1
|
Han J, Cherry C, Mejías JC, Krishnan K, Ruta A, Maestas DR, Peña AN, Nguyen HH, Nagaraj S, Yang B, Gray-Gaillard EF, Rutkowski N, Browne M, Tam AJ, Fertig EJ, Housseau F, Ganguly S, Moore EM, Pardoll DM, Elisseeff JH. Age-associated Senescent - T Cell Signaling Promotes Type 3 Immunity that Inhibits the Biomaterial Regenerative Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310476. [PMID: 38087458 DOI: 10.1002/adma.202310476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Indexed: 12/30/2023]
Abstract
Aging is associated with immunological changes that compromise response to infections and vaccines, exacerbate inflammatory diseases and can potentially mitigate tissue repair. Even so, age-related changes to the immune response to tissue damage and regenerative medicine therapies remain unknown. Here, it is characterized how aging induces changes in immunological signatures that inhibit tissue repair and therapeutic response to a clinical regenerative biological scaffold derived from extracellular matrix. Signatures of inflammation and interleukin (IL)-17 signaling increased with injury and treatment both locally and regionally in aged animals, and computational analysis uncovered age-associated senescent-T cell communication that promotes type 3 immunity in T cells. Local inhibition of type 3 immune activation using IL17-neutralizing antibodies improves healing and restores therapeutic response to the regenerative biomaterial, promoting muscle repair in older animals. These results provide insights into tissue immune dysregulation that occurs with aging that can be targeted to rejuvenate repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Christopher Cherry
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Joscelyn C Mejías
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Kavita Krishnan
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Anna Ruta
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - David R Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Alexis N Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Helen Hieu Nguyen
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Sushma Nagaraj
- Department of Neurology, Brain Science Institute, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Brenda Yang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Elise F Gray-Gaillard
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Natalie Rutkowski
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Maria Browne
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Ada J Tam
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Elana J Fertig
- Department of Biomedical Engineering and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Franck Housseau
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sudipto Ganguly
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Erika M Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Drew M Pardoll
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| |
Collapse
|
2
|
Chen F, Li J, Li L, Tong L, Wang G, Zou X. Multidimensional biological characteristics of ground glass nodules. Front Oncol 2024; 14:1380527. [PMID: 38841161 PMCID: PMC11150621 DOI: 10.3389/fonc.2024.1380527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
The detection rate of ground glass nodules (GGNs) has increased in recent years because of their malignant potential but relatively indolent biological behavior; thus, correct GGN recognition and management has become a research focus. Many scholars have explored the underlying mechanism of the indolent progression of GGNs from several perspectives, such as pathological type, genomic mutational characteristics, and immune microenvironment. GGNs have different major mutated genes at different stages of development; EGFR mutation is the most common mutation in GGNs, and p53 mutation is the most abundant mutation in the invasive stage of GGNs. Pure GGNs have fewer genomic alterations and a simpler genomic profile and exhibit a gradually evolving genomic mutation profile as the pathology progresses. Compared to advanced lung adenocarcinoma, GGN lung adenocarcinoma has a higher immune cell percentage, is under immune surveillance, and has less immune escape. However, as the pathological progression and solid component increase, negative immune regulation and immune escape increase gradually, and a suppressive immune environment is established gradually. Currently, regular computer tomography monitoring and surgery are the main treatment strategies for persistent GGNs. Stereotactic body radiotherapy and radiofrequency ablation are two local therapeutic alternatives, and systemic therapy has been progressively studied for lung cancer with GGNs. In the present review, we discuss the characterization of the multidimensional molecular evolution of GGNs that could facilitate more precise differentiation of such highly heterogeneous lesions, laying a foundation for the development of more effective individualized treatment plans.
Collapse
Affiliation(s)
- Furong Chen
- Department of Oncology, The First People’s Hospital of Shuangliu District/West China (Airport) Hospital, Sichuan University, Chengdu, China
| | - Jiangtao Li
- Department of Oncology, The First People’s Hospital of Shuangliu District/West China (Airport) Hospital, Sichuan University, Chengdu, China
| | - Lei Li
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
- Department of State Key Laboratory of Respiratory Health and Multimobidity, West China Hospital, Sichuan University, Chengdu, China
| | - Lunbing Tong
- Department of Respiratory Medicine, Chengdu Seventh People’s Hospital/Affiliated Cancer Hospital of Chengdu Medical College, Chengdu, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
- Department of State Key Laboratory of Respiratory Health and Multimobidity, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelin Zou
- Department of Respiratory Medicine, Chengdu Seventh People’s Hospital/Affiliated Cancer Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
3
|
Sundararaj R, Mathimaran A, Prabhu D, Ramachandran B, Jeyaraman J, Muthupandian S, Asmelash T. In silico approaches for the identification of potential allergens among hypothetical proteins from Alternaria alternata and its functional annotation. Sci Rep 2024; 14:6696. [PMID: 38509156 PMCID: PMC10954717 DOI: 10.1038/s41598-024-55463-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Direct exposure to the fungal species Alternaria alternata is a major risk factor for the development of asthma, allergic rhinitis, and inflammation. As of November 23rd 2020, the NCBI protein database showed 11,227 proteins from A. alternata genome as hypothetical proteins (HPs). Allergens are the main causative of several life-threatening diseases, especially in fungal infections. Therefore, the main aim of the study is to identify the potentially allergenic inducible proteins from the HPs in A. alternata and their associated functional assignment for the complete understanding of the complex biological systems at the molecular level. AlgPred and Structural Database of Allergenic Proteins (SDAP) were used for the prediction of potential allergens from the HPs of A. alternata. While analyzing the proteome data, 29 potential allergens were predicted by AlgPred and further screening in SDAP confirmed the allergic response of 10 proteins. Extensive bioinformatics tools including protein family classification, sequence-function relationship, protein motif discovery, pathway interactions, and intrinsic features from the amino acid sequence were used to successfully predict the probable functions of the 10 HPs. The functions of the HPs are characterized as chitin-binding, ribosomal protein P1, thaumatin, glycosyl hydrolase, and NOB1 proteins. The subcellular localization and signal peptide prediction of these 10 proteins has further provided additional information on localization and function. The allergens prediction and functional annotation of the 10 proteins may facilitate a better understanding of the allergenic mechanism of A. alternata in asthma and other diseases. The functional domain level insights and predicted structural features of the allergenic proteins help to understand the pathogenesis and host immune tolerance. The outcomes of the study would aid in the development of specific drugs to combat A. alternata infections.
Collapse
Affiliation(s)
- Rajamanikandan Sundararaj
- Department of Biochemistry, Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Amala Mathimaran
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, 630 004, India
| | - Dhamodharan Prabhu
- Department of Biotechnology, Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Balajee Ramachandran
- Department of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston University, 700 Albany Street, Boston, MA, 02118, USA
| | - Jeyakanthan Jeyaraman
- Structural Biology and Biocomputing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, 630 004, India
| | - Saravanan Muthupandian
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600077, India
| | - Tsehaye Asmelash
- Department of Medical Microbiology and Immunology, College of Health Sciences, Mekelle University, Mekelle, Tigray, Ethiopia.
| |
Collapse
|
4
|
Ahmed RH, Rashad Ahmed R, Galaly SR, Moustafa N, Abourehab MAS, Abdelgawad MA, Ahmed OM, Abdul-Hamid M. Mesenchymal Stem Cells and Curcumin Effectively Mitigate Freund's Adjuvant- induced Arthritis via their Anti-inflammatory and Gene Expression of COX-1, IL-6 and IL-4. Endocr Metab Immune Disord Drug Targets 2024; 24:468-488. [PMID: 36825726 DOI: 10.2174/1871530323666230223143011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 02/25/2023]
Abstract
BACKGROUND AND OBJECTIVES Rheumatoid arthritis (RA) is a type of arthritis that damages joints and can affect the thymus and the spleen. RA is an autoimmune disorder in which the immune system targets the body's own tissues. The causes of RA are unknown, although a genetic link is thought to be involved. The objective of this research was to evaluate the effect of curcumin, mesenchymal stem cells (MSCs), and their combination on the disruption of serum cytokines, ankle joint, thymus and spleen histopathology, and affected genes in complete Freund's adjuvant (CFA)-induced arthritis in male and female Wistar rats. METHODS Experimental animals were organized into 16 groups (6 animals for each), eight groups including male rats and the other eight groups including females rats. The groups are normal control, CMC, curcumin, MSCs, CFA, CFA/curcumin, CFA/ MSCs and the arthritic group treated with MSCs and curcumin. One subcutaneous injection of 0.1 mL CFA was given to rats into the right hind leg footpad to induce RA. The arthritic rats were intravenously injected three times with bone marrow-derived MSCs (BM-MSCs) and/or treated orally with curcumin daily (100 mg per kg body weight per day) for 21 days. RESULTS Curcumin and BM-MSCs work together to dramatically (P < 0.05) restore the high serum PGE2 and IL-17 levels and lower the IL-13 level in arthritic rats to normal levels. Deleterious effects on the spleen and thymus histological structure were counteracted. Gene expression of COX-1 and IL-6 was increased and IL-4 was decreased; these changes were improved by the combination treatment (P < 0.05). CONCLUSION Based on these findings, additive therapeutic effects on RA occur from the combined treatment of curcumin and BM-MSCs compared with their individual use (P < 0.05). Thus, it can be said that both curcumin and BM-MSCs are effective at reducing inflammation while also having beneficial effects on the ankle joint, thymus and spleen.
Collapse
Affiliation(s)
- Rania Hamed Ahmed
- Department of Zoology, Faculty of Science, Histology, Cell Biology and Genetic Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Rasha Rashad Ahmed
- Department of Zoology, Faculty of Science, Histology, Cell Biology and Genetic Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Sanaa Rida Galaly
- Department of Zoology, Faculty of Science, Histology, Cell Biology and Genetic Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Nadia Moustafa
- Department of Zoology, Faculty of Science, Histology, Cell Biology and Genetic Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Mohammed Abdelwahab Sayed Abourehab
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | | | - Osama Mohamed Ahmed
- Department of Zoology, Faculty of Science, Physiology Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Manal Abdul-Hamid
- Department of Zoology, Faculty of Science, Histology, Cell Biology and Genetic Division, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
5
|
Pan Z, Dai Y, Akar-Ghibril N, Simpson J, Ren H, Zhang L, Hou Y, Wen X, Chang C, Tang R, Sun JL. Impact of Air Pollution on Atopic Dermatitis: A Comprehensive Review. Clin Rev Allergy Immunol 2023; 65:121-135. [PMID: 36853525 DOI: 10.1007/s12016-022-08957-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 03/01/2023]
Abstract
Air pollution is associated with multiple health problems worldwide, contributing to increased morbidity and mortality. Atopic dermatitis (AD) is a common allergic disease, and increasing evidence has revealed a role of air pollution in the development of atopic dermatitis. Air pollutants are derived from several sources, including harmful gases such as nitrogen dioxide (NO2), sulfur dioxide (SO2), and carbon monoxide (CO), as well as particulate matter (PM) of various sizes, and bioaerosols. Possible mechanisms linking air pollution to atopic dermatitis include damage to the skin barrier through oxidative stress, increased water loss, physicochemical injury, and an effect on skin microflora. Furthermore, oxidative stress triggers immune dysregulation, leading to enhanced sensitization to allergens. There have been multiple studies focusing on the association between various types of air pollutants and atopic dermatitis. Since there are many confounders in the current research, such as climate, synergistic effects of mixed pollutants, and diversity of study population, it is not surprising that inconsistencies exist between different studies regarding AD and air pollution. Still, it is generally accepted that air pollution is a risk factor for AD. Future studies should focus on how air pollution leads to AD as well as effective intervention measures.
Collapse
Affiliation(s)
- Zhouxian Pan
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yimin Dai
- Eight-Year Clinical Medicine System, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Nicole Akar-Ghibril
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA
| | - Jessica Simpson
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA
| | - Huali Ren
- Department of Allergy, Beijing Electric Power Hospital of State Grid Company of China, Electric Power Teaching Hospital of Capital Medical University, Beijing, 100073, China
| | - Lishan Zhang
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yibo Hou
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xueyi Wen
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Christopher Chang
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA.
