1
|
Zhang S, Zhou R, Xie X, Xiong S, Li L, Li Y. Polysaccharides from Lycium barbarum, yam, and sunflower ameliorate colitis in a structure and intrinsic flora-dependent manner. Carbohydr Polym 2025; 349:122905. [PMID: 39643421 DOI: 10.1016/j.carbpol.2024.122905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Polysaccharides have been suggested to ameliorate metabolic diseases. However, their differential colitis-mitigating effects in mouse models with different colony structures remain poorly understood. Therefore, this study investigated the effects of polysaccharides from Lycium barbarum (LBP), sunflower (SP), and yam (YP) on colitis in C57BL/6 J (B6) mice born via vaginal delivery (VD) and in both caesarean section (CS)- and VD-born Institute of Cancer Research (ICR) mice. LBP was mainly composed of glucose (30.2 %), galactose (27.5 %), and arabinose (26.9 %). The main components of SP and YP were galacturonic acid (75.8 %) and glucose (98.1 %), respectively. Interestingly, LBP effectively alleviated body weight loss, reduced inflammatory cytokine levels, and restored intestinal barrier function in all three mouse models. Moreover, LBP decreased the abundance of norank_f__norank_o__Clostridia_UCG-014, Coriobacteriaceae_UCG-002, and norank_f_Eubacterium_coprostanoligenes_group in B6 mice, and the abundance of these genera positively correlated with pro-inflammatory cytokine levels. LBP increased the abundance of Lactobacillus, which was positively correlated with the levels of the protective factor, IL-10, in CS-born ICR mice. Collectively, our study suggests the potential application of LBP in the treatment of ulcerative colitis. We also provide an alternative method for restoring intestinal homeostasis in CS-born offspring.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ruchen Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoran Xie
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shanshan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Liu S, Li J, Zhang Y, Wang C, Zhang L. IL-10: the master immunomodulatory cytokine in allergen immunotherapy. Expert Rev Clin Immunol 2025; 21:17-28. [PMID: 39323099 DOI: 10.1080/1744666x.2024.2406894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Allergen immunotherapy (AIT) is the only disease-modifying treatment for patients with IgE-mediated allergic diseases. Successful AIT can induce long-term immune tolerance to the common allergen, which provides clinical benefits for years after discontinuation. The cytokine interleukin (IL)-10, as a key anti-inflammatory mediator with strong immunoregulatory functions, has drawn increasing attention over the past decades. AREAS COVERED After an extensive search of PubMed, EMBASE, and Web of Science databases, covering articles published from 1989 to 2024, our review aims to emphasize the key common information from previous reviews on the crucial involvement of IL-10 in allergen immunotherapy (AIT) induced immunological tolerance. In this review, we discuss the regulation of IL-10 expression and the molecular pathways associated with IL-10 function. We also further summarize mechanisms of immune tolerance induced by AIT, especially the indispensable role of IL-10 in AIT. EXPERT OPINION IL-10 plays an indispensable role in immune tolerance induced by AIT. Understanding the importance of the role of IL-10 in AIT would help us comprehend the mechanisms thoroughly and develop targeted therapeutics for allergic diseases.
Collapse
Affiliation(s)
- Shixian Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyun Li
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Chengshuo Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Wei TT, Gao K, Tai JH, Wei YJ, Zhan B. A study of specific immunoglobulin G4 expression in allergic rhinitis and its value in assessing efficacy and in predicting prognosis of sublingual immunotherapy. Kaohsiung J Med Sci 2024:e12916. [PMID: 39739782 DOI: 10.1002/kjm2.12916] [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: 08/15/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Allergic rhinitis (AR) is a widespread health issue with a rising global prevalence, and sublingual immunotherapy (SLIT) has shown efficacy in AR treatment. We examined specific immunoglobulin G4 (sIgG4) expression in AR and its role in evaluating SLIT efficacy and predicting patient prognosis. We compared total nasal symptom score (TNSS), total medication score (TMS), visual analogue scale (VAS) score, inflammatory cytokines, and immune function markers in AR patients before and after SLIT. SLIT reduced TNSS, TMS, VAS scores, IL-4, IL-17, eosinophilia percentage (EOS%), and specific immunoglobulin E (sIgE) levels, while increasing INF-γ, IL-10, and sIgG4. The sIgG4 level at pre-treatment and 12 months post-treatment was negatively correlated with TNSS, TMS, VAS score, IL-4, IL-17, EOS%, and sIgE, and positively correlated with IFN-γ and IL-10. Most patients showed symptomatic improvement. After 12 months, sIgG4 level demonstrated an area under the curve (AUC) of 0.867 for assessing SLIT as effective. Pre-treatment sIgG4 level showed an AUC of 0.869 for predicting SLIT as effective. Collectively, sIgG4 has strong potential assessing SLIT efficacy and prognosis in AR patients, with correlations to TNSS, TMS, VAS score, and IL-4, IL-10, IL-17, INF-γ, EOS% and sIgE levels.
Collapse
Affiliation(s)
- Ting-Ting Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian Province, China
| | - Kai Gao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian Province, China
| | - Jun-Hu Tai
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian Province, China
| | - Yong-Jun Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian Province, China
| | - Bin Zhan
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian Province, China
| |
Collapse
|
4
|
Cui Q, Zheng X, Bai Y, Guo Y, Liu S, Lu Y, Liu L, Song J, Liu Y, Heng BC, You F, Xu M, Deng X, Zhang X. Manipulation of Surface Potential Distribution Enhances Osteogenesis by Promoting Pro-Angiogenic Macrophage Polarization via Activation of the PI3K-Akt Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2414278. [PMID: 39739591 DOI: 10.1002/advs.202414278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/07/2024] [Indexed: 01/02/2025]
Abstract
Regulation of the immune response is key to promoting bone regeneration by electroactive biomaterials. However, how electrical signals at the micro- and nanoscale regulate the immune response and subsequent angiogenesis during bone regeneration remains to be elucidated. Here, the distinctly different surface potential distributions on charged poly(vinylidene fluoridetrifluoroethylene) (P(VDF-TrFE)) matrix surfaces are established by altering the dimensions of ferroelectric nanofillers from 0D BaTiO3 nanoparticles (homogeneous surface potential distribution, HOPD) to 1D BaTiO3 nanofibers (heterogeneous surface potential distribution, HEPD). Compared to HOPD, HEPD is significantly better at inducing the M2 polarization of macrophages and promoting neovascularization, which results in accelerated bone regeneration in vivo. The transcriptomic analysis reveals that macrophages modulated by HEPD display high expression levels of pro-angiogenic genes, which is corroborated by tube-formation assays, RT-qPCR, and western blot analyses in vitro. Mechanistic explorations elucidate activation of the PI3K-Akt signaling pathway, which in turn induces the polarization of macrophages toward a pro-angiogenic phenotype. This study elucidates the cascade of biological processes by which heterogeneous electrical signals at the micro- and nanoscale modulate macrophage functions to promote vascularization and bone regeneration. Hence, this study provides new insights into how the micro- and nanoscale distribution characteristics of electrical signals facilitate bone regeneration.
Collapse
Affiliation(s)
- Qun Cui
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xiaona Zheng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Oral Translational Medicine Research Center, Joint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial Repair, Reconstruction and Regeneration, The First People's Hospital of Jinzhong, Jinzhong, Shanxi, 030600, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Shuo Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Lulu Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Jia Song
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yang Liu
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Fuping You
- Institute of Systems Biomedicine, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xuliang Deng
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Oral Translational Medicine Research Center, Joint Training base for Shanxi Provincial Key Laboratory in Oral and Maxillofacial Repair, Reconstruction and Regeneration, The First People's Hospital of Jinzhong, Jinzhong, Shanxi, 030600, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| |
Collapse
|
5
|
El-Samad LM, Maklad AM, Elkady AI, Hassan MA. Unveiling the mechanism of sericin and hydroxychloroquine in suppressing lung oxidative impairment and early carcinogenesis in diethylnitrosamine-induced mice by modulating PI3K/Akt/Nrf2/NF-κB signaling pathway. Biomed Pharmacother 2024; 182:117730. [PMID: 39671723 DOI: 10.1016/j.biopha.2024.117730] [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: 07/13/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024] Open
Abstract
This study sheds light on the ameliorative influence of combined sericin and hydroxychloroquine (HQ) on mitigating diethylnitrosamine (DEN)-induced lung oxidative impairment and inflammation, thereby precluding early carcinogenic episodes in mice. Besides, the pivotal role of sericin and HQ in controlling the PI3K/Akt/Nrf2/NF-κB signaling pathway was probed. Therefore, male Swiss albino mice were assigned to different groups and treated with different drugs. Oxidative stress and inflammatory biomarkers, in addition to the expression of PI3K and Akt genes were evaluated in lung tissues. Treatment with DEN disturbed the redox homeostasis associated with inflammation in the lungs. Conversely, sericin combined with HQ remarkably upregulated Nrf2 expression in the lungs associated with significant ameliorations of antioxidant factors, including SOD, GST, GSH, and MDA. Furthermore, sericin and HQ abated inflammation instigated by DEN through downregulating NF-κB and inflammatory biomarkers, including TNF-α and IL-6, with an increase in IL-10. Importantly, sericin and HQ treatment significantly downregulated PI3K and Akt expression. Immunohistochemical investigations demonstrated marked diminutions in Ki-67 and p53 expressions in animals cotreated with sericin and HQ compared to the DEN-treated group, inhibiting lung cancer progression. Histopathological and ultrastructural anomalies were detected in lung tissues from the DEN group, while significant enhancements were perceived in lung tissues treated with sericin and HQ. Our findings emphasized that the combinatorial therapy of sericin and HQ could orchestrate the PI3K/Akt/Nrf2/NF-κB signaling pathway in the lungs, counteracting oxidative stress, inflammation, and uncontrolled cellular proliferation and sustaining lung structures. Furthermore, they could serve as anticancer agents, hindering lung cancer progression.
Collapse
Affiliation(s)
- Lamia M El-Samad
- Department of Zoology, Faculty of Science, Alexandria University, Egypt
| | - Alaa M Maklad
- Department of Zoology, Faculty of Science, Alexandria University, Egypt
| | - Ayman I Elkady
- Department of Zoology, Faculty of Science, Alexandria University, Egypt; Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohamed A Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria 21934, Egypt.
| |
Collapse
|
6
|
Niu Q, Liang LM, Ye SY, Lian CY, Li Q, Feng X, Chen SJ, Wang M, Zheng YY, Cui XL, Zhao LQ, Jia ZH, Hu SH, Cheng PP, Cai PC, Ye H, Ma WL. IL-10 mediates pleural remodeling in systemic lupus erythematosus. Cell Commun Signal 2024; 22:554. [PMID: 39563376 PMCID: PMC11575414 DOI: 10.1186/s12964-024-01911-4] [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: 04/17/2024] [Accepted: 10/27/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Interleukin-10 (IL-10), a pivotal anti-inflammatory cytokine, has gotten attention for its involvement in tissue remodeling and organ fibrosis. Pleurisy and subsequent pleural remodeling are recognized as quantifiable indicators of systemic lupus erythematosus (SLE) activity. However, the role of IL-10 in SLE-associated pleural remodeling remains unknown. In this study, we investigated role of IL-10 in SLE-associated pleural remodeling and the underlying mechanism. METHODS Clinical data and serum specimens were obtained from SLE patients, while pleural mesothelial cells and mouse models served as primary experimental subjects. The protein expression-related technologies, histopathological staining, and other experimental methods were used in the study. RESULTS Our investigation got several key findings. Firstly, serum obtained from SLE patients with pleural thickening was found to induce pleural mesothelial cell remodeling. Subsequently, heightened levels of IL-10 were found in serum from SLE patients with pleural thickening compared to that of SLE patients without pleural thickening. Secondly, administration of recombinant IL-10 was confirmed its ability to induce pleural mesothelial cell remodeling, on the contrary, this remodeling was effectively mitigated by IL-10 inhibition. Notably, blockade of IL-10 significantly prevented collagen deposition and prevented thickening in pleura of SLE mouse models. Lastly, the IL-10/JAK2/STAT3/HIF1α/TMEM45A/P4HA1 signaling axis was elucidated to mediate pleural remodeling and thickening. CONCLUSIONS Our study uncovered that IL-10 mediated pleural remodeling in SLE. We suggested that serum IL-10 level exceeding 6.32 pg/mL was a potential reference threshold for predicting pleural thickening in SLE patients.
Collapse
Affiliation(s)
- Qian Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, 430030, China
| | - Shu-Yi Ye
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen-Yue Lian
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuai-Jun Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan-Yi Zheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Lin Cui
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Qin Zhao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zi-Heng Jia
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shi-He Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peng-Cheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hong Ye
- Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, 430030, China.
| |
Collapse
|
7
|
Zhao J, Zhang G, Yang J, Qi X, Yao F, Gao Y, Li C, Liu L, Kang L. Surface proteins of Bifidobacterium bifidum DNG6 growing in 2'-fucosyllactose alleviating lipopolysaccharide-induced intestinal barrier injury in vitro. J Dairy Sci 2024; 107:8865-8873. [PMID: 38969003 DOI: 10.3168/jds.2024-25019] [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: 04/17/2024] [Accepted: 06/07/2024] [Indexed: 07/07/2024]
Abstract
Human milk oligosaccharides promote the growth and adhesion of Bifidobacteria, thus exerting multiple biological functions on intestinal epithelial cells. Bacterial surface proteins play an important role in bacterial-host intestinal epithelial interactions. In this study, we aimed to investigate the effects of surface proteins extracted from Bifidobacterium bifidum DNG6 (B. bifidum DNG6) consuming 2'-fucosyllactose (2'-FL) on Caco-2 cells monolayer barrier injury induced by lipopolysaccharide, compared with lactose and galacto-oligosaccharides. Our results indicated that 2'-FL may promote the surface proteins of B. bifidum DNG6 to improve intestinal barrier injury by positively regulating the NF-κB signaling pathway, reducing inflammation (TNF-α reduced by 50.34%, IL-6 reduced by 22.83%, IL-1β reduced by 37.91%, and IL-10 increased by 63.47%) and strengthening tight junction proteins (ZO,1 2.39×; claudin,1 2.79×; and occluding, 4.70×). The findings of this study indicate that 2'-FL can further regulate intestinal barrier damage by promoting the alteration of B. bifidum DNG6 surface proteins. The findings of this research will also provide theoretical support for the development of synbiotic formulations.
Collapse
Affiliation(s)
- Jingjing Zhao
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Guofang Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jingbo Yang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoxi Qi
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Fei Yao
- Synaura Biotechnology (Shanghai) Co., Ltd., Shanghai 200120, China
| | - Yunfeng Gao
- Heilongjiang Agricultural Products and Veterinary Medicine and Feed Technology Identification Station, 150036, China
| | - Chun Li
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| | - Libo Liu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| | - Linhui Kang
- Synaura Biotechnology (Shanghai) Co., Ltd., Shanghai 200120, China
| |
Collapse
|
8
|
Bohlen J, Bagarić I, Vatovec T, Ogishi M, Ahmed SF, Cederholm A, Buetow L, Sobrino S, Le Floc’h C, Arango-Franco CA, Seabra L, Michelet M, Barzaghi F, Leardini D, Saettini F, Vendemini F, Baccelli F, Catala A, Gambineri E, Veltroni M, Aguilar de la Red Y, Rice GI, Consonni F, Berteloot L, Largeaud L, Conti F, Roullion C, Masson C, Bessot B, Seeleuthner Y, Le Voyer T, Rinchai D, Rosain J, Neehus AL, Erazo-Borrás L, Li H, Janda Z, Cho EJ, Muratore E, Soudée C, Lainé C, Delabesse E, Goulvestre C, Ma CS, Puel A, Tangye SG, André I, Bole-Feysot C, Abel L, Erlacher M, Zhang SY, Béziat V, Lagresle-Peyrou C, Six E, Pasquet M, Alsina L, Aiuti A, Zhang P, Crow YJ, Landegren N, Masetti R, Huang DT, Casanova JL, Bustamante J. Autoinflammation in patients with leukocytic CBL loss of heterozygosity is caused by constitutive ERK-mediated monocyte activation. J Clin Invest 2024; 134:e181604. [PMID: 39403923 PMCID: PMC11475086 DOI: 10.1172/jci181604] [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: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Patients heterozygous for germline CBL loss-of-function (LOF) variants can develop myeloid malignancy, autoinflammation, or both, if some or all of their leukocytes become homozygous for these variants through somatic loss of heterozygosity (LOH) via uniparental isodisomy. We observed an upregulation of the inflammatory gene expression signature in whole blood from these patients, mimicking monogenic inborn errors underlying autoinflammation. Remarkably, these patients had constitutively activated monocytes that secreted 10 to 100 times more inflammatory cytokines than those of healthy individuals and CBL LOF heterozygotes without LOH. CBL-LOH hematopoietic stem and progenitor cells (HSPCs) outgrew the other cells, accounting for the persistence of peripheral monocytes homozygous for the CBL LOF variant. ERK pathway activation was required for the excessive production of cytokines by both resting and stimulated CBL-LOF monocytes, as shown in monocytic cell lines. Finally, we found that about 1 in 10,000 individuals in the UK Biobank were heterozygous for CBL LOF variants and that these carriers were at high risk of hematological and inflammatory conditions.
Collapse
Affiliation(s)
- Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Ivan Bagarić
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Taja Vatovec
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Syed F. Ahmed
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lori Buetow
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Steicy Sobrino
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Chromatin and Gene Regulation during Development, Paris Cité University, INSERM U1163, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Corentin Le Floc’h
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Carlos A. Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Luis Seabra
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Marine Michelet
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Leardini
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Francesco Saettini
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Francesco Baccelli
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Albert Catala
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Eleonora Gambineri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Marinella Veltroni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | | | - Gillian I. Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Filippo Consonni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
- “Mario Serio” Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Laureline Berteloot
- Department of Pediatric Imaging, Necker Hospital for Sick Children, Paris, France
- INSERM U1163, Paris, France
| | - Laetitia Largeaud
- Laboratory of Hematology, Hospital Center of the University of Toulouse, Toulouse, France
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Cécile Roullion
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Cécile Masson
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Bioinformatic Plateform, INSERM U1163 and INSERM US24/CNRS UAR3633, Paris Cité University, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Lucia Erazo-Borrás
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Zarah Janda
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - En-Jui Cho
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Edoardo Muratore
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Candice Lainé
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Eric Delabesse
- Department of Hematology, CHU and Centre de Recherche de Cancérologie de Toulouse, Paul-Sabatier University, Toulouse, France
| | | | - Cindy S. Ma
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Isabelle André
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Christine Bole-Feysot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Emmanuelle Six
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Marlène Pasquet
- Department of Pediatric Hematology and Oncology, Centre Hospitalo–Universitaire de Toulouse, Toulouse, France
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Yanick J. Crow
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Riccardo Masetti
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Danny T. Huang
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Necker Hospital for Sick Children–AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| |
Collapse
|
9
|
Xu Y, Zhu F, Zhou Z, Ma S, Zhang P, Tan C, Luo Y, Qin R, Chen J, Pan P. A novel mRNA multi-epitope vaccine of Acinetobacter baumannii based on multi-target protein design in immunoinformatic approach. BMC Genomics 2024; 25:791. [PMID: 39160492 PMCID: PMC11334330 DOI: 10.1186/s12864-024-10691-7] [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: 05/13/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
Acinetobacter baumannii is a gram-negative bacillus prevalent in nature, capable of thriving under various environmental conditions. As an opportunistic pathogen, it frequently causes nosocomial infections such as urinary tract infections, bacteremia, and pneumonia, contributing to increased morbidity and mortality in clinical settings. Consequently, developing novel vaccines against Acinetobacter baumannii is of utmost importance. In our study, we identified 10 highly conserved antigenic proteins from the NCBI and UniProt databases for epitope mapping. We subsequently screened and selected 8 CTL, HTL, and LBL epitopes, integrating them into three distinct vaccines constructed with adjuvants. Following comprehensive evaluations of immunological and physicochemical parameters, we conducted molecular docking and molecular dynamics simulations to assess the efficacy and stability of these vaccines. Our findings indicate that all three multi-epitope mRNA vaccines designed against Acinetobacter baumannii are promising; however, further animal studies are required to confirm their reliability and effectiveness.
Collapse
Affiliation(s)
- Yizhong Xu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fei Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyou Zhou
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shiyang Ma
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peipei Zhang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Caixia Tan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuying Luo
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rongliu Qin
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China.
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China.
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Sarmiento-Ortega VE, Moroni-González D, Diaz A, Brambila E, Treviño S. Curcumin Treatment Ameliorates Hepatic Insulin Resistance Induced by Sub-chronic Oral Exposure to Cadmium LOAEL Dose via NF-κB and Nrf2 Pathways. Biol Trace Elem Res 2024:10.1007/s12011-024-04314-1. [PMID: 39103711 DOI: 10.1007/s12011-024-04314-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/12/2024] [Indexed: 08/07/2024]
Abstract
Cadmium (Cd) is a global pollutant, and its accumulation in the liver causes oxidative stress, inflammation, insulin resistance (IR), and metabolic complications. This study investigated whether curcumin treatment could alleviate hepatic IR in Wistar rats exposed to sub-chronic cadmium and explored the underlying molecular pathways. Male Wistar rats were divided into a control group (standard normocaloric diet + cadmium-free water) and a cadmium group (standard normocaloric diet + drinking water with 32.5 ppm CdCl2) for 30 days. Oral glucose tolerance, insulin response, and IR were assessed using mathematical models. Liver tissue was analyzed for markers of oxidative stress, inflammation, and key regulatory pathways, including NF-κB, Nrf2, MAPKs (JNK and p38), and the IRS1-Akt pathway. We established an effective curcumin dose of 250 mg/kg for 5 days orally. Results demonstrated that after 30 days of exposure, cadmium accumulated in the liver, inducing an oxidative and inflammatory state. This was characterized by increased expression of NF-κB, JNK, and p38, along with diminished Nrf2 expression, hepatic IR, hyperglycemia, and hyperinsulinemia. Curcumin treatment effectively alleviated these metabolic disorders by restoring the balance between NF-κB and Nrf2 in the liver, modulating the MAPK pathway, and, consequently, improving oxidative and inflammatory balance. In conclusion, this study suggests that cadmium induces hepatic IR through an imbalance between NF-κB and Nrf2 signaling pathways. Curcumin treatment appears to improve these pathways, thereby ameliorating hepatic IR.
Collapse
Affiliation(s)
- Victor Enrique Sarmiento-Ortega
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Chemistry Department, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, 72560, Puebla, C.P, Mexico
| | - Diana Moroni-González
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Chemistry Department, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, 72560, Puebla, C.P, Mexico
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, Meritorious Autonomous University of Puebla, 22 South. FCQ9, Ciudad Universitaria, 72560, Puebla, C.P, Mexico
| | - Eduardo Brambila
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Chemistry Department, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, 72560, Puebla, C.P, Mexico
| | - Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Chemistry Department, Meritorious Autonomous University of Puebla, 14 Sur. FCQ1, Ciudad Universitaria, 72560, Puebla, C.P, Mexico.
| |
Collapse
|
11
|
Prah DA, Laryea-Akrong E. Asymptomatic Low-Density Plasmodium falciparum Infections: Parasites Under the Host's Immune Radar? J Infect Dis 2024; 229:1913-1918. [PMID: 38349649 PMCID: PMC11175676 DOI: 10.1093/infdis/jiad581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/18/2023] [Indexed: 06/15/2024] Open
Abstract
A large body of evidence suggests that low parasite carriage in Plasmodium falciparum asymptomatic infection is required for the maintenance of malaria immunity. However, the fact that treating such infections has little to no impact on subsequent clinical malaria is rarely noted. In this paper, we review data and argue that low-density parasite carriage in asymptomatic infection may not support host immune processes and that parasites are virtually under the host's immunological radar. We also discuss factors that may be constraining parasitemia in asymptomatic infections from reaching the threshold required to cause clinical symptoms. A thorough understanding of this infectious reservoir is essential for malaria control and eradication because asymptomatic infections contribute significantly to Plasmodium transmission.
Collapse
Affiliation(s)
- Diana Ahu Prah
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana
- Faculty of Applied Sciences, Department of Science Laboratory Technology, Accra Technical University, Accra, Ghana
| | - Elizabeth Laryea-Akrong
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| |
Collapse
|
12
|
Alvarez-Martinez M, Cox LS, Pearson CF, Branchett WJ, Chakravarty P, Wu X, Slawinski H, Al-Dibouni A, Samelis VA, Gabryšová L, Priestnall SL, Suárez-Bonnet A, Mikolajczak A, Briscoe J, Powrie F, O'Garra A. Blimp-1 and c-Maf regulate immune gene networks to protect against distinct pathways of pathobiont-induced colitis. Nat Immunol 2024; 25:886-901. [PMID: 38609547 PMCID: PMC11065689 DOI: 10.1038/s41590-024-01814-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: 02/07/2022] [Accepted: 03/13/2024] [Indexed: 04/14/2024]
Abstract
Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.
Collapse
Affiliation(s)
| | - Luke S Cox
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Claire F Pearson
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - William J Branchett
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Probir Chakravarty
- Computational Biology Laboratory, The Francis Crick Institute, London, UK
| | - Xuemei Wu
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Hubert Slawinski
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Alaa Al-Dibouni
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Vasileios A Samelis
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Leona Gabryšová
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
| | - Simon L Priestnall
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
- Experimental Histopathology, The Francis Crick Institute, London, UK
| | - Alejandro Suárez-Bonnet
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
- Experimental Histopathology, The Francis Crick Institute, London, UK
| | - Anna Mikolajczak
- Experimental Histopathology, The Francis Crick Institute, London, UK
| | - James Briscoe
- Developmental Dynamics Laboratory, The Francis Crick Institute, London, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Anne O'Garra
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
13
|
Cen Y, Li F, Li Y, Zhang K, Riaz F, Zhao K, Wei P, Pan F. Dimethyl fumarate alleviates allergic asthma by strengthening the Nrf2 signaling pathway in regulatory T cells. Front Immunol 2024; 15:1375340. [PMID: 38711519 PMCID: PMC11070462 DOI: 10.3389/fimmu.2024.1375340] [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: 01/23/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Allergic asthma is a widely prevalent inflammatory condition affecting people across the globe. T cells and their secretory cytokines are central to the pathogenesis of allergic asthma. Here, we have evaluated the anti-inflammatory impact of dimethyl fumarate (DMF) in allergic asthma with more focus on determining its effect on T cell responses in allergic asthma. By utilizing the ovalbumin (OVA)-induced allergic asthma model, we observed that DMF administration reduced the allergic asthma symptoms and IgE levels in the OVA-induced mice model. Histopathological analysis showed that DMF treatment in an OVA-induced animal model eased the inflammation in the nasal and bronchial tissues, with a particular decrease in the infiltration of immune cells. Additionally, RT-qPCR analysis exhibited that treatment of DMF in an OVA-induced model reduced the expression of inflammatory cytokine (IL4, IL13, and IL17) while augmenting anti-inflammatory IL10 and Foxp3 (forkhead box protein 3). Mechanistically, we found that DMF increased the expression of Foxp3 by exacerbating the expression of nuclear factor E2-related factor 2 (Nrf2), and the in-vitro activation of Foxp3+ Tregs leads to an escalated expression of Nrf2. Notably, CD4-specific Nrf2 deletion intensified the allergic asthma symptoms and reduced the in-vitro iTreg differentiation. Meanwhile, DMF failed to exert protective effects on OVA-induced allergic asthma in CD4-specific Nrf2 knock-out mice. Overall, our study illustrates that DMF enhances Nrf2 signaling in T cells to assist the differentiation of Tregs, which could improve the anti-inflammatory immune response in allergic asthma.
Collapse
Affiliation(s)
- Yanhong Cen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Otolaryngology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Fangfang Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yikui Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kaimin Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Farooq Riaz
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ping Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Otolaryngology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
14
|
Hou T, Zhu L, Wang Y, Peng L. Oxidative stress is the pivot for PM2.5-induced lung injury. Food Chem Toxicol 2024; 184:114362. [PMID: 38101601 DOI: 10.1016/j.fct.2023.114362] [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: 09/21/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Fine particulate matter (PM2.5) is a primary air pollutant recognized worldwide as a serious threat to public health. PM2.5, which has a diameter of less than 2.5 μm, is known to cause various diseases, including cardiovascular, respiratory, metabolic, and neurological diseases. Studies have shown that the respiratory system is particularly susceptible to PM2.5 as it is the first line of defense against external pollutants. PM2.5 can cause oxidative stress, which is triggered by the catalyzation of biochemical reactions, the activation of oxidases and metabolic enzymes, and mitochondrial dysfunction, all of which can lead to lung injury and aggravate various respiratory diseases including chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis, and cancer. Oxidative stress plays a crucial role in the harmful effects and mechanisms of PM2.5 on the respiratory system by activating several detrimental pathways related to inflammation and cellular damage. However, experimental studies have shown that antioxidative therapy methods can effectively cure PM2.5-induced lung injury. This review aims to clarify how PM2.5 induces oxidative stress and the mechanisms by which it is involved in the aggravation of various lung diseases. Additionally, we have listed antioxidant treatments to protect against PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Tianhua Hou
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, 130001, China
| | - Laiyu Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, 130001, China
| | - Yusheng Wang
- Department of Otolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130001, China.
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, 130001, China.
| |
Collapse
|
15
|
Milani A, Akbari E, Pordanjani PM, Jamshidi F, Ghayoumi S, Sadeghi SA, Bolhassani A. Immunostimulatory effects of Hsp70 fragments and Hsp27 in design of novel HIV-1 vaccine formulations. HIV Med 2024; 25:276-290. [PMID: 37936563 DOI: 10.1111/hiv.13576] [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: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Heat shock proteins (HSPs) as an adjuvant induce antigen-specific immunity through facilitating antigen presentation and stimulating T cells. In this study, the immunostimulatory properties of two major fragments of Hsp70 (N-Hsp70(aa 1-387) with ATPase property and C-Hsp70 (aa 508-641) with peptide-binding capacity) and the full length of Hsp27 as vaccine adjuvants were evaluated to boost HIV-1 Nef antigen-specific immunity in both in vitro and in vivo experiments. METHODS At first, the nanoparticles harbouring DNA fusion constructs (i.e. N-Hsp70-Nef, C-Hsp70-Nef and Hsp27-Nef) complexed with HIV Rev (34-50) cell-penetrating peptide were generated to deliver DNA into the cells. Then, the recombinant Nef, Hsp27-Nef, N-Hsp70-Nef and C-Hsp70-Nef proteins were generated in E.coli expression system. Next, the immunostimulatory properties of these fusion constructs were evaluated in both in vitro and in vivo studies. Finally, the secretion of main cytokines from single-cycle replicable (SCR) HIV-1 virion-exposed splenocytes was investigated. RESULTS Our data showed that the stable and non-toxic DNA/Rev nanoparticles could successfully deliver the genes of interest into the cells. Moreover, higher secretion of antibodies and cytokines was detected in mice receiving the Hsp-Nef constructs than in mice receiving Nef antigen. The C-Hsp70 was also superior for inducing Nef-specific Th1 and CTL immunity compared with N-Hsp70 and Hsp27. The T-cell activity was maintained in the SCR-exposed splenocytes, especially the splenocytes of mice receiving the C-Hsp70-Nef regimen. CONCLUSION Altogether, these findings demonstrate the significance of Hsps as enhancers of antigen-specific immunity. Notably, the C-Hsp70 region showed better adjuvant properties for inducing cellular immunity in the improvement of HIV-1 therapeutic vaccines.
Collapse
Affiliation(s)
- Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Elahe Akbari
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fateme Jamshidi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Shahrzad Ghayoumi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Seyed Amir Sadeghi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
16
|
Zhao Y, Wang H, Jin L, Zhang Z, Liu L, Zhou M, Zhang X, Zhang L. Targeting fusion proteins of the interleukin family: A promising new strategy for the treatment of autoinflammatory diseases. Eur J Pharm Sci 2024; 192:106647. [PMID: 37984595 DOI: 10.1016/j.ejps.2023.106647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
As a means of communication between immune cells and non-immune cells, Interleukins (ILs) has the main functions of stimulating the proliferation and activation of inflammatory immune cells such as dendritic cells and lymphocytes, promote the development of blood cells and so on. However, dysregulation of ILs expression is a major feature of autoinflammatory diseases. The drugs targeting ILs or IL-like biologics have played an important role in the clinical treatment of autoinflammatory diseases. Nevertheless, the widespread use of IL products may result in significant off-target adverse reactions. Thus, there is a clear need to develop next-generation ILs products in the biomedical field. Fusion proteins are proteins created through the joining of two or more genes that originally coded for separate proteins. Over the last 30 years, there has been increasing interest in the use of fusion protein technology for developing anti-inflammatory drugs. In comparison to single-target drugs, fusion proteins, as multiple targets drugs, have the ability to enhance the cytokine therapeutic index, resulting in improved efficacy over classical drugs. The strategy of preparing ILs or their receptors as fusion proteins is increasingly used in the treatment of autoimmune and chronic inflammation. This review focuses on the efficacy of several fusion protein drugs developed with ILs or their receptors in the treatment of autoinflammatory diseases, in order to illustrate the prospects of this new technology as an anti-inflammatory drug development protocol in the future.
Collapse
Affiliation(s)
- Yuchen Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Han Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Ziwei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Lianghu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Mengqi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui 230032, China; Anti-inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui 230032, China.
| |
Collapse
|
17
|
Chen X, Wu Y, Li J, Jiang S, Sun Q, Xiao L, Jiang X, Xiao X, Li X, Mu Y. Lycium barbarum Ameliorates Oral Mucositis via HIF and TNF Pathways: A Network Pharmacology Approach. Curr Pharm Des 2024; 30:2718-2735. [PMID: 39076092 DOI: 10.2174/0113816128312694240712072959] [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: 02/26/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Oral mucositis is the most common and troublesome complication for cancer patients receiving radiotherapy or chemotherapy. Recent research has shown that Lycium barbarum, an important economic crop widely grown in China, has epithelial protective effects in several other organs. However, it is unknown whether or not Lycium barbarum can exert a beneficial effect on oral mucositis. Network pharmacology has been suggested to be applied in "multi-component-multi-target" functional food studies. The purpose of this study is to evaluate the effect of Lycium barbarum on oral mucositis through network pharmacology, molecular docking and experimental validation. AIMS To explore the biological effects and molecular mechanisms of Lycium barbarum in the treatment of oral mucositis through network pharmacology and molecular docking combined with experimental validation. METHODS Based on network pharmacology methods, we collected the active components and related targets of Lycium barbarum from public databases, as well as the targets related to oral mucositis. We mapped protein- protein interaction (PPI) networks, performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment, and constructed a 'components-disease-targets' network and 'components-pathways-targets' network using Cytoscape to further analyse the intrinsic molecular mechanisms of Lycium barbarum against oral mucositis. The affinity and stability predictions were performed using molecular docking strategies, and experiments were conducted to demonstrate the biological effects and possible mechanisms of Lycium barbarum against oral mucositis. RESULTS A network was established between 49 components and 61 OM targets. The main active compounds were quercetin, beta-carotene, palmatine, and cyanin. The predicted core targets were IL-6, RELA, TP53, TNF, IL10, CTNNB1, AKT1, CDKN1A, HIF1A and MYC. The enrichment analysis predicted that the therapeutic effect was mainly through the regulation of inflammation, apoptosis, and hypoxia response with the involvement of TNF and HIF pathways. Molecular docking results showed that key components bind well to the core targets. In both chemically and radiation-induced OM models, Lycium barbarum significantly promoted healing and reduced inflammation. The experimental verification showed Lycium barbarum targeted the key genes (IL-6, RELA, TP53, TNF, IL10, CTNNB1, AKT1, CDKN1A, HIF1A, and MYC) through regulating the HIF and TNF signaling pathways, which were validated using the RT-qPCR, immunofluorescence staining and western blotting assays. CONCLUSION In conclusion, the present study systematically demonstrated the possible therapeutic effects and mechanisms of Lycium barbarum on oral mucositis through network pharmacology analysis and experimental validation. The results showed that Lycium barbarum could promote healing and reduce the inflammatory response through TNF and HIF signaling pathways.
Collapse
Affiliation(s)
- Xun Chen
- School of Stomatology, Southwest Medical University, Luzhou 646699, China
| | - Yanhui Wu
- School of Stomatology, Southwest Medical University, Luzhou 646699, China
| | - Jing Li
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Sijing Jiang
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qiang Sun
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Li Xiao
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiliang Jiang
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Xiao
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xianxian Li
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yandong Mu
- School of Stomatology, Southwest Medical University, Luzhou 646699, China
- Stomatology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
18
|
Chufistova AV, Shabaldina EV, Bedareva AV, Vakhrameev IN, Abramova NA, Shabaldin AV. [Features of inflammatory endotypes and phenotypes in chronic rhinosinusitis]. Vestn Otorinolaringol 2024; 89:60-67. [PMID: 39171879 DOI: 10.17116/otorino20248904160] [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: 08/23/2024]
Abstract
Recently, significant progress has been made in identifying the cellular and molecular mechanisms responsible for the pathogenesis of chronic rhinosinusitis (CRS). Cohort studies of CRS have led to advances in the clinical understanding of this disease. New therapeutic agents have been approved or are undergoing clinical trials to expand treatment options for this disease. One of the promising areas in medicine is the provision of personalized clinical care. From this perspective, CRS can be divided into three different endotypes depending on the type of underlying inflammatory response. In the United States, CRS with and without nasal polyps is predominantly characterized as the second inflammatory endotype. CRS with nasal polyps (about 17%) and without nasal polyps (up to 20%) belongs to the 1st and 3rd inflammatory endotypes, respectively. And if for the second inflammatory endotype the effectiveness of targeted biological therapy is beyond doubt, then for the first and third inflammatory endotypes the principles of such conservative therapy are under active development. Moreover, large validated studies to confirm associations between CRS phenotypes and endotypes, as well as to find effective biological markers of inflammatory endotypes, remain to be performed.
Collapse
Affiliation(s)
| | | | | | - I N Vakhrameev
- Podgorbunsky Kuzbass Clinical Hospital of Emergency Medical Care, Kemerovo, Russia
| | - N A Abramova
- Belyaev Kuzbass Regional Clinical Hospital, Kemerovo, Russia
| | - A V Shabaldin
- Kemerovo State Medical University, Kemerovo, Russia
- Kemerovo State University, Kemerovo, Russia
| |
Collapse
|
19
|
Cox LS, Alvarez-Martinez M, Wu X, Gabryšová L, Luisier R, Briscoe J, Luscombe NM, O'Garra A. Blimp-1 and c-Maf regulate Il10 and negatively regulate common and unique proinflammatory gene networks in IL-12 plus IL-27-driven T helper-1 cells. Wellcome Open Res 2023; 8:403. [PMID: 38074197 PMCID: PMC10709690 DOI: 10.12688/wellcomeopenres.19680.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Background CD4 + Th1 cells producing IFN-γ are required to eradicate intracellular pathogens, however if uncontrolled these cells can cause immunopathology. The cytokine IL-10 is produced by multiple immune cells including Th1 cells during infection and regulates the immune response to minimise collateral host damage. In this study we aimed to elucidate the transcriptional network of genes controlling the expression of Il10 and proinflammatory cytokines, including Ifng in Th1 cells differentiated from mouse naive CD4 + T cells. Methods We applied computational analysis of gene regulation derived from temporal profiling of gene expression clusters obtained from bulk RNA sequencing (RNA-seq) of flow cytometry sorted naïve CD4 + T cells from mouse spleens differentiated in vitro into Th1 effector cells with IL-12 and IL-27 to produce Ifng and Il10, compared to IL-27 alone which express Il10 only , or IL-12 alone which express Ifng and no Il10, or medium control driven-CD4 + T cells which do not express effector cytokines . Data were integrated with analysis of active genomic regions from these T cells using an assay for transposase-accessible chromatin with sequencing (ATAC)-seq, integrated with literature derived-Chromatin-immunoprecipitation (ChIP)-seq data and the RNA-seq data, to elucidate the transcriptional network of genes controlling expression of Il10 and pro-inflammatory effector genes in Th1 cells. The co-dominant role for the transcription factors, Prdm1 (encoding Blimp-1) and Maf (encoding c-Maf) , in cytokine gene regulation in Th1 cells, was confirmed using T cells obtained from mice with T-cell specific deletion of these transcription factors. Results We show that the transcription factors Blimp-1 and c-Maf each have unique and common effects on cytokine gene regulation and not only co-operate to induce Il10 gene expression in IL-12 plus IL-27 differentiated mouse Th1 cells, but additionally directly negatively regulate key proinflammatory cytokines including Ifng, thus providing mechanisms for reinforcement of regulated Th1 cell responses. Conclusions These data show that Blimp-1 and c-Maf positively and negatively regulate a network of both unique and common anti-inflammatory and pro-inflammatory genes to reinforce a Th1 response in mice that will eradicate pathogens with minimum immunopathology.
Collapse
Affiliation(s)
- Luke S. Cox
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Marisol Alvarez-Martinez
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Xuemei Wu
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Leona Gabryšová
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Raphaëlle Luisier
- Computational Biology Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - James Briscoe
- Developmental Dynamics Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Nicholas M. Luscombe
- Computational Biology Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, England, UK
| | - Anne O'Garra
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, England, NW1 1AT, UK
- National Heart and Lung Institute, Imperial College London, London, England, UK
| |
Collapse
|
20
|
Moniruzzaman M, Maiti AK, Chakraborty SB, Saha I, Saha NC. Melatonin ameliorates lipopolysaccharide induced brain inflammation through modulation of oxidative status and diminution of cytokine rush in Danio rerio. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 96:103983. [PMID: 36182043 DOI: 10.1016/j.etap.2022.103983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
Lipopolysaccharide (LPS) is known to induce inflammation and immunonomodulation in a piscine model of Danio rerio. Present study aimed to explore the ability of melatonin in attenuating LPS-induced oxidative damages using this model. In LPS-exposed fish, activation of stress marker MDA was observed in brain with corresponding augmentation of multiple pro-inflammatory cytokines (IL1β, IL6, IL10 and TNFα). In addition, it also showed marked increase in the levels of heat shock factor (HSF) and heat shock proteins (HSPs) in association with transcription factors (NF-kB and NRF2) and mitogen-activated protein kinases (MAPKs). The changes in the levels of these mediators are highly correlated with the induction of pro-inflammatory cytokines. In melatonin-treated fishes, significant amelioration of oxidative stress was observed with reduced levels of MDA and pro-inflammatory cytokines. Melatonin also modulated expression of HSPs that facilitated the brain to overcome inflammation-induced stress by directly initiating NFkB/NRF2 translocation. In summary, melatonin effectively functions to reduce stress induced inflammatory signalling through modulation of oxidative stress and protects the brain from the neuropathological insult.
Collapse
Affiliation(s)
| | - Arpan Kumar Maiti
- Department of Zoology, University of North Bengal, Darjeeling 734013, India
| | | | - Ishita Saha
- Department of Physiology, Medical College and Hospital, Kolkata, India
| | - Nimai Chandra Saha
- Fisheries and Ecotoxicology Research Laboratory Vice Chancellor's Research Group, Department of Zoology, The University of Burdwan, Purba bardhaman, Burdwan 713104, West Bengal, India.
| |
Collapse
|
21
|
Zhang B, Wang J, Liu M, Zhao Q, Yu G, Zhang B, Hua H, Xu J, Li J, Yu Q, Koda S, Xu YH, Jiang Z, Yan C, Zheng KY. IL-10 regulates Th17 response to inhibit hepatobiliary injury caused by Clonorchis sinensis infection in C57BL/6J mice. Front Cell Infect Microbiol 2022; 12:994838. [PMID: 36310865 PMCID: PMC9606589 DOI: 10.3389/fcimb.2022.994838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Clonorchiasis caused by Clonorchis sinensis is a mainly foodborne parasitic disease. It can lead to hepatobiliary duct inflammation, fibrosis, obstructive jaundice, liver cirrhosis, and even cholangiocarcinoma. Interleukin (IL)-10 is an immune-regulatory cytokine which plays an immunosuppressive role during infection. Our previous study found that IL-10 was increased in mice with C. sinensis infection. However, the role and mechanism of IL-10 playing in hepatobiliary injury induced by C. sinensis infection remain unknown. Herein, Il10+/+ mice and Il10+/- C57BL/6J mice were infected with C. sinensis. It was found that IL-10 deficiency aggravated biliary hyperplasia and exacerbated periductal fibrosis induced by C. sinensis infection. Moreover, IL-10 deficiency increased CD4+T cells and CD8+T cells but not macrophages in the liver of mice with infection. There were no apparent differences in Th1 and Treg cells between Il10+/+ and Il10+/- mice infected with C. sinensis. However, the proportion of Th17 cells in CD4+T cells in Il10+/- infected mice was significantly higher than that in Il10+/+ infected mice. IL-10 deficiency also enhanced the increase of Th17 cells induced by ESPs stimulation in vitro. Taken together, our results suggest that IL-10 plays a protective role in hepatobiliary injury in C57BL/6J mice induced by C. sinensis infection via inhibiting Th17 cells, which could deepen our understanding of the immunopathology of clonorchiasis.
Collapse
Affiliation(s)
- Beibei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Jianling Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Man Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qianqian Zhao
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Guozhi Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Bo Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Hui Hua
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Jinyao Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Jing Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qian Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Stephane Koda
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yin-Hai Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhihua Jiang
- Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Chao Yan, ; Kui-Yang Zheng,
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Chao Yan, ; Kui-Yang Zheng,
| |
Collapse
|
22
|
Yi M, Wang G, Niu J, Peng M, Liu Y. Pterostilbene attenuates the proliferation and differentiation of TNF‑α‑treated human periodontal ligament stem cells. Exp Ther Med 2022; 23:304. [PMID: 35340874 PMCID: PMC8931590 DOI: 10.3892/etm.2022.11233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/05/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Min Yi
- Department of Integrative Therapy, Shanghai Huangpu District 2nd Dental Disease Prevention and Treatment Institute, Shanghai 200001, P.R. China
| | - Guanglei Wang
- Department of Stomatology, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Jianhua Niu
- Department of Integrative Therapy, Shanghai Huangpu District 2nd Dental Disease Prevention and Treatment Institute, Shanghai 200001, P.R. China
| | - Minghui Peng
- Department of Integrative Therapy, Shanghai Huangpu District 2nd Dental Disease Prevention and Treatment Institute, Shanghai 200001, P.R. China
| | - Yi Liu
- Department of Stomatology, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| |
Collapse
|
23
|
Rodrigues de Souza I, Savio de Araujo-Souza P, Morais Leme D. Genetic variants affecting chemical mediated skin immunotoxicity. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2022; 25:43-95. [PMID: 34979876 DOI: 10.1080/10937404.2021.2013372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The skin is an immune-competent organ and this function may be impaired by exposure to chemicals, which may ultimately result in immune-mediated dermal disorders. Interindividual variability to chemical-induced skin immune reactions is associated with intrinsic individual characteristics and their genomes. In the last 30-40 years, several genes influencing susceptibility to skin immune reactions were identified. The aim of this review is to provide information regarding common genetic variations affecting skin immunotoxicity. The polymorphisms selected for this review are related to xenobiotic-metabolizing enzymes (CYPA1 and CYPB1 genes), antioxidant defense (GSTM1, GSTT1, and GSTP1 genes), aryl hydrocarbon receptor signaling pathway (AHR and ARNT genes), skin barrier function transepidermal water loss (FLG, CASP14, and SPINK5 genes), inflammation (TNF, IL10, IL6, IL18, IL31, and TSLP genes), major histocompatibility complex (MHC) and neuroendocrine system peptides (CALCA, TRPV1, ACE genes). These genes present variants associated with skin immune responses and diseases, as well as variants associated with protecting skin immune homeostasis following chemical exposure. The molecular and association studies focusing on these genetic variants may elucidate their functional consequences and contribution in the susceptibility to skin immunotoxicity. Providing information on how genetic variations affect the skin immune system may reduce uncertainties in estimating chemical hazards/risks for human health in the future.
Collapse
Affiliation(s)
| | | | - Daniela Morais Leme
- Graduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba, Brazil
- National Institute for Alternative Technologies of Detection, Toxicological Evaluation and Removal of Micropollutants and Radioactives (INCT-DATREM), Institute of Chemistry, Araraquara, Brazil
| |
Collapse
|
24
|
Nahalka J. Transcription of the Envelope Protein by 1-L Protein-RNA Recognition Code Leads to Genes/Proteins That Are Relevant to the SARS-CoV-2 Life Cycle and Pathogenesis. Curr Issues Mol Biol 2022; 44:791-816. [PMID: 35723340 PMCID: PMC8928949 DOI: 10.3390/cimb44020055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/21/2022] [Accepted: 02/03/2022] [Indexed: 12/02/2022] Open
Abstract
The theoretical protein-RNA recognition code was used in this study to research the compatibility of the SARS-CoV-2 envelope protein (E) with mRNAs in the human transcriptome. According to a review of the literature, the spectrum of identified genes showed that the virus post-transcriptionally promotes or represses the genes involved in the SARS-CoV-2 life cycle. The identified genes/proteins are also involved in adaptive immunity, in the function of the cilia and wound healing (EMT and MET) in the pulmonary epithelial tissue, in Alzheimer's and Parkinson's disease and in type 2 diabetes. For example, the E-protein promotes BHLHE40, which switches off the IL-10 inflammatory "brake" and inhibits antiviral THαβ cells. In the viral cycle, E supports the COPII-SCAP-SREBP-HSP90α transport complex by the lowering of cholesterol in the ER and by the repression of insulin signaling, which explains the positive effect of HSP90 inhibitors in COVID-19 (geldanamycin), and E also supports importin α/β-mediated transport to the nucleus, which explains the positive effect of ivermectin, a blocker of importins α/β. In summary, transcription of the envelope protein by the 1-L protein-RNA recognition code leads to genes/proteins that are relevant to the SARS-CoV-2 life cycle and pathogenesis.
Collapse
Affiliation(s)
- Jozef Nahalka
- Centre for Glycomics, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84538 Bratislava, Slovakia
- Centre of Excellence for White-Green Biotechnology, Institute of Chemistry, Slovak Academy of Sciences, Trieda Andreja Hlinku 2, SK-94976 Nitra, Slovakia
| |
Collapse
|
25
|
Jia Y, Liu Y, Zhu J, Liu L, Ma X, Liu D, Han S, Zhang L, Ling ZQ, Wang Y. DEC1 promotes progression of Helicobacter pylori-positive gastric cancer by regulating Akt/NF-κB pathway. J Cell Mol Med 2022; 26:1943-1954. [PMID: 35122398 PMCID: PMC8980912 DOI: 10.1111/jcmm.17219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/23/2021] [Accepted: 01/22/2022] [Indexed: 11/30/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection plays a crucial role in the initiation and progression of gastric cancer (GC). Differentiated embryo‐chondrocyte expressed gene 1 (DEC1) is dysregulated in some cancers and may regulate cell proliferation in specific contexts. Of note, DEC1 is emerging as one of the important factors regulating cellular responses in microenvironment. However, the triggers and precise regulation mechanism for DEC1 during inflammatory carcinoma transformation of GC are unclear. In this study, we identified DEC1 was upregulated in both H. pylori‐infected gastric tissues and GC cells. DEC1 expression was positively associated with H. pylori infection status and GC progression. DEC1‐positive expression indicated a poorer prognosis in H. pylori‐positive GC. DEC1 was required for H. pylori‐induced GC cells proliferation. Mechanistically, H. pylori infection significantly activated Akt/NF‐κB signal pathway and this induction depend on DEC1 expression level in GC cells. Importantly, their interaction pathway was further verified by H. pylori‐positive gastritis mice model. Taken together, our findings identified a novel function of DEC1 in GC. H. pylori infection induce DEC1 expression, and which leading to the progression of GC through activating Akt/ NF‐κB signalling pathway. Blocking DEC1/Akt/NF‐κB, therefore, presents a promising novel therapeutic strategy for H. pylori‐positive GC.
Collapse
Affiliation(s)
- Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China.,Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanyan Liu
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, China
| | - Jingyu Zhu
- Department of Gastroenterology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Liang Liu
- Department of Gastroenterology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China.,Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Duanrui Liu
- Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuyi Han
- Medical Research & Laboratory Diagnostic Center, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Lulu Zhang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China.,Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Qiang Ling
- Zhejiang Cancer Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China.,Research Center of Basic Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
26
|
Du E, Fan Q, Zhao N, Zhang W, Wei J, Chen F, Huang S, Guo W. Supplemental magnolol improves the antioxidant capacity and intestinal health of broiler chickens. Anim Sci J 2021; 92:e13665. [PMID: 34874084 DOI: 10.1111/asj.13665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/19/2021] [Accepted: 11/12/2021] [Indexed: 11/30/2022]
Abstract
Magnolol is a multifunctional polyphenol rich in Magnolia officinalis. The objective of this study was to investigate the effects of magnolol on growth performance, carcass traits, antioxidant capacity, and gut health of broiler chickens. A total of 240 1-day-old broilers were randomly allocated into five dietary treatments: control (Ctrl); control diet supplemented with 100, 200, or 300 mg/kg of magnolol (M100, M200, and M300); and control diet supplemented with 200 mg/kg of bacitracin zinc (PC). The results showed that magnolol linearly decreased the feed conversion ratio between d 0 and d 14, linearly decreased the amount of malondialdehyde and increased the activity of total superoxide dismutase (T-SOD) in both serum and ileal mucosa on d 42 with increasing magnolol levels (p < 0.05). Moreover, the ileal villus height, the ileal villus height to crypt depth ratio, and the jejunal gene expressions of SOD1, glutathione peroxidase, and Claudin1 were linearly up-regulated with increasing magnolol levels (p < 0.05). The supplementation of magnolol had no effect on carcass traits or cecal short chain fatty acids (p > 0.05). The results indicated that magnolol could be applied in the diet of broiler chickens to benefit their antioxidant capacity and intestinal health.
Collapse
Affiliation(s)
- Encun Du
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China.,State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiwen Fan
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Na Zhao
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Wei Zhang
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Jintao Wei
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Fang Chen
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Shaowen Huang
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Wanzheng Guo
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
27
|
Kava R, Peripolli E, Berton MP, Lemos M, Lobo RB, Stafuzza NB, Pereira AS, Baldi F. Genome-wide structural variations in Brazilian Senepol cattle, a tropically adapted taurine breed. Livest Sci 2021. [DOI: 10.1016/j.livsci.2021.104708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Keaton SA, Arnetz J, Jamil H, Dhalimi A, Stemmer PM, Ruden DM, Yamin J, Achtyes E, Smart L, Brundin L, Arnetz BB. IL-10: A possible immunobiological component of positive mental health in refugees. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2021; 8:100097. [PMID: 35757662 PMCID: PMC9216633 DOI: 10.1016/j.cpnec.2021.100097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 01/23/2023] Open
Abstract
Objective As the number of refugees continues to rise, there is growing concern about the impact from trauma exposures on their mental health. However, there is a limited understanding of possible biological mechanisms contributing to the substantial inter-individual differences in trauma-related outcomes, especially as it relates to positive mental health. Only sparse work has focused on the biology of positive mental health, including energy and sleep, in trauma-exposed persons. In this study, we analyzed cytokines in blood from newly arrived refugees with differential trauma exposures in relationship to self-reported energy, as a key marker of positive mental health. Methods Within the first month of arrival in the USA, 64 refugees from Iraq and Syria were interviewed. Refugees completed the clinical DSM-IV PTSD-Checklist Civilian Version (PCL-C), the Beck Anxiety Inventory (BAI), and the Hospital Anxiety and Depression Scale (HADS). Ten psychiatrically healthy non-refugee persons were used as healthy controls to compare levels of cytokines. Blood samples were collected at the time of the interview and subsequently analyzed for IL-1β, IL-6, IL-8, IL-10, and TNF-α concentrations. Results Energy correlated positively with current concentration ability and sleep quality, and negatively with stress, PCL-C, BAI and HADS scores (Spearman correlations, all p<0.05). Refugees had lower levels of IL-10 compared to controls (p<0.05). IL-10 levels in refugees correlated with higher energy levels (p<0.01). Conclusions Results suggest that self-reported energy is a key component of positive mental health in newly arrived traumatized refugees. Additionally, the anti-inflammatory cytokine IL-10 could be a marker of, or causally associated with positive mental health. A better understanding of the balance between pro- and anti-inflammatory states in highly traumatized individuals has the potential to create more targeted and effective treatments with implications for long-term health outcomes.
Collapse
Affiliation(s)
- Sarah A. Keaton
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Judy Arnetz
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Hikmet Jamil
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Abir Dhalimi
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Paul M. Stemmer
- Institute of Environmental Health Science, Wayne State University, Detroit, MI, USA
| | - Douglas M. Ruden
- Institute of Environmental Health Science, Wayne State University, Detroit, MI, USA
| | - Jolin Yamin
- Institute of Environmental Health Science, Wayne State University, Detroit, MI, USA
| | - Eric Achtyes
- Pine Rest Christian Mental Health, Grand Rapids, MI, USA
- Division of Psychiatry & Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - LeAnn Smart
- Pine Rest Christian Mental Health, Grand Rapids, MI, USA
| | - Lena Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
- Division of Psychiatry & Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Bengt B. Arnetz
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
29
|
Yuan L, You H, Qin N, Zuo W. Interleukin-10 Modulates the Metabolism and Osteogenesis of Human Dental Pulp Stem Cells. Cell Reprogram 2021; 23:270-276. [PMID: 34491831 DOI: 10.1089/cell.2021.0044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The osteogenic differentiation of mesenchymal stem cells (MSCs) is strongly related with the inflammatory microenvironment. The ability of osteogenic differentiation of MSCs is vital for the bone tissue engineering. Interleukin (IL)-10, a well-known anti-inflammatory factor, plays a key role in tissue repair. Dental pulp stem cells (DPSCs), with the advantage of convenience of extraction, are suitable for the bone tissue engineering. Therefore, it is meaning to explore the effects of IL-10 on the osteogenic differentiation of DPSCs. The proliferation activity of DPSCs were evaluated by MTS assay (CellTiter 96® Aqueous One Solution Cell Proliferation Assay [Promega]) and real-time polymerase chain reaction (RT-PCR). The osteogenic differentiation of DPSCs were determined by Alizarin Red staining, RT-PCR, and alkaline phosphatase activity test. The glucose metabolism was detected by Mito Stress test and glycolysis assay. IL-10 (10 or 20 nM) could enhance the osteogenic differentiation of DPSCs and promoted the metabolic switch from glycolysis to oxidative phosphorylation (OXPHOS), whereas IL-10 (5 and 50 nM) has no obvious effects on the osteogenic differentiation of DPSCs. The OXPHOS inhibitor restrained the promotion of osteogenic differentiation induced by IL-10. These findings show that IL-10 can promote the osteogenesis of DPSCs through the activation of OXPHOS, which provides a potential way for enhancing the osteogenic differentiation of DPSCs in bone tissue engineering.
Collapse
Affiliation(s)
- Li Yuan
- Department of Stomatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Hongxia You
- Department of Stomatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Nianhong Qin
- Department of Stomatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Wenxin Zuo
- Department of Stomatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
30
|
Xu L, Ye X, Wang Q, Xu B, Zhong J, Chen Y, Wang L. T-cell infiltration, contribution and regulation in the central nervous system post-traumatic injury. Cell Prolif 2021; 54:e13092. [PMID: 34189783 PMCID: PMC8349661 DOI: 10.1111/cpr.13092] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022] Open
Abstract
T cells participate in the repair process and immune response in the CNS post-traumatic injury and play both a beneficial and harmful role. Together with nerve cells and other immune cells, they form a microenvironment in the CNS post-traumatic injury. The repair of traumatic CNS injury is a long-term process. T cells contribute to the repair of the injury site to influence the recovery. Recently, with the advance of new techniques, such as mass spectrometry-based flow cytometry, modern live-cell imaging, etc, research focusing on T cells is becoming one of the valuable directions for the future therapy of traumatic CNS injury. In this review, we summarized the infiltration, contribution and regulation of T cells in post-traumatic injury, discussed the clinical significance and predicted the future research direction.
Collapse
Affiliation(s)
- Lvwan Xu
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Xin Ye
- Department of NeurosurgerySir Run Run Shaw Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Qingyi Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Bihan Xu
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Jinjie Zhong
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Ying‐ying Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lin‐lin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
31
|
Zhang G, Ma L, Bai L, Li M, Guo T, Tian B, He Z, Fu Q. Inflammatory microenvironment-targeted nanotherapies. J Control Release 2021; 334:114-126. [PMID: 33887284 DOI: 10.1016/j.jconrel.2021.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory microenvironments (IMEs) are common pathological characteristics and drive the development of multiple chronic diseases. Thus, IME-targeted therapies exhibit potential for the treatment of inflammatory diseases. Nanoplatforms have significant advantages in improving the efficiency of anti-inflammatory treatments. Owing to their improved therapeutic effects and reduced side effects, IME-targeted nanotherapies have recently drawn interest from the research community. This review introduces IMEs and discusses the application of IME-targeted nanotherapies for inflammatory diseases. The development of rational targeting strategies tailored to IMEs in damaged tissues can help promote therapies for chronic diseases.
Collapse
Affiliation(s)
- Guangshuai Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Lixue Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Lijun Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Tiange Guo
- Laboratory Animal Department, General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
32
|
Wang Y, Wang N, Cui L, Li Y, Cao Z, Wu X, Wang Q, Zhang B, Ma C, Cheng Y. Long Non-coding RNA MEG3 Alleviated Ulcerative Colitis Through Upregulating miR-98-5p-Sponged IL-10. Inflammation 2021; 44:1049-1059. [PMID: 33394187 DOI: 10.1007/s10753-020-01400-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
Ulcerative colitis (UC) is a refractory chronic colitis disease with the particularly complex cause. Recently, long noncoding RNAs (lncRNAs) have been reported to be related to the development of UC. LncRNA MEG3 has been proved to play an anti-inflammatory role in a variety of inflammatory diseases, which share similar pathogenesis with UC, indicating the potential involvement of lncRNA MEG3 in UC. This study aims to investigate the functional role and underlying mechanism of lncRNA MEG3 in UC. Gradient concentration of H2O2 (0, 20, 50, 100, and 200 μM) was used to induce Caco-2 damage models in vitro. Cell viability was detected by cell counting kit-8 (CCK-8) assay. LncRNA MEG3, miR-98-5p, and IL-10 levels in H2O2-treated Caco-2 cells were assessed by performing real-time quantitative polymerase chain reaction (RT-qPCR). Moreover, the binding relationship between lncRNA MEG3 and miR-98-5p, as well as the binding relationship between miR-98-5p and IL-10, was validated using dual-luciferase reporter assay. 2, 4, 6-Trinitrobenzenesulfonic acid solution (TNBS) was applied to induce ulcerative colitis in young rats. The body weight, disease activity index (DAI), length and weight of the colons, pathological scores of UC rats, reactive oxygen species (ROS), and inflammatory cytokines were determined to evaluate the effects of lncRNA MEG3 on the progression of UC. Besides, hematoxylin-eosin (HE) staining was exploited to observe histological changes of UC rat colons. In addition, western blotting analysis was also performed to evaluate the apoptosis and pyroptosis-related protein levels. Moreover, lncRNA MEG3, miR-98-5p, and IL-10 levels in UC rat colons were further assessed by RT-qPCR. Meanwhile, IL-10 expression was determined using immunohistochemistry. LncRNA MEG3 and IL-10 levels were distinctly decreased while miR-98-5p was increased in Caco-2 damage models and UC rats. Bioinformatics analysis predicted the binding sites of lncRNA MEG3 to miR-98-5p and miR-98-5p to IL-10. Besides, dual-luciferase reporter assay validated the negative correlation between lncRNA MEG3 and miR-98-5p, miR-98-5p, and IL-10. Overexpressed lncRNA MEG3 reduced. DAI scores and colon weight/length ratio improved UC ulceration. In addition, upregulation of lncRNA MEG3 relieved oxidative stress, inflammatory response, apoptosis, and pyroptosis of UC rat colons. LncRNA MEG3 overexpression alleviates the serve ulceration of UC rat colons by upregulating IL-10 expression via sponging miR-98-5p. To sum up, this study reveals the protective role of lncRNA MEG3 in the development of UC and may provide potential therapeutic targets for UC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Nan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Lianlian Cui
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Yan Li
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Zhenfeng Cao
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Xing Wu
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Qianhan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Bo Zhang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Caixia Ma
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China
| | - Yanbo Cheng
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China.
| |
Collapse
|
33
|
Huang G, Ge Y, Gui Z, Zhu M, Liu J, Wang H. Toxicity of Melastoma dodecandrum Lour. and its effects on lipopolysaccharide-induced inflammation and oxidative stress. Exp Ther Med 2021; 22:807. [PMID: 34093763 DOI: 10.3892/etm.2021.10239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Melastoma dodecandrum Lour. (MDL) is component used in traditional Chinese medicine that is widely distributed throughout southern China. MDL has been long utilized in clinical treatment for various conditions, such as inflammation. However, the toxicity and underlying anti-inflammatory mechanism of MDL remain to be elucidated. In the present study, Sprague-Dawley rats received intragastric administration of MDL for 2 months, and the toxicity of MDL was investigated. The rats were treated with lipopolysaccharide (LPS) for 8 h to determine the potential anti-inflammatory mechanism of MDL. The results demonstrated that MDL alone did not affect the expression levels of factors associated with inflammation (IL-1β, IL-6 and TNF-α) and oxidative stress [malondialdehyde (MDA), superoxide dismutase (SOD) and nitric oxide (NO)] in the rat serum and exerted no effects on rat liver and kidneys. By contrast, MDL attenuated LPS-induced inflammation and oxidative stress by regulating specific cytokines, such as IL-1β, IL-6, TNF-α, MDA, SOD and NO in the rat serum and alleviated LPS-induced liver and kidney damage. Additionally, compared with the LPS group, MDL inhibited CD4+ T cell differentiation into Th1 and Th17 cells and enhanced CD4+ T cell differentiation into Th2 and Treg cells. MDL also suppressed reactive oxygen species (ROS) production and mitochondrial apoptosis by modulating mitochondrial apoptosis-related proteins in spleen CD4+ T cells. In conclusion, the results of the present study demonstrated the non-toxic nature of MDL and revealed that it alleviated LPS-induced inflammation and oxidative stress by regulating differentiation and ROS production in CD4+ T cells.
Collapse
Affiliation(s)
- Gang Huang
- Department of Traditional Chinese Medicine, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Yuli Ge
- Department of Infection, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Zhihong Gui
- Department of Nephrology, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Meixiao Zhu
- Department of Pharmacy, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Jin Liu
- Department of Scientific Research Office, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| | - Huafu Wang
- Department of Clinical Pharmacology, Lishui People's Hospital, Lishui, Zhejiang 323000, P.R. China
| |
Collapse
|
34
|
Kozma GT, Mészáros T, Bakos T, Hennies M, Bencze D, Uzonyi B, Győrffy B, Cedrone E, Dobrovolskaia MA, Józsi M, Szebeni J. Mini-Factor H Modulates Complement-Dependent IL-6 and IL-10 Release in an Immune Cell Culture (PBMC) Model: Potential Benefits Against Cytokine Storm. Front Immunol 2021; 12:642860. [PMID: 33995361 PMCID: PMC8113956 DOI: 10.3389/fimmu.2021.642860] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
Cytokine storm (CS), an excessive release of proinflammatory cytokines upon overactivation of the innate immune system, came recently to the focus of interest because of its role in the life-threatening consequences of certain immune therapies and viral diseases, including CAR-T cell therapy and Covid-19. Because complement activation with subsequent anaphylatoxin release is in the core of innate immune stimulation, studying the relationship between complement activation and cytokine release in an in vitro CS model holds promise to better understand CS and identify new therapies against it. We used peripheral blood mononuclear cells (PBMCs) cultured in the presence of autologous serum to test the impact of complement activation and inhibition on cytokine release, testing the effects of liposomal amphotericin B (AmBisome), zymosan and bacterial lipopolysaccharide (LPS) as immune activators and heat inactivation of serum, EDTA and mini-factor H (mfH) as complement inhibitors. These activators induced significant rises of complement activation markers C3a, C4a, C5a, Ba, Bb, and sC5b-9 at 45 min of incubation, with or without ~5- to ~2,000-fold rises of IL-1α, IL-1β, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13 and TNFα at 6 and 18 h later. Inhibition of complement activation by the mentioned three methods had differential inhibition, or even stimulation of certain cytokines, among which effects a limited suppressive effect of mfH on IL-6 secretion and significant stimulation of IL-10 implies anti-CS and anti-inflammatory impacts. These findings suggest the utility of the model for in vitro studies on CS, and the potential clinical use of mfH against CS.
Collapse
Affiliation(s)
- Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Tamás Bakos
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | | | - Dániel Bencze
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Barbara Uzonyi
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs Győrffy
- Second Department of Bioinformatics and Pediatrics, Semmelweis University, Budapest, Hungary
- Lendület Cancer Biomarker Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Edward Cedrone
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Marina A. Dobrovolskaia
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Mihály Józsi
- MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary
| |
Collapse
|
35
|
Sun H, Wu Y, Zhang Y, Ni B. IL-10-Producing ILCs: Molecular Mechanisms and Disease Relevance. Front Immunol 2021; 12:650200. [PMID: 33859642 PMCID: PMC8042445 DOI: 10.3389/fimmu.2021.650200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
Innate lymphoid cells (ILCs) are mainly composed of natural killer (NK) cells and helper-like lymphoid cells, which play a vital role in maintaining tissue homeostasis, enhancing adaptive immunity and regulating tissue inflammation. Alteration of the distribution and function of ILCs subgroups are closely related to the pathogenesis of inflammatory diseases and cancers. Interleukin-10 (IL-10) is a highly pleiotropic cytokine, and can be secreted by several cell types, among of which ILCs are recently verified to be a key source of IL-10. So far, the stable production of IL-10 can only be observed in certain NK subsets and ILC2s. Though the regulatory mechanisms for ILCs to produce IL-10 are pivotal for understanding ILCs and potential intervenes of diseases, which however is largely unknown yet. The published studies show that ILCs do not share exactly the same mechanisms for IL-10 production with helper T cells. In this review, the molecular mechanisms regulating IL-10 production in NK cells and ILC2s are discussed in details for the first time, and the role of IL-10-producing ILCs in diseases such as infections, allergies, and cancers are summarized.
Collapse
Affiliation(s)
- Hui Sun
- Department of Pathophysiology, Third Military Medical University, Chongqing, China
| | - Yuzhang Wu
- Chongqing International Institute for Immunology, Chongqing, China
| | - Yi Zhang
- Chongqing International Institute for Immunology, Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
36
|
Yoneyama T, Nakano N, Hara M, Yamada H, Izawa K, Uchida K, Kaitani A, Ando T, Kitaura J, Ohtsuka Y, Ogawa H, Okumura K, Shimizu T. Notch signaling contributes to the establishment of sustained unresponsiveness to food allergens by oral immunotherapy. J Allergy Clin Immunol 2021; 147:1063-1076.e9. [DOI: 10.1016/j.jaci.2020.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 06/02/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
|
37
|
Zhou X, Jin N, Chen B. Human cytomegalovirus infection: A considerable issue following allogeneic hematopoietic stem cell transplantation. Oncol Lett 2021; 21:318. [PMID: 33692850 PMCID: PMC7933754 DOI: 10.3892/ol.2021.12579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cytomegalovirus (CMV) is an opportunistic virus, whereby recipients are most susceptible following allogeneic hematopoietic stem cell transplantation (allo-HSCT). With the development of novel immunosuppressive agents and antiviral drugs, accompanied with the widespread application of prophylaxis and preemptive treatment, significant developments have been made in transplant recipients with human (H)CMV infection. However, HCMV remains an important cause of short- and long-term morbidity and mortality in transplant recipients. The present review summarizes the molecular mechanism and risk factors of HCMV reactivation following allo-HSCT, the diagnosis of CMV infection following allo-HSCT, prophylaxis and treatment of HCMV infection, and future perspectives. All relevant literature were retrieved from PubMed and have been reviewed.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Nan Jin
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
38
|
Ohya S, Matsui M, Kajikuri J, Endo K, Kito H. Increased Interleukin-10 Expression by the Inhibition of Ca 2+-Activated K + Channel K Ca3.1 in CD4 +CD25 + Regulatory T Cells in the Recovery Phase in an Inflammatory Bowel Disease Mouse Model. J Pharmacol Exp Ther 2021; 377:75-85. [PMID: 33504590 DOI: 10.1124/jpet.120.000395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract arising from abnormal responses of the innate and adaptative immune systems. Interleukin (IL)-10-producing CD4+CD25+ regulatory T (Treg) cells play a protective role in the recovery phase of IBD. In the present study, the effects of the administration of the selective Ca2+-activated K+ channel KCa3.1 inhibitor TRAM-34 on disease activities were examined in chemically induced IBD model mice. IBD disease severity, as assessed by diarrhea, visible fecal blood, inflammation, and crypt damage in the colon, was significantly lower in mice administered 1 mg/kg TRAM-34 than in vehicle-administered mice. Quantitative real-time polymerase chain reaction examinations showed that IL-10 expression levels in the recovery phase were markedly increased by the inhibition of KCa3.1 in mesenteric lymph node (mLN) Treg cells of IBD model mice compared with vehicle-administered mice. Among several positive and negative transcriptional regulators (TRs) for IL-10, three positive TRs-E4BP4, KLF4, and Blimp1-were upregulated by the inhibition of KCa3.1 in the mLN Treg cells of IBD model mice. In mouse peripheral CD4+CD25+ Treg cells induced by lectin stimulation, IL-10 expression and secretion were enhanced by the treatment with TRAM-34, together with the upregulation of E4BP4, KLF4, and Blimp1. Collectively, the present results demonstrated that the pharmacological inhibition of KCa3.1 decreased IBD symptoms in the IBD model by increasing IL-10 production in peripheral Treg cells and that IL-10high Treg cells produced by the treatment with KCa3.1 inhibitor may contribute to efficient Treg therapy for chronic inflammatory disorders, including IBD. SIGNIFICANCE STATEMENT: Pharmacological inhibition of Ca2+-activated K+ channel KCa3.1 increased IL-10 expression in peripheral Treg cells, together with the upregulation of the transcriptional regulators of IL-10: Krüppel-like factor 4, E4 promoter-binding protein 4, and/or B lymphocyte-induced maturation protein 1. The manipulation of IL-10high-producing Treg cells by the pharmacological inhibition of KCa3.1 may be beneficial in the treatment of chronic inflammatory diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Miki Matsui
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kyoko Endo
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
39
|
Nanda A, K T, Banerjee P, Dutta M, Wangdi T, Sharma P, Chaudhury K, Jana SK. Cytokines, Angiogenesis, and Extracellular Matrix Degradation are Augmented by Oxidative Stress in Endometriosis. Ann Lab Med 2021; 40:390-397. [PMID: 32311852 PMCID: PMC7169633 DOI: 10.3343/alm.2020.40.5.390] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/26/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
Background The effect of the interplay among inflammation, angiogenesis, extracellular matrix (ECM) degradation, and oxidative stress (OS) on the pathogenesis of endometriosis remains unclear. Previously, we demonstrated the role of OS in endometriosis. Here, we performed a comprehensive investigation of several molecules involved in inflammation, angiogenesis, and ECM degradation in women with endometriosis to study their interplay with OS. Methods Blood samples were collected from women with endometriosis (N=80), as well as from women with tubal factor infertility as controls (N=80). Interleukin (IL)-1β, tumor necrosis factor-alpha, interferon-gamma, transforming growth factor-beta, IL-4, −10, −2, −6, −8, vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-2, −9, tissue inhibitor of metalloproteinases (TIMP)-1, −2, and cyclooxygenase (COX)-2 levels in serum samples were measured using an ELISA. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in peripheral blood mononuclear cells was measured using flow cytometry. Results Cytokines, VEGF, MMPs, and COX-2 were significantly higher and TIMPs were significantly lower in patients with endometriosis. Multivariate statistical analysis indicated that IL-10 was the most significant variable capable of discriminating endometriosis samples from controls. Conclusions Deregulation of NF-κB activation by OS affects the expression of various cytokines in endometriosis. Elevated cytokine levels further up-regulate IL-10, which subsequently activates the MMPs, leading to excessive ECM degradation and angiogenesis. Moreover, IL-10 emerged as the most important molecule involved in the pathogenesis of endometriosis. Measurement of these molecules may help in better management of the patients with endometriosis.
Collapse
Affiliation(s)
- Amalesh Nanda
- Department of Biotechnology, National Institute of Technology, Yupia, Papum Pare, Arunachal Pradesh, India
| | - Thangapandi K
- Department of Biotechnology, National Institute of Technology, Yupia, Papum Pare, Arunachal Pradesh, India
| | - Priyanka Banerjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India
| | - Mainak Dutta
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani, Dubai Campus, Dubai International Academic City, Dubai, UAE
| | - Tsering Wangdi
- Department of Obstetrics and Gynecology, Pratiksha Hospital, Borbari, Guwahati, Assam, India
| | - Pramod Sharma
- Department of Obstetrics and Gynecology, Pratiksha Hospital, Borbari, Guwahati, Assam, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India
| | - Saikat Kumar Jana
- Department of Biotechnology, National Institute of Technology, Yupia, Papum Pare, Arunachal Pradesh, India
| |
Collapse
|
40
|
Dong Z, Liu Z, Dai H, Liu W, Feng Z, Zhao Q, Gao Y, Liu F, Zhang N, Dong X, Zhou X, Du J, Huang G, Tian X, Liu B. The Potential Role of Regulatory B Cells in Idiopathic Membranous Nephropathy. J Immunol Res 2020; 2020:7638365. [PMID: 33426094 PMCID: PMC7772048 DOI: 10.1155/2020/7638365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/22/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Regulatory B cells (Breg) are widely regarded as immunomodulatory cells which play an immunosuppressive role. Breg inhibits pathological autoimmune response by secreting interleukin-10 (IL-10), transforming growth factor-β (TGF-β), and adenosine and through other ways to prevent T cells and other immune cells from expanding. Recent studies have shown that different inflammatory environments induce different types of Breg cells, and these different Breg cells have different functions. For example, Br1 cells can secrete IgG4 to block autoantigens. Idiopathic membranous nephropathy (IMN) is an autoimmune disease in which the humoral immune response is dominant and the cellular immune response is impaired. However, only a handful of studies have been done on the role of Bregs in this regard. In this review, we provide a brief overview of the types and functions of Breg found in human body, as well as the abnormal pathological and immunological phenomena in IMN, and propose the hypothesis that Breg is activated in IMN patients and the proportion of Br1 can be increased. Our review aims at highlighting the correlation between Breg and IMN and proposes potential mechanisms, which can provide a new direction for the discovery of the pathogenesis of IMN, thus providing a new strategy for the prevention and early treatment of IMN.
Collapse
Affiliation(s)
- Zhaocheng Dong
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Zhiyuan Liu
- Shandong First Medical University, No. 619 Changcheng Road, Tai'an City, Shandong 271016, China
| | - Haoran Dai
- Shunyi Branch, Beijing Traditional Chinese Medicine Hospital, Station East 5, Shunyi District, Beijing 101300, China
| | - Wenbin Liu
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
| | - Zhendong Feng
- Beijing Chinese Medicine Hospital Pinggu Hospital, No. 6, Pingxiang Road, Pinggu District, Beijing 101200, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Yu Gao
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Fei Liu
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Na Zhang
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Xuan Dong
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
- Capital Medical University, No. 10, Xitoutiao, You'anmenwai, Fengtai District, Beijing 100069, China
| | - Xiaoshan Zhou
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Jieli Du
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| | - Guangrui Huang
- Beijing University of Chinese Medicine, No. 11, North Third Ring Road, Chaoyang District, Beijing 100029, China
| | - Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, No. 23 Meishuguanhou Street, Dongcheng District, Beijing 100010, China
| |
Collapse
|
41
|
Abstract
Autoimmune diseases are one of the dreadful group of human diseases that have always been of keen interest to researchers. Due to complex and broad-spectrum nature, scientists are not yet able to pinpoint the pathogenesis of and delineate effective therapy against this group of diseases. However, it is becoming clear that a decrease in number and function of T regulatory cells (Treg), an increase in autoreactive Th1/Th17 cells and associated immunomodulation and inflammation participate in the pathogenesis of many autoimmune diseases. Cinnamon (Cinnamonum verum or Cinnamonum cassia) is a widely used natural spice and flavoring ingredient and its metabolite sodium benzoate (NaB) is a food-additive and FDA-approved drug against nonketotic hyperglycinemia (NKH) and urea cycle disorders (UCD). Recent studies indicate that cinnamon either in powder or extract form and NaB are capable of modulating different autoimmune pathways as well as protecting animals from different autoimmune disorders. Here, we have made an honest attempt to delineate such pieces of evidence with available anti-autoimmune mechanisms and analyze whether cinnamon supplements could be used to control the fury of autoimmune disorders.
Collapse
Affiliation(s)
- Swarupa Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| |
Collapse
|
42
|
Ariyoshi T, Hagihara M, Eguchi S, Fukuda A, Iwasaki K, Oka K, Takahashi M, Yamagishi Y, Mikamo H. Clostridium butyricum MIYAIRI 588-Induced Protectin D1 Has an Anti-inflammatory Effect on Antibiotic-Induced Intestinal Disorder. Front Microbiol 2020; 11:587725. [PMID: 33193245 PMCID: PMC7661741 DOI: 10.3389/fmicb.2020.587725] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
Metabolites are thought as the end products in cellular regulatory processes and their levels show the strongest relationships with the phenotype. Previously, we showed that the administration of Clostridium butyricum MIYAIRI 588 (CBM 588) upregulated protectin D1, an anti-inflammatory lipid metabolite, in colon tissue under antibiotic therapy. However, how CBM 588 induces protectin D1 expression and whether the metabolite has anti-inflammatory effects on antibiotic-induced inflammation are unclear. Therefore, here, we evaluated the effect of CBM 588 on lipid metabolism and protectin D1 in gut protection from antibiotic-induced intestinal disorders. In the CBM 588 treatment group, expression levels of genes encoding lipid receptors related to the conversion of DHA to protectin D1, such as polyunsaturated fatty acid (PUFA) receptors, G-protein coupled receptor 120 (GPR120), and 15-lipoxygenase (LOX), were increased in colon tissue. CD4+ cells producing interleukin (IL)-4, the main component of T helper type 2 (Th2) cells that can activate 15-LOX, also increased in CBM 588-treated groups even after clindamycin co-administration. In addition, similar to CBM 588, exogenously administered protectin D1 reduced inflammatory cytokines, while IL-10 and TGF-β1, works as anti-inflammatory cytokines, were increased. Our data revealed that CBM 588 activated 15-LOX to enhance protectin D1 production by increasing IL-4-producing CD4+ cell population in the intestinal tract. Additionally, CBM 588-induced protectin D1 clearly upregulated IL-10-producing CD4+ cells to control antibiotic-induced gut inflammation. We provide new insights into CBM 588-mediated lipid metabolism induction for the treatment of gut inflammatory diseases.
Collapse
Affiliation(s)
- Tadashi Ariyoshi
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Mao Hagihara
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan.,Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University, Nagakute, Japan
| | | | - Aiki Fukuda
- Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Kenta Iwasaki
- Departments of Kidney Disease and Transplant Immunology, Aichi Medical University, Nagakute, Japan
| | - Kentaro Oka
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan.,Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University, Nagakute, Japan
| | - Motomichi Takahashi
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan.,Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University Graduate School of Medicine, Nagakute, Japan
| |
Collapse
|
43
|
Matsubara Y, Kiwan G, Fereydooni A, Langford J, Dardik A. Distinct subsets of T cells and macrophages impact venous remodeling during arteriovenous fistula maturation. JVS Vasc Sci 2020; 1:207-218. [PMID: 33748787 PMCID: PMC7971420 DOI: 10.1016/j.jvssci.2020.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Patients with end-stage renal failure depend on hemodialysis indefinitely without renal transplantation, requiring a long-term patent vascular access. While the arteriovenous fistula (AVF) remains the preferred vascular access for hemodialysis because of its longer patency and fewer complications compared with other vascular accesses, the primary patency of AVF is only 50-60%, presenting a clinical need for improvement. AVF mature by developing a thickened vascular wall and increased diameter to adapt to arterial blood pressure and flow volume. Inflammation plays a critical role during vascular remodeling and fistula maturation; increased shear stress triggers infiltration of T-cells and macrophages that initiate inflammation, with involvement of several different subsets of T-cells and macrophages. We review the literature describing distinct roles of the various subsets of T-cells and macrophages during vascular remodeling. Immunosuppression with sirolimus or prednisolone reduces neointimal hyperplasia during AVF maturation, suggesting novel approaches to enhance vascular remodeling. However, M2 macrophages and CD4+ T-cells play essential roles during AVF maturation, suggesting that total immunosuppression may suppress adaptive vascular remodeling. Therefore it is likely that regulation of inflammation during fistula maturation will require a balanced approach to coordinate the various inflammatory cell subsets. Advances in immunosuppressive drug development and delivery systems may allow for more targeted regulation of inflammation to improve vascular remodeling and enhance AVF maturation.
Collapse
Affiliation(s)
- Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| |
Collapse
|
44
|
Han X, Wu D, Sun Z, Sun H, Lv Q, Zhang L, Wei Y. Type 1/type 2 inflammatory cytokines correlate with olfactory function in patients with chronic rhinosinusitis. Am J Otolaryngol 2020; 41:102587. [PMID: 32516657 DOI: 10.1016/j.amjoto.2020.102587] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/09/2020] [Accepted: 05/25/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND Olfactory dysfunction secondary to chronic rhinosinusitis (CRS) has been highly associated with impaired quality of life. Asian CRS patients showed a distinct inflammatory profile, with less type 2 endotype compared with European and North American. This study aimed to explore the pattern of the inflammatory cytokines in CRS patients from China and their association with olfactory function. METHODS Institutional review board-approved prospective study in which the olfactory function of 71 CRS patients was assessed with Sniffin' Sticks before the nasal endoscopic surgery. A set of cytokines and inflammatory mediators including type 1 and type 2 inflammatory cytokines were measured in nasal mucus by using a multiplex flow cytometric bead assay (CBA). Baseline characteristics in CRS patients were collected and the Spearman r statistic was performed to assess the association of olfactory function with cytokines and inflammatory mediators. RESULTS A total of 71 nasal mucus samples of CRS patients, including 25 chronic rhinosinusitis without nasal polyposis (CRSsNP) patients and 46 chronic rhinosinusitis with nasal polyposis (CRSwNP) patients, were evaluated in this study. The nasal mucus levels of type 1 inflammatory cytokine IFN-γ (interferon-γ), type 2 inflammatory cytokines including IL-4, IL-5 and GM-CSF (granulocyte-macrophage colony-stimulating factor) and anti-inflammatory cytokine IL-10 were significantly and inversely correlated with olfactory function in total patients with CRS (r = -0.308, p = 0.009; r = -0.250, p = 0.036; r = -0.399, p = 0.001; r = -0.269, p = 0.023; r = -0.273, p = 0.021, respectively). In CRSsNP, the olfactory function was inversely correlated with levels of type 1 inflammatory cytokine TNF-α (tumor necrosis factor-α) (r = -0.637, p = 0.001) and IL-10 (r = -0.468, p = 0.018). Nevertheless, the olfactory function in CRSwNP was inversely correlated with type 2 inflammatory cytokines including IL-4 (r = -0.303, p = 0.041) and IL-5 (r = -0.383, p = 0.009). CONCLUSION Both type 1 and type 2 inflammatory cytokines may contribute to the pathogenesis of CRS-associated olfactory dysfunction in the Chinese population.
Collapse
Affiliation(s)
- Xingyu Han
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Dawei Wu
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Zhifu Sun
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Haili Sun
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China; Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, No. 2 Anzhen Road, Beijing 100029, China
| | - Qianwen Lv
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Lichuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Yongxiang Wei
- Department of Otolaryngology-Head and Neck Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
45
|
Li R, Wu K, Li Y, Liang X, Tse WKF, Yang L, Lai KP. Revealing the targets and mechanisms of vitamin A in the treatment of COVID-19. Aging (Albany NY) 2020; 12:15784-15796. [PMID: 32805728 PMCID: PMC7467385 DOI: 10.18632/aging.103888] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), an epidemic disease characterized by rapid infection and a high death toll. The clinical diagnosis of patients with COVID-19 has risen sharply, especially in Western countries. Globally, an effective treatment for COVID-19 is still limited. Vitamin A (VA) exhibits pharmacological activity in the management of pneumonia. Thus, we reason that VA may potentially serve as an anti-SARS-CoV-2 regimen. In this study, bioinformatics analysis and computation assays using a network pharmacology method were conducted to explore and uncover the therapeutic targets and mechanisms of VA for treating COVID-19. We identified candidate targets, pharmacological functions, and therapeutic pathways of VA against SARS-CoV-2. Bioinformatics findings indicate that the mechanisms of action of VA against SARS-CoV-2 include enrichment of immunoreaction, inhibition of inflammatory reaction, and biological processes related to reactive oxygen species. Furthermore, seven core targets of VA against COVID-19, including MAPK1, IL10, EGFR, ICAM1, MAPK14, CAT, and PRKCB were identified. With this bioinformatics-based report, we reveal, for the first time, the anti-SARS-CoV-2 functions and mechanisms of VA and suggest that VA may act as a potent treatment option for COVID-19, a deadly global epidemic.
Collapse
Affiliation(s)
- Rong Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Ka Wu
- Department of Pharmacy, The Second People's Hospital of Nanning City, The Third Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Li
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Xiao Liang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - William Ka Fai Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Lu Yang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Keng Po Lai
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| |
Collapse
|
46
|
Lin X, Ma X, Cui X, Zhang R, Pan H, Gao W. Effects of Erythropoietin on Lung Injury Induced by Cardiopulmonary Bypass After Cardiac Surgery. Med Sci Monit 2020; 26:e920039. [PMID: 32310911 PMCID: PMC7191960 DOI: 10.12659/msm.920039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Lung injury after cardiopulmonary bypass (CPB) is a serious postoperative complication and can affect the postoperative recovery. The purpose of this study was to explore whether erythropoietin (EPO) has an effect on lung injury caused by CPB. MATERIAL AND METHODS Sixty patients who received the CPB were randomly divided into a saline group and the EPO group. All the patients received saline or EPO preoperatively, respectively. The ventilation function, including dynamic compliance, peak airway pressure, and plateau pressure, were recorded. The level of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1ß, and IL-10 in serum and arterial blood gas were analyzed. The mechanical ventilation time in the intensive care unit (ICU), the length of time spent in the ICU, the time from operation to discharge, and the total time of hospitalization were recorded. Adverse events in the ICU were monitored and recorded. RESULTS EPO significantly decreased the level of TNF-alpha and IL-1ß, but increased the level of IL-10 after CPB. EPO significantly improved pulmonary ventilated function and gas exchange function after CPB. EPO significantly shortened the mechanical ventilation time and stay in the ICU. CONCLUSIONS Preoperative EPO injection reduced lung injury and promoted lung function in patients who underwent CPB. The protection effect of EPO may be associated with inhibition of inflammatory response.
Collapse
Affiliation(s)
- Xue Lin
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xiaobei Ma
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xiaoguang Cui
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Ruiqin Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Hong Pan
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Wei Gao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
47
|
Depletion of Blautia Species in the Microbiota of Obese Children Relates to Intestinal Inflammation and Metabolic Phenotype Worsening. mSystems 2020; 5:5/2/e00857-19. [PMID: 32209719 PMCID: PMC7093825 DOI: 10.1128/msystems.00857-19] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Child obesity constitutes a risk factor for developing insulin resistance which, if sustained, could lead to more severe conditions like type 2 diabetes (T2D) in adulthood. Our study identified previously unknown species whose depletion (Blautia luti and Blautia wexlerae) is associated with insulin resistance in obese individuals. Our results also indicate that these bacterial species might help to reduce inflammation causally linked to obesity-related complications. Childhood is considered a window of opportunity to tackle obesity. These new findings provide, therefore, valuable information for the future design of microbiota-based strategies for the early prevention of obesity-related complications. Cross-sectional studies conducted with obese and control subjects have suggested associations between gut microbiota alterations and obesity, but the links with specific disease phenotypes and proofs of causality are still scarce. The present study aimed to profile the gut microbiota of lean and obese children with and without insulin resistance to characterize associations with specific obesity-related complications and understand the role played in metabolic inflammation. Through massive sequencing of 16S rRNA gene amplicons and data analysis using a novel permutation approach, we have detected decreased incidence of Blautia species, especially Blautia luti and B. wexlerae, in the gut microbiota of obese children, which was even more pronounced in cases with both obesity and insulin resistance. There was also a parallel increase in proinflammatory cytokines and chemokines (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and monocyte chemoattractant protein 1 [MCP-1]) in feces of obese children compared to those of lean ones. B. luti and B. wexlerae were also shown to exert an anti-inflammatory effect in peripheral blood mononuclear cell cultures in vitro, compared to non-obesity-associated species. We suggest that the depletion of B. luti and B. wexlerae species in the gut ecosystem may occur in cases of obesity and contribute to metabolic inflammation leading to insulin resistance. IMPORTANCE Child obesity constitutes a risk factor for developing insulin resistance which, if sustained, could lead to more severe conditions like type 2 diabetes (T2D) in adulthood. Our study identified previously unknown species whose depletion (Blautia luti and Blautia wexlerae) is associated with insulin resistance in obese individuals. Our results also indicate that these bacterial species might help to reduce inflammation causally linked to obesity-related complications. Childhood is considered a window of opportunity to tackle obesity. These new findings provide, therefore, valuable information for the future design of microbiota-based strategies for the early prevention of obesity-related complications.
Collapse
|
48
|
Chen X, Wang R, Meng W, Zhang X. Exploration of the Molecular Mechanism of FUZI (Aconiti Lateralis Radix Praeparata) in Allergic Rhinitis Treatment Based on Network Pharmacology. Med Sci Monit 2020; 26:e920872. [PMID: 32114589 PMCID: PMC7065509 DOI: 10.12659/msm.920872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
FUZI (Aconiti Lateralis Radix Praeparata) is a traditional Chinese medicine herb used extensively for nourishing yang (regarded as the positive, male universal force), which is critical in treatment of allergic rhinitis. In this paper, FUZI was explored based on network pharmacology. The active components of FUZI were screened out, its protein targets were assessed, and the protein interaction network map was built with the differential protein of allergic rhinitis, as an attempt to determine the critical targets of FUZI for treating allergic rhinitis. Subsequently, DAVID was employed to explore the biological function and pathway enrichment to determine the biological pathway of FUZI for treating allergic rhinitis. As suggested by the results, FUZI is likely to affect the inhibition of inflammation and the regulation of immunity, probably reducing the incidence of allergic rhinitis, or alleviating nasal discomfort attributed to allergic inflammation. The targets and pathways of FUZI for treating allergic rhinitis assessed by network pharmacology provided a direction for our subsequent studies and may be a novel therapeutic target.
Collapse
Affiliation(s)
- Xiangjing Chen
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Renzhong Wang
- Department of Otorhinolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Wei Meng
- Department of Otorhinolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|