1
|
Gao Z, Shao S, Xu Z, Nie J, Li C, Du C. IDO1 induced macrophage M1 polarization via ER stress-associated GRP78-XBP1 pathway to promote ulcerative colitis progression. Front Med (Lausanne) 2025; 12:1524952. [PMID: 40370742 PMCID: PMC12075526 DOI: 10.3389/fmed.2025.1524952] [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: 11/08/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disorder distinguished by alternating phases of remission and exacerbation. Restoring immune balance through the modulation of M1 macrophage polarization represents a potentially valuable therapeutic strategy for UC. Indoleamine 2,3-dioxygenase-1 (IDO1) has been shown to contribute to macrophage plasticity, but its role in the pathogenesis of UC via regulating M1 macrophage polarization has not been studied yet. For the clinical component, we analyzed IDO1 expression in UC using bioinformatics analysis of Gene Expression Omnibus (GEO) datasets and validated the result using western blotting of colonic tissues from new recruited UC patients. Colitis was induced in mice via dextran sulfate sodium (DSS) treatment and subsequently treated with oral administration of 1-methyl-DL-tryptophan (1-MT), an inhibitor of IDO1 pathway. The results indicated that IDO1 expression was significantly elevated in UC patients and correlated with M1 macrophage polarization observed in both human data and colitis mice. Furthermore, 1-MT markedly ameliorated DSS-induced weight loss, colonic shortening and disease severity via inhibiting IDO1 expression level, downregulating GRP78-XBP1 pathway and reducing M1 proportion. Notably, in vitro study revealed that overexpressing IDO1 in RAW264.7 cells induced macrophage M1 polarization with increased expression levels of GRP78 and XBP1, which was attenuated by 1-MT treatment. Additionally, the catalytic effect exerted by IDO1 overexpression on M1 polarization was neutralized by employing an inhibitor targeting the endoplasmic reticulum (ER) stress pathway. Thus, our findings suggest that IDO1 may promote UC progression by skewing macrophages towards M1 polarization through ER stress-associated GRP78-XBP1 pathway.
Collapse
Affiliation(s)
- Zijian Gao
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Shuai Shao
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Zhen Xu
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Jiao Nie
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chenglin Li
- Department of Oncology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chao Du
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Weihai Municipal Hospital, Shandong University, Weihai, China
| |
Collapse
|
2
|
Zheng T, Huang KY, Tang XD, Wang FY, Lv L. Endoplasmic reticulum stress in gut inflammation: Implications for ulcerative colitis and Crohn's disease. World J Gastroenterol 2025; 31:104671. [PMID: 40248056 PMCID: PMC12001174 DOI: 10.3748/wjg.v31.i13.104671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
Eukaryotic cells contain the endoplasmic reticulum (ER), a prevalent and intricate membranous structural system. During the development of inflammatory bowel disease (IBD), the stress on the ER and the start of the unfolded protein response are very important. Some chemicals, including 4μ8C, small molecule agonists of X-box binding protein 1, and ISRIB, work on the inositol-requiring enzyme 1, turn on transcription factor 6, and activate protein kinase RNA-like ER kinase pathways. This may help ease the symptoms of IBD. Researchers investigating the gut microbiota have discovered a correlation between ER stress and it. This suggests that changing the gut microbiota could help make new medicines for IBD. This study looks at how ER stress works and how it contributes to the emergence of IBD. It also talks about its possible clinical importance as a therapeutic target and looks into new ways to treat this condition.
Collapse
Affiliation(s)
- Ting Zheng
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Kai-Yue Huang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Xu-Dong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Feng-Yun Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lin Lv
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
3
|
Lin L, Liao Z, Li C. Exploring Ribosomal Genes as Potential Biomarkers of the Immune Microenvironment in Respiratory Syncytial Virus Infection. Biochem Genet 2025; 63:1839-1861. [PMID: 38630357 DOI: 10.1007/s10528-024-10778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/09/2024] [Indexed: 03/23/2025]
Abstract
Respiratory syncytial virus (RSV) is the most common pathogen causing acute lower respiratory tract infection in infants and children. Due to limited knowledge of the pathological and molecular mechanisms of immunodeficiency underlying RSV disease, there is currently a lack of an approved and effective RSV vaccine to combat RSV infections. This study aimed to identify genes associated with immune dysfunction using bioinformatics methods to gain insights into the role of dysregulated immune genes in RSV disease progression, and to predict potential therapeutic drugs by targeting dysregulated immune-related genes. 423 immune-related differential genes (DEIRGs) were filtered from the blood samples of 87 healthy individuals and 170 RSV patients. According to CIBERSORT analysis, the blood of RSV patients showed increased infiltration of various immune cells. Subsequently, ten immune-related hub genes were screened via Protein-Protein Interaction Networks. Six signature immune-related genes (RPS2, RPS5, RPS13, RPS14, RPS18, and RPS4X) as candidate characteristic genes for the diagnostic model were identified by Lasso regression. The AUC value of the ROC curve of the six signature genes was 0.884. This result, intriguingly, suggested that all six immune-related genes with a good internal validation effect were ribosome family genes. Finally, through molecular docking analyses targeting these differential immune genes, ADO and fluperlapine were found to have high stable binding to major proteins of important immune-related genes in nine drug-protein interactions. Overall, the present study screened immune-related genes that are dysregulated in the development of RSV disease to investigate the pathogenesis of RSV infection from the standpoint of immune disorders. Unexpectedly, bioinformatics analysis revealed that ribosome family genes may be involved in the immune dysregulation of RSV disease, and these genes as targets formed the basis for potential drug modification candidates in RSV disease.
Collapse
Affiliation(s)
- Lu Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zenghua Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chaoqian Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
Feng L, Xu L, Huang J, Wang Y, Xia Q, Meng J, Wang R, Liu K. Cardiotoxicity induced by xanthatin via activating apoptosis and ERS pathways in zebrafish. Drug Chem Toxicol 2025:1-12. [PMID: 40125983 DOI: 10.1080/01480545.2025.2481863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Xanthatin, a sesquiterpene lactone compound, isolated from Chinese herb, Xanthium strumarium L, has various activities, including anti-inflammatory, anti-tumor, anti-ulcer effects. However, it has been less studied in terms of its toxicity, especially the potential toxicity on heart. This study is mainly aimed to assess the cardiotoxicity of xanthatin in vivo using zebrafish larva and in vitro using cardiomyocytes H9C2. The cardiotoxicity in zebrafish was assessed by the pericardial edema, blood flow dynamics, SV-BA distance, and sub-intestinal vein. The apoptosis was determined by AO staining, the blood red cell reduction and distribution was detected by O-dianisidine staining, histopathological evaluations were detected by HE staining. The anti-proliferative and pro-apoptotic activities in H9C2 cells were assessed by EdU staining and Hoechst 33342/PI double staining. The in vivo results showed that xanthatin caused cardiac malformations and dysfunctions, including decreased heart rate, reduced red blood cell count, hemodynamics, stroke volume, increased SV-BA distance and sub-intestinal vein congestion. Furthermore, apoptosis occurred in the heart of the zebrafish after xanthatin exposure. Additionally, cat, Mn-sod, chop, perk, and hspa5 related to oxidative stress and ERS also changed by xanthatin. Apoptotic genes caspase3 and caspase9 were also increased. Moreover, the in vitro results showed that xanthatin had proapoptotic and antiproliferative effects. To sum up, these results suggest that xanthatin has cardiotoxicity and the oxidative stress, ERS and apoptosis pathways are involved in the cardiotoxicity induced by xanthatin. This finding will be helpful for the better understanding of the potential cardiotoxicity of xanthatin and the underlying mechanism.