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, 95616, USA.
| | - Rui Tang
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Jin-Lyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
6
|
Kim KW, Koh SJ, Kang HW, Park H, Ha H, Park J, Kim K, Jun Y, Han YM, Lee HJ, Yoon H, Im JP, Park YS, Kim JW, Kim JS. Atopic dermatitis is associated with the clinical course of inflammatory bowel disease. Scand J Gastroenterol 2023; 58:1115-1121. [PMID: 37165647 DOI: 10.1080/00365521.2023.2209688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/23/2023] [Indexed: 05/12/2023]
Abstract
OBJECTIVES There are a few studies about the relationship between inflammatory bowel disease (IBD) and atopic dermatitis (AD). It is implied that both diseases have common pathophysiologic mechanisms and can affect each other. However, little information is available on the effect of AD on the clinical course of patients with IBD. METHODS This is a multi-center, retrospective, observational study. We define AD as a chronic eczematoid dermatosis diagnosed by dermatologists. Patients with concurrent IBD and AD were defined as a case group. Age, gender, and IBD subtype-matched patients without AD were included as a reference group. RESULTS The numbers of patients in the case and reference groups were 61 and 122 respectively. There was a significantly shorter biologics-free survival in the case group than that in the reference group according to the multivariable-adjusted Cox regression analysis with the onset age, disease duration, smoking status, use of steroid, use of immunomodulator, initial C-reactive protein, initial erythrocyte sedimentation rate, presence of other allergic diseases and initial disease severity [hazard ratio (HR) 1.828, 95% confidence interval (CI) 1.022-3.271, p = .042]. The trend was consistent in the subgroup analysis with ulcerative colitis (HR 3.498, 95% CI 1.066-11.481, p = .039), but not with Crohn's disease (HR 1.542, 95% CI 0.720-3.301, p = .265). CONCLUSIONS AD showed a significant effect on the biologics-free survival of patients with IBD and especially the UC subtype. Further mechanistic research is required to elucidate the pathogenesis of AD on the clinical course of IBD.
Collapse
Affiliation(s)
- Kwang Woo Kim
- Division of Gastroenterology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong-Joon Koh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyoun Woo Kang
- Division of Gastroenterology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunsun Park
- Department of Dermatology, Boramae Medical Center, Seoul Metropolitan Government Seoul National University, Seoul, Republic of Korea
| | - Hyeonjin Ha
- Department of Education and Training, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Kyunghoon Kim
- Division of Gastroenterology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yukyung Jun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Yoo Min Han
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine and Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Hyun Jung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Ji Won Kim
- Division of Gastroenterology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Li J, Chen Y, Shi Q, Sun J, Zhang C, Liu L. Omega-3 polyunsaturated fatty acids ameliorate PM2.5 exposure induced lung injury in mice through remodeling the gut microbiota and modulating the lung metabolism. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:40490-40506. [PMID: 36609968 PMCID: PMC9822699 DOI: 10.1007/s11356-022-25111-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
Short-term or long-term exposure to fine particulate matter (PM2.5) is related to increased incidences of respiratory diseases. This study aimed to investigate the influences of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) supplementation on oxidative stress, inflammation, lung metabolic profile, and gut microbiota in PM2.5-induced lung injury mice. Mice were divided into four groups (n = 15, per group): two unsupplemented groups, control group and PM2.5 group, and two supplemented groups with ω-3 PUFAs, ω-3 PUFAs group, and ω-3 PUFAs + PM2.5 group. Mice in the supplemented groups were placed on an ω-3 PUFAs-enriched diet (ω-3 PUFAs, 21 g/kg). During the 5th to 6th week of dietary supplementation, mice were exposed to PM2.5 by intra-tracheal instillation. ω-3 PUFAs ameliorate lung histopathological injury, reduce inflammatory responses and oxidative stress, affect lung metabolite profile, and modulate gut microbiota in PM2.5-induced lung injury mice. Thus, supplementary ω-3 PUFAs showed effectiveness in attenuation of PM2.5-induced lung injury, indicating that the interventions exhibited preventive and therapeutic potential.
Collapse
Affiliation(s)
- Jingli Li
- Department of Pulmonary and Critical Care Medicine, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Yang Chen
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Qiangqiang Shi
- Department of Respiratory Medicine, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, Zhejiang, China
| | - Jian Sun
- Department of Pulmonary and Critical Care Medicine, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Chunyi Zhang
- Department of Pulmonary and Critical Care Medicine, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China
| | - Lingjing Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
8
|
Ansari MA, Singh PK, Dar SA, Rai G, Akhter N, Pandhi D, Gaurav V, Bhattacharya SN, Banerjee BD, Ahmad A, Das S. Deregulated phenotype of autoreactive Th17 and Treg clone cells in pemphigus vulgaris after in-vitro treatment with desmoglein antigen (Dsg-3). Immunobiology 2023; 228:152340. [PMID: 36689824 DOI: 10.1016/j.imbio.2023.152340] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
The loss of balance between regulatory T (Treg) and T helper 17 (Th17) causes loss of tolerance against desmoglein (Dsg)-3 leading to pemphigus vulgaris (PV), an autoimmune bullous skin disorder associated with autoantibodies against Dsg-3. We aimed to elucidate the complex relationship of Th17 and Treg cells, their molecules, and the underlying mechanism in the development of PV disease. Using cytokine secretion assays, Th17 and Treg cells were sorted by FACS Aria-III within Dsg-3-responsive PBMC population and homogeneous T cell clones were generated in-vitro. Different cell surface molecules like CD25, GITR, CD122, CD152, CD45RO, IL-23R, STAT3, STAT5, CD127, HLA-DR, CCR4, CCR5, CCR6 and CCR7 were studied. The functional response of Th17 and Treg cells were elucidated by measuring the levels of various cytokines released by IL-10 and IL-17 T cells. The mRNA expression of transcription factors (FoxP3 and RORγt) was also analyzed. IL-17 secreting (Th17) cells with phenotype CD4+IL-17+ were greatly increased and IL-10 secreting (Treg) cells with phenotype CD4+IL-10+ were reduced in PV cases than healthy controls. The qPCR analysis showing high expression of retinoic acid receptor-related orphan receptor gamma (RORγt) mRNA in comparison to forkhead box P3 (FoxP3) mRNA confirmed the development of pro-inflammatory Th17 response in PV. Further, the cytokine profile of pro-inflammatory and anti-inflammatory cytokines suggested defective suppressive functions in Treg cells with high inflammatory response. Our findings indicate that autoantigen Dsg-3 specifically allows the proliferation of IL-17 secreting T cells though has a negative effect on IL-10 secreting T cells leading to dysregulation of immunity in PV patients. This antagonistic relationship between Dsg-3-specific Th17 and Treg cells may be critical for the onset and persistence of inflammation in PV cases.
Collapse
Affiliation(s)
- Mohammad Ahmad Ansari
- Multidisciplinary Research Unit (Department of Health Research), University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Praveen Kumar Singh
- Department of Microbiology, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Sajad Ahmad Dar
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
| | - Gargi Rai
- Department of Microbiology, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Naseem Akhter
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha 65731, Saudi Arabia
| | - Deepika Pandhi
- Department of Dermatology & STD, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Vishal Gaurav
- Department of Dermatology & STD, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Sambit Nath Bhattacharya
- Department of Dermatology & STD, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Basu Dev Banerjee
- Department of Biochemistry, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shukla Das
- Department of Microbiology, University College of Medical Sciences (University of Delhi) & GTB Hospital, Delhi 110095, India.
| |
Collapse
|
9
|
Zhou H, Wang F, Wan J, Su S, Shi Y, Li X, Wu T, Liang J. Systematic Review and Meta-Analysis of Observational Studies on the Effectiveness and Safety of Ustekinumab among Patients with Inflammatory Bowel Disease in Eastern and Western Countries. J Clin Med 2023; 12:jcm12051894. [PMID: 36902681 PMCID: PMC10004158 DOI: 10.3390/jcm12051894] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Ustekinumab (UST) is an IL12/23 inhibitor utilized for altering inflammatory responses in inflammatory bowel disease (IBD). Clinical trials and case reports suggested that the effectiveness and safety of UST may differ among IBD patients in Eastern and Western countries. However, related data have not been systematically reviewed and analyzed. METHODS This systematic review and meta-analysis of the safety and effectiveness of UST in IBD included relevant literature from the Medline and Embase databases. The main outcomes were clinical response, clinical remission, endoscopic response, endoscopic remission, and adverse events in IBD. RESULTS We analyzed 49 real-world studies, most of which included patients with biological failure (89.1% CD and 97.1% UC). In UC patients, clinical remission rates were 34% at 12 weeks, 40% at 24 weeks, and 37% at 1 year. In CD patients, clinical remission rates were 46% at 12 weeks, 51% at 24 weeks, and 47% at 1 year. Clinical remission rates of CD patients were 40% at 12 weeks and 44% at 24 weeks in Western countries, versus 63% and 72% in Eastern countries, respectively. CONCLUSION UST is an effective drug for IBD with a promising safety profile. Although no RCTs have been performed in Eastern countries, the effectiveness of UST on CD patients is not inferior to that in Western countries based on the existing data.
Collapse
Affiliation(s)
- He Zhou
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Fang Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Jian Wan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Song Su
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanting Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Xiaofei Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
| | - Tong Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (T.W.); (J.L.)
| | - Jie Liang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (T.W.); (J.L.)
| |
Collapse
|
10
|
Hanif T, Ivaska LE, Ahmad F, Tan G, Mikola E, Puhakka T, Palomares O, Akdis CA, Toppila-Salmi S, Jartti T. Tonsillar transcriptional profiles in atopic and non-atopic subjects. Allergy 2023; 78:522-536. [PMID: 35899482 PMCID: PMC10087516 DOI: 10.1111/all.15458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/29/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Emerging research suggests that local lymphatic tissue such as tonsils have important role in regulating the immune responses. However, allergen sensitization-induced alterations in transcriptome of tonsils are not known. OBJECTIVES To examine the key differences in tonsillar gene expression between atopic and non-atopic subjects and further by type of sensitization. METHODS RNA-sequencing was performed on 52 tonsillar samples from atopic and non-atopic tonsillectomy patients. Sensitization to common food- and aero-allergen was defined by allergen specific IgE. Following groups were studied: (1) aero- and food-allergen sensitized (AS+FS) versus non-sensitized (NS), (2) aeroallergen-sensitized (AS) versus food-allergen sensitized (FS), (3) AS versus NS, (4) FS versus NS. Bioinformatics analysis was done using DESeq2(v3.10.2), WGCNA and GATK pipeline in R software (v3.3.1). Protein-protein interaction network was made from String database. RESULTS We studied 13 aeroallergen-sensitized, 6 food-allergen sensitized, 4 both food-and aero-allergen-sensitized and 29 non-sensitized tonsillectomy patients. Overall, 697 unique differentially expressed genes (DEGs) were detected in all sensitized subgroups including chemokines (CXCL2, CXCL8, CXCL10, CXCL11), IL-20RA, MUC1 and MUC20. When comparing different groups, the gene expression profiles overlapped except the AS versus FS group comparison, suggesting significantly different gene expression between the two sensitization subgroups. Furthermore, aeroallergen-sensitized subjects had more prominent immune responses compared with non-sensitized and food-allergen sensitized subjects including gene expression for IL-17 pathway and Toll-like receptor signalling pathway. CONCLUSION Allergic sensitization is associated with extensive tonsillar transcriptomic alterations and changes in immune related genes and pathways. Distinct differences were found between aero-allergen and food-allergen sensitization.
