1
|
Joshi S, Garlapati C, Nguyen T, Sharma S, Chandrashekar DS, Bhattarai S, Varambally S, Krishnamurti U, Li X, Aneja R. C/EBPβ increases tumor aggressiveness by enhancing KIFC1 expression in androgen receptor negative triple negative breast cancer. Cell Commun Signal 2025; 23:255. [PMID: 40448099 DOI: 10.1186/s12964-025-02243-7] [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: 03/02/2025] [Accepted: 05/09/2025] [Indexed: 06/02/2025] Open
Abstract
Quadruple-negative breast cancers (also known as AR-triple negative (TN) BC) lack the expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and androgen receptor (AR). AR-TNBC exhibits aggressive characteristics and a poor prognosis. Because of the lack of expression of therapeutic targets, limited therapeutic options exist for patients with AR-TNBC. Hence, new therapeutic targets and risk-predictive biomarkers are required for patients with AR-TNBC. In this study, we investigated the role of kinesin-like protein 1 (KIFC1) in AR-TNBC. We found that C/EBPβ binds to the KIFC1 promoter and induces its expression in the AR-TNBC cells. Notably, AR status was negatively correlated with KIFC1 levels. We also found that AR transcriptionally repressed the transcription factor C/EBPβ, which regulates the expression of KIFC1. The lack of AR expression in AR-TNBC led to C/EBPβ upregulation, thereby enhancing KIFC1 expression. Moreover, upregulation of KIFC1 in AR-TNBC increased cancer cell proliferation and promoted epithelial-mesenchymal transition (EMT), contributing to the aggressive characteristics of AR-TNBC. Inhibiting KIFC1 using the small molecule inhibitor CW069 significantly reduced tumor volume in mice bearing AR-TNBC xenografts, but not in those with triple-negative breast tumors. These data suggest that upregulation of C/EBPβ and KIFC1 contributes to the aggressive characteristics and poor prognosis of AR-TNBC, providing strong evidence that targeting KIFC1 using kinesin inhibitors could be a viable therapeutic approach for patients with AR-TNBC.
Collapse
Affiliation(s)
- Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Alkermes Inc, Waltham, MA, 02451, USA
- Discovery and Development Sciences, Leads Discovery and Optimization, Bristol Myers Squibb, Cambridge, MA, 02141, USA
| | - Chakravarthy Garlapati
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Alkermes Inc, Waltham, MA, 02451, USA.
| | - Thi Nguyen
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shaligram Sharma
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Chemical Insights Research Institute of UL Research Institutes, Marietta, GA, 30067, USA
| | | | - Shristi Bhattarai
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Uma Krishnamurti
- Department of Pathology, Yale University, New Haven, CT, 06510, USA
| | - Xiaoxian Li
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
2
|
Allen B, Bottomly D, Köhnke T, Wang A, Lin HY, Johnson K, Kenna I, Streltsova A, Martin E, Chen R, Savoy L, Long N, Ryabinin P, Kurtz SE, Eide CA, Carlos A, Kaempf A, Liu T, Tognon C, Searles R, Piehowski PD, Gosline SJC, Agarwal A, Chang BH, Barton M, Druker BJ, McWeeney SK, Majeti R, Tyner JW, Zhang H. A CEBPB/IL-1β/TNF-α feedback loop drives drug resistance to venetoclax and MDM2 inhibitors in monocytic leukemia. Blood 2025; 145:2488-2506. [PMID: 40009487 DOI: 10.1182/blood.2024028239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRAT MDM2 inhibitors are promising therapeutics for acute myeloid leukemia (AML) with wild-type TP53. Through an integrated analysis of functional genomic data from primary patient samples, we found that an MDM2 inhibitor, idasanutlin, like venetoclax, is ineffective against monocytic leukemia (French-American-British [FAB] subtype M4/M5). To dissect the underlying resistance mechanisms, we explored both intrinsic and extrinsic factors. We found that monocytic leukemia cells express elevated levels of CEBPB, which promote monocytic differentiation, suppress CASP3 and CASP6, and upregulate MCL1, BCL2A1, and the interleukin (IL-1)/tumor necrosis factor alpha (TNF-α)/NF-κB pathway members, thereby conferring drug resistance to a broad range of MDM2 inhibitors, BH3 mimetics, and venetoclax combinations. In addition, aberrant monocytes in M4/M5 leukemia produce elevated levels of IL-1 and TNF-α, which promote monocytic differentiation and upregulate inflammatory cytokines and receptors, thereby extrinsically protecting leukemia blasts from venetoclax and MDM2 inhibition. Interestingly, IL-1β and TNF-α only increase CEBPB levels and protect M4/M5 cells from these drugs but not M0/M1 leukemia cells. Treatment with venetoclax and idasanutlin induces compensatory upregulation of CEBPB and the IL-1/TNF-α/NF-κB pathway independent of the FAB subtype, indicating drug-induced compensatory protection mechanisms. The combination of venetoclax or idasanutlin with inhibitors that block the IL-1/TNF-α pathway demonstrates synergistic cytotoxicity in M4/M5 AML. As such, we uncovered a targetable positive feedback loop that involves CEBPB, IL-1/TNF-α, and monocyte differentiation in M4/M5 leukemia and promotes both intrinsic and extrinsic drug resistance and drug-induced protection against venetoclax and MDM2 inhibitors.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Drug Resistance, Neoplasm/drug effects