Collapse
Affiliation(s)
- Lixin Feng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Liyan Xu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Jing Huang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Yuxin Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Jin Meng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, Jinan, PR China
| |
Collapse
|
5
|
Yan B, Liao P, Zhang W, Han Z, Wang C, Chen F, Lei P. Identification of Key Fatty Acid Metabolism-Related Genes in Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04857-x. [PMID: 40108056 DOI: 10.1007/s12035-025-04857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, and the role of fatty acid metabolism in its pathogenesis remains incompletely understood. Using AD transcriptome sequencing data from the GEO database, we initially screened for differentially expressed genes and applied Weighted Gene Correlation Network Analysis (WGCNA) to identify crucial gene modules. By intersecting these genes with fatty acid metabolism-related genes (FAMRGs), we obtained AD-related fatty acid metabolism genes (AD-FAMRGs). Subsequently, we conducted KEGG, GO, and Single-sample Gene Set Enrichment Analysis (ssGSEA). Furthermore, we employed three machine learning algorithms to determine the key AD-FAMRGs. Risk genes were thus identified, leading to the construction of a risk model which was subsequently validated through receiver operating characteristic (ROC) curve analysis. Additionally, protein docking studies were performed to assess interactions between key AD-FAMRGs and Tau as well as amyloid beta (Aβ) proteins. To explore potential therapeutic avenues, we searched the DrugBank database for agents targeting these AD-FAMRGs, followed by molecular docking and dynamics simulations. Our investigations highlighted three key AD-FAMRGs: DLD, ELOVL5, and HMGCS1. Functional enrichment analysis indicated their association with metabolism, oxidative stress, and AD pathogenesis. ZDOCK analysis further suggested their interactions with Tau and Aβ proteins, pointing to their possible involvement in AD's pathological processes. ROC analysis demonstrated the predictive accuracy of these AD-FAMRGs, with AUC values ranging from 0.764 to 0.876. Molecular docking and dynamic simulations confirmed the favorable binding of predicted therapeutic agents to these key AD-FAMRGs. Our findings suggest that fatty acid metabolism may be involved in AD pathogenesis, and DLD, ELOVL5, and HMGCS1 may serve as potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Bo Yan
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Pan Liao
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
- School of Medicine, Nankai University, Tianjin, 300192, China
| | - Wei Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Conglin Wang
- First Department of General Medicine, Tianjin First Central Hospital, Tianjin, 300190, China
| | - Fanglian Chen
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
- School of Medicine, Nankai University, Tianjin, 300192, China.
| |
Collapse
|
6
|
Du F, Wu X, He Y, Zhao S, Xia M, Zhang B, Tong J, Xia T. Identification of an Amino Acid Metabolism Reprogramming Signature for Predicting Prognosis, Immunotherapy Efficacy, and Drug Candidates in Colon Cancer. Appl Biochem Biotechnol 2025; 197:714-734. [PMID: 39222169 DOI: 10.1007/s12010-024-05049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Colon cancer ranked third among the most frequently diagnosed cancers worldwide. Amino acid metabolic reprogramming was related to the occurrence and development of colon cancer. We looked for the amino acid metabolism genes (AMGs) associated with amino acid metabolism from molecular signatures database as prognostic markers and constructed amino acid metabolism scoring model (AMS). According to AMS, the patients were divided into high AMS and low AMS groups, and the prognostic characteristics, molecular phenotypes, somatic cell mutation characteristics, immune cell infiltration characteristics, and immunotherapy effect of the two groups were systematically analyzed. Finally, the compounds targeting AMGs were also screened. We screen out 6 prognostic AMGs (P < 0.05) and construct an AMS model based on them. K-M curve indicated that OS in low AMS group was significantly higher than that in high group (P < 0.05), which were validated in multiple datasets. And different AMS groups had different molecular phenotypes, somatic cell mutation characteristics and immune cell infiltration characteristics. Low AMS group had a better effect for immunotherapy. In addition, we predicted potential therapeutic compounds that could bind to AMGs target proteins. AMS model can be used as a hierarchical tool to evaluate the prognosis, immune infiltration characteristics and immunotherapy response ability of colon cancer. And the compounds screened based on AMGs may become new anti-tumor drugs.
Collapse
Affiliation(s)
- Fenqi Du
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xiangxin Wu
- Ganzhou Cancer Hospital, Ganzhou, Jiangxi Province, People's Republic of China
| | - Yibo He
- Department of Acupuncture Massage & Rehabilitation, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong Province, People's Republic of China
| | - Shihui Zhao
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Mingyu Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Bomiao Zhang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinxue Tong
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| | - Tianyi Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
7
|
Kong D, Cheng H, Wang M. Novel pyroptosis-immune-related lncRNA signature exhibits a distinct immune cell infiltration landscape in breast cancer. Front Immunol 2025; 15:1522327. [PMID: 39906743 PMCID: PMC11790642 DOI: 10.3389/fimmu.2024.1522327] [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: 11/04/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction This study investigated pyroptosis- and immunity-related long non-coding RNAs (lncRNAs) to identify promising therapeutic targets for breast cancer (BC), and constructed lncRNA signatures to determine the prognosis and immunotherapy responses of BC patients. Methods Pearson's correlation coefficient was used to identify pyroptosis- and immune-related differentially expressed lncRNAs (DE-pyrolncRNAs and DE-ImmlncRNAs, respectively). The Cancer Genome Atlas dataset was allocated to training and testing subsets. Prognostic lncRNA signatures were derived based on the training subset using univariate Cox regression analysis and Least Absolute Shrinkage and Selection Operator methods. Stepwise Cox regression was used to refine these signatures and to select the optimal lncRNA signature. The median risk score of the training subset was applied as a threshold to divide patients into high-risk (HR) and low-risk (LR) groups. The Wilcoxon test was used to reveal differences in immune scores, cell types, functions, and checkpoint genes between these groups. Single-cell sequencing data from GSE176078 were used to validate the immune cell infiltration landscape of the identified lncRNA signatures. Results We identified a six-lncRNA pyroptosis-immune signature comprising MAPT.AS1, CTA.384, D8.34, RP11.561, I11.3, HID1.AS1, AC097713.3, and USP2.AS1. Patients in the HR group demonstrated inferior prognoses in the training, testing, and full datasets (P=3.622e-07, P=3.736e-03, and P=1.151e-08, respectively). Immune scores were significantly enhanced in the LR group, whereas tumor purity was elevated in the HR group. Fifty-eight immune scores showed significant differences between the groups (P<0.05). Immune function (APC coinhibition, CCR, and checkpoints) more significantly impaired in the HR group. Expression levels of 38 immune checkpoint genes, including KIR2DS4, KIR3DL2, CD40LG, KIR3DL1, and PDCD1, were significantly higher in the LR group. Conversely, the TDO2, PVR, and CD276 levels were elevated in the HR group. Single-cell sequencing data from GSE176078 showed sparse T cell, B cell, myeloid, and plasmablast clusters in the HR group, whereas the LR group displayed significant clustering of B cells, myeloids, and plasmablasts. Conclusion The six-lncRNA pyroptosis-immune signature effectively predicted BC prognosis and highlighted distinct immune cell infiltration patterns. This holds promise for evaluating immunotherapy responses and guiding therapeutic target identification in BC.
Collapse
Affiliation(s)
- Dedi Kong
- Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Breast and Thyroid Surgery, Jining No 1 People’s Hospital, Jining, Shandong, China
| | - Hongju Cheng
- Physiology Teaching and Research Office, Jining Medical College, Jining, Shandong, China
| | - Meihong Wang
- Hematology Department, Jining No 1 People’s Hospital, Jining, Shandong, China
| |
Collapse
|
8
|
Xie Y, Yang M, Huang J, Jiang Z. Identification and Characterization of Genes Associated with Intestinal Ischemia-Reperfusion Injury and Oxidative Stress: A Bioinformatics and Experimental Approach Integrating High-Throughput Sequencing, Machine Learning, and Validation. J Inflamm Res 2025; 18:701-722. [PMID: 39835298 PMCID: PMC11745141 DOI: 10.2147/jir.s500360] [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: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Purpose Intestinal ischemia-reperfusion injury (IIRI) occurs as a result of temporary blood flow interruption, leading to tissue damage upon reperfusion. Oxidative stress plays a critical role in this process, instigating inflammation and cell death. Identifying and characterizing genes associated with the oxidative stress response can offer valuable insights into potential therapeutic targets for managing IIRI. Patients and Methods The IIRI dataset was sourced from the NCBI Gene Expression Omnibus Database (GEO), while oxidative stress genes were obtained from the Genecards database. Following the acquisition of differentially expressed genes in IIRI, they were cross-linked with oxidative stress genes to yield IIRI oxidative stress related genes (IOSRGs). The least absolute shrinkage and selection operator, as well as the support vector machine with random forest algorithm, were utilized for machine learning. Subsequently, the PPI network was established, and the Degree and MNC algorithms of the Cytohuba plugin were integrated with the genes obtained through the machine learning algorithms to identify hub IOSRGs (HIOSRGs). A mouse IIRI model and ROC curve were employed to verify the accuracy of HIOSRGs. Finally, siRNA was utilized to suppress the expression of HDAC3 in Caco2 cells, and the changes in oxidative stress levels before and after hypoxia-reoxygenation in Caco2 cells were observed. Results A total of 277 OSRGs and 4 HIOSRGs were obtained. Concurrently, in vivo experimental results of IIRI in C57BL/6 mice, and the establishment of ROC curves, reflected the accuracy and specificity of HIOSRGs. The knockdown of HDAC3 in Caco2 cells resulted in increased oxidative stress levels before and after hypoxia-reoxygenation, underscoring the significant role of HDAC3 in IIRI. Conclusion This study elucidates the interplay between oxidative stress genes and IIRI, offering novel insights into the potential pathogenesis of IIRI and medical interventions for IIRI.
Collapse
Affiliation(s)
- Yongguo Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| | - Mingpu Yang
- General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| | - Juanjuan Huang
- Department of Microbiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Zongbin Jiang
- Department of Pain Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530007, People’s Republic of China
| |
Collapse
|
9
|
Lin L, Liao Z, Li Y, Pan S, Wu S, Sun QX, Li C. Transcriptomic analysis and validation study of key genes and the HIF‑1α/HO‑1 pathway associated with ferroptosis in neutrophilic asthma. Exp Ther Med 2024; 28:433. [PMID: 39347495 PMCID: PMC11425779 DOI: 10.3892/etm.2024.12722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 10/01/2024] Open
Abstract
Ferroptosis, as a unique form of cell death caused by iron overload and lipid peroxidation, is involved in the pathogenesis of various inflammatory diseases of the airways. Inhibition of ferroptosis has become a novel strategy for reducing airway epithelial cell death and improving airway inflammation. The aim of the present study was to analyze and validate the key genes and signaling pathways associated with ferroptosis by bioinformatic methods combined with experimental analyzes in vitro and in vivo to aid the diagnosis and treatment of neutrophilic asthma. A total of 1,639 differentially expressed genes (DEGs) were identified in the transcriptome dataset. After overlapping with ferroptosis-related genes, 11 differentially expressed ferroptosis-related genes (DE-FRGs) were obtained. A new diagnostic model was constructed by these DE-FRGs from the transcriptome dataset with those from the GSE108417 dataset. The receiver operating characteristic curve analysis indicated that the area under the curve had good diagnostic performance (>0.8). As a result, four key DE-FRGs (CXCL2, HMOX1, IL-6 and SLC7A5) and biological pathway [hypoxia-inducible factor 1 (HIF-1) signaling pathway] associated with ferroptosis in neutrophilic asthma were identified by the bioinformatics analysis combined with experimental validation. The upstream regulatory network of key DE-FRGs and target drugs were predicted and the molecular docking results from screened 37 potential therapeutic drugs revealed that the 13 small-molecule drugs exhibited a higher stable binding to the primary proteins of key DE-FRGs. The results suggested that four key DE-FRGs and the HIF-1α/heme oxygenase 1 pathway associated with ferroptosis have potential as novel markers or targets for the diagnosis or treatment of neutrophilic asthma.
Collapse
Affiliation(s)
- Lu Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zenghua Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yinghua Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Shitong Pan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Sihui Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qi-Xiang Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chaoqian Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
10
|
Chen T, Zhang Y, Zhang D, Zhou H. Immune-based subgroups uncover diverse tumor immunogenicity and implications for prognosis and precision therapy in acute myeloid leukemia. Front Immunol 2024; 15:1451486. [PMID: 39582863 PMCID: PMC11581856 DOI: 10.3389/fimmu.2024.1451486] [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: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although a considerable proportion of acute myeloid leukemia (AML) patients achieve remission through chemotherapy, relapse remains a recurring and significant event leading to treatment failure. This study aims to investigate the immune landscape in AML and its potential implications for prognosis and chemo-/immune-therapy. Methods Integrated analyses based on multiple sequencing datasets of AML were performed. Various algorithms estimated immune infiltration in AML samples. A subgroup prediction model was developed, and comprehensive bioinformatics and machine learning algorithms were applied to compare immune-based subgroups in relation to clinical features, mutational landscapes, immune characterizations, drug sensitivities, and cellular hierarchies at the single-cell level. Results Two immune-based AML subgroups, G1 and G2, were identified. G1 demonstrated higher immune infiltration, a more monocytic phenotype, increased proportions of monocytes/macrophages, and higher FLT3, DNMT3A, and NPM1 mutation frequencies. It was associated with a poorer prognosis, lower proportions of various immune cell types and a lower T cell infiltration score (TIS). AML T-cell-based immunotherapy target antigens, including CLEC12A, Folate receptor β, IL1RAP and TIM3, showed higher expression levels in G1, while CD117, CD244, CD96, WT and TERT exhibited higher expression levels in G2. G1 samples demonstrated higher sensitivity to elesclomol and panobinostat but increased resistance to venetoclax compared to G2 samples. Moreover, we observed a positive correlation between sample immune infiltration and sample resistance to elesclomol and panobinostat, whereas a negative correlation was found with venetoclax resistance. Conclusion Our study enriches the current AML risk stratification and provides guidance for precision medicine in AML.
Collapse
Affiliation(s)
| | | | | | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Jiang W, Zhao Y, Han M, Xu J, Chen K, Liang Y, Yin J, Hu J, Shen Y. N4BP3 facilitates NOD2-MAPK/NF-κB pathway in inflammatory bowel disease through mediating K63-linked RIPK2 ubiquitination. Cell Death Discov 2024; 10:440. [PMID: 39420190 PMCID: PMC11487068 DOI: 10.1038/s41420-024-02213-x] [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: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
The NOD2 signaling pathway, which plays an important role in the mechanisms of inflammatory bowel disease (IBD) development, has been closely associated with ubiquitination. It was revealed in this study that NOD2 receptor activation could obviously affect the expression of 19 ubiquitination-related genes, with N4BP3 being the most prominently expressed and upregulated. In addition, N4BP3 knockdown was found to reduce the mRNA levels of MDP-induced inflammatory factors, while N4BP3 overexpression elevated their mRNA levels as well as the levels of phospho-ERK1/2, phospho-JNK, phospho-P38 and phospho-NF-κB P65 proteins. Immunoprecipitation tests showed that N4BP3 could pull down RIPK2 and promote its K63-linked ubiquitination. In human tissue specimen assays and mouse experiments, we found that the expression of N4BP3 was significantly elevated in Crohn's disease (CD) patients and IBD mice, and N4BP3 knockdown reduced the dextran sulfate sodium-induced pathological score and the expression of inflammatory factors in the mouse colon tissue. In conclusion, N4BP3 is able to interact with RIPK2 and promote its K63-linked ubiquitination, to further promote the NOD2-MAPK/NF-κB pathway, thereby increasing promoting the release of inflammation factors and the degree of IBD inflammation.
Collapse
Affiliation(s)
- Wang Jiang
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China
| | - Yan Zhao
- Department of Pathology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China
| | - Min Han
- Department of Cardiovascular Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China
| | - Jiafan Xu
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China
| | - Kun Chen
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Yi Liang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, 35 Jiefang Road, Hengyang, 421000, China
| | - Jie Yin
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China
| | - Jinyue Hu
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Yueming Shen
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha, 410000, China.
| |
Collapse
|
12
|
Qi XL, Luo GQ, Tuersun A, Chen M, Wu GB, Zheng L, Li HJ, Lou XL, Luo M. Construction of an endoplasmic reticulum stress and cuproptosis -related lncRNAs signature in chemosensitivity in hepatocellular carcinoma by comprehensive bioinformatics analysis. Heliyon 2024; 10:e38342. [PMID: 39398070 PMCID: PMC11471205 DOI: 10.1016/j.heliyon.2024.e38342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) and cuproptosis have remarkable effects on hepatocellular carcinoma (HCC) leading to a poor prognosis. The current study aimed to explore credible signature for predicting the prognosis of HCC based on ERS and cuproptosis-related lncRNAs. In our study, clinical and transcriptomic profiles of HCC patients were obtained from the Cancer Genome Atlas (TCGA) database. An ERS and cuproptosis-related lncRNA prognostic signature, including NRAV, SNHG3, LINC00839 and AC004687.1, was determined by correlation tests, Cox regression analysis, least absolute shrinkage, and selection operator (LASSO) methods. Survival and predictive value were evaluated using Kaplan-Meier and receiver operating characteristic (ROC) curves, while calibration and nomograms curves were developed. Besides the enrichment analyses for ERS and cuproptosis-related lncRNAs, mutational status and immune status were assessed with TMB and ESTIMATE. Additionally, consensus cluster analysis was employed to compare cancer subtype differences, while drug sensitivity and immunologic efficacy were evaluated for further exploration. qRT-PCR and CCK-8 were utilized to verify the alteration of the prognostic lncRNAs expression and proliferation in vitro. High-risk groups exhibited poorer prognosis. The signature exhibited robust predictive value as an independent prognostic indicator and showed significant correlation with clinicopathological features. In the enriched analysis, biological membrane pathways were enriched. Low-risk patients had lower TMB and higher immune status. A cluster analysis revealed that cluster 2 had the best clinical immunological efficacy and most active immune function. In brief, our constructed signature with ERS and cuproptosis-related lncRNAs was associated survival outcomes of HCC, and can be used to predict the clinical classification and curative effect.
Collapse
Affiliation(s)
- Xiao-Liang Qi
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gu-Qing Luo
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Abudukadier Tuersun
- Department of General Surgery, Kashgar Prefecture Second People's Hospital of Xinjiang Uygur Autonomous Regions, Kashgar, Xinjiang, China
| | - Min Chen
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-Bo Wu
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Jie Li
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lou Lou
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Wang J, Gu J, Ma F, Wei Y, Wang P, Yang S, Yan X, Xiao Y, Xing K, Lou A, Zheng L, Cao T, Zhu D, Li J, Zhang L, Li Y, Chen T. Melatonin Induces Analgesic Effects through MT 2 Receptor-Mediated Neuroimmune Modulation in the Mice Anterior Cingulate Cortex. RESEARCH (WASHINGTON, D.C.) 2024; 7:0493. [PMID: 39381792 PMCID: PMC11458856 DOI: 10.34133/research.0493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024]
Abstract
Neuropathic pain (NP) represents a considerable clinical challenge, profoundly impacting patients' quality of life. Presently, pharmacotherapy serves as a primary approach for NP alleviation, yet its efficacy often remains suboptimal. Melatonin (MLT), a biologically active compound secreted by the pineal gland, has long been associated with promoting and maintaining sleep. Although recent studies suggest analgesic effects of MLT, the underlying mechanism remains largely unknown, particularly its impact on the cortex. In this study, we induced an NP model in mice through spared nerve injury (SNI) and observed a considerable, dose-dependent alleviation in NP symptoms following intraperitoneal or anterior cingulate cortex (ACC) administration of MLT. Our findings further indicated that the NP management of MLT is selectively mediated by MLT-related receptor 2 (MT2R), rather than MT1R, on neurons and microglia within the ACC. Transcriptome sequencing, complemented by bioinformatics analysis, implicated MLT in the modulation of Gα(i) and immune-inflammatory signals. Specifically, MLT inhibited the excitability level of pyramidal cells in the ACC by activating the Gα(i) signaling pathway. Simultaneously, MLT attenuated M1 polarization and promoted M2 polarization of microglia, thereby mitigating the inflammatory response and type II interferon response within the ACC. These findings unveil a hitherto unrecognized molecular mechanism: an MLT-mediated neuroimmune modulation pathway in the ACC mediated by MT2R. This elucidation sheds light on the regulatory character of MLT in chronic nociceptive pain conditions, offering a prospective therapeutic strategy for NP management.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- Department of Neurosurgery, Tangdu Hospital,
Fourth Military Medical University, Xi’an 710038, China
| | - Junxiang Gu
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- Department of Neurosurgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Fujuan Ma
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- School of Medicine, Northwest University, Xi’an 710069, China
| | - Yi Wei
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- School of Medicine, Northwest University, Xi’an 710069, China
| | - Pan Wang
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Shanming Yang
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- School of Medicine, Northwest University, Xi’an 710069, China
| | - Xianxia Yan
- Department of Neurosurgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yifan Xiao
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
- School of Medicine, Northwest University, Xi’an 710069, China
| | - Keke Xing
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Anxin Lou
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Liru Zheng
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Tingting Cao
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Dayu Zhu
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Jinlian Li
- School of Medicine, Northwest University, Xi’an 710069, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yunqing Li
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| | - Tao Chen
- Department of Anatomy and K.K. Leung Brain Research Centre,
Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
14
|
Wang Y, Liang C, Liu X, Cheng SQ. A novel tumor-derived exosomal gene signature predicts prognosis in patients with pancreatic cancer. Transl Cancer Res 2024; 13:4324-4340. [PMID: 39262474 PMCID: PMC11384923 DOI: 10.21037/tcr-23-2354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 06/02/2024] [Indexed: 09/13/2024]
Abstract
Background Pancreatic cancer is a devastating disease with poor prognosis. Accumulating evidence has shown that exosomes and their cargo have the potential to mediate the progression of pancreatic cancer and are promising non-invasive biomarkers for the early detection and prognosis of this malignancy. This study aimed to construct a gene signature from tumor-derived exosomes with high prognostic capacity for pancreatic cancer using bioinformatics analysis. Methods Gene expression data of solid pancreatic cancer tumors and blood-derived exosome tissues were downloaded from The Cancer Genome Atlas (TCGA) and ExoRBase 2.0. Overlapping differentially expressed genes (DEGs) in the two datasets were analyzed, followed by functional enrichment analysis, protein-protein interaction networks, and weighted gene co-expression network analysis (WGCNA). Using the least absolute shrinkage and selection operator (LASSO) regression of prognosis-related exosomal DEGs, a tumor-derived exosomal gene signature was constructed based on the TCGA dataset, which was validated by an external validation dataset, GSE62452. The prognostic power of this gene signature and its relationship with various pathways and immune cell infiltration were analyzed. Results A total of 166 overlapping DEGs were identified from the two datasets, which were markedly enriched in functions and pathways associated with the cell cycle. Two key modules and corresponding 70 exosomal DEGs were identified using WGCNA. Using LASSO Cox regression of prognosis-related exosomal DEGs, a tumor-derived exosomal gene signature was built using six exosomal DEGs (ARNTL2, FHL2, KRT19, MMP1, CDCA5, and KIF11), which showed high predictive performance for prognosis in both the training and validation datasets. In addition, this prognostic signature is associated with the differential activation of several pathways, such as the cell cycle, and the infiltration of some immune cells, such as Tregs and CD8+ T cells. Conclusions This study established a six-exosome gene signature that can accurately predict the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Yang Wang
- Department of Hepatopancreatobiliary Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Liang
- Department of Hepatopancreatobiliary Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinbo Liu
- Department of Hepatopancreatobiliary Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
15
|
Ma X, Hu Q, Jiang T, Chen Y, Zhang W, Gao P, Zeng J, Efferth T. Dehydroevodiamine Alleviates Ulcerative Colitis by Inhibiting the PI3K/AKT/NF-κB Signaling Pathway via Targeting AKT1 and Regulating Gut Microbes and Serum Metabolism. Molecules 2024; 29:4031. [PMID: 39274878 PMCID: PMC11397320 DOI: 10.3390/molecules29174031] [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: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Ulcerative colitis (UC) is a typical inflammatory bowel disease (IBD), impairing the quality of life of patients. Dehydroevodiamine (DHE) is an active alkaloid isolated from Tetradium ruticarpum that exerts significant anti-inflammatory effects in gastrointestinal diseases. However, the effect and mechanisms of DHE on UC remain unclear. We performed a DSS-induced experimental UC rat model to reveal the efficacy and potential mechanisms of DHE on UC. HE and AB-PAS staining were used for the evaluation of pathologies, and 16S rRNA sequencing was used to detect changes in gut microbes. Metabolomics was used to detect changes in serum metabolites. Network pharmacology and transcriptomics were conducted to reveal the underlying mechanisms of DHE for UC. HuProt proteome microarrays, molecular docking, and SPR were used to reveal the targets of action of DHE. WB, RT-qPCR, and IHC were used to assess the action effects of DHE. DHE demonstrated significant alleviation of DSS-induced colitis symptoms in rats by suppressing inflammatory and oxidative stress responses, amending colonic barrier injury, and inhibiting apoptosis. In terms of gut microbial modulation, DHE decreased the abundance of Allobaculum, Clostridium, Escherichia, Enterococcus, and Barnesiella and increased the abundance of Lactobacillus, Bifidobacterium, and SMB5. Moreover, metabolomics suggested that the regulation of DHE in DSS-induced UC rats mainly involved aminoacyl-tRNA biosynthesis, vitamin B6 metabolism, phenylalanine, tyrosine, and so on. Mechanically, DHE alleviated UC in rats by targeting AKT1, thereby inhibiting the PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Tao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Pan Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Q.H.); (T.J.); (Y.C.); (W.Z.); (P.G.)
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
16
|
Dong S, Zhang Y, Ye L, Cao Q. Identification of a Novel Activated NK-Associated Gene Score Associated with Diagnosis and Biological Therapy Response in Ulcerative Colitis. Digestion 2024; 106:1-22. [PMID: 39182484 PMCID: PMC11825133 DOI: 10.1159/000540939] [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: 02/28/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Natural killer (NK) cells are associated with the pathogenesis of ulcerative colitis (UC); however, their precise contributions remain unclear. The present study aimed to investigate the diagnostic value of the activated NK-associated gene (ANAG) score in UC and evaluate its predictive value in response to biological therapy. METHODS Bulk RNA-seq and scRNA-seq datasets were obtained from the Gene Expression Omnibus (GEO) and Single Cell Portal (SCP) databases. In the bulk RNA-seq, differentially expressed genes (DEGs) were screened by the "Batch correction" and "Robust rank aggregation" (RRA) methods. The immune infiltration landscape was estimated using single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT. DEGs that correlated with activated NK cells were identified as activated NK-associated genes (ANAGs). Protein-protein interaction (PPI) analysis and least absolute shrinkage and selection operator (LASSO) regression were used to screen key ANAGs and establish an ANAG score. The expression levels of the four key ANAGs were validated in human samples by real-time quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence. The potential therapeutic drugs for UC were identified using the DSigDB database. Through scRNA-seq data analysis, the cell scores based on the ANAGs were calculated by "AddModuleScore" and "AUCell." RESULTS Immune infiltration analysis revealed a higher abundance of activated NK cells in noninflamed UC tissues (ssGSEA, p < 0.001; CIBERSORT, p < 0.01). Fifty-four DEGs correlated with activated NK cells were identified as ANAGs. The ANAG score was established using four key ANAGs (SELP, TIMP1, MMP7, and ABCG2). The ANAG scores were significantly higher in inflamed tissues (p < 0.001) and in biological therapy nonresponders (NR) tissues before treatment (golimumab, p < 0.05; ustekinumab, p < 0.001). The ANAG score demonstrated an excellent diagnostic value (AUC = 0.979). Patients with higher ANAG scores before treatment were more likely to experience a lack of response to golimumab or ustekinumab (golimumab, p < 0.05; ustekinumab, p < 0.001). CONCLUSION This study established a novel ANAG score with the ability to precisely diagnose UC and distinguish the efficacy of biological treatment. INTRODUCTION Natural killer (NK) cells are associated with the pathogenesis of ulcerative colitis (UC); however, their precise contributions remain unclear. The present study aimed to investigate the diagnostic value of the activated NK-associated gene (ANAG) score in UC and evaluate its predictive value in response to biological therapy. METHODS Bulk RNA-seq and scRNA-seq datasets were obtained from the Gene Expression Omnibus (GEO) and Single Cell Portal (SCP) databases. In the bulk RNA-seq, differentially expressed genes (DEGs) were screened by the "Batch correction" and "Robust rank aggregation" (RRA) methods. The immune infiltration landscape was estimated using single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT. DEGs that correlated with activated NK cells were identified as activated NK-associated genes (ANAGs). Protein-protein interaction (PPI) analysis and least absolute shrinkage and selection operator (LASSO) regression were used to screen key ANAGs and establish an ANAG score. The expression levels of the four key ANAGs were validated in human samples by real-time quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence. The potential therapeutic drugs for UC were identified using the DSigDB database. Through scRNA-seq data analysis, the cell scores based on the ANAGs were calculated by "AddModuleScore" and "AUCell." RESULTS Immune infiltration analysis revealed a higher abundance of activated NK cells in noninflamed UC tissues (ssGSEA, p < 0.001; CIBERSORT, p < 0.01). Fifty-four DEGs correlated with activated NK cells were identified as ANAGs. The ANAG score was established using four key ANAGs (SELP, TIMP1, MMP7, and ABCG2). The ANAG scores were significantly higher in inflamed tissues (p < 0.001) and in biological therapy nonresponders (NR) tissues before treatment (golimumab, p < 0.05; ustekinumab, p < 0.001). The ANAG score demonstrated an excellent diagnostic value (AUC = 0.979). Patients with higher ANAG scores before treatment were more likely to experience a lack of response to golimumab or ustekinumab (golimumab, p < 0.05; ustekinumab, p < 0.001). CONCLUSION This study established a novel ANAG score with the ability to precisely diagnose UC and distinguish the efficacy of biological treatment.
Collapse
Affiliation(s)
- Siyuan Dong
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lingna Ye
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Feng B, Zhang Y, Qiao L, Tang Q, Zhang Z, Zhang S, Qiu J, Zhou X, Huang C, Liang Y. Evaluating the significance of ECSCR in the diagnosis of ulcerative colitis and drug efficacy assessment. Front Immunol 2024; 15:1426875. [PMID: 39170615 PMCID: PMC11335526 DOI: 10.3389/fimmu.2024.1426875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Background The main challenge in diagnosing and treating ulcerative colitis (UC) has prompted this study to discover useful biomarkers and understand the underlying molecular mechanisms. Methods In this study, transcriptomic data from intestinal mucosal biopsies underwent Robust Rank Aggregation (RRA) analysis to identify differential genes. These genes intersected with UC key genes from Weighted Gene Co-expression Network Analysis (WGCNA). Machine learning identified UC signature genes, aiding predictive model development. Validation involved external data for diagnostic, progression, and drug efficacy assessment, along with ELISA testing of clinical serum samples. Results RRA integrative analysis identified 251 up-regulated and 211 down-regulated DEGs intersecting with key UC genes in WGCNA, yielding 212 key DEGs. Subsequently, five UC signature biomarkers were identified by machine learning based on the key DEGs-THY1, SLC6A14, ECSCR, FAP, and GPR109B. A logistic regression model incorporating these five genes was constructed. The AUC values for the model set and internal validation data were 0.995 and 0.959, respectively. Mechanistically, activation of the IL-17 signaling pathway, TNF signaling pathway, PI3K-Akt signaling pathway in UC was indicated by KEGG and GSVA analyses, which were positively correlated with the signature biomarkers. Additionally, the expression of the signature biomarkers was strongly correlated with various UC types and drug efficacy in different datasets. Notably, ECSCR was found to be upregulated in UC serum and exhibited a positive correlation with neutrophil levels in UC patients. Conclusions THY1, SLC6A14, ECSCR, FAP, and GPR109B can serve as potential biomarkers of UC and are closely related to signaling pathways associated with UC progression. The discovery of these markers provides valuable information for understanding the molecular mechanisms of UC.
Collapse
Affiliation(s)
- Bin Feng
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanqiu Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui, China
| | - Longwei Qiao
- Center for Reproduction and Genetics, School of Gusu, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Qingqin Tang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sheng Zhang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Qiu
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xianping Zhou
- Department of Laboratory, Bozhou Hospital Affiliated to Anhui Medical University, Bozhou, Anhui, China
- Department of Laboratory, Anhui Medical University, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chao Huang
- Center for Reproduction and Genetics, School of Gusu, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yuting Liang
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
18
|
Xu Y, Wang S, Ye Z, Zhang H. Identifying hub genes in response to ustekinumab and the impact of ustekinumab treatment on fibrosis in Crohn's disease. Front Immunol 2024; 15:1401733. [PMID: 38840917 PMCID: PMC11150586 DOI: 10.3389/fimmu.2024.1401733] [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: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Crohn's disease (CD) is a chronic inflammatory disease. Approximately 50% of patients with CD progressed from inflammation to fibrosis. Currently, there are no effective drugs for treating intestinal fibrosis. Biologic therapies for CD such as ustekinumab have benefited patients; however, up to 30% of patients with CD have no response to initial treatment, and the effect of ustekinumab on intestinal fibrosis is still uncertain. Therefore, it is of great significance to explore the predictive factors of ustekinumab treatment response and the effect of ustekinumab on intestinal fibrosis. Materials and methods Public datasets-GSE207465 (blood samples) and GSE112366 and GSE207022 (intestinal samples)-were downloaded and analyzed individually (unmerged) based on the treatment response. Differentially expressed genes (DEGs) were identified by the "limma" R package and changes in immune cell infiltration were determined by the "CIBERSORT" R package in both blood and intestinal samples at week 0 (before treatment). To find predictive factors of ustekinumab treatment response, the weighted gene co-expression network analysis (WGCNA) R package was used to identify hub genes in GSE112366. Hub genes were then verified in GSE207022, and a prediction model was built by random forest algorithm. Furthermore, fibrosis-related gene changes were analyzed in ileal samples before and after treatment with ustekinumab. Results (1) Our analysis found that MUC1, DUOX2, LCN2, and PDZK1IP1 were hub genes in GSE112366. GSE207022 revealed that MUC1 (AUC:0.761), LCN2 (AUC:0.79), and PDZK1IP1 (AUC:0.731) were also lower in the response group. Moreover, the random forest model was shown to have strong predictive capabilities in identifying responders (AUC = 0.875). To explore the relationship between intestinal tissue and blood, we found that ITGA4 had lower expression in the intestinal and blood samples of responders. The expression of IL18R1 is also lower in responders' intestines. IL18, the ligand of IL18R1, was also found to have lower expression in the blood samples from responders vs. non-responders. (2) GSE112366 revealed a significant decrease in fibrosis-related module genes (COL4A1, TUBB6, IFITM2, SERPING1, DRAM1, NAMPT, MMP1, ZEB2, ICAM1, PFKFB3, and ACTA2) and fibrosis-related pathways (ECM-receptor interaction and PI3K-AKT pathways) after ustekinumab treatment. Conclusion MUC1, LCN2, and PDZK1IP1 were identified as hub genes in intestinal samples, with lower expression indicating a positive prediction of ustekinumab treatment response. Moreover, ITGA4 and IL18/IL18R1 may be involved in the treatment response in blood and intestinal samples. Finally, ustekinumab treatment was shown to significantly alter fibrotic genes and pathways.
Collapse
Affiliation(s)
| | | | | | - Hongjie Zhang
- Department of Gastroenterology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
周 伟, 聂 军, 胡 佳, 蒋 艺, 张 大. [Differential expressions of endoplasmic reticulum stress-associated genes in aortic dissection and their correlation with immune cell infiltration]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:859-866. [PMID: 38862443 PMCID: PMC11166729 DOI: 10.12122/j.issn.1673-4254.2024.05.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To explore differentially expressed endoplasmic reticulum stress-associated genes (ERSAGs) in aortic dissection (AD) and their correlations with immune cell infiltration to identify new therapeutic targets for AD. METHODS Two AD mRNA expression datasets (GSE190635 and GSE98770) were downloaded from GEO database for analysis of differentially expressed genes between the aorta of AD patients and normal aorta using R software. ERSAGs dataset was downloaded from GeneCards website, and GeneMANIA database was used to analyze the protein-protein interaction network of the differentially expressed ERSAGs and the proteins interacting with these genes. Based on GSE98770 dataset we analyzed the distributions of 22 immune cells within the aortic wall of AD patients using CIBERSORT package of R software. Surgical aortic wall specimens were obtained from 10 AD patients and 10 non-AD patients for detecting AGER mRNA expression using qRTPCR, and the upstream transcriptional factors, miRNAs, and chemicals targeting AGER were analyzed using the TRRUST database and NetworkAnalyst database. RESULTS Bioinformatic analysis suggested significant differential expression of AGER in AD, which interacted with 20 proteins involved in pattern recognition receptor signaling pathway, positive regulation of DNA-binding transcription factor activity, myeloid leukocyte migration, leukocyte migration, and regulation of the I-κB kinase/NF-κB signaling. In AD, AGER expression level was positively correlated with Treg cell abundance (r=0.59, P < 0.05). The results of qRT-PCR demonstrated significantly lower expression of AGER mRNA in AD than in non-AD patients (1.00±0.30 vs 1.76±0.68, P < 0.05). ROC curve analysis showed that at the cut-off value of 1.335, AGER had an AUC of 0.86 (95% CI: 0.67-1.00, P= 0.0073) for predicting AD. Three transcriptional factors, 3 miRNAs, and 27 chemicals were predicted in the AGER regulatory network. CONCLUSION AGER is lowly expressed in the aorta of AD patients and may influence the occurrence of AD through Treg cells.
Collapse
|
20
|
Zhou W, Nie J, Zhang D. Comprehensive Analysis of Key Endoplasmic Reticulum Stress-Related Genes and Immune Infiltrates in Stanford Type A Aortic Dissection. Anatol J Cardiol 2024; 28:236-244. [PMID: 38445624 PMCID: PMC11059230 DOI: 10.14744/anatoljcardiol.2024.4251] [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: 01/09/2024] [Accepted: 02/07/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Type A aortic dissection is a fatal disease. However, the role of endoplasmic reticulum stress-related genes (ERSRGs) in type A aortic dissection has not yet been fully clarified. METHODS Differentially expressed genes in the aorta of type A aortic dissection patients were analyzed based on the GSE52093 database. The ERSRGs were downloaded from the GeneCards website. Functional enrichment analysis and protein-protein interaction analysis were performed on the acquired differentially expressed ERSRGs. The mRNA expression of the 5 top key differentially expressed ERSRGs was further explored in GSE153434 and clinical samples. Immune infiltration correlation analysis was performed on the validated key genes. Finally, we constructed regulatory networks of transcription factors, miRNAs, and chemicals. RESULTS Twelve differentially expressed ERSRGs were identified, of which 8 genes were downregulated and 4 genes were upregulated. GeneMANIA was adopted to analyze these genes and their interacting proteins, and the results showed that the main function was calcium ion transport. Four key genes, ACTC1, CASQ2, SPP1, and REEP1, were validated in GSE153434 and clinical samples. The area under the ROC curve values for ACTC1, CASQ2, SPP1, and REEP1 were 0.92, 0.96, 0.89, and 1.00, respectively. ACTC1 and REEP1 correlated with multiple immune cells. Many transcription factors, microRNAs, and chemicals were identified with the potential to regulate these 4 key genes. CONCLUSION In this study, we identified 12 differentially expressed ERSRGs by analyzing the Gene Expression Omnibus database. Four key genes may influence the development of type A aortic dissection by regulating endoplasmic reticulum stress. These results expand our understanding of type A aortic dissection, and the 4 key genes are expected to be diagnostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jun Nie
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Dafa Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
21
|
Li D, Zhang J, Wang L, Yan X, Zi J, Du X, Yu L, Jiang Y. Identification of Pyroptosis-Related Genes Regulating the Progression of Chronic Rhinosinusitis with Nasal Polyps. Int Arch Allergy Immunol 2024; 185:411-424. [PMID: 38402873 DOI: 10.1159/000536371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024] Open
Abstract
INTRODUCTION Chronic rhinosinusitis with nasal polyps (CRSwNP) is an immunologic disease, and pyroptosis, an inflammation-based cellular death, strictly modulates CRSwNP pathology, whereas the pyroptosis genes and mechanisms involved in CRSwNP remain unclear. Herein, we explored disease biomarkers and potential therapeutic targets for pyroptosis and immune regulation in CRSwNP using bioinformatics analysis and tissue-based verification. METHODS We retrieved the transcriptional profiles of the high-throughput dataset GSE136825 from the Gene Expression Omnibus database, as well as 170 pyroptosis-related gene expressions from GeneCards. Using R, we identified differentially expressed pyroptosis-related genes and examined the potential biological functions of the aforementioned genes using Gene Ontology, Kyoto Encyclopedia of the Genome pathway, immune infiltration, and protein-protein interaction (PPI) network analyses, thereby generating a list of hub genes. The hub genes were, in turn, verified using real-time quantitative polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), and Western blotting (WB). Ultimately, using the StarBase and miRTarBase databases, we estimated the targeting microRNAs and long chain non-coding RNAs. RESULTS We demonstrated that the identified pyroptosis-related genes primarily modulated bacterial defense activities, as well as inflammasome immune response and assembly. Moreover, they were intricately linked to neutrophil and macrophage infiltration. Furthermore, we validated the tissue contents of hub genes AIM2, NLPR6, and CASP5 and examined potential associations with clinical variables. We also developed a competitive endogenous RNA (ceRNA) modulatory axis to examine possible underlying molecular mechanisms. CONCLUSION We found AIM2, CASP5, and NLRP6, three hub genes for pyroptosis in chronic rhinosinusitis with nasal polyps, by biological analysis, experimental validation, and clinical variable validation.
Collapse
Affiliation(s)
- Danyang Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China,
| | - Jisheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Yan
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiajia Zi
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyun Du
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Longgang Yu
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Hua R, Qiao G, Chen G, Sun Z, Jia H, Li P, Zhang B, Qi F. Single-Cell RNA-Sequencing Analysis of Colonic Lamina Propria Immune Cells Reveals the Key Immune Cell-Related Genes of Ulcerative Colitis. J Inflamm Res 2023; 16:5171-5188. [PMID: 38026254 PMCID: PMC10649030 DOI: 10.2147/jir.s440076] [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: 09/13/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Background Ulcerative colitis (UC) is a severe threat to humans worldwide. Single-cell RNA sequencing (scRNA-seq) can be used to screen gene expression patterns of each cell in the intestine, provide new insights into the potential mechanism of UC, and analyze the development of immune cell changes. These findings can provide new ideas for the diagnosis and treatment of intestinal diseases. In this study, bioinformatics analysis combined with experiments applied in dextran sulfate sodium (DSS)-induced colitis mice was used to explore new diagnostic genes for UC and their potential relationship with immune cells. Methods We downloaded microarray datasets (GSE75214, GSE87473, GSE92415) from the Gene Expression Omnibus and used these datasets to screen differentially expressed genes (DEGs) and conduct Weighted Gene Co-expression Network Analysis (WGCNA) after quality control. The hub genes were screened, and ROC curves were drawn to verify the reliability of the results in both training set (GSE75214, GSE87473, GSE92415) and validation cohort (GSE87466). Also, we explored the relation of diagnostic genes and immune cells by CIBERSORT algorithm and single-cell analysis. Finally, the expression of hub genes and their relation with immune cells were verified in DSS-induced colitis mice. Results Diagnostic genes (ANXA5, MMP7, NR1H4, CYP3A4, ABCG2) were identified. In addition, we found these five genes firmly related to immune infiltration. The DSS-induced colitis mice confirm that the expression of ANXA5 mainly increased in the intestinal macrophages and had a strong negative correlation with M2 macrophages, which indicated its possible influence on the polarization of macrophages in UC patients. Conclusion We identified ANXA5, MMP7, NR1H4, CYP3A4, and ABCG2 as diagnostic genes of UC that are closely related to immune infiltration and ANXA5 maintains a negative correlation with M2 macrophages which indicated its possible influence on the polarization of macrophage in UC patients.
Collapse
Affiliation(s)
- Run Hua
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Gangjie Qiao
- Department of Colorectal Surgery, Shanxi Provincial People’s Hospital, Shanxi, People’s Republic of China
| | - Guoshan Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Zhaonan Sun
- General Surgery Department, Tianjin Medical University General Hospital Airport Hospital, Tianjin, People’s Republic of China
| | - Haowen Jia
- General Surgery Department, Tianjin Medical University General Hospital Airport Hospital, Tianjin, People’s Republic of China
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Baotong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| |
Collapse
|