Collapse
Affiliation(s)
- Tanzeela Hanif
- Department of Pediatrics and Adolescent Medicine, University of Turku, Turku, Finland.,Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Lotta E Ivaska
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Freed Ahmad
- Department of Biology University of Turku, Turku, Finland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zürich/University of Zürich, Zurich, Switzerland.,Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Mikola
- Department of Otorhinolaryngology, Satakunta Central Hospital, Pori, Finland
| | - Tuomo Puhakka
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Oscar Palomares
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.,Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Sanna Toppila-Salmi
- Haartman Institute, University of Helsinki & Skin and Allergy Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine, University of Turku, Turku, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
11
|
Adnan M, Siddiqui AJ, Noumi E, Hannachi S, Ashraf SA, Awadelkareem AM, Snoussi M, Badraoui R, Bardakci F, Sachidanandan M, Patel M, Patel M. Integrating Network Pharmacology Approaches to Decipher the Multi-Target Pharmacological Mechanism of Microbial Biosurfactants as Novel Green Antimicrobials against Listeriosis. Antibiotics (Basel) 2022; 12:5. [PMID: 36671206 PMCID: PMC9854906 DOI: 10.3390/antibiotics12010005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Listeria monocytogenes (L. monocytogenes) is a serious food-borne pathogen that can cause listeriosis, an illness caused by eating food contaminated with this pathogen. Currently, the treatment or prevention of listeriosis is a global challenge due to the resistance of bacteria against multiple commonly used antibiotics, thus necessitating the development of novel green antimicrobials. Scientists are increasingly interested in microbial surfactants, commonly known as "biosurfactants", due to their antimicrobial properties and eco-friendly nature, which make them an ideal candidate to combat a variety of bacterial infections. Therefore, the present study was designed to use a network pharmacology approach to uncover the active biosurfactants and their potential targets, as well as the signaling pathway(s) involved in listeriosis treatment. In the framework of this study, 15 biosurfactants were screened out for subsequent studies. Among 546 putative targets of biosurfactants and 244 targets of disease, 37 targets were identified as potential targets for treatment of L. monocytogenes infection, and these 37 targets were significantly enriched in a Gene Ontology (GO) analysis, which aims to identify those biological processes, cellular locations, and molecular functions that are impacted in the condition studied. The obtained results revealed several important biological processes, such as positive regulation of MAP kinase activity, protein kinase B signaling, ERK1 and ERK2 cascade, ERBB signaling pathway, positive regulation of protein serine/threonine kinase activity, and regulation of caveolin-mediated endocytosis. Several important KEGG pathways, such as the ERBBB signaling pathway, TH17 cell differentiation, HIF-1 signaling pathway, Yersinia infection, Shigellosis, and C-type lectin receptor signaling pathways, were identified. The protein-protein interaction analysis yielded 10 core targets (IL2, MAPK1, EGFR, PTPRC, TNF, ITGB1, IL1B, ERBB2, SRC, and mTOR). Molecular docking was used in the latter part of the study to verify the effectiveness of the active biosurfactants against the potential targets. Lastly, we found that a few highly active biosurfactants, namely lichenysin, iturin, surfactin, rhamnolipid, subtilisin, and polymyxin, had high binding affinities towards IL2, MAPK1, EGFR, PTPRC, TNF, ITGB1, IL1B, ERBB2, SRC, and mTOR, which may act as potential therapeutic targets for listeriosis. Overall, based on the integrated network pharmacology and docking analysis, we found that biosurfactants possess promising anti-listeriosis properties and explored the pharmacological mechanisms behind their effect, laying the groundwork for further research and development.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Emira Noumi
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Sami Hannachi
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, La Rabta 1007, Tunis, Tunisia
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Manojkumar Sachidanandan
- Department of Oral Radiology, College of Dentistry, University of Hail, Hail P.O. Box 2440, Saudi Arabia
| | - Mirav Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| |
Collapse
|
12
|
Cheong PK, Ho TM, Chan KL, Lo CW, Leung SB, Hon KL, Leung KC, Siu THC, Song TH, Zhang H, Ching JYL, Chow TY, Sum CH, Chia CP, Lin ZX. The efficacy and safety of Yupingfeng Powder with variation in the treatment of allergic rhinitis: Study protocol for a randomized, double-blind, placebo-controlled trial. Front Pharmacol 2022; 13:1058176. [PMID: 36588672 PMCID: PMC9800420 DOI: 10.3389/fphar.2022.1058176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Allergic rhinitis (AR) is an upper airways chronic inflammatory disease mediated by IgE, which affects 10%-20% of the population. The mainstay for allergic rhinitis nowadays include steroids and antihistamines, but their effects are less than ideal. Many patients therefore seek Chinese medicine for treatment and Yupingfeng Powder is one of the most common formulae prescribed. In this study, we aim to investigate the efficacy and safety of Yupingfeng Powder with variation for the treatment of allergic rhinitis. Study design: This is a double-blind, randomized, placebo-controlled trial. A 2-week screening period will be implemented, and then eligible subjects with allergic rhinitis will receive interventions of either "Yupingfeng Powder with variation" granules or placebo granules for 8 weeks, followed by post treatment visits at weeks 12 and 16. The change in the Total Nasal Symptom Score (TNSS) will be used as the primary outcome. Discussion: This trail will evaluate the efficacy and safety of Yupingfeng Powder in treating allergic rhinitis. The study may provide the solid evidence of Yupingfeng Powder with variation can produce better clinical efficacy than the placebo granules. Trial registration: ClinicalTrials.gov, identifier NCT04976023.
Collapse
Affiliation(s)
- Pui Kuan Cheong
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,S. H. Ho Centre for Digestive Health, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tin Muk Ho
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Kam Leung Chan
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Cho Wing Lo
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,S. H. Ho Centre for Digestive Health, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Sin Bond Leung
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China,Department of Medicine and Geriatric, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China
| | - Kam Lun Hon
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Chun Leung
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tony Hon Chung Siu
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tian-He Song
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongwei Zhang
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jessica Yuet Ling Ching
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,S. H. Ho Centre for Digestive Health, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tak Yee Chow
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Him Sum
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chon Pin Chia
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhi-Xiu Lin
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, China,School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China,*Correspondence: Zhi-Xiu Lin,
| |
Collapse
|
13
|
Aguilar-Cazares D, Chavez-Dominguez R, Marroquin-Muciño M, Perez-Medina M, Benito-Lopez JJ, Camarena A, Rumbo-Nava U, Lopez-Gonzalez JS. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:929572. [PMID: 36072935 PMCID: PMC9441602 DOI: 10.3389/fendo.2022.929572] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment is a dynamic, complex, and redundant network of interactions between tumor, immune, and stromal cells. In this intricate environment, cells communicate through membrane-membrane, ligand-receptor, exosome, soluble factors, and transporter interactions that govern cell fate. These interactions activate the diverse and superfluous signaling pathways involved in tumor promotion and progression and induce subtle changes in the functional activity of infiltrating immune cells. The immune response participates as a selective pressure in tumor development. In the early stages of tumor development, the immune response exerts anti-tumor activity, whereas during the advanced stages, the tumor establishes mechanisms to evade the immune response, eliciting a chronic inflammation process that shows a pro-tumor effect. The deregulated inflammatory state, in addition to acting locally, also triggers systemic inflammation that has repercussions in various organs and tissues that are distant from the tumor site, causing the emergence of various symptoms designated as paraneoplastic syndromes, which compromise the response to treatment, quality of life, and survival of cancer patients. Considering the tumor-host relationship as an integral and dynamic biological system, the chronic inflammation generated by the tumor is a communication mechanism among tissues and organs that is primarily orchestrated through different signals, such as cytokines, chemokines, growth factors, and exosomes, to provide the tumor with energetic components that allow it to continue proliferating. In this review, we aim to provide a succinct overview of the involvement of cancer-related inflammation at the local and systemic level throughout tumor development and the emergence of some paraneoplastic syndromes and their main clinical manifestations. In addition, the involvement of these signals throughout tumor development will be discussed based on the physiological/biological activities of innate and adaptive immune cells. These cellular interactions require a metabolic reprogramming program for the full activation of the various cells; thus, these requirements and the by-products released into the microenvironment will be considered. In addition, the systemic impact of cancer-related proinflammatory cytokines on the liver-as a critical organ that produces the leading inflammatory markers described to date-will be summarized. Finally, the contribution of cancer-related inflammation to the development of two paraneoplastic syndromes, myelopoiesis and cachexia, will be discussed.
Collapse
Affiliation(s)
- Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Rodolfo Chavez-Dominguez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Marroquin-Muciño
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Human Leukocyte Antigen (HLA), Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Uriel Rumbo-Nava
- Clinica de Neumo-Oncologia, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| |
Collapse
|
14
|
Nguyen HH, Shinkuma S, Hayashi R, Katsumi T, Nishiguchi T, Natsuga K, Fujita Y, Abe R. New insight of itch mediators and proinflammatory cytokines in epidermolysis bullosa. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2022. [DOI: 10.1002/cia2.12230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Hong Ha Nguyen
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Satoru Shinkuma
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
- Department of Dermatology Nara Medical University Kashihara Japan
| | - Ryota Hayashi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Tatsuya Katsumi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Tomoki Nishiguchi
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| | - Ken Natsuga
- Department of Dermatology Faculty of Medicine and Graduate School of Medicine Hokkaido University Sapporo Japan
| | - Yasuyuki Fujita
- Department of Dermatology Faculty of Medicine and Graduate School of Medicine Hokkaido University Sapporo Japan
- Department of Dermatology Sapporo City General Hospital Sapporo Japan
| | - Riichiro Abe
- Division of Dermatology Niigata University Graduate School of Medical and Dental Science Niigata Japan
| |
Collapse
|
15
|
Soh H, Lee HJ, Han K, Park S, Hong SW, Moon JM, Kang EA, Lee J, Chun J, Im JP, Kim JS. Atopic Diseases Are Associated With Development of Inflammatory Bowel Diseases in Korea: A Nationwide Population-based Study. Clin Gastroenterol Hepatol 2021; 19:2072-2081.e6. [PMID: 32712393 DOI: 10.1016/j.cgh.2020.07.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The association between atopic diseases and inflammatory bowel diseases (IBD) is unclear. We conducted a nationwide population-based study in Korea to investigate the effect of atopic diseases on the development of IBD. METHODS A total of 9,923,521 participants, who received a medical check-up in 2009, were included and followed through 2017. The presence of any atopic diseases, including atopic dermatitis (AD), allergic rhinitis (AR), and asthma, was evaluated. Patients who developed IBD, including Crohn's disease (CD) and ulcerative colitis (UC), were identified using claims data from National Health Insurance; the association between atopic diseases and the risk of IBD was evaluated using Cox proportional hazard models, and presented as adjusted hazard ratios (aHRs) with 95% CIs. RESULTS During a mean follow-up period of 7.3 years, 1419 patients (0.014%) developed CD and 5897 patients (0.059%) developed UC. The incidences of CD (per 100,000 person-years) were 3.756, 2.248, and 2.346 in patients with AD, AR, or asthma, respectively. The incidences of UC were 11.952, 9.818, and 9.358 in patients with AD, AR, or asthma, respectively. Multivariable analysis revealed that the aHRs for incident CD in patients with AD, AR, or asthma were 2.02, 1.33, and 1.60 (95% CIs, 1.118-3.663, 1.149-1.529, and 1.193-2.136, respectively) compared with controls. The risks of incident UC in patients with AD, AR, or asthma were 1.51, 1.32, and 1.29 (95% CIs, 1.082-2.104, 1.229-1.410, and 1.115-1.491, respectively) compared with controls. Moreover, an increase in the number of atopic diseases gradually increased the risk for CD and UC; for 1 or 2 or more atopic diseases, the aHRs for CD were 1.35 and 1.65 (95% CIs, 1.171-1.560 and 1.146-2.376), and the aHRs for UC were 1.30 and 1.49 (95% CIs, 1.211-1.392 and 1.249-1.774), respectively. CONCLUSIONS Based on a nationwide population-based study in Korea, patients with any atopic disease, including AD, AR, or asthma, have an increased risk for CD and UC. The risk for IBD increases with the increase in the number of atopic diseases.
Collapse
Affiliation(s)
- Hosim Soh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Seona Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Min Moon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Ae Kang
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jooyoung Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Republic of Korea
| | - Jaeyoung Chun
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Naringin and Hesperidin Counteract Diclofenac-Induced Hepatotoxicity in Male Wistar Rats via Their Antioxidant, Anti-Inflammatory, and Antiapoptotic Activities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9990091. [PMID: 34422219 PMCID: PMC8376442 DOI: 10.1155/2021/9990091] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/27/2021] [Accepted: 07/14/2021] [Indexed: 01/18/2023]
Abstract
This study is aimed at evaluating the preventive effect and at suggesting the mode of actions of naringin and hesperidin and their combination in diclofenac-induced hepatotoxicity. Male Wistar rats, intraperitoneally injected with diclofenac sodium (3 mg/kg b.wt/day), were orally treated with naringin (20 mg/kg b.wt/day) and hesperidin (20 mg/kg b.wt/day) and their combination for 4 weeks. The administrations of naringin and hesperidin to diclofenac-injected rats led to a significant decrease in the elevated serum ALT, AST, LDH, ALP, GGT, total bilirubin, TNF-α, and IL-17 levels as well as liver lipid peroxidation and liver p53 and caspase-3 mRNA expressions. In contrast, serum IL-4 level, liver GSH content, and liver GPx and SOD activities increased. In association, diclofenac-induced deleterious histological alterations including hydropic degeneration, cytoplasmic vacuolization, apoptosis, and focal hepatic necrosis of hepatocytes associated with inflammatory cells' infiltration were remarkably improved by treatments with naringin and hesperidin. In conclusion, naringin, hesperidin, and their combination, which was the most potent, counteract diclofenac-induced liver injury via antioxidant, anti-inflammatory, and antiapoptotic actions. Thus, this study recommends the use of naringin and hesperidin or their combination to resolve the side effects of drugs like diclofenac on the liver.
Collapse
|
17
|
Hassan RA, Hozayen WG, Abo Sree HT, Al-Muzafar HM, Amin KA, Ahmed OM. Naringin and Hesperidin Counteract Diclofenac-Induced Hepatotoxicity in Male Wistar Rats via Their Antioxidant, Anti-Inflammatory, and Antiapoptotic Activities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1-14. [DOI: https:/doi.org/10.1155/2021/9990091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
This study is aimed at evaluating the preventive effect and at suggesting the mode of actions of naringin and hesperidin and their combination in diclofenac-induced hepatotoxicity. Male Wistar rats, intraperitoneally injected with diclofenac sodium (3 mg/kg b.wt/day), were orally treated with naringin (20 mg/kg b.wt/day) and hesperidin (20 mg/kg b.wt/day) and their combination for 4 weeks. The administrations of naringin and hesperidin to diclofenac-injected rats led to a significant decrease in the elevated serum ALT, AST, LDH, ALP, GGT, total bilirubin, TNF-α, and IL-17 levels as well as liver lipid peroxidation and liver p53 and caspase-3 mRNA expressions. In contrast, serum IL-4 level, liver GSH content, and liver GPx and SOD activities increased. In association, diclofenac-induced deleterious histological alterations including hydropic degeneration, cytoplasmic vacuolization, apoptosis, and focal hepatic necrosis of hepatocytes associated with inflammatory cells’ infiltration were remarkably improved by treatments with naringin and hesperidin. In conclusion, naringin, hesperidin, and their combination, which was the most potent, counteract diclofenac-induced liver injury via antioxidant, anti-inflammatory, and antiapoptotic actions. Thus, this study recommends the use of naringin and hesperidin or their combination to resolve the side effects of drugs like diclofenac on the liver.
Collapse
Affiliation(s)
- Rasha A. Hassan
- Department of Biochemistry, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Walaa G. Hozayen
- Department of Biochemistry, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Haidy T. Abo Sree
- Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef, Egypt
| | - Hessah M. Al-Muzafar
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Kamal A. Amin
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
- Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Osama M. Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
18
|
Dejima H, Hu X, Chen R, Zhang J, Fujimoto J, Parra ER, Haymaker C, Hubert SM, Duose D, Solis LM, Su D, Fukuoka J, Tabata K, Pham HHN, Mcgranahan N, Zhang B, Ye J, Ying L, Little L, Gumbs C, Chow CW, Estecio MR, Godoy MCB, Antonoff MB, Sepesi B, Pass HI, Behrens C, Zhang J, Vaporciyan AA, Heymach JV, Scheet P, Lee JJ, Wu J, Futreal PA, Reuben A, Kadara H, Wistuba II, Zhang J. Immune evolution from preneoplasia to invasive lung adenocarcinomas and underlying molecular features. Nat Commun 2021; 12:2722. [PMID: 33976164 PMCID: PMC8113327 DOI: 10.1038/s41467-021-22890-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanism by which anti-cancer immunity shapes early carcinogenesis of lung adenocarcinoma (ADC) is unknown. In this study, we characterize the immune contexture of invasive lung ADC and its precursors by transcriptomic immune profiling, T cell receptor (TCR) sequencing and multiplex immunofluorescence (mIF). Our results demonstrate that anti-tumor immunity evolved as a continuum from lung preneoplasia, to preinvasive ADC, minimally-invasive ADC and frankly invasive lung ADC with a gradually less effective and more intensively regulated immune response including down-regulation of immune-activation pathways, up-regulation of immunosuppressive pathways, lower infiltration of cytotoxic T cells (CTLs) and anti-tumor helper T cells (Th), higher infiltration of regulatory T cells (Tregs), decreased T cell clonality, and lower frequencies of top T cell clones in later-stages. Driver mutations, chromosomal copy number aberrations (CNAs) and aberrant DNA methylation may collectively impinge host immune responses and facilitate immune evasion, promoting the outgrowth of fit subclones in preneoplasia into dominant clones in invasive ADC.
Collapse
Affiliation(s)
- Hitoshi Dejima
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Hu
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Runzhe Chen
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiexin Zhang
- Department of Bioinformatics & Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cara Haymaker
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shawna M Hubert
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dzifa Duose
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M Solis
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Su
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Junya Fukuoka
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kazuhiro Tabata
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hoa H N Pham
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Nicholas Mcgranahan
- Cancer Research United Kingdom-University College London Lung Cancer Centre of Excellence, London, UK
| | - Baili Zhang
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Ye
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisha Ying
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Cancer Research Institute, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Latasha Little
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Curtis Gumbs
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chi-Wan Chow
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcos Roberto Estecio
- Department of Epigenetics and Molecular Carcinogenesis, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center of Cancer Epigenetics, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Scheet
- Department of Epidemiology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Humam Kadara
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jianjun Zhang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
19
|
Studies of molecular pathways associated with blood neutrophil corticosteroid insensitivity in equine asthma. Vet Immunol Immunopathol 2021; 237:110265. [PMID: 33989854 DOI: 10.1016/j.vetimm.2021.110265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 12/25/2022]
Abstract
Severe equine asthma is characterized by airway hyperresponsiveness, neutrophilic inflammation and structural alterations of the lower airways. In asthmatic horses with neutrophilic inflammation, there is insensitivity to corticosteroids characterized by the persistence of neutrophils within the airways with therapy. We hypothesized that hypoxia or oxidative stress in the microenvironment of the lung contributes to this insensitivity of neutrophils to corticosteroids in asthmatic horses. Blood neutrophils isolated from horses with severe asthma (N = 8) and from healthy controls (N = 8) were incubated under different cell culture conditions simulating hypoxia and oxidative stress and, in the presence, or absence of dexamethasone. The pro-inflammatory gene and protein expression of neutrophils were studied. In both groups, pyocyanin-induced oxidative stress increased the mRNA expression of IL-8, IL-1β, and TNF-α. While IL-1β and TNF-α were downregulated by dexamethasone under these conditions, IL-8 was not. Simulated hypoxic conditions did not enhance pro-inflammatory gene expression in neutrophils from either group of horses. In conclusion, oxidative stress but not hypoxia may contribute to corticosteroid insensitivity via a selective gene regulation pathway. Equine neutrophil responses were similar in both heathy and asthmatic horses, indicating that it is not specific to asthmatic inflammation.
Collapse
|
20
|
Li W, Shi W, Yin Y, Chen J, Luo L. <p>Association of <em>IL-17</em> and <em>IL-23</em> Gene Variants with Plasma Levels and Risk of Vulvovaginal Candidiasis in a Chinese Han Population</p>. Pharmgenomics Pers Med 2020; 13:725-733. [PMID: 33364812 PMCID: PMC7751320 DOI: 10.2147/pgpm.s275073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/16/2020] [Indexed: 12/02/2022] Open
Abstract
Background Vulvovaginal candidiasis (VVC) is a common vaginal inflammatory disease in females. The interleukin (IL)-23/IL-17 axis was involved in vaginal inflammation. Nevertheless, the relationship between gene polymorphisms in the IL-23/IL-17 axis and VVC risk is still unexplored. Methods We enrolled 217 VCC cases and 326 controls in this study. The genotyping of all polymorphisms was implemented by PCR-RFLP methods. Results Data indicated that IL-17F gene rs763780, IL-17A gene rs2275913, and IL-23R rs11209026 polymorphisms were linked with an elevated risk of VVC in Chinese ethnicity. Subgroup analyses uncovered that IL-23R rs11209026, IL-17A rs10484879 and IL-17F rs763780 polymorphisms increased the risk of VVC among smokers or individuals with BMI ≥25 kg/m2. Additionally, IL-17F rs763780 polymorphism was shown to increase the risk of recurrent VVC (RVVC). Furthermore, IL-23 and IL-17 serum levels were higher among VVC cases than controls. We also observed that IL-23 and IL-17 gene polymorphisms were related to their serum levels. Receiver operating characteristics (ROC) curve analysis found that IL-17 and IL-23 serum levels were associated with the relapse of VVC. Conclusion In conclusion, this study indicates that polymorphisms in the IL-23/IL-17 axis increase the risk of VVC.
Collapse
Affiliation(s)
- Wei Li
- Department of Gynecology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Wenyin Shi
- Department of Gynecology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Yujun Yin
- Department of Gynaecology and Obstetrics, Dantu District People’s Hospital, Zhenjiang, Jiangsu, People’s Republic of China
| | - Juan Chen
- Department of Gynecology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Lanlan Luo
- Department of Obstetrics and Gynecology, Taizhou People’s Hospital, Taizhou, People’s Republic of China
- Correspondence: Lanlan Luo Tel/Fax +86-523-86606267 Email
| |
Collapse
|
21
|
Yu X, Wang M, Cao Z. Reduced CD4 +T Cell CXCR3 Expression in Patients With Allergic Rhinitis. Front Immunol 2020; 11:581180. [PMID: 33224143 PMCID: PMC7669911 DOI: 10.3389/fimmu.2020.581180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/12/2020] [Indexed: 01/23/2023] Open
Abstract
While T cells are considered to play a primary role in IgE-mediated atopic diseases, little is known about the systemic variations of T cell subsets from patients with allergic rhinitis (AR). To elucidate the characteristics of peripheral T cells, we analyzed natural killer, B cell, and T cell populations, performed T cell subset construction, and assessed chemokine receptor and associated serum cytokine expression in 25 AR patients and 20 healthy controls. Our results revealed increased levels of CD4+T cells, serum interleukin (IL)-10, IL-6, and interferon (IFN)-γ, and reduced Th1 and Th17 subsets, identified by their chemokine receptors, in AR patients. These results suggest a systemic activation of T cell responses in AR. We further demonstrated that AR patients exhibit significantly reduced CD4+T cell CXCR3 expression, especially in patients with moderate-severe disease severity, demonstrating that CXCR3 is a potential key molecule that hinders the Th1/Th2 balance in AR pathology. Overall, systemic T cell activation occurred in AR patients and CXCR3 dramatically decreased in CD4+T cells, which may ultimately be used as a potential disease and/or therapeutic target.
Collapse
Affiliation(s)
- Xiaofeng Yu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Meng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiwei Cao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Li J, Lin X, Liu X, Ma Z, Li Y. Baicalin regulates Treg/Th17 cell imbalance by inhibiting autophagy in allergic rhinitis. Mol Immunol 2020; 125:162-171. [PMID: 32688118 DOI: 10.1016/j.molimm.2020.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/12/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Baicalin has many biological properties such as anti-oxidation and anti-allergy. The current study aimed to explore the effect of Baicalin on allergic rhinitis (AR) and its potential mechanism of action. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation. Expression levels of Th17 and Treg cells-related proteins in nasal mucosa and peripheral blood cells were detected by real-time quantitative PCR, flow cytometry and Western blot. The mice were randomly divided into Control, ovalbumin (OVA), l-Baicalin, H-Baicalin, DSGC, 3-MA, and H-Baicalin + Rapa groups. Changes of allergic rhinitis conditions and eosinophil infiltration of the mice were detected and scored by Diff-Quik staining, and histological changes were observed by Hematoxylin & Eosin (H&E) staining and Periodate Schiff (PAS) staining. Serological changes, expression levels of interleukin-17A (IL-17A), interleukin-10 (IL-10), eosinophilic cationic protein (ECP) and anti-OVA-specific antibodies were detected by Enzyme-linked immunosorbent assay (ELISA). RESULTS Clinical case analysis found that AR patients had a Th17/Treg imbalance and activated autophagy, however, Baicalin restored Th17/Treg cell balance and inhibited autophagy in vitro. in vivo experiments demonstrated that Baicalin inhibited OVA-induced allergic nasal symptoms and the activation of autophagy pathway, which was the same as the regulation of 3-MA, while Rapa could weaken the effects of H-baicalin. Moreover, Baicalin reduced the infiltration of different inflammatory cells of the nasal lavage fluid, prevented the damages to epithelial cells, and improved nasal mucosal thickness and mucus secretion. In addition, Baicalin regulated the balance of mouse anti-OVA-specific antibody levels and expressions of Th17/Treg-associated cytokines. CONCLUSION Our study revealed that Baicalin can be used to treat AR, and the effect is realized through inhibiting autophagy to regulate Th17/Treg cell differentiation.
Collapse
Affiliation(s)
- Jing Li
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Shangcheng District, Hangzhou, Zhejiang Province, 310006, China
| | - Xiaoyan Lin
- The Affiliated Hangzhou Hospital of Nanjing Medical University, No.261, Huansha Road, Shangcheng District, Hangzhou, Zhejiang Province, 310006, China
| | - Xiang Liu
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Shangcheng District, Hangzhou, Zhejiang Province, 310006, China
| | - Zhiqi Ma
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Shangcheng District, Hangzhou, Zhejiang Province, 310006, China
| | - Yong Li
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No. 261, Huansha Road, Shangcheng District, Hangzhou, Zhejiang Province, 310006, China.
| |
Collapse
|
23
|
Moh’d Al-Rawashdeh B, Sada Alhanjori A, Ali E, Zihlif M. Association of IL-4 Polymorphisms with Allergic Rhinitis in Jordanian Population. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E179. [PMID: 32295284 PMCID: PMC7230575 DOI: 10.3390/medicina56040179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Background and objectives: Allergic rhinitis has complex patterns of inheritance, and single nucleotide polymorphisms, a common genetic variation in a population, exert a significant role in allergic rhinitis pathology. The current study aimed to investigate the association of Interleukin-4 (IL-4) polymorphisms with allergic rhinitis. Materials and Methods: Our study included 158 patients with allergic rhinitis and 140 healthy controls from Jordan that were genotyped for IL-4 single nucleotide polymorphisms (SNPs) C-589T (rs2243250) and T-2979G (rs2227284) using restriction fragment length polymorphism-polymerase chain reaction. Statistical analysis was conducted using IBM SPSS Statistics version 24 software. Results: The results showed that the allelic frequency of the minor alleles was 0.19 and 0.67 for C-589T (rs2243250) and T-2979G (rs2227284) in the allergic rhinitis patients, respectively, while it was 0.18 for C-589T (rs2243250) and 0.64 T-2979G (rs2227284) in the control group. The homozygous (TT) genotype of C-589T (rs2243250) was significantly associated with allergic rhinitis (p < 0.05), while there was no association of any of T-2979G (rs2227284) genotypes with allergic rhinitis. Conclusions: The results of this study indicate that genetic inter-population variation precipitates the differences in the percentages of many diseases among populations, including allergic rhinitis.
Collapse
Affiliation(s)
- Baeth Moh’d Al-Rawashdeh
- Department of Special Surgery, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (B.M.A.-R.); (E.A.)
| | - Ahmed Sada Alhanjori
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Elnagi Ali
- Department of Special Surgery, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; (B.M.A.-R.); (E.A.)