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- CCAAT-Enhancer-Binding Protein-beta/genetics
- Leukemia, Monocytic, Acute/drug therapy
- Leukemia, Monocytic, Acute/metabolism
- Leukemia, Monocytic, Acute/pathology
- Leukemia, Monocytic, Acute/genetics
- Signal Transduction/drug effects
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Monocytes
Collapse
Affiliation(s)
- Basil Allen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Thomas Köhnke
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Anthony Wang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Hsin-Yun Lin
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Kara Johnson
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Isabel Kenna
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Anastatia Streltsova
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Emma Martin
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Reid Chen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Lindsey Savoy
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Nicola Long
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Peter Ryabinin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Stephen E Kurtz
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Christopher A Eide
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Amy Carlos
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Andy Kaempf
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Tingting Liu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Cristina Tognon
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Robert Searles
- Massively Parallel Sequencing Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Paul D Piehowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Sara J C Gosline
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA
| | - Anupriya Agarwal
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Bill H Chang
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Michelle Barton
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR
| | - Brian J Druker
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute and Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA
| | - Jeffrey W Tyner
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Haijiao Zhang
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| |
Collapse
|
3
|
Wang M, Li B, Nie S, Meng X, Wang G, Yang M, Dang W, He K, Sun T, Xu P, Yang X, Ye K. Asparagine endopeptidase cleaves apolipoprotein A1 and accelerates pathogenesis of atherosclerosis. J Clin Invest 2025; 135:e185128. [PMID: 40371638 PMCID: PMC12077905 DOI: 10.1172/jci185128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/12/2025] [Indexed: 05/16/2025] Open
Abstract
Atherosclerosis is a slowly progressing inflammatory disease characterized with cholesterol disorder and intimal plaques. Asparagine endopeptidase (AEP) is an endolysosomal protease that is activated under acidic conditions and is elevated substantially in both plasma and plaques of patients with atherosclerosis. However, how AEP accelerates atherosclerosis development remains incompletely understood, especially from the view of cholesterol metabolism. This project aims to reveal the crucial substrate of AEP during atherosclerosis plaque formation and to lay the foundation for developing novel therapeutic agents for Atherosclerosis. Here, we show that AEP is augmented in the atherosclerosis plaques obtained from patients and proteolytically cuts apolipoprotein A1 (APOA1) and impairs cholesterol efflux and high-density lipoprotein (HDL) formation, facilitating atherosclerosis pathologies. AEP is activated in the liver and aorta of apolipoprotein E-null (APOE-null) mice, and deletion of AEP from APOE-/- mice attenuates atherosclerosis. APOA1, an essential lipoprotein in HDL for cholesterol efflux, is cleaved by AEP at N208 residue in the liver and atherosclerotic macrophages of APOE-/- mice. Blockade of APOA1 cleavage by AEP via N208A mutation or its specific inhibitor, #11a, substantially diminishes atherosclerosis in both APOE-/- and LDLR-/- mice. Hence, our findings support that AEP disrupts cholesterol metabolism and accelerates the development of atherosclerosis.
Collapse
Affiliation(s)
- Mengmeng Wang
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Bowei Li
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Shuke Nie
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xin Meng
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Guangxing Wang
- Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghan Yang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Wenxin Dang
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Kangning He
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, China
| | - Tucheng Sun
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020–2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Keqiang Ye
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), University of Chinese Academy of Science, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Ji A, Meredith LW, Shridas P. Serum Amyloid A: A Double-Edged Sword in Health and Disease. Int J Mol Sci 2025; 26:4528. [PMID: 40429677 PMCID: PMC12110822 DOI: 10.3390/ijms26104528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Despite more than fifty years since its discovery in the 1970s, Serum Amyloid A (SAA)'s true biological functions remain enigmatic. The research so far has primarily associated SAA with chronic inflammatory conditions such as cardiovascular disease, obesity, and type 2 diabetes; its role in acute inflammation is less understood. Unlike the modest elevations observed in chronic conditions, SAA levels surge dramatically during acute inflammatory responses. Notably, approximately 2.5% of hepatic protein synthesis is devoted to SAA production during acute inflammation-despite the high energy demands required for synthesizing pro-inflammatory cytokines and immune cell activation-leaving its precise necessity unclear. Elucidating SAA's physiological role in acute inflammation is crucial to determine the therapeutic potential of SAA inhibition for chronic inflammatory diseases, such as atherosclerosis and abdominal aortic aneurysms. The evidence suggests that SAA may play a protective role in acute inflammation, positioning it as a "double-edged sword": detrimental in chronic inflammation, yet potentially beneficial in acute settings. This review explores the divergent roles of SAA in chronic versus acute inflammation, proposing that while SAA inhibition could offer therapeutic benefits for chronic conditions, it might pose risks during acute inflammation. As the primary transporter of SAA in circulation, high-density lipoprotein (HDL) has been shown to become dysfunctional in chronic inflammation, at least partly due to SAA's effects. However, we propose that SAA may confer functional properties to HDL during acute inflammatory states, such as sepsis, thereby highlighting the context-dependent nature of its impact.
Collapse
Affiliation(s)
- Ailing Ji
- Saha Cardiovascular Research Center, University of Kentucky, 567 Charles T Wethington Building, 900 S. Limestone Street, Lexington, KY 40536-0200, USA; (A.J.); (L.W.M.)
| | - Luke W. Meredith
- Saha Cardiovascular Research Center, University of Kentucky, 567 Charles T Wethington Building, 900 S. Limestone Street, Lexington, KY 40536-0200, USA; (A.J.); (L.W.M.)
| | - Preetha Shridas
- Saha Cardiovascular Research Center, University of Kentucky, 567 Charles T Wethington Building, 900 S. Limestone Street, Lexington, KY 40536-0200, USA; (A.J.); (L.W.M.)
- Department of Internal Medicine, University of Kentucky, 567 Charles T Wethington Building, 900 S. Limestone Street, Lexington, KY 40536-0200, USA
| |
Collapse
|
5
|
Vajeethaveesin N, Kanitwithayanun J, Suriyo T, Chujan S, Satayavivad J. Perfluorooctane sulfonic acid: a possible risk factor of endothelial dysfunction based on in silico and in vitro studies. Arch Toxicol 2025:10.1007/s00204-025-04047-7. [PMID: 40244404 DOI: 10.1007/s00204-025-04047-7] [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: 12/11/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Perfluorooctane sulfonic acid (PFOS) is a fluorinated chemical utilized in a variety of industrial and household products. PFOS has been detected in human serum and is associated with multiple human adverse health effects. Epidemiological evidence has linked PFOS exposure to endothelial dysfunction, which is a key contributor to atherosclerosis. However, the underlying mechanisms of PFOS-induced endothelial dysfunction associated atherosclerosis has not been investigated. In the present study, human microvascular endothelial cells (HMEC-1) exposed to PFOS (15 μM) for 72 h, mimicking long-term exposure. We further employed integrated RNA-sequencing (RNA-seq) and transcriptomic analysis to identify differentially expressed genes (DEGs) for biological alterations: gene ontology (GO), pathway enrichment analysis (KEGG), protein-protein interaction network and modular clustering analysis. Furthermore, the Metascape database was used for disease association, tissue specificity, and transcription factor analysis. Hub genes were verified using atherosclerosis patient data sets from the GEO dataset. Alteration of hub genes in patients was then validated using immunoblotting and ELISA. Our results revealed that PFOS altered amino acid biosynthesis, lipid metabolism and induced the ER-stress response through the HRI/eIF2α/ATF4 pathway, leading to endothelial dysfunction. Interestingly, we found that PFOS induced inflammation by increasing COX-2, ICAM-1 and IL-6 expression through NF-κB and JAK2/STAT3 pathway in endothelial cells. Moreover, up-regulated C/EBPβ and ATF4 were observed in both patients and PFOS-exposed endothelium, which may use as an early biomarker and may have a potential role in PFOS-induced endothelial dysfunction. These findings provide novel insight into the underlying molecular mechanisms of PFOS-induced endothelial dysfunction associated with atherosclerosis.
Collapse
Affiliation(s)
- Nutsira Vajeethaveesin
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand
| | - Jantamas Kanitwithayanun
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence On Environmental Health and Toxicology, Bangkok, 10400, Thailand
| | - Tawit Suriyo
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence On Environmental Health and Toxicology, Bangkok, 10400, Thailand
| | - Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand.
- Center of Excellence On Environmental Health and Toxicology, Bangkok, 10400, Thailand.
| | - Jutamaad Satayavivad
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand.
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand.
- Center of Excellence On Environmental Health and Toxicology, Bangkok, 10400, Thailand.
| |
Collapse
|
6
|
Niu S, Li M, Wang J, Zhong P, Wen X, Huang F, Yin L, Liao Y, Zhou J. Identify the potential target of efferocytosis in knee osteoarthritis synovial tissue: a bioinformatics and machine learning-based study. Front Immunol 2025; 16:1550794. [PMID: 40083558 PMCID: PMC11903261 DOI: 10.3389/fimmu.2025.1550794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Knee osteoarthritis (KOA) is a degenerative joint disease characterized by the progressive deterioration of cartilage and synovial inflammation. A critical mechanism in the pathogenesis of KOA is impaired efferocytosis in synovial tissue. The present study aimed to identify and validate key efferocytosis-related genes (EFRGs) in KOA synovial tissue by using comprehensive bioinformatics and machine learning approaches. Methods We integrated three datasets (GSE55235, GSE55457, and GSE12021) from the Gene Expression Omnibus database to screen differentially expressed genes (DEGs) associated with efferocytosis and performed weighted gene co-expression network analysis. Subsequently, we utilized univariate logistic regression analysis, least absolute shrinkage and selection operator regression, support vector machine, and random forest algorithms to further refine these genes. The results were then inputted into multivariate logistic regression analysis to construct a diagnostic nomogram. Public datasets and quantitative real-time PCR experiments were employed for validation. Additionally, immune infiltration analysis was conducted with CIBERSORT using the combined datasets. Results Analysis of the intersection between DEGs and EFRGs identified 12 KOA-related efferocytosis DEGs. Further refinement through machine learning algorithms and multivariate logistic regression revealed UCP2, CX3CR1, and CEBPB as hub genes. Immune infiltration analysis demonstrated significant correlations between immune cell components and the expression levels of these hub genes. Validation using independent datasets and experimental approaches confirmed the robustness of these findings. Conclusions This study successfully identified three hub genes (UCP2, CX3CR1, and CEBPB) with significant expression alterations in KOA, demonstrating high diagnostic potential and close associations with impaired efferocytosis. These targets may modulate synovial efferocytosis-related immune processes, offering novel therapeutic avