure system or filtered air-equipped chambers (control) for 16 wk with or without NMN supplementation in drinking water (regarded as Con-H2O, Exp-H2O, Con-NMN and Exp-NMN groups, respectively) in Shijiazhuang City, China (n=20/group). The effects of NMN supplementation (500mg/kg) on PM-induced chronic pulmonary inflammation were assessed, and its mechanism was characterized using single-cell transcriptomic sequencing (scRNA-seq) analysis of whole lung cells. RESULTS The NMN-treated mice exhibited higher NAD+ levels in multiple tissues. Following 16-wk PM exposure, slightly less pulmonary inflammation and less collagen deposition were noted in mice with NMN supplementation in response to real-ambient PM exposure (Exp-NMN group) compared with the Exp-H2O group (all p<0.05). Mouse lung tissue isolated from the Exp-NMN group was characterized by fewer neutrophils, monocyte-derived cells, fibroblasts, and myeloid-derived suppressor cells induced by subchronic PM exposure as detected by scRNA-seq transcriptomic analysis. The improved immune functions were further characterized by interleukin-17 signaling pathway inhibition and lower secretion of profibrotic cytokines in the Exp-NMN group compared with the Exp-H2O group. In addition, reduced proportions of differentiated myofibroblasts and profibrotic interstitial macrophages were identified in the NMN-supplemented mice in response to PM exposure. Furthermore, less immune function suppression and altered differentiation of pathological cell phenotypes NMN was related to intracellular lipid metabolism activation. DISCUSSION Our novel findings suggest that NMN supplementation mitigated PM-induced lung injury by regulating immune functions and improving lipid metabolism in male mice, providing a putative intervention method for prevention of human health effects associated with PM exposure. https://doi.org/10.1289/EHP12259.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ziwei Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Lizhu Ye
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yue Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Miao Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xinhang Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhanyu Guo
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yujie Niu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Cheng TY, Chang CC, Luo CS, Chen KY, Yeh YK, Zheng JQ, Wu SM. Targeting Lung-Gut Axis for Regulating Pollution Particle-Mediated Inflammation and Metabolic Disorders. Cells 2023; 12:901. [PMID: 36980242 PMCID: PMC10047528 DOI: 10.3390/cells12060901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Cigarette smoking (CS) or ambient particulate matter (PM) exposure is a risk factor for metabolic disorders, such as insulin resistance (IR), increased plasma triglycerides, hyperglycemia, and diabetes mellitus (DM); it can also cause gut microbiota dysbiosis. In smokers with metabolic disorders, CS cessation decreases the risks of serious pulmonary events, inflammation, and metabolic disorder. This review included recent studies examining the mechanisms underlying the effects of CS and PM on gut microbiota dysbiosis and metabolic disorder development; one of the potential mechanisms is the disruption of the lung-gut axis, leading to gut microbiota dysbiosis, intestinal dysfunction, systemic inflammation, and metabolic disease. Short-chain fatty acids (SCFAs) are the primary metabolites of gut bacteria, which are derived from the fermentation of dietary fibers. They activate G-protein-coupled receptor (GPCR) signaling, suppress histone deacetylase (HDAC) activity, and inhibit inflammation, facilitating the maintenance of gut health and biofunction. The aforementioned gut microbiota dysbiosis reduces SCFA levels. Treatment targeting SCFA/GPCR signaling may alleviate air pollution-associated inflammation and metabolic disorders, which involve lung-gut axis disruption.
Collapse
Affiliation(s)
- Tzu-Yu Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yun-Kai Yeh
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jing-Quan Zheng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
7
|
Bifidobacterium animalis A12 and Lactobacillus salivarius M18-6 Alleviate Alcohol Injury by keap1-Nrf2 Pathway and Thioredoxin System. Foods 2023; 12:foods12030439. [PMID: 36765968 PMCID: PMC9914461 DOI: 10.3390/foods12030439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Excessive drinking can significantly damage people's health and well-being. Although some lactic acid bacterial strains have been previously shown to alleviate the symptoms of alcohol injury, the mechanism underlying these effects remains unclear. The aim of this study was to establish an alcohol injury model and examine the protective effect and mechanism of B. animalis A12 and L. salivarius M18-6. The results showed that A12 freeze-dried powder could maintain the survival rate of mice with alcohol injury at 100%. Compared with Alco group, L. salivarius M18-6 dead cell improved the survival rate of mice, attenuated liver steatosis, and significantly down-regulated serum Alanine transaminase (ALT) level; at the same time, it activated keap1-Nrf2 signaling pathway and up-regulated Superoxide dismutase (SOD), it protects mouse liver cells from oxidative stress induced by alcohol injury. In addition, B. animalis A12 can reduce the stress response to short-term alcohol intake and improve the ability of anti-oxidative stress by upregulating the level of isobutyric acid, reducing the level of keap1 protein in the liver of mice and upregulating the expression of thioredoxin genes (Txnrd1, Txnrd3, Txn1). Taken together, the results showed that B. animalis A12 and L. salivarius M18-6 alleviate alcohol injury in mice through keap1-Nrf2 signaling pathway and thioredoxin system.
Collapse
|
8
|
Chiu YH, Chiu HP, Lin MY. Synergistic effect of probiotic and postbiotic on attenuation of PM2.5-induced lung damage and allergic response. J Food Sci 2023; 88:513-522. [PMID: 36463413 DOI: 10.1111/1750-3841.16398] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022]
Abstract
To date, few studies have been conducted on the relationship between postbiotics and air pollution, and there is limited knowledge if postbiotic and probiotic have synergistic effects. Therefore, we created a PM-induced lung inflammation mice model and demonstrated the effect of probiotic, postbiotic, and their combination treatment on attenuation of PM2.5-induced lung damage and allergic response. The mice were intratracheally given PM2.5 triggering conditions of acute lung damage and allergic response. Our results showed that individual treatment of probiotic and postbiotic reduced body weight loss by 47.1% and 48.9%, but the results did not show any effect on polarizing IFN-γ/IL-4 ratio. In addition, PM2.5-induced overactive expression of IgE treated by probiotic and postbiotic was reduced by 33.2% and 30.4%, respectively. While combination treatment of probiotic and postbiotic exerted a synergistic effect, especially considerably on improving IgE reduction by 57.1%, body weight loss by 78.3%, and IFN-γ/IL-4 ratio boost by 87.5%. To sum up the above functionality, these research findings may help establish a novel platform for postbiotic application, formulation, and mechanistic selection with regard to PM2.5-induced lung injury. PRACTICAL APPLICATION: Allergic inflammation caused by PM2.5 is not like common allergens (ex. Pollens, ovalbumin, dust mites), which simply skewing Th1/Th2 polarization to Th2. Thus using probiotics screened by Th1-skewing criteria might not be the best choice to treat on PM2.5-induced symptoms. This research proposed a combination of probiotics and postbiotics on modulating immunity homeostasis, and consequently attenuating complications of PM2.5-induced lung damage. These research findings may help establish a novel platform for postbiotic application, formulation and mechanistic selection with regard to PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Yi-Heng Chiu
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, ROC, Taiwan.,Chambio Co., Ltd., Taichung, ROC, Taiwan
| | | | - Meei-Yn Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, ROC, Taiwan
| |
Collapse
|
9
|
The Role of Gut Bacteriome in Asthma, Chronic Obstructive Pulmonary Disease and Obstructive Sleep Apnoea. Microorganisms 2022; 10:microorganisms10122457. [PMID: 36557710 PMCID: PMC9781820 DOI: 10.3390/microorganisms10122457] [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: 10/29/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The human body contains a very complex and dynamic ecosystem of bacteria. The bacteriome interacts with the host bi-directionally, and changes in either factor impact the entire system. It has long been known that chronic airway diseases are associated with disturbances in the lung bacteriome. However, less is known about the role of gut bacteriome in the most common respiratory diseases. Here, we aim to summarise the evidence concerning the role of the intestinal bacteriome in the pathogenesis and disease course of bronchial asthma, chronic obstructive pulmonary disease, and obstructive sleep apnea. Furthermore, we discuss the consequences of an altered gut bacteriome on the most common comorbidities of these lung diseases. Lastly, we also reflect on the therapeutic potential of influencing the gut microbiome to improve disease outcomes.
Collapse
|
10
|
Rocchi G, Giovanetti M, Benedetti F, Borsetti A, Ceccarelli G, Zella D, Altomare A, Ciccozzi M, Guarino MPL. Gut Microbiota and COVID-19: Potential Implications for Disease Severity. Pathogens 2022; 11:1050. [PMID: 36145482 PMCID: PMC9503814 DOI: 10.3390/pathogens11091050] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 pandemic resulted in an unprecedented global crisis. SARS-CoV-2 primarily causes lung infection trough the binding of the virus with the ACE-2 cell receptor located on the surface of the alveolar epithelial cells. Notably, ACE-2 cell receptors are also expressed in the epithelial cells of the intestinal tract (GI). Recent data showed that the microbial communities of the GI might act as local and systematic inflammatory modulators. Gastrointestinal symptoms, including diarrhea, are frequently observed in infected individuals, and recent released data indicate that SARS-CoV-2 may also spread by fecal-oral transmission. Moreover, the gut microbiota's ecosystem can regulate and be regulated by invading pathogens, including viruses, facilitating an effective immune response, which in turn results in less severe diseases. In this regard, increased SARS-CoV-2 mortality and morbidities appear to be frequently observed in elderly immunocompromised patients and in people with essential health problems, such as diabetes, who, indeed, tend to have a less diverse gut microbiota (dysbiosis). Therefore, it is important to understand how the interaction between the gut microbiota and SARS-CoV-2 might shape the intensity of the infection and different clinical outcomes. Here, we provide insights into the current knowledge of dysbiosis during SARS-CoV-2 infection and methods that may be used to re-establish a more correct microbiota composition.
Collapse
Affiliation(s)
- Giulia Rocchi
- Department of Science and Engineering for Human and the Environment, University of Campus Bio-Medico, 00128 Rome, Italy
| | - Marta Giovanetti
- Laboratorio de Flavivirus, lnstituto Oswaldo Cruz/Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Department of Science and Technology for Humans and the Environment, University of Campus Bio-Medico, 00128 Rome, Italy
| | - Francesca Benedetti
- Institute of Human Virology and Global Virus Network Center, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, Italy
| | - Davide Zella
- Institute of Human Virology and Global Virus Network Center, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Annamaria Altomare
- Department of Science and Technology for Humans and the Environment, University of Campus Bio-Medico, 00128 Rome, Italy
- Unit of Digestive Disease, Campus Bio-Medico University, 00128 Rome, Italy
| | - Massimo Ciccozzi
- Medical Statistic and Molecular Epidemiology Unit, University of Biomedical Campus, 00128 Rome, Italy
| | | |
Collapse
|
11
|
Yan X, Pan S, Dong X, Tan B, Li T, Huang W, Suo X, Li Z, Yang Y. Vitamin E amelioration of oxidative stress and low immunity induced by high-lipid diets in hybrid grouper (♀ Epinephelus fuscoguttatus × ♂ E. lanceolatu). FISH & SHELLFISH IMMUNOLOGY 2022; 124:156-163. [PMID: 35395411 DOI: 10.1016/j.fsi.2022.03.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/02/2022] [Accepted: 03/27/2022] [Indexed: 06/14/2023]
Abstract
The experiment was conducted to investigate the effects of vitamin E (VE) on growth, oxidative stress and immunity for hybrid grouper (♀ Epinephelus fuscoguttatus × ♂ E. lanceolatu) fed high-lipid diet. Six groups of iso-protein (50.23%) and iso-lipidic high-lipid (15.36%) experimental diets were prepared by adding 0 (basic diet control), 0.01%, 0.02%, 0.03%, 0.04%, 0.05% α-tocopherol respectively in basic diet. Each treatment consisted of 3 replicates and 30 fish (10.20 ± 0.02 g) in each replicate for 8 weeks. The results showed that: 1) compared with the control group, the growth performance of grouper was not affected by the addition of VE in high-lipid diet, but the specific growth rate (SGR) in high VE dose (0.6%) group were significantly decreased compared with 0.02% and 0.03% groups. 2) Adding VE to high-lipid diet can alleviate the hepatic oxidative damage caused by high-lipid diet, and significantly improve the serum and liver antioxidant enzyme activity. 3) Compared with the control group, appropriate VE significantly increased the expression of liver anti-inflammatory factors TGF-β and IL10, and significantly decreased the expression of proinflammatory factors IL8 and IL6. In conclusion, adding appropriate amount of VE into high-lipid diet can improve antioxidant capacity and immunity of grouper, we speculated that VE may alleviate lipid peroxidation by improving antioxidant capacity to reduce the inflammatory response. In combination with the results of the current study, we recommend an additional dose of 0.02%-0.03% of α-tocopherol in this experiment under high-lipid conditions.
Collapse
Affiliation(s)
- Xiaobo Yan
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Simiao Pan
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Xiaohui Dong
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, Guangdong, 524000, PR China.
| | - Beiping Tan
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China; Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang, Guangdong, 524000, PR China
| | - Tao Li
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Weibin Huang
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Xiangxiang Suo
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Zhihao Li
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China; Aquatic Animals Precision Nutrition and High Efficiency Feed Engineering Research Center of Guangdong Province, Zhanjiang, 524088, PR China
| | - Yuanzhi Yang
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, PR China
| |
Collapse
|
12
|
Qin Y, Zhang H, Jiang B, Chen J, Zhang T. Food bioactives lowering risks of chronic diseases induced by fine particulate air pollution: a comprehensive review. Crit Rev Food Sci Nutr 2022; 63:7811-7836. [PMID: 35317688 DOI: 10.1080/10408398.2022.2051162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Airborne particulate matter (PM) exerts huge negative impacts on human health worldwide, not only targeting the respiratory system but more importantly inducing and aggravating associated chronic diseases like asthma, lung cancer, atherosclerosis, diabetes mellitus and Alzheimer diseases. Food-derived bioactive compounds like vitamins, dietary polyphenols, omega-3 polyunsaturated fatty acids and sulforaphane are feasible alternative therapeutic approaches against PM-mediated potential health damages, drawing great attention in recent years. In this review, the association between PM exposure and risks of developing chronic diseases, and the detailed mechanisms underlying the detrimental effects of PM will be discussed. Subsequently, principal food-derived bioactive compounds, with emphasize on the preventative or protective effects against PM, along with potential mechanisms will be elucidated. This comprehensive review will discuss and present current research findings to reveal the nutritional intervention as a preventative or therapeutic strategy against ambient air pollution, thereby lowering the risk of developing chronic diseases.
Collapse
Affiliation(s)
- Yang Qin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Hua Zhang
- College of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Bo Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Jingjing Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
13
|
Chiu YHM, Carroll KN, Coull BA, Kannan S, Wilson A, Wright RJ. Prenatal Fine Particulate Matter, Maternal Micronutrient Antioxidant Intake, and Early Childhood Repeated Wheeze: Effect Modification by Race/Ethnicity and Sex. Antioxidants (Basel) 2022; 11:366. [PMID: 35204249 PMCID: PMC8868511 DOI: 10.3390/antiox11020366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/20/2023] Open
Abstract
Fine particulate matter (PM2.5) potentiates in utero oxidative stress influencing fetal development while antioxidants have potential protective effects. We examined associations among prenatal PM2.5, maternal antioxidant intake, and childhood wheeze in an urban pregnancy cohort (n = 530). Daily PM2.5 exposure over gestation was estimated using a satellite-based spatiotemporally resolved model. Mothers completed the modified Block98 food frequency questionnaire. Average energy-adjusted percentile intake of β-carotene, vitamins (A, C, E), and trace minerals (zinc, magnesium, selenium) constituted an antioxidant index (AI). Maternal-reported child wheeze was ascertained up to 4.1 ± 2.8 years. Bayesian distributed lag interaction models (BDLIMs) were used to examine time-varying associations between prenatal PM2.5 and repeated wheeze (≥2 episodes) and effect modification by AI, race/ethnicity, and child sex. Covariates included maternal age, education, asthma, and temperature. Women were 39% Black and 33% Hispanic, 36% with ≤high school education; 21% of children had repeated wheeze. Higher AI was associated with decreased wheeze in Blacks (OR = 0.37 (0.19-0.73), per IQR increase). BDLIMs identified a sensitive window for PM2.5 effects on wheeze among boys born to Black mothers with low AI (at 33-40 weeks gestation; OR = 1.74 (1.19-2.54), per µg/m3 increase in PM2.5). Relationships among prenatal PM2.5, antioxidant intake, and child wheeze were modified by race/ethnicity and sex.
Collapse
Affiliation(s)
- Yueh-Hsiu Mathilda Chiu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1057, New York, NY 10029, USA; (Y.-H.M.C.); (K.N.C.)
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kecia N. Carroll
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1057, New York, NY 10029, USA; (Y.-H.M.C.); (K.N.C.)
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brent A. Coull
- Department of Biostatistics, Harvard TH Chan School of Public Health, Harvard University, Boston, MA 02115, USA;
| | - Srimathi Kannan
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ander Wilson
- Department of Statistics, Colorado State University, Fort Collins, CO 80523, USA;
| | - Rosalind J. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, P.O. Box 1057, New York, NY 10029, USA; (Y.-H.M.C.); (K.N.C.)
- Kravis Children’s Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
14
|
Yiang G, Chen T, Chen C, Hung Y, Hsueh K, Wu T, Pan Y, Chien Y, Chen C, Yu Y, Wei C. Antioxidant vitamins promote anticancer effects on low-concentration methotrexate-treated glioblastoma cells via enhancing the caspase-3 death pathway. Food Sci Nutr 2021; 9:3308-3316. [PMID: 34136195 PMCID: PMC8194871 DOI: 10.1002/fsn3.2298] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/02/2023] Open
Abstract
Vitamin C and vitamin E are well-known antioxidant vitamins, both of which are also applied as adjunct treatments for cancer therapy. Methotrexate (MTX) is a clinical drug that is used widely for rheumatoid arthritis and cancer treatment. Human glioblastoma multiforme (GBM) is an aggressive malignant brain tumor; the mean survival time for GBM patients is <2 years with traditional therapies. Developing and investigating novel treatments are important for clinical GBM therapy. Therefore, the aim of this study was to investigate whether combined treatment with vitamin C/E and MTX can display anticancer activities on GBM. Our studies showed that MTX displays anticancer effects on GBM in a dose-dependent manner, while vitamins C and E are not cytotoxic to glioblastoma. Importantly, this study showed that vitamins C and E can promote anticancer effects on low-concentration methotrexate-treated glioblastoma. Additionally, this study suggested that MTX alone or combined with vitamins C/E inhibits GBM cell growth via the caspase-3 death pathway.
Collapse
Affiliation(s)
- Giou‐Teng Yiang
- Department of Emergency MedicineTaipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew TaipeiTaiwan
- Department of Emergency MedicineSchool of MedicineTzu Chi UniversityHualienTaiwan
| | - Tsu‐Yi Chen
- Department of Emergency MedicineTaipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew TaipeiTaiwan
| | - Cian Chen
- Department of Emergency MedicineTaipei Tzu Chi HospitalBuddhist Tzu Chi Medical FoundationNew TaipeiTaiwan
- Master Program of Biomedical NutritionDepartment of NutritionHung kuang UniversityTaichungTaiwan
| | - Yu‐Ting Hung
- Master Program of Biomedical NutritionDepartment of NutritionHung kuang UniversityTaichungTaiwan
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
| | - Kuan‐Chun Hsueh
- Department of SurgeryTungs' Taichung MetroHarbor HospitalTaichungTaiwan
| | - Tsai‐Kun Wu
- Division of Renal MedicineTungs' Taichung MetroHarbor HospitalTaichungTaiwan
| | - Ying‐Ru Pan
- Division of Renal MedicineTungs' Taichung MetroHarbor HospitalTaichungTaiwan
| | - Yi‐Chung Chien
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
- Drug Development CenterResearch Center for Cancer BiologyChina Medical UniversityTaichungTaiwan
- Center for Molecular MedicineChina Medical University HospitalTaichungTaiwan
| | - Chao‐Hsuan Chen
- Department of NeurosurgeryChina Medical University HospitalTaichungTaiwan
| | - Yung‑Lung Yu
- Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
- Drug Development CenterResearch Center for Cancer BiologyChina Medical UniversityTaichungTaiwan
- Center for Molecular MedicineChina Medical University HospitalTaichungTaiwan
- Ph.D. Program for Translational MedicineChina Medical UniversityTaichungTaiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichungTaiwan
- Department of Medical Laboratory Science and BiotechnologyAsia UniversityTaichungTaiwan
| | - Chyou‐Wei Wei
- Master Program of Biomedical NutritionDepartment of NutritionHung kuang UniversityTaichungTaiwan
| |
Collapse
|
15
|
Trushna T, Tripathi AK, Rana S, Tiwari RR. Nutraceuticals with anti-inflammatory and anti-oxidant properties as intervention for reducing the health effects of fine particulate matter: Potential and Prospects. Comb Chem High Throughput Screen 2021; 25:1639-1660. [PMID: 33845731 DOI: 10.2174/1386207324666210412121226] [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: 11/26/2020] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 11/22/2022]
Abstract
Air pollution, especially particulate matter pollution adversely affects human health. A growing pool of evidence has emerged which underscores the potential of individual-level nutritional interventions in attenuating the adverse health impact of exposure to PM2.5. Although controlling emission and reducing the overall levels of air pollution remains the ultimate objective globally, the sustainable achievement of such a target and thus consequent protection of human health will require a substantial amount of time and concerted efforts worldwide. In the meantime, smaller-scale individual-level interventions that can counter the inflammatory or oxidative stress effects triggered by exposure to particulate matter may be utilized to ameliorate the health effects of PM2.5 pollution. One such intervention is incorporation of nutraceuticals in the diet. Here, we present a review of the evidence generated from various in vitro, in vivo and human studies regarding the effects of different anti-inflammatory and antioxidant nutraceuticals in ameliorating the health effects of particulate matter air pollution. The studies discussed in this review suggest that these nutraceuticals when consumed as a part of the diet, or as additional supplementation, can potentially negate the cellular level adverse effects of exposure to particulate pollution. The potential benefits of adopting a non-pharmacological diet-based approach to air pollution-induced disease management have also been discussed. We argue that before a nutraceuticals-based approach can be used for widespread public adoption, further research, especially human clinical trials, is essential to confirm the beneficial action of relevant nutraceuticals and to explore the safe limits of human supplementation and the risk of side effects. Future research should focus on systematically translating bench-based knowledge regarding nutraceuticals gained from in-vitro and in-vivo studies into clinically usable nutritional guidelines.
Collapse
Affiliation(s)
- Tanwi Trushna
- Department of Environmental Health and Epidemiology, ICMR- National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Amit K Tripathi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Sindhuprava Rana
- Department of Bioinformatics, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Rajnarayan R Tiwari
- ICMR- National Institute for Research in Environmental Health (NIREH), Bhopal-462030, Madhya Pradesh. India
| |
Collapse
|
16
|
Asthma and air pollution: recent insights in pathogenesis and clinical implications. Curr Opin Pulm Med 2021; 26:10-19. [PMID: 31724961 DOI: 10.1097/mcp.0000000000000644] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Air pollution has adverse effects on the onset and morbidity of respiratory diseases, including asthma. In this review, we discuss recent insights into the effects of air pollution on the incidence and exacerbation of asthma. We focus on epidemiological studies that describe the association between air pollution exposure and development, mortality, persistence and exacerbations of asthma among different age groups. Moreover, we also provide an update on translational studies describing the mechanisms behind this association. RECENT FINDINGS Mechanisms linking air pollutants such as particulate matter, nitrogen dioxide (NO2) and ozone to the development and exacerbation of asthma include the induction of both eosinophilic and neutrophilic inflammation driven by stimulation of airway epithelium and increase of pro-inflammatory cytokine production, oxidative stress and DNA methylation changes. Although exposure during foetal development is often reported as a crucial timeframe, exposure to air pollution is detrimental in people of all ages, thus influencing asthma onset as well as increase in asthma prevalence, mortality, persistence and exacerbation. SUMMARY In conclusion, this review highlights the importance of reducing air pollution levels to avert the progressive increase in asthma incidence and morbidity.
Collapse
|
17
|
Daniel S, Phillippi D, Schneider LJ, Nguyen KN, Mirpuri J, Lund AK. Exposure to diesel exhaust particles results in altered lung microbial profiles, associated with increased reactive oxygen species/reactive nitrogen species and inflammation, in C57Bl/6 wildtype mice on a high-fat diet. Part Fibre Toxicol 2021; 18:3. [PMID: 33419468 PMCID: PMC7796587 DOI: 10.1186/s12989-020-00393-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Exposure to traffic-generated emissions is associated with the development and exacerbation of inflammatory lung disorders such as chronic obstructive pulmonary disorder (COPD) and idiopathic pulmonary fibrosis (IPF). Although many lung diseases show an expansion of Proteobacteria, the role of traffic-generated particulate matter pollutants on the lung microbiota has not been well-characterized. Thus, we investigated the hypothesis that exposure to diesel exhaust particles (DEP) can alter commensal lung microbiota, thereby promoting alterations in the lung's immune and inflammatory responses. We aimed to understand whether diet might also contribute to the alteration of the commensal lung microbiome, either alone or related to exposure. To do this, we used male C57Bl/6 mice (4-6-week-old) on either regular chow (LF) or high-fat (HF) diet (45% kcal fat), randomly assigned to be exposed via oropharyngeal aspiration to 35 μg DEP, suspended in 35 μl 0.9% sterile saline or sterile saline only (control) twice a week for 30 days. A separate group of study animals on the HF diet was concurrently treated with 0.3 g/day of Winclove Ecologic® Barrier probiotics in their drinking water throughout the study. RESULTS Our results show that DEP-exposure increases lung tumor necrosis factor (TNF)-α, interleukin (IL)-10, Toll-like receptor (TLR)-2, TLR-4, and the nuclear factor kappa B (NF-κB) histologically and by RT-qPCR, as well as Immunoglobulin A (IgA) and Immunoglobulin G (IgG) in the bronchoalveolar lavage fluid (BALF), as quantified by ELISA. We also observed an increase in macrophage infiltration and peroxynitrite, a marker of reactive oxygen species (ROS) + reactive nitrogen species (RNS), immunofluorescence staining in the lungs of DEP-exposed and HF-diet animals, which was further exacerbated by concurrent DEP-exposure and HF-diet consumption. Histological examinations revealed enhanced inflammation and collagen deposition in the lungs DEP-exposed mice, regardless of diet. We observed an expansion of Proteobacteria, by qPCR of bacterial 16S rRNA, in the BALF of DEP-exposed mice on the HF diet, which was diminished with probiotic-treatment. CONCLUSIONS Our findings suggest that exposure to DEP causes persistent and sustained inflammation and bacterial alterations in a ROS-RNS mediated fashion, which is exacerbated by concurrent consumption of an HF diet.
Collapse
Affiliation(s)
- Sarah Daniel
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Leah J Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Kayla N Nguyen
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Julie Mirpuri
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Amie K Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA.
| |
Collapse
|
18
|
Naringenin inhibits migration of breast cancer cells via inflammatory and apoptosis cell signaling pathways. Inflammopharmacology 2019; 27:1021-1036. [PMID: 30941613 DOI: 10.1007/s10787-018-00556-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Naringenin, a flavonoid compound, has a wide variety of uses in the pharmaceutical industry for its antioxidant and anti-inflammatory potential. OBJECTIVES The current experiment aimed to investigate the anticancer effect of naringenin in triple-negative human breast cancer cells (MDA-MR-231) and an animal model with 7,12-dimethylbenz[a] anthracene (DMBA)-induced breast cancer in female rats to determine the mechanisms and molecular targets. METHODS The cytotoxic effects of naringenin against MDA-MB-231 cells were assessed by MTT assay. Apoptosis and cell cycle alterations were analyzed via flow cytometry. Morphological and biochemical changes in DMBA-induced cancer with naringenin treatment were assayed using our protocol. The potential mechanisms of action were verified via qRT-PCR. RESULTS Naringenin was found to inhibit cell proliferation in a time- and concentration-dependent manner. This effect was associated with cell cycle arrest at the G0/G1 phase, along with apoptosis and deposition at the sub-G1 phase (75%). Treatment with naringenin reduced tumor incidence (45.55, 40, and 27.67%) and tumor burden (78.7, 35.4, and 1.2 g) in a dose-dependent manner. Naringenin treatment altered the biochemical and antioxidant parameters related to inflammation necessary for anticancer activity. The qRT-PCR studies further confirmed the mitochondrial-mediated apoptotic effects of naringenin. CONCLUSION On the basis of these results, we can conclude that naringenin exerts an anticancer effect in the MDA-MB-231 cell line that arrests cell development at the G0/G1 phase, and in vivo it alters the mitochondrial-mediated intrinsic pathway responsible for apoptosis.
Collapse
|