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan;
| |
Collapse
|
24
|
Looman KIM, Meel ER, Grosserichter‐Wagener C, Vissers FJM, Klingenberg JH, Jong NW, Jongste JC, Pasmans SGMA, Duijts L, Zelm MC, Moll HA. Associations of Th2, Th17, Treg cells, and IgA + memory B cells with atopic disease in children: The Generation R Study. Allergy 2020; 75:178-187. [PMID: 31385614 DOI: 10.1111/all.14010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/10/2019] [Accepted: 06/18/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND New insights into immune cells could contribute to treatment and monitoring of atopic disease. Because nongenetic factors shape the human immune system, we here studied these immune cells in a large cohort with atopic children with adjustment for prenatal and postnatal confounders. METHODS Information on atopic dermatitis, inhalant- and food-allergic sensitization, asthma lung function scores was obtained from 855 10-year-old children within the Generation R cohort. 11-color flow cytometry was performed to determine CD27+ and CD27- IgG+ , IgE+ and IgA+ memory B cells, Th1, Th2, Th17, and Treg-memory cells from venous blood. Associations between any atopic disease, the individual atopic diseases, and immune cell numbers were determined. RESULTS Children with any atopic disease had higher Th2, Treg, Treg-memory, and CD27+ IgA+ memory B-cell numbers compared to children without atopic disease. When studying the individual diseases compared to children without the individual diseases, children with atopic dermatitis, inhalant-, and food-allergic sensitization had higher memory Treg cell numbers 12.3% (95% CI 4.2; 21.0), (11.1% (95% CI 3.0; 19.8), (23.7% (95% CI 7.9; 41.8), respectively. Children with food-allergic sensitization had higher total B and CD27+ IgA+ memory B-cell numbers (15.2% [95% CI 3.2; 28.7], 22.5% [95% CI 3.9; 44.3], respectively). No associations were observed between asthma and B- or T-cell numbers. CONCLUSION Children with any atopic disease and children with inhalant- and food-allergic sensitization or atopic dermatitis had higher circulating memory Treg cells, but not higher IgE+ B-cell numbers. The associations of higher Treg and CD27+ IgA+ B-cell numbers in children with food-allergic sensitization are suggestive of TGF-β-mediated compensation for chronic inflammation.
Collapse
Affiliation(s)
- Kirsten I. M. Looman
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Evelien R. Meel
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | | | - Floor J. M. Vissers
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Janice H. Klingenberg
- Generation R Study Group Erasmus MC, University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Nicolette W. Jong
- Department of Internal Medicine, Division of Allergology Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Johan C. Jongste
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | | | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Pediatrics, Division of Neonatology Erasmus MC, University Medical Center Rotterdam Rotterdam the Netherlands
| | - Menno C. Zelm
- Department of Immunology and Pathology, Central Clinical School Monash University and Alfred Hospital Melbourne Victoria Australia
| | - Henriëtte A. Moll
- Department of Pediatrics Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam the Netherlands
| |
Collapse
|
25
|
Zhang L, Gao L, Xu C, Li X, Wang P, Zhang C, Zhao C. Porphyromonas gingivalis lipopolysaccharide promotes T- helper 17 cell differentiation from human CD4+ naïve T cells via toll-like receptor-2 in vitro. Arch Oral Biol 2019; 107:104483. [DOI: 10.1016/j.archoralbio.2019.104483] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/28/2019] [Accepted: 07/14/2019] [Indexed: 12/20/2022]
|
26
|
Huang Q, Han L, Lv R, Ling L. Magnolol exerts anti-asthmatic effects by regulating Janus kinase-signal transduction and activation of transcription and Notch signaling pathways and modulating Th1/Th2/Th17 cytokines in ovalbumin-sensitized asthmatic mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:251-261. [PMID: 31297009 PMCID: PMC6609269 DOI: 10.4196/kjpp.2019.23.4.251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/15/2022]
Abstract
Allergic asthma, is a common chronic inflammatory disease of the airway presenting with airway hyperresponsiveness and airway remodelling. T helper cells-derived cytokines are critically associated with asthma pathogenesis. Janus kinase-signal transduction and activation of transcription (JAK/STAT) signaling is found to be involved in asthma. Magnolol is a plant-derived bioactive compound with several pharmacological effects. The study aimed to assess the effects of magnolol in ovalbumin (OVA)-induced asthmatic model. BALB/c mice were sensitized and challenged with OVA. Magnolol (12.5, 25, or 50 mg/kg body weight) was administered to separate groups of animals. Dexamethasone was used as the positive control. Cellular infiltration into the bronchoalveolar lavage fluid (BALF) were reduced on magnolol treatment. The levels of Th2 and Th17 cytokines were reduced with noticeably raised levels of interferon gamma. Lung function was improved effectively along with restoration of bronchial tissue architecture. OVA-specific immunoglobulin E levels in serum and BALF were decreased by magnolol. Magnolol reduced Th17 cell population and effectively modulated the JAK-STAT and Notch 1 signaling. The results suggest the promising use of magnolol in therapy for allergic asthma.
Collapse
Affiliation(s)
- Qi Huang
- Department of Gerontology, Wujiang Hospital Affiliated to Nantong University, Suzhou, Jiangsu 215505, China
| | - Lele Han
- Department of Gerontology, Wujiang Hospital Affiliated to Nantong University, Suzhou, Jiangsu 215505, China
| | - Rong Lv
- Department of Gerontology, Wujiang Hospital Affiliated to Nantong University, Suzhou, Jiangsu 215505, China
| | - Ling Ling
- Department of Gerontology, Wujiang Hospital Affiliated to Nantong University, Suzhou, Jiangsu 215505, China
| |
Collapse
|
27
|
Ren Q, Li X, Li Q, Yang H, Wang H, Zhang H, Zhao L, Jiang‐yong S, Meng X, Zhang Y, Shen X. Total flavonoids from sea buckthorn ameliorates lipopolysaccharide/cigarette smoke‐induced airway inflammation. Phytother Res 2019; 33:2102-2117. [PMID: 31209984 DOI: 10.1002/ptr.6404] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/01/2019] [Accepted: 05/18/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Qing‐cuo Ren
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University HospitalSichuan University Chengdu China
| | - Xuan‐hao Li
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Qiu‐yue Li
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Hai‐ling Yang
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Hong‐ling Wang
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Hai Zhang
- College of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University HospitalSichuan University Chengdu China
| | - Si‐lang Jiang‐yong
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Xian‐li Meng
- College of PharmacyChengdu University of Traditional Chinese Medicine Chengdu China
| | - Yi Zhang
- College of Ethnic MedicineChengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiao‐fei Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University HospitalSichuan University Chengdu China
| |
Collapse
|
28
|
Kuroda R, Higuchi H, Yoshida K, Yonejima Y, Hisa K, Utsuyama M, Osawa K, Hirokawa K. Effects of chocolate containing Leuconostoc mesenteroides strain NTM048 on immune function: a randomized, double-blind, placebo-controlled trial. IMMUNITY & AGEING 2018; 15:29. [PMID: 30479641 PMCID: PMC6247524 DOI: 10.1186/s12979-018-0139-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/08/2018] [Indexed: 11/21/2022]
Abstract
Background Previous reports showed that oral administration of Leuconostoc mesenteroides strain NTM048 increases IgA levels and CD4+ T cell population in feces and mice, respectively, as revealed by flow cytometric analysis of splenocytes. This study aimed to evaluate the effect of chocolate supplemented with L. mesenteroides strain NTM048 (> 1.00 × 109 CFU/day, NTM048) on the immune parameters of healthy subjects, using a randomized, placebo-controlled, double-blinded study design. Methods Participants (mean age: 46.3 years) ingested 28 g of test food daily, at a time of their own choice, for 4 weeks. The immunological parameters of all participants were evaluated two times (pre- and post- ingestion). At the end of the study, various immunological parameters of the participants were measured and scoring of immunological vigor (SIV) was performed using a comprehensive algorithm. Results Ingestion of NTM048-supplemented chocolate significantly improved SIV in the NTM048 group (18.6 ± 1.6) compared to that in the placebo group (17.8 ± 2.0) after 4 weeks (p = 0.049). Several immunological parameters (CD8+T cells, CD8+CD28+ T cells, and memory T cells) were significantly elevated in the NTM048 group as compared to the placebo group (all p < 0.05). In addition, T cell proliferation index at post-ingestion significantly increased compared with that at pre-ingestion in the NTM048 (p = 0.017) and placebo groups (p = 0.037), although no differences were observed between the two groups. Conclusion Our results suggest that ingestion of chocolate supplemented with NTM048 is effective against the age-related decline in T cell-related immune functions. Trial registration UMIN Clinical Trials Registry UMIN000021989. Registered 19 April 2016, https://upload.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000025321
Collapse
Affiliation(s)
- Reiko Kuroda
- Central Laboratory, LOTTE Co., Ltd., 1-1, Numakage 3-chome, Minami-ku, Saitama, 336-8601 Japan
| | - Hiroaki Higuchi
- Central Laboratory, LOTTE Co., Ltd., 1-1, Numakage 3-chome, Minami-ku, Saitama, 336-8601 Japan
| | - Keishirou Yoshida
- Central Laboratory, LOTTE Co., Ltd., 1-1, Numakage 3-chome, Minami-ku, Saitama, 336-8601 Japan
| | - Yasunori Yonejima
- Nitto Pharmaceutical Industries, Ltd., 35-3, Minamibiraki, Kamiueno-cho, Muko, Kyoto, 617-0006 Japan
| | - Keiko Hisa
- Nitto Pharmaceutical Industries, Ltd., 35-3, Minamibiraki, Kamiueno-cho, Muko, Kyoto, 617-0006 Japan
| | - Masanori Utsuyama
- 3Institute for Health and Life Science, Tokyo Medical & Dental University, 3-10, Kandasurugadai 10-chome, Chiyoda-ku, Tokyo, 101-0062 Japan.,4Department of Comprehensive Pathology, Tokyo Medical & Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Kenji Osawa
- Central Laboratory, LOTTE Co., Ltd., 1-1, Numakage 3-chome, Minami-ku, Saitama, 336-8601 Japan
| | - Katsuiku Hirokawa
- 3Institute for Health and Life Science, Tokyo Medical & Dental University, 3-10, Kandasurugadai 10-chome, Chiyoda-ku, Tokyo, 101-0062 Japan.,4Department of Comprehensive Pathology, Tokyo Medical & Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
29
|
Lyons DO, Plewes MR, Pullen NA. Soluble transforming growth factor beta-1 enhances murine mast cell release of Interleukin 6 in IgE-independent and Interleukin 13 in IgE-dependent settings in vitro. PLoS One 2018; 13:e0207704. [PMID: 30444930 PMCID: PMC6239331 DOI: 10.1371/journal.pone.0207704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/03/2018] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION For immune cells transforming growth factor beta-1 (TGF-β1) can enhance or repress effector functions. Here, we characterize the effects of TGF-β1 on IgE-mediated and IL-33-mediated activation of primary murine mast cells derived from hematopoietic stem cells (bone marrow derived mast cells; BMMC). We also investigated potential interactions between TGF-β1 and stem cell factor (SCF). We conclude TGF-β1 plays a selectively stimulatory role for mast cell cultures in vitro. METHODS BMMCs from C57BL/6 mice were differentiated with IL-3 and then treated with TGF-β1. BMMCs were exposed to TGF-β1, primed with IgE, activated with antigen, and then IL-6 and IL-13 cytokine release was quantified using ELISA. Additionally, the effects of TGF-β1 on both IgE and IL-33-mediated short term activation were observed via flow cytometric analysis of both surface LAMP-1 expression and intracellular IL-6. Receptor colocalization was visualized using fluorescence confocal microscopy and individual receptor expression levels were also quantified. RESULTS Resting IL-6 production increased with TGF-β1 but significance was lost following BMMC activation via IgE receptor (FcεRI) crosslinking. This was similar to a comparison effect due to SCF treatment alone, which also enhanced resting levels of IL-6. TGF-β1 treatment enhanced release of IL-13 only with FcεRI-IgE-mediated activation. TGF-β1 suppressed mobilization of IL-6 with short-term BMMC activation when stimulated with IL-33. Lastly, colocalization patterns of the SCF receptor (CD117) and FcεRI with IgE crosslinking were unaffected by TGF-β1 treatment, but individual expression levels for FcεRI, CD117, and TGFβRII were all reduced following either IgE activation or TGF-β1 treatment; this reduction was partially recovered in BMMCs that were both activated by IgE and treated with TGF-β1. DISCUSSION These data reveal a novel positive effect of soluble TGF-β1 on mast cell activation in vitro, suggesting mast cells may be activated through a non-canonical pathway by TGF-β1. Understanding this interaction will provide insight into the potential role of mast cells in settings where TGF-β1 is produced in an aberrant manner, such as in and around high grade tumors.
Collapse
Affiliation(s)
- David O. Lyons
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| | - Michele R. Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Nicholas A. Pullen
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States of America
| |
Collapse
|
30
|
Zhai C, Li S, Feng W, Shi W, Wang J, Wang Q, Chai L, Zhang Q, Yan X, Li M. Association of interleukin-17a rs2275913 gene polymorphism and asthma risk: a meta-analysis. Arch Med Sci 2018; 14:1204-1211. [PMID: 30393475 PMCID: PMC6209699 DOI: 10.5114/aoms.2018.73345] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 11/14/2016] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Interleukin-17A (IL-17A), a pro-inflammatory cytokine, plays an important role in the pathogenesis of asthma. A number of studies have investigated the relationship between IL-17A rs2275913 polymorphism and risk of asthma. However, the results obtained are inconclusive. The aim of this meta-analysis is to clarify the relationship between IL-17A rs2275913 polymorphism and asthma risk. MATERIAL AND METHODS Searches were conducted in PubMed, Web of Science, Elsevier, Google Scholar, Wanfang and Chinese National Knowledge Infrastructure (CNKI) databases, and data were extracted from eligible studies by two independent reviewers. The pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were calculated. Publication bias, heterogeneity and sensitivity analysis were also assessed. RESULTS Ten studies with a total of 5016 subjects were included. Overall, the results indicated a significant association between the IL-17A rs2275913 polymorphism and the risk of asthma (G vs. A: OR = 0.866, 95% CI: 0.789-0.951, p = 0.003; GG+GA vs. AA: OR = 0.752, 95% CI: 0.633-0.895, p = 0.001). In subgroup analysis by age and ethnicity, the G allele of rs2275913 in IL-17A was significantly associated with a reduced risk of asthma in children and Asians. CONCLUSIONS The results of this meta-analysis indicate that the G allele of rs2275913 in IL-17A is a protective factor for the development of asthma.
Collapse
Affiliation(s)
- Cui Zhai
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
31
|
Liu LL, Zhang Y, Zhang XF, Li FH. Influence of rutin on the effects of neonatal cigarette smoke exposure-induced exacerbated MMP-9 expression, Th17 cytokines and NF-κB/iNOS-mediated inflammatory responses in asthmatic mice model. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:481-491. [PMID: 30181695 PMCID: PMC6115346 DOI: 10.4196/kjpp.2018.22.5.481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/30/2017] [Accepted: 06/01/2017] [Indexed: 11/15/2022]
Abstract
Allergic asthma is one of the most enduring diseases of the airway. The T-helper cells and regulatory T-cells are critically involved in inflammatory responses, mucus hypersecretion, airway remodelling and in airway hyper-responsiveness. Cigarette smoke (CS) has been found to aggravate inflammatory responses in asthma. Though currently employed drugs are effective, associated side effects demand identification and development of novel drugs with negligible or no adverse effects. Rutin, plant-derived flavonoid has been found to possess antioxidant and anti-inflammatory effects. We investigated the ability of rutin to modulate T-cells and inhibit inflammation in experimentally-induced asthma in cigarette smoke exposed mice. Separate groups of neonatal mice were exposed to CS for 10 days from post-natal days 2 to 11. After 2 weeks, the mice were sensitized and challenged with ovalbumin (OVA). Treatment group were given rutin (37.5 or 75 mg/kg body weight) during OVA sensitization and challenge. Rutin treatment was found to significantly inhibit cellular infiltration in the airways and Th2 and Th17 cytokine levels as well. Flow cytometry revealed effectively raised CD4+CD25+Fox3+ Treg cells and supressed Th17 cell population on rutin treatment. Airway hyper-responsiveness observed following CS and OVA challenge were inhibited by rutin. NF-κB and iNOS, chief regulators of inflammatory responses robustly activated by CS and OVA were down-regulated by rutin. Rutin also inhibited the expression of matrix metalloproteinase 9, thereby aiding in prevention of airway remodelling in asthma thereby revealing to be a potent candidate in asthma therapy.
Collapse
Affiliation(s)
- Li-Li Liu
- Children's Medical Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R.China
| | - Yan Zhang
- Children's Medical Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R.China
| | - Xiao-Fang Zhang
- Department of Pathology, Shandong University of Medicine, Jinan, Shandong 250012, P.R.China
| | - Fu-Hai Li
- Children's Medical Center, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R.China
| |
Collapse
|
32
|
Shen Y, Zhang ZH, Hu D, Ke X, Gu Z, Zou QY, Hu GH, Song SH, Kang HY, Hong SL. The airway inflammation induced by nasal inoculation of PM2.5 and the treatment of bacterial lysates in rats. Sci Rep 2018; 8:9816. [PMID: 29959403 PMCID: PMC6026196 DOI: 10.1038/s41598-018-28156-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Particulate matter (PM) is one of the most important environmental issues in China. This study aimed to explore the correlation between PM2.5 and airway inflammation in healthy rats. The PM2.5 group was given an intranasal instillation of PM2.5 suspension on 15 consecutive days, and each received oral saline from day 16 to 90. The BV intervention group was treated as the PM2.5 exposure group, except that BV instead of saline was given daily. A histopathologic examination was performed to evaluate the airway inflammation. The prevalence and function of Th1/Th2/Treg/Th17 cells were detected by flow cytometry and ELISA. The expression of AhR was detected by western blot and real-time PCR. We found that epithelial damage and increased infiltration of inflammatory cell were present in the airways after PM2.5 exposure; there was an immune imbalance of Th cells in the PM2.5 group; the expression of AhR was increased in the airways after PM2.5 exposure. In the PM2.5 + BV group, we demonstrated alleviated immune imbalance and reduced inflammatory cell infiltration in the airways. Our study showed that exposure to PM2.5 induced airway inflammation. The imbalance of Th1/Th2/Treg/Th17 in PM2.5-induced airway inflammation might be associated with activation of the AhR pathway. Oral BV reduces PM2.5-induced airway inflammation and regulates systemic immune responses in rats.
Collapse
Affiliation(s)
- Yang Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhi-Hai Zhang
- Chongqing Key Laboratory of Ophthalmology, Chongqing, People's Republic of China
| | - Di Hu
- Chongqing Key Laboratory of Ophthalmology, Chongqing, People's Republic of China
| | - Xia Ke
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zheng Gu
- Chongqing Key Laboratory of Ophthalmology, Chongqing, People's Republic of China
| | - Qi-Yuan Zou
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guo-Hua Hu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shang-Hua Song
- Chongqing Key Laboratory of Ophthalmology, Chongqing, People's Republic of China
| | - Hou-Yong Kang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Su-Ling Hong
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
33
|
Abstract
Neutrophilic bronchiolitis is the primary lesion in asthma-affected horses. Neutrophils are key actors in host defense, migrating toward sites of inflammation and infection, where they act as early responder cells toward external insults. However, neutrophils can also mediate tissue damage in various non-infectious inflammatory processes. Within the airways, these cells likely contribute to bronchoconstriction, mucus hypersecretion, and pulmonary remodeling by releasing pro-inflammatory mediators, including the cytokines interleukin (IL)-8 and IL-17, neutrophil elastase, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs). The mechanisms that regulate neutrophil functions in the tissues are complex and incompletely understood. Therefore, the inflammatory activity of neutrophils must be regulated with exquisite precision and timing, a task achieved through a complex network of mechanisms that regulates neutrophil survival. The discovery and development of compounds that can help regulate ROS, NET formation, cytokine release, and clearance would be highly beneficial in the design of therapies for this disease in horses. In this review, neutrophil functions during inflammation will be discussed followed by a discussion of their contribution to airway tissue injury in equine asthma.
Collapse
|
34
|
Dong M, Ma C, Wang WQ, Chen J, Wei Y. Regulation of the IL-33/ST2 pathway contributes to the anti-inflammatory effect of acupuncture in the ovalbumin-induced murine asthma model. Acupunct Med 2018; 36:319-326. [PMID: 29581139 DOI: 10.1136/acupmed-2017-011377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Bronchial asthma is a chronic airway inflammatory disease which has three main pathological features: airway hyperresponsiveness (AHR), airway remodelling, and chronic inflammation. Acupuncture is known to be an effective integrative medical therapy that has been used in the treatment of several chronic diseases, including bronchial asthma. The aim of the current study was to evaluate the effects of acupuncture on inflammation and regulation of the IL-33/ST2 pathway in a mouse model of asthma. METHODS The murine asthma model was established by both injection and inhalation of ovalbumin (OVA). Within 24 hours of the last OVA challenge, lung function was assessed by measurement of the airway resistance (RL) and lung dynamic compliance (Cdyn). Pulmonary tissues were collected for the detection of pathological changes and mucus secretion. Serum levels of tumour necrosis factor α (TNF-α), interleukin (IL)-1β, IL-33 and sST2 (secreted ST2) were detected by ELISA. Th17 cell proportions and counts in bronchoalveolar lavage fluid (BALF) were analysed by flow cytometry. RESULTS The results showed that AHR, chronic inflammation and mucus secretion were significantly suppressed by acupuncture treatment. RL decreased while Cdyn increased after acupuncture treatment. There was an apparent decrease in the serum concentrations of certain pro-inflammatory cytokines, such as TNF-α, IL-1β and IL-33, and an increase in sST2 level compared with untreated asthmatic mice. Acupuncture also reduced the CD4 +IL-17A+ cell proportion and counts in BALF. CONCLUSION Acupuncture effectively protects lung function and attenuates airway inflammation in the OVA-induced mouse model of asthma, which supports the role of acupuncture as a potential therapy in asthma treatment.
Collapse
Affiliation(s)
- Ming Dong
- Gumei Community Health Center of Minhang District of Shanghai, Shanghai, China
| | - Cheng Ma
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| | - Wen-Qian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| | - Juan Chen
- Department of Pediatric Neurological Rehabilitation, Maternal and Child Health Hospital of Dengfeng, Henan, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine of Fudan University, Shanghai, China
| |
Collapse
|
35
|
Guo W, Yu D, Wang X, Luo C, Chen Y, Lei W, Wang C, Ge Y, Xue W, Tian Q, Gao X, Yao W. Anti-inflammatory effects of interleukin-23 receptor cytokine-binding homology region rebalance T cell distribution in rodent collagen-induced arthritis. Oncotarget 2017; 7:31800-13. [PMID: 27177334 PMCID: PMC5077977 DOI: 10.18632/oncotarget.9309] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/28/2016] [Indexed: 12/31/2022] Open
Abstract
IL-23 is an important cytokine to regulate Th17 cell differentiation and promote the proliferation of inflammatory cells in Th17-mediated autoimmune diseases. The collagen-induced arthritis (CIA) in rat is a model of rheumatoid arthritis characterized by pronounced inflammatory auto-responses from B and T cells, especially Th17 cells in lesions. In the present study, we used rhIL23R-CHR to block the IL-23 signaling pathway to probe the importance of IL-23 in misbalancing the ratio of Th17/Th9/Treg cells in CIA rats. After treatments with rhIL23R-CHR, the CIA rats showed a significant decrease of secretions of IL-17 and IL-9, whereas FoxP3 was activated in the process, indicating that IL-23 can manipulate the balance of Th17/Th9/Treg cells. Similar to the animal model, IL-23 also possessed remarkable proinflammatory effects on human fibroblast-like synoviocyte cells (HFLS), showing synergetic outcomes with TNF-α. Together, IL-23 could act as a modulator to imbalance the ratio of Th17/Th9/Treg cells, and rhIL23R-CHR could serve as a potential therapeutic agent for RA patients.
Collapse
Affiliation(s)
- Wei Guo
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dongmei Yu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xin Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Cheng Luo
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yucong Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wen Lei
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yaoyao Ge
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenyao Xue
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qiqi Tian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiangdong Gao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
36
|
Barin JG, Talor MV, Diny NL, Ong S, Schaub JA, Gebremariam E, Bedja D, Chen G, Choi HS, Hou X, Wu L, Cardamone AB, Peterson DA, Rose NR, Čiháková D. Regulation of autoimmune myocarditis by host responses to the microbiome. Exp Mol Pathol 2017; 103:141-152. [PMID: 28822770 PMCID: PMC5721523 DOI: 10.1016/j.yexmp.2017.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/27/2022]
Abstract
The extensive, diverse communities that constitute the microbiome are increasingly appreciated as important regulators of human health and disease through inflammatory, immune, and metabolic pathways. We sought to elucidate pathways by which microbiota contribute to inflammatory, autoimmune cardiac disease. We employed an animal model of experimental autoimmune myocarditis (EAM), which results in inflammatory and autoimmune pathophysiology and subsequent maladaptive cardiac remodeling and heart failure. Antibiotic dysbiosis protected mice from EAM and fibrotic cardiac dysfunction. Additionally, mice derived from different sources with different microbiome colonization profiles demonstrated variable susceptibility to disease. Unexpectedly, it did not track with segmented filamentous bacteria (SFB)-driven Th17 programming of CD4+ T cells in the steady-state gut. Instead, we found disease susceptibility to track with presence of type 3 innate lymphoid cells (ILC3s). Ablating ILCs by antibody depletion or genetic tools in adoptive transfer variants of the EAM model demonstrated that ILCs and microbiome profiles contributed to the induction of CCL20/CCR6-mediated inflammatory chemotaxis to the diseased heart. From these data, we conclude that sensing of the microbiome by ILCs is an important checkpoint in the development of inflammatory cardiac disease processes through their ability to elicit cardiotropic chemotaxis.
Collapse
Affiliation(s)
- Jobert G Barin
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Monica V Talor
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Nicola L Diny
- The Johns Hopkins Bloomberg School of Public Health, The W. Harry Feinstone Dept. of Molecular Microbiology & Immunology, United States
| | - SuFey Ong
- The Johns Hopkins Bloomberg School of Public Health, The W. Harry Feinstone Dept. of Molecular Microbiology & Immunology, United States
| | - Julie A Schaub
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Elizabeth Gebremariam
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Djahida Bedja
- The Johns Hopkins University School of Medicine, Dept. of Cardiology, United States
| | - Guobao Chen
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Hee Sun Choi
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Xuezhou Hou
- The Johns Hopkins Bloomberg School of Public Health, The W. Harry Feinstone Dept. of Molecular Microbiology & Immunology, United States
| | - Lei Wu
- The Johns Hopkins Bloomberg School of Public Health, The W. Harry Feinstone Dept. of Molecular Microbiology & Immunology, United States
| | - Ashley B Cardamone
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Daniel A Peterson
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States
| | - Noel R Rose
- Brigham & Women's Hospital, Harvard Medical School, Dept. of Pathology, Boston, MA 02115, United States
| | - Daniela Čiháková
- The Johns Hopkins University School of Medicine, Dept. of Pathology, Div. of Immunology, Baltimore, MD 21205, United States; The Johns Hopkins Bloomberg School of Public Health, The W. Harry Feinstone Dept. of Molecular Microbiology & Immunology, United States.
| |
Collapse
|
37
|
Abstract
The biology of the T cell cytokines Interleukin (IL-)17 and IL-22 has been a main focus in the field of clinical immunology in the last decade. This intensive interest in both cytokines has resulted in almost 5,000 scientific publications (www.pubmed.com) dealing with the molecular structure, extra- and intracellular signaling pathways, specific transcription factors and the function of IL-17 and IL-22. This review article highlights the main findings concerning IL-17 and IL-22 in the last years.
Collapse
|
38
|
Avoiding contact allergens: from basic research to the in vitro identification of contact allergens. Allergol Select 2017; 1:77-84. [PMID: 30402606 PMCID: PMC6039996 DOI: 10.5414/alx01440e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 06/21/2011] [Indexed: 11/18/2022] Open
Abstract
Allergic contact dermatitis (ACD) is a chemical-induced inflammatory skin disease. Contact allergens are low-molecular-weight chemicals that must react with proteins in order to become immunogenic. This interaction leads to the activation of innate immune and stress responses and to the formation of antigenic epitopes for T cells which are the effector cells of ACD. Due to the multitude of chemicals that surround us in our daily life and their potential sensitizing capacity, it is crucial to identify contact sensitizers before these chemicals are used in consumer products. Appropriate in vitro assays for hazard identification are urgently needed to replace animal-based assays. The EU-wide ban on sensitization testing of cosmetic ingredients in animals is in effect since March 2009 and the necessity to test more than 30,000 already marketed chemicals for their sensitizing potential under the EU regulation REACh has intensified the worldwide efforts to replace animal testing. We summarize here the current strategies to develop a battery of assays which allows the identification of contact allergens by in vitro alternatives to animal testing. Our main focus lies on the test systems recently developed within the EU project Sens-it-iv in which we participate.
Collapse
|
39
|
Chaparro M, Ramas M, Benítez JM, López-García A, Juan A, Guardiola J, Mínguez M, Calvet X, Márquez L, Fernández Salazar LI, Bujanda L, García C, Zabana Y, Lorente R, Barrio J, Hinojosa E, Iborra M, Cajal MD, Van Domselaar M, García-Sepulcre MF, Gomollón F, Piqueras M, Alcaín G, García-Sánchez V, Panés J, Domènech E, García-Esquinas E, Rodríguez-Artalejo F, Gisbert JP. Extracolonic Cancer in Inflammatory Bowel Disease: Data from the GETECCU Eneida Registry. Am J Gastroenterol 2017; 112:1135-1143. [PMID: 28534520 DOI: 10.1038/ajg.2017.96] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The objective of this study was (a) To know the prevalence and distribution of extracolonic cancer (EC) in patients with inflammatory bowel disease (IBD); (b) To estimate the incidence rate of EC; (c) To evaluate the association between EC and treatment with immunosuppressants and anti-tumor necrosis factor (TNF) agents. METHODS This was an observational cohort study. INCLUSION CRITERIA IBD and inclusion in the ENEIDA Project (a prospectively maintained registry) from GETECCU. EXCLUSION CRITERIA Patients with EC before the diagnosis of IBD, lack of relevant data for this study, and previous treatment with immunosuppressants other than corticosteroids, thiopurines, methotrexate, or anti-TNF agents. The Kaplan-Meier method was used to evaluate the impact of several variables on the risk of EC, and any differences between survival curves were evaluated using the log-rank test. Stepwise multivariate Cox regression analysis was used to investigate factors potentially associated with the development of EC, including drugs for the treatment of IBD, during follow-up. RESULTS A total of 11,011 patients met the inclusion criteria and were followed for a median of 98 months. Forty-eight percent of patients (5,303) had been exposed to immunosuppressants or anti-TNF drugs, 45.8% had been exposed to thiopurines, 4.7% to methotrexate, and 21.6% to anti-TNF drugs. The prevalence of EC was 3.6%. In the multivariate analysis, age (HR=1.05, 95% CI=1.04-1.06) and having smoked (hazards ratio (HR)=1.47, 95% confidence interval (CI)=1.10-1.80) were the only variables associated with a higher risk of EC. CONCLUSIONS Neither immunosuppressants nor anti-TNF drugs seem to increase the risk of EC. Older age and smoking were associated with a higher prevalence of EC.
Collapse
Affiliation(s)
- María Chaparro
- Hospital de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - M Ramas
- Hospital de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - J M Benítez
- Hospital Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | | | - A Juan
- Hospital Germans Trias i Pujol (CIBERehd), Badalona, Spain
| | - J Guardiola
- Badalona, Hospital Bellvitge, Barcelona, Spain
| | - M Mínguez
- Hospital Clínico de Valencia, Valencia, Spain
| | - X Calvet
- Hospital Parc Taulí, Barcelona, Spain
| | | | | | - L Bujanda
- Hospital Donostia (CIBERehd), Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - C García
- Hospital Ramón y Cajal, Madrid, Spain
| | - Y Zabana
- Hospital Mutua de Terrassa (CIBERehd), Terrassa, Spain
| | - R Lorente
- Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - J Barrio
- Hospital Universitario Río Hortega, Valladolid, Spain
| | | | - M Iborra
- Hospital La Fe (CIBERehd), Valencia, Spain
| | | | | | | | - F Gomollón
- ISS Aragón, Hospital Clínico "Lozano Blesa" (CIBERehd), Zaragoza, Spain
| | | | - G Alcaín
- Hospital Clínico Universitario de Málaga, Málaga, Spain
| | - V García-Sánchez
- Hospital Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - J Panés
- Hospital Clinic (CIBERehd), Barcelona, Spain
| | - E Domènech
- Hospital Germans Trias i Pujol (CIBERehd), Badalona, Spain
| | - E García-Esquinas
- Department of Preventive Medicine and Public Health, Universidad Autónoma de Madrid/IdiPaz and CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - F Rodríguez-Artalejo
- Department of Preventive Medicine and Public Health, Universidad Autónoma de Madrid/IdiPaz and CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - J P Gisbert
- Hospital de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| |
Collapse
|
40
|
Munde EO, Raballah E, Okeyo WA, Ong'echa JM, Perkins DJ, Ouma C. Haplotype of non-synonymous mutations within IL-23R is associated with susceptibility to severe malaria anemia in a P. falciparum holoendemic transmission area of Kenya. BMC Infect Dis 2017; 17:291. [PMID: 28427357 PMCID: PMC5397818 DOI: 10.1186/s12879-017-2404-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/13/2017] [Indexed: 12/17/2022] Open
Abstract
Background Improved understanding of the molecular mechanisms involved in pediatric severe malarial anemia (SMA) pathogenesis is a crucial step in the design of novel therapeutics. Identification of host genetic susceptibility factors in immune regulatory genes offers an important tool for deciphering malaria pathogenesis. The IL-23/IL-17 immune pathway is important for both immunity and erythropoiesis via its effects through IL-23 receptors (IL-23R). However, the impact of IL-23R variants on SMA has not been fully elucidated. Methods Since variation within the coding region of IL-23R may influence the pathogenesis of SMA, the association between IL-23R rs1884444 (G/T), rs7530511 (C/T), and SMA (Hb < 6.0 g/dL) was examined in children (n = 369, aged 6–36 months) with P. falciparum malaria in a holoendemic P. falciparum transmission area. Results Logistic regression analysis, controlling for confounding factor of anemia, revealed that individual genotypes of IL-23R rs1884444 (G/T) [GT; OR = 1.34, 95% CI = 0.78–2.31, P = 0.304 and TT; OR = 2.02, 95% CI = 0.53–7.74, P = 0.286] and IL-23R rs7530511 (C/T) [CT; OR = 2.6, 95% CI = 0.59–11.86, P = 0.202 and TT; OR = 1.66, 95% CI = 0.84–3.27, P = 0.142] were not associated with susceptibility to SMA. However, carriage of IL-23R rs1884444T/rs7530511T (TT) haplotype, consisting of both mutant alleles, was associated with increased susceptibility to SMA (OR = 1.12, 95% CI = 1.07–4.19, P = 0.030). Conclusion Results presented here demonstrate that a haplotype of non-synonymous IL-23R variants increase susceptibility to SMA in children of a holoendemic P. falciparum transmission area. Electronic supplementary material The online version of this article (doi:10.1186/s12879-017-2404-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elly O Munde
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya.,University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Evans Raballah
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Medical Laboratory Science, School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Winnie A Okeyo
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - John M Ong'echa
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Douglas J Perkins
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.,Department of Internal Medicine, Centre for Global Health, Health Sciences Centre, University of New Mexico, Albuquerque, NM, USA
| | - Collins Ouma
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya. .,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya. .,Ideal Research Centre, Kisumu, Kenya.
| |
Collapse
|
41
|
Involvement of lncRNA-1700040D17Rik in Th17 cell differentiation and the pathogenesis of EAE. Int Immunopharmacol 2017; 47:141-149. [PMID: 28395256 DOI: 10.1016/j.intimp.2017.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 02/22/2017] [Accepted: 03/10/2017] [Indexed: 02/08/2023]
Abstract
IL-23/STAT3 signaling pathway is a key process in Th17 cell differentiation, and Th17 cells are closely related to the development of autoimmune diseases. We previously designed and prepared rhIL23R-CHR protein to antagonize endogenous IL-23, showing effectiveness in the treatment of experimental autoimmune encephalomyelitis (EAE) in mice. To further elucidate the mechanism of action, mouse lncRNA microarray was used to screen expression profiles of lncRNAs, and a particular lncRNA, 1700040D17Rik was found to down-regulate in EAE model and its expression was significantly increased after the treatment by rhIL23R-CHR. The function of 1700040D17Rik was revealed to associate with the differentiation of Th17 cells through the regulation of the key transcription factor RORγt. Together, regulation of Th17 cells through lncRNA is responsible for the effects of rhIL23R-CHR to balance the immune responses, and 1700040D17Rik has the potential to serve as a therapeutic target or a biomarker for autoimmune diseases.
Collapse
|
42
|
Gu ZW, Wang YX, Cao ZW. Neutralization of interleukin-17 suppresses allergic rhinitis symptoms by downregulating Th2 and Th17 responses and upregulating the Treg response. Oncotarget 2017; 8:22361-22369. [PMID: 28423590 PMCID: PMC5410229 DOI: 10.18632/oncotarget.15652] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 02/15/2017] [Indexed: 01/05/2023] Open
Abstract
Allergic rhinitis (AR) has long been considered to predominantly involve the actions of Th2 cells, with relatively small contributions from Th1 cells. In recent years, the discovery of Th17 and regulatory T (Treg) cells has rendered the Th1/Th2 balance paradigm more complex and expanded our understanding of the pathogenesis of AR. IL-17, a key cytokine produced by Th17 cells, is known to induce allergen-specific Th2 cell activation, eosinophil and neutrophil accumulation, and serum IgE production in asthma; all of these features may play important roles in AR. To the best of our knowledge, only a few studies have assessed the feasibility of using IL-17 antagonists to treat AR. Thus, the principal objectives of the present study were, first, to determine the status of Th17 and Treg cells in the nasal mucosa of a mouse model of AR, and, second, to investigate the effects of IL-17 on such cells and the therapeutic efficacy of anti-IL-17 antibodies (Abs) in the context of AR. Anti-IL-17 Abs were given intranasally during the re-challenge of BALB/c mice with ovalbumin (OVA)-induced AR. We measured the numbers of nasal rubbing motions and sneezes, eosinophil and neutrophil levels, Th1, Th2, Th17, and Treg parameters in the nasal mucosa. Anti-IL-17 Abs markedly reduced the number of nasal rubbing motions and sneezes, decreased eosinophil and neutrophil infiltration, reduced Th2 and Th17 responses, and increased the Treg response. Anti-IL-17 Ab treatment protects against AR. These results will improve our understanding of AR pathogenesis and may lead to the development of novel therapeutic approaches for management of the condition.
Collapse
Affiliation(s)
- Zhao Wei Gu
- Department of Otorhinolaryngology, China Medical University Affiliated Shengjing Hospital, Shenyang, Liaoning, China
| | - Yun Xiu Wang
- Department of Medical Insurance, China Medical University Affiliated Shengjing Hospital, Shenyang, Liaoning, China
| | - Zhi Wei Cao
- Department of Otorhinolaryngology, China Medical University Affiliated Shengjing Hospital, Shenyang, Liaoning, China
| |
Collapse
|
43
|
Tian J, Liu Y, Jiang Y, Zhou H, Zhu T, Zhao X, Peng L, Yan C. Association of single nucleotide polymorphisms of IL23R and IL17 with necrotizing enterocolitis in premature infants. Mol Cell Biochem 2017; 430:201-209. [PMID: 28224332 DOI: 10.1007/s11010-017-2972-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/02/2017] [Indexed: 12/30/2022]
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal inflammatory disease in neonates, particularly in preterm infants. The interleukin (IL) 23/IL17 axis has been shown to play an important role in the gastrointestinal inflammation. However, the association of gene polymorphisms in the IL23/IL17 axis and the development of NEC remains unknown. In this study, we aimed to explore a possible genetic role of IL23R and IL17 in the development of NEC. We identified single nucleotide polymorphisms (SNPs) in IL23R (rs10889677), IL17A (rs2275913), and IL17F (rs763780) by polymerase chain reaction and Sanger sequencing. A total of 102 NEC patients (stage II, n = 75; and stage III, n = 27) and 120 control subjects were recruited for the study. All of the participants were premature (gestational age < 37 weeks). Our results revealed that the combination of the IL17F rs763780 (TC + CC) genotype and the C allele both significantly increased the risk of NEC [odds ratio (OR) 1.89, 95% confidence interval (CI) 1.04-3.43, P = 0.035; OR 1.82, 95% CI 1.06-3.13, P = 0.028, respectively]. Furthermore, the rs763780 (TC + CC) genotype was associated with increased severity of NEC and the incidence of NEC-related perforation [OR 2.80, 95% CI 1.10-7.12, P = 0.031; OR 3.86, 95% CI 1.10-13.53, P = 0.035, respectively]. However, IL23R rs10889677 and IL17A rs2275913 were not associated with the susceptibility to NEC. In conclusion, our data suggest that a variant of IL17F (rs763780) may contribute to the development of NEC.
Collapse
Affiliation(s)
- Jiayi Tian
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.,Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Yanjun Liu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.,Division of Endocrinology, Metabolism, and Molecular Medicine, Charles R. Drew University of Medicine and Sciences, University of California Los Angeles (UCLA) School of Medicine, Los Angeles, CA, USA
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Haohan Zhou
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China.,Department of Orthopedics Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tong Zhu
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoqi Zhao
- Department of Neonatology, The Second Hospital of Jilin University, Changchun, China
| | - Liping Peng
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, China. .,Department of Respirology, The First Hospital of Jilin University, Changchun, China.
| | - Chaoying Yan
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
44
|
Wang X, Hui Y, Zhao L, Hao Y, Guo H, Ren F. Oral administration of Lactobacillus paracasei L9 attenuates PM2.5-induced enhancement of airway hyperresponsiveness and allergic airway response in murine model of asthma. PLoS One 2017; 12:e0171721. [PMID: 28199353 PMCID: PMC5310903 DOI: 10.1371/journal.pone.0171721 10.1371/journal.pone.0171721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This study investigated allergy immunotherapy potential of Lactobacillus paracasei L9 to prevent or mitigate the particulate matter 2.5 (PM2.5) enhanced pre-existing asthma in mice. Firstly, we used a mouse model of asthma (a 21-day ovalbumin (OVA) sensitization and challenge model) followed by PM2.5 exposure twice on the same day of the last challenge. PM2.5 was collected from the urban area of Beijing and underwent analysis for metals and polycyclic aromatic hydrocarbon contents. The results showed that PM2.5 exposure enhanced airway hyper-responsiveness (AHR) and lead to a mixed Th2/ IL-17 response in asthmatic mice. Secondly, the PM2.5 exposed asthmatic mice were orally administered with L9 (4×107, 4×109 CFU/mouse, day) from the day of first sensitization to the endpoint, for 20 days, to investigate the potential mitigative effect of L9 on asthma. The results showed that L9 ameliorated PM2.5 exposure enhanced AHR with an approximate 50% decrease in total airway resistance response to methacholine (48 mg/ml). L9 also prevented the exacerbated eosinophil and neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and decreased the serum level of total IgE and OVA-specific IgG1 by 0.44-fold and 0.3-fold, respectively. Additionally, cytokine production showed that L9 significantly decreased T-helper cell type 2 (Th2)-related cytokines (IL-4, -5, -13) and elevated levels of Th1 related IFN-γ in BALF. L9 also reduced the level of IL-17A and increased the level of TGF-β. Taken together, these results indicate that L9 may exert the anti-allergic benefit, possibly through rebalancing Th1/Th2 immune response and modulating IL-17 pro-inflammatory immune response. Thus, L9 is a promising candidate for preventing PM exposure enhanced pre-existing asthma.
Collapse
Affiliation(s)
- Xifan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yan Hui
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Huiyuan Guo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
- Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
- * E-mail:
| |
Collapse
|
45
|
Wang X, Hui Y, Zhao L, Hao Y, Guo H, Ren F. Oral administration of Lactobacillus paracasei L9 attenuates PM2.5-induced enhancement of airway hyperresponsiveness and allergic airway response in murine model of asthma. PLoS One 2017; 12:e0171721. [PMID: 28199353 PMCID: PMC5310903 DOI: 10.1371/journal.pone.0171721] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 11/22/2022] Open
Abstract
This study investigated allergy immunotherapy potential of Lactobacillus paracasei L9 to prevent or mitigate the particulate matter 2.5 (PM2.5) enhanced pre-existing asthma in mice. Firstly, we used a mouse model of asthma (a 21-day ovalbumin (OVA) sensitization and challenge model) followed by PM2.5 exposure twice on the same day of the last challenge. PM2.5 was collected from the urban area of Beijing and underwent analysis for metals and polycyclic aromatic hydrocarbon contents. The results showed that PM2.5 exposure enhanced airway hyper-responsiveness (AHR) and lead to a mixed Th2/ IL-17 response in asthmatic mice. Secondly, the PM2.5 exposed asthmatic mice were orally administered with L9 (4×107, 4×109 CFU/mouse, day) from the day of first sensitization to the endpoint, for 20 days, to investigate the potential mitigative effect of L9 on asthma. The results showed that L9 ameliorated PM2.5 exposure enhanced AHR with an approximate 50% decrease in total airway resistance response to methacholine (48 mg/ml). L9 also prevented the exacerbated eosinophil and neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and decreased the serum level of total IgE and OVA-specific IgG1 by 0.44-fold and 0.3-fold, respectively. Additionally, cytokine production showed that L9 significantly decreased T-helper cell type 2 (Th2)-related cytokines (IL-4, -5, -13) and elevated levels of Th1 related IFN-γ in BALF. L9 also reduced the level of IL-17A and increased the level of TGF-β. Taken together, these results indicate that L9 may exert the anti-allergic benefit, possibly through rebalancing Th1/Th2 immune response and modulating IL-17 pro-inflammatory immune response. Thus, L9 is a promising candidate for preventing PM exposure enhanced pre-existing asthma.
Collapse
Affiliation(s)
- Xifan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yan Hui
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanling Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Huiyuan Guo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Co-constructed by ministry of Education and Beijing Government, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
- Beijing Higher Institution Engineering Research Center of Animal Product, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
46
|
Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol 2016; 138:984-1010. [DOI: 10.1016/j.jaci.2016.06.033] [Citation(s) in RCA: 450] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022]
|
47
|
Abstract
This review focuses on conjunctival goblet cells and their essential function in the maintenance of eye health. The main function of goblet cells is to produce and secrete mucins that lubricate the ocular surface. An excess or a defect in those mucins leads to several alterations that makes goblet cells central players in maintaining the proper mucin balance and ensuring the correct function of ocular surface tissues. A typical pathology that occurs with mucous deficiency is dry eye disease, whereas the classical example of mucous hyperproduction is allergic conjunctivitis. In this review, we analyze how goblet cell number and function can be altered in these diseases and in contact lens (CL) wearers. We found that most published studies focused exclusively on the goblet cell number. However, recent advances have demonstrated that, along with mucin secretion, goblet cells are also able to secrete cytokines and respond to them. We describe the effect of different cytokines on goblet cell proliferation and secretion. We conclude that it is important to further explore the effect of CL wear and cytokines on conjunctival goblet cell function.
Collapse
|
48
|
Liu J, Wei Y, Luo Q, Xu F, Zhao Z, Zhang H, Lu L, Sun J, Liu F, Du X, Li M, Wei K, Dong J. Baicalin attenuates inflammation in mice with OVA-induced asthma by inhibiting NF-κB and suppressing CCR7/CCL19/CCL21. Int J Mol Med 2016; 38:1541-1548. [PMID: 27666000 DOI: 10.3892/ijmm.2016.2743] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 09/09/2016] [Indexed: 11/06/2022] Open
Abstract
Baicalin, extracted and purified from the Chinese medicinal plant, Scutellaria baicalensis Georgi (Huang qin in Chinese), exhibits potent anti-inflammatory activity against asthma. However, it remains unknown whether baicalin inhibits the activity of CC chemokine receptor 7 (CCR7) and its ligands, which are crucial for the initiation of airway inflammation. In the present study, we investigated the effects of baicalin on CCR7 and its ligands, CCL19 and CCL21, as well as on the nuclear factor-κB (NF-κB) pathway in a mouse model of asthma. A mouse model of acute asthma was established by exposing the mice to ovalbumin (OVA) (by intraperitoneal injection and inhalational challenge). Within 24 h of the final OVA challenge, lung function was detected by direct airway resistance analysis. Lung tissues were examined for pathological changes. Inflammatory cell counts in bronchoalveolar lavage fluid (BALF) were assessed. ELISA was utilized to evaluate the OVA-IgE, CCL19 and CCL21 levels in BALF. The interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels in serum were also detected by ELISA. The protein expression levels of CCR7, as well as that of phosphorylated IκBα (p-IκBα) and phosphorylated p65 (p-p65) were determined by western blot analysis and RT-qPCR was used to determine the CCR7 mRNA levels. Our data demonstrated that the oral administration of baicalin significantly improved pulmonary function and attenuated inflammatory cell infiltration into the lungs. Baicalin also decreased the levels of OVA-IgE, IL-6, TNF-α and CCR7, as well as those of its ligand, CCL19; the levels of NF-κB were also markedly suppressed by baicalin. The CCR7 mRNA level was substantially decreased. Our results thus suggest that baicalin exerts an inhibitory effect on airway inflammation, and this effect may be associated with the inhibition of CCR7 and CCL19/CCL21, which may provide new mechanistic insight into the anti‑inflammatory effects of baicalin.
Collapse
Affiliation(s)
- Jiaqi Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Fei Xu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhengxiao Zhao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hongying Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Feng Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xin Du
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Mihui Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Kai Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
49
|
Abstract
One key approach to increase the efficacy and the safety of immunotherapy is the use of adjuvants. However, many of the adjuvants currently in use can cause adverse events, raising concerns regarding their clinical use, and are geared toward productive immune responses but not necessarily tolerogenic responses. Thus, novel adjuvants for immunotherapy are needed and are being developed. Essential is their potential to boost appropriate tolerogenic adaptive immune responses to allergens while limiting side effects. This review provides an overview of adjuvants currently in clinical use or under development and discusses their therapeutic effect in enhancing allergen-induced tolerance.
Collapse
|
50
|
Abstract
Allergy is a common hypersensitivity disorder of the immune system, which, along with other factors, is also subjected to regulation by microRNAs. The most common allergic diseases are allergic rhinitis, asthma, atopic dermatitis, and food allergy, which all are multifactorial and very heterogeneous conditions, highlighting the need for more individualized treatment techniques. More particular key questions in relation to allergic diseases are how microRNAs influence the differentiation, polarization, plasticity and functions of T helper and other immune cells, as well as the development of immune tolerance. In addition, microRNAs can affect allergic inflammation and tissue remodeling through their functions in epithelial and other tissue cells. Among immune system-related microRNAs, miR-21, miR-146a, and miR-155 are the most intensively studied and have convincingly been demonstrated to regulate immune responses and tissue inflammation in allergic diseases. Further characterization of microRNA functions is important, as similar to other conditions, the modulation of microRNA expression could potentially be used for therapeutic purposes in allergic diseases in the future. In addition, miRNAs could be implemented as biomarkers for endotyping complex allergic conditions.
Collapse
|