1
|
Jin J, Nguyen TV, Jiang Y, Yu ZN, Song CH, Lee SY, Shin HS, Chai OH. Hydrangea serrata extract attenuates PM-exacerbated airway inflammation in the CARAS model by modulating the IL-33/ST2/NF-κB signaling pathway. Biomed Pharmacother 2024; 174:116596. [PMID: 38631146 DOI: 10.1016/j.biopha.2024.116596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Particulate matter (PM) significantly contributes to the global health crisis of respiratory diseases. It is known to induce and exacerbate conditions such as asthma and respiratory infections. Long exposure to PM can increase the risk of combined allergic rhinitis and asthma syndrome (CARAS). Although therapeutic drugs can be used to improve symptoms of respiratory diseases caused by PM, their usage is often accompanied by side effects. Therefore, many studies are being conducted to discover functional food materials that can more effectively treat respiratory diseases while minimizing the side effects of these therapeutic drugs. This study was conducted to investigate the efficacy of Hydrangea serrata extract (HSE) in airway inflammation in a mouse model of CARAS exacerbated by PM. In the CARAS mouse model worsened by PM, the airway inflammation improvement effect of HSE was evaluated by analyzing allergic nasal symptoms, changes in inflammatory cells, OVA-specific immunoglobulin (Ig) levels, cytokines, mast cell activation, and histopathological findings of both nasal mucosa and lung tissue. HSE effectively reduced OVA-specific IgE and IgG1 and inhibited the production of T helper type 2 (Th2)-related cytokines such as IL-4 and IL-5. Importantly, HSE reduced IL-33 and ST2 expression and inhibited the activation of the NF-κB signaling pathway. In addition, HSE inhibited airway hypersensitivity, mucus production, and inflammatory cell infiltration. These results suggest that HSE may inhibit airway inflammation in CARAS/PM mice by regulating the IL-33/ST2/NF-κB signaling pathway, opening avenues for considering HSE as a potential material for treating allergic airway inflammation diseases in the future.
Collapse
Affiliation(s)
- Juan Jin
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| | - Thi Van Nguyen
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| | - Yuna Jiang
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| | - Zhen Nan Yu
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| | - Chang Ho Song
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea; Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea
| | - So-Young Lee
- Department of Food Biotechnology, University of Science and Technology (UST), Daejeon 34113, South Korea; Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, South Korea
| | - Hee Soon Shin
- Department of Food Biotechnology, University of Science and Technology (UST), Daejeon 34113, South Korea; Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, South Korea
| | - Ok Hee Chai
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea; Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54896, South Korea.
| |
Collapse
|
2
|
Liu K, Huang H, Xiong M, Wang Q, Chen X, Feng Y, Ma H, Chen W, Li X, Ye X. IL-33 Accelerates Chronic Atrophic Gastritis through AMPK-ULK1 Axis Mediated Autolysosomal Degradation of GKN1. Int J Biol Sci 2024; 20:2323-2338. [PMID: 38617533 PMCID: PMC11008276 DOI: 10.7150/ijbs.93573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a complex disease characterized by atrophy and inflammation in gastric mucosal tissue, especially with high expression of interleukins. However, the interaction and mechanisms between interleukins and gastric mucosal epithelial cells in CAG remain largely elusive. Here, we elucidate that IL-33 stands out as the predominant inflammatory factor in CAG, and its expression is induced by H. pylori and MNNG through the ROS-STAT3 signaling pathway. Furthermore, our findings reveal that the IL-33/ST2 axis is intricately involved in the progression of CAG. Utilizing phosphoproteomics mass spectrometry, we demonstrate that IL-33 enhances autophagy in gastric epithelial cells through the phosphorylation of AMPK-ULK1 axis. Notably, inhibiting autophagy alleviates CAG severity, while augmentation of autophagy exacerbates the disease. Additionally, ROS scavenging emerges as a promising strategy to ameliorate CAG by reducing IL-33 expression and inhibiting autophagy. Intriguingly, IL-33 stimulation promotes GKN1 degradation through the autolysosomal pathway. Clinically, the combined measurement of IL-33 and GKN1 in serum shows potential as diagnostic markers. Our findings unveil an IL-33-AMPK-ULK1 regulatory mechanism governing GKN1 protein stability in CAG, presenting potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Kewei Liu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hongxia Huang
- Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qiaojiao Wang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xiantao Chen
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Yinqiong Feng
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Hang Ma
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400038, China
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Xuegang Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400038, China
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
3
|
Qin Z, Xie L, Li W, Wang C, Li Y. New Insights into Mechanisms Traditional Chinese Medicine for Allergic Rhinitis by Regulating Inflammatory and Oxidative Stress Pathways. J Asthma Allergy 2024; 17:97-112. [PMID: 38405022 PMCID: PMC10888064 DOI: 10.2147/jaa.s444923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Allergy rhinitis (AR) is becoming more common and has serious medical and societal consequences. Sneezing, paroxysmal nasal blockage, nasal itching, mucosal edema, coughing, and rhinorrhea are symptoms of this type I allergic immunological illness. Immunoglobulin E-mediated inflammation is the cause of it. Because AR is prone to recurrent attacks, extended medication therapy may impair its effectiveness. In addition to negatively affecting the patients' physical health, this can also negatively impact their mental health. During AR development, there are inflammatory and oxidative stress responses that are linked to problems in a number of signal transduction pathways. By using the terms "allergic rhinitis", "traditional Chinese medicine", "inflammation", and "oxidative stress", we screened for pertinent research published over the previous five years in databases like PubMed. We saw that NF-KB, TLR, IL-33/ST2, PI3K/AKT, MAPK, and Nrf2 are some of the most important inflammatory and oxidative stress pathways in AR. Studies have revealed that antioxidant and anti-inflammatory therapy reduced the risk of AR and was therapeutic; however, the impact of the therapy varies widely. The Chinese medical system places a high value on traditional Chinese medicine (TCM), which has been there for virtually all of China's 5000-year history. By influencing signaling pathways related to inflammation and oxidative stress, Chinese herbal medicine and its constituent compounds have been shown to prevent allergic rhinitis. This review will focus on this evidence and provide references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Zhu Qin
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Liangzhen Xie
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Wentao Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Chao Wang
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Yan Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
4
|
Jin J, Fan YJ, Nguyen TV, Yu ZN, Song CH, Lee SY, Shin HS, Chai OH. Chaenomeles sinensis Extract Ameliorates Ovalbumin-Induced Allergic Rhinitis by Inhibiting the IL-33/ST2 Axis and Regulating Epithelial Cell Dysfunction. Foods 2024; 13:611. [PMID: 38397588 PMCID: PMC10888344 DOI: 10.3390/foods13040611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Chaenomeles sinensis has traditionally been used as an herbal medicine due to its characteristics that protect against inflammation, hypertension, and mutagenesis. However, the effect of Chaenomeles sinensis extract (CSE) on allergic rhinitis (AR) and its underlying mechanisms have yet to be thoroughly investigated. The current study explored the likely effect of CSE on AR in an ovalbumin (OVA)-induced AR mouse model. To this end, OVA-specific immunoglobulins, nasal symptoms, cytokine production, the infiltration of inflammatory cells, and nasal histopathology were assessed to determine the role of CSE against AR. The supplementation of CSE was found to suppress OVA-specific IgE, while OVA-specific IgG2a was increased in the serum. Further, CSE ameliorated the production of T helper type 2 (Th2) cytokines whereas it increased Th1 cytokine levels in nasal lavage fluid. Moreover, the CSE treatment group exhibited significant inhibition of IL-33/ST2 signaling. Subsequently, CES reversed the OVA-induced enhancement of epithelial permeability and upregulated E-cadherin, thus indicating that CES plays a protective role on epithelial barrier integrity. Altogether, the oral administration of CSE effectively controlled allergic response by restricting the buildup of inflammatory cells, enhancing nasal and lung histopathological traits, and regulating cytokines associated with inflammation. Collectively, the results show that the supplementation of CSE at different doses effectively regulated AR, thus suggesting the therapeutic efficiency of CSE in suppressing airway diseases.
Collapse
Affiliation(s)
- Juan Jin
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
| | - Yan Jing Fan
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
| | - Thi Van Nguyen
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
| | - Zhen Nan Yu
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
| | - Chang Ho Song
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
- Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
| | - So-Young Lee
- Department of Food Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Hee Soon Shin
- Department of Food Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Ok Hee Chai
- Department of Anatomy, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (J.J.)
- Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
| |
Collapse
|
5
|
Porfire (Irimia) IM, Berindan-Neagoe I, Budisan L, Leucuta DC, Gata A, Minoiu AC, Georgescu BA, Covaliu BF, Albu S. Tissue Interleukin-33: A Novel Potential Regulator of Innate Immunity and Biomarker of Disease Severity in Chronic Rhinosinusitis with Nasal Polyps. J Clin Med 2023; 12:7537. [PMID: 38137606 PMCID: PMC10743505 DOI: 10.3390/jcm12247537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Chronic rhinosinusitis with nasal polyps (CRSwNP) is a disease of real interest for researchers due to its heterogenicity and complex pathophysiological mechanisms. Identification of the factors that ensure success after treatment represents one of the main challenges in CRSwNP research. No consensus in this direction has been reached so far. Biomarkers for poor outcomes have been noted, but nonetheless, their prognostic value has not been extensively investigated, and needs to be sought. We aimed to evaluate the correlation between potential prognostic predictors for recalcitrant disease in patients with CRSwNP. METHODS The study group consisted of CRSwNP patients who underwent surgical treatment and nasal polyp (NP) tissue sampling. The preoperative workup included Lund-Mackay assessment, nasal endoscopy, eosinophil blood count, asthma, and environmental allergy questionnaire. Postoperatively, in subjects with poor outcomes, imagistic osteitis severity was evaluated, and IL-33 expression was measured. RESULTS IL-33 expression in NP was positively and significantly correlated with postoperative osteitis on CT scans (p = 0.01). Furthermore, high osteitis CT scores were related to high blood eosinophilia (p = 0.01). A positive strong correlation was found between postoperative osteitis and the Lund-Mackay preoperative score (p = 0.01), as well as the nasal endoscopy score (p = 0.01). CONCLUSIONS Our research analyzed the levels of polyp IL-33, relative to blood eosinophilia, overall disease severity score, and osteitis severity, in patients with CRSwNP. These variables are prognostic predictors for poor outcomes and recalcitrant disease. Considering the importance of bone involvement in CRSwNP, this research aims to provide a better insight into the correlations of osteitis with clinical and biological factors.
Collapse
Affiliation(s)
- Ioana Maria Porfire (Irimia)
- IInd Department of Otorhinolaryngology, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.B.-N.); (L.B.)
| | - Livia Budisan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (I.B.-N.); (L.B.)
| | - Daniel-Corneliu Leucuta
- Medical Informatics and Biostatistics Department, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400349 Cluj-Napoca, Romania;
| | - Anda Gata
- IInd Department of Otorhinolaryngology, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Aurelian Costin Minoiu
- Diagnostical and Interventional Radiology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | | | - Bogdan Florin Covaliu
- IVth Department of Community Medicine, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Silviu Albu
- IInd Department of Otorhinolaryngology, ‘Iuliu Hatieganu’ University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
6
|
Mou J, Xie L, Xu Y, Zhou T, Liu Y, Huang Q, Tang K, Tian Z, Xing H, Qiu S, Rao Q, Wang M, Wang J. 2B4 inhibits the apoptosis of natural killer cells through phosphorylated extracellular signal-related kinase/B-cell lymphoma 2 signal pathway. Cytotherapy 2023; 25:1080-1090. [PMID: 37516949 DOI: 10.1016/j.jcyt.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND AIMS Decades after the identification of natural killer (NK) cells as potential effector cells against malignantly transformed cells, an increasing amount of research suggests that NK cells are a prospective choice of immunocytes for cancer immunotherapy in addition to T lymphocytes for cancer immunotherapy. Recent studies have led to a breakthrough in the combination of hematopoietic stem-cell transplantation with allogeneic NK cells infusion for the treatment of malignant tumors. However, the short lifespan of NK cells in patients is the major impediment, limiting their efficacy. Therefore, prolonging the survival of NK cells will promote the application of NK-cell immunotherapy. As we have known, NK cells use a "missing-self" mechanism to lyse target cells and exert their functions through a wide array of activating, co-stimulatory and inhibitory receptors. Our previous study has suggested that CD244 (2B4), one of the co-stimulatory receptors, can improve the function of chimeric antigen receptor NK cells. However, the underlying mechanism of how 2B4 engages in the function of NK cells requires further investigation. Overall, we established a feeder cell with the expression of CD48, the ligand of 2B4, to investigate the function of 2B4-CD48 axis in NK cells, and meanwhile, to explore whether the newly generated feeder cell can improve the function of ex vivo-expanded NK cells. METHODS First, K562 cells overexpressing 4-1BBL and membrane-bound IL-21 (mbIL-21) were constructed (K562-41BBL-mbIL-21) and were sorted to generate the single clone. These widely used feeder cells (K562-41BBL-mbIL-21) were named as Basic Feeder hereinafter. Based on the Basic feeder, CD48 was overexpressed and named as CD48 Feeder. Then, the genetically modified feeder cells were used to expand primary NK cells from peripheral blood or umbilical cord blood. In vitro experiments were performed to compare proliferation ability, cytotoxicity, survival and activation/inhibition phenotypes of NK cells stimulated via different feeder cells. K562 cells were injected into nude mice subcutaneously with tail vein injection of NK cells from different feeder system for the detection of NK in vivo persistence and function. RESULTS Compared with Basic Feeders, CD48 Feeders can promote the proliferation of primary NK cells from peripheral blood and umbilical cord blood and reduce NK cell apoptosis by activating the p-ERK/BCL2 pathway both in vitro and in vivo without affecting overall phenotypes. Furthermore, NK cells expanded via CD48 Feeders showed stronger anti-tumor capability and infiltration ability into the tumor microenvironment. CONCLUSIONS In this preclinical study, the engagement of the 2B4-CD48 axis can inhibit the apoptosis of NK cells through the p-ERK/BCL2 signal pathway, leading to an improvement in therapeutic efficiency.
Collapse
Affiliation(s)
- Junli Mou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Leling Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tong Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qianqian Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Tianjin, China; Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| |
Collapse
|
7
|
Wang Y, Li J, Wang F, Cui Y, Song L, Ruan B, Yu Y. Analysis of ceRNA Regulatory Mechanism of Rape Pollen Allergy Based on Whole-Transcriptome Sequencing of Peripheral Blood Mononuclear Cells. J Asthma Allergy 2023; 16:775-788. [PMID: 37534326 PMCID: PMC10390718 DOI: 10.2147/jaa.s416772] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/12/2023] [Indexed: 08/04/2023] Open
Abstract
Background Rape pollen allergy is a common allergic reaction disorder that affects the health and life of patients seriously. The research on ceRNA regulatory network in rape pollen allergy is poor. Methods High throughput whole-transcriptome sequencing was conducted on rape pollen allergic samples and non-allergic samples. Differentially expressed microRNAs (DEmiRNAs), circRNAs (DEcircRNAs), long non-coding RNA (DElncRNAs), mRNA (DEmRNAs) were identified and a ceRNA regulatory network was constructed by Cytoscape. Functional enrichment analyses were performed on DEmRNAs in the ceRNA network. Then, the least absolute shrinkage and selection operator (LASSO) regression model was used to identify characteristic genes for rape pollen allergy. The receiver operating characteristic (ROC) curve was used to evaluate the diagnostic ability of characteristic genes. Results A total of 25 DEmiRNAs, 258 DEcircRNAs, 304 DElncRNAs, and 383 DEmRNAs in the allergic group compared with the non-allergic group were uncovered, respectively. A ceRNA network containing 21 miRNAs, 57 circRNAs, 28 lncRNAs, and 33 mRNAs was generated with 139 nodes and 160 edges. The signal transduction-related processes, immune-related processes, the ion, inorganic substance, and hormone regulation processes were associated with mRNAs in the ceRNA network. The results of pathway enrichment illustrated that mRNAs in the ceRNA were significantly linked to IL-17 signaling pathway, inflammatory mediator regulation of trp channels, GMP-PKG signaling pathway, signaling by GPCR, and GPCR downstream signaling pathway. Then, five characteristic genes (KCNQ3, CCR5, FOSB, CFAP43, and PRKG1) were defined by the LASSO algorithm. The AUC values of these genes indicated that these genes had a powerful discrimination ability in discriminating allergic samples from non-allergic samples. Conclusion Taken together, we revealed the ceRNA regulatory network in rape pollen allergy and excavated five characteristic genes (KCNQ3, CCR5, FOSB, CFAP43, and PRKG1) with the diagnostic value that may be a potential target in diagnosis and treatment.
Collapse
Affiliation(s)
- Ya Wang
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Jianhua Li
- Department of Otolaryngology, the Sixth Affiliated Hospital of Kunming Medical University, Yuxi, People’s Republic of China
| | - Fang Wang
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Yunhua Cui
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Li Song
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Biao Ruan
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Yongmei Yu
- Department of Otolaryngology, the First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
8
|
Zhang H, Li F, Cao J, Wang X, Cheng H, Qi K, Wang G, Xu K, Zheng J, Fu YX, Yang X. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity. Sci Transl Med 2021; 13:13/578/eaba7308. [PMID: 33504651 DOI: 10.1126/scitranslmed.aba7308] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 08/24/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Although chimeric antigen receptor (CAR)-modified T cells have shown great success in the treatment of B cell malignancies, this approach has limited efficacy in patients with solid tumors. Various modifications in CAR structure have been explored to improve this efficacy, including the incorporation of two costimulatory domains. Because costimulatory signals are transduced together with T cell receptor signals during T cell activation, we engineered a type of CAR-T cells with a costimulatory signal that was activated independently from the tumor antigen to recapitulate physiological stimulation. We screened 12 costimulatory receptors to identify OX40 as the most effective CAR-T function enhancer. Our data indicated that these new CAR-T cells showed superior proliferation capability compared to current second-generation CAR-T cells. OX40 signaling reduced CAR-T cell apoptosis through up-regulation of genes encoding Bcl-2 family members and enhanced proliferation through increased activation of the NF-κB (nuclear factor κB), MAPK (mitogen-activated protein kinase), and PI3K-AKT (phosphoinositide 3-kinase to the kinase AKT) pathways. OX40 signaling not only enhanced the cytotoxicity of CAR-T cells but also reduced exhaustion markers, thereby maintaining their function in immunosuppressive tumor microenvironments. In mouse tumor models and in patients with metastatic lymphoma, these CAR-T cells exhibited robust amplification and antitumor activity. Our findings provide an alternative option for CAR-T optimization with the potential to overcome the challenge of treating solid tumors.
Collapse
Affiliation(s)
- Huihui Zhang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.,Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fanlin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.,Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.,Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiang Cao
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Xin Wang
- Shanghai Longyao Biotechnology Limited, Shanghai 201203, China
| | - Hai Cheng
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Kunming Qi
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Kailin Xu
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xuanming Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China. .,Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.,Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
9
|
Zhang J, Sun X, Zhong L, Shen B. IL-32 exacerbates adenoid hypertrophy via activating NLRP3-mediated cell pyroptosis, which promotes inflammation. Mol Med Rep 2021; 23:226. [PMID: 33495843 PMCID: PMC7851829 DOI: 10.3892/mmr.2021.11865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Adenoid hypertrophy (AH) is a common pediatric disease caused by inflammatory stimulation. The pro-inflammatory cytokine IL-32 has been reported to promote airway inflammation and also be involved in the pyroptosis pathway. However, whether IL-32 can contribute to AH by mediating pyroptosis remains to be elucidated. The present study aimed to investigate the role of IL-32 in AH and determine the potential underlying mechanisms. Adenoid tissues were collected from healthy children and children with AH, and the expression of IL-32, NACHT LRR and PYD domains-containing protein 3 (NLRP3) and IL-1β in normal and hypertrophic tissues were measured. Human nasal epithelial cells (HNEpCs) were exposed to a series of IL-32 concentrations. HNEpCs with or without IL-32 silencing were stimulated with lipopolysaccharide (LPS), and cell proliferation, cell apoptosis, gasdermin D (GSDMD) activation, production of inflammatory cytokines and the expression levels of proteins related to the potential mechanisms were evaluated by Cell Counting Kit-8, flow cytometry, immunofluorescence staining, ELISA and western blot assays, respectively. The results showed that IL-32, NLRP3 and IL-1β exhibited higher expression in adenoid tissues with AH compared with normal tissues. In HNEpC cells, treatment with IL-32 (2 and 10 ng/ml) promoted cell proliferation, while 50 ng/ml IL-32 inhibited cell proliferation at 12, 24 and 48 h post-treatment. IL-32 (2, 10 and 50 ng/ml) also resulted in differing degrees of apoptosis, GSDMD activation, release of IL-1β, IL-6 and TNF-α, and increased protein expression levels of NLRP3, cleaved-caspase-1, activated GSDMD, nucleotide-binding oligomerization domain-containing protein (NOD) 1/2 and Toll-like receptor (TLR)4 in a concentration-dependent manner. In addition, compared with the LPS group, IL-32 knockdown significantly inhibited LPS-induced enhancement of cell proliferation, cell apoptosis, GSDMD activation and production of inflammatory cytokines, and reversed the increased protein expression of NLRP3, cleaved-caspase-1, activated GSDMD, NOD1/2 and TLR4. In conclusion, IL-32 may play a role in the progression of AH via promoting inflammation, and the potential mechanism may involve the activation of NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Junmei Zhang
- Department of Otolaryngology, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| | - Xuyuan Sun
- Department of Otolaryngology, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| | - Lingling Zhong
- Department of Otolaryngology, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| | - Bei Shen
- Department of Otolaryngology, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| |
Collapse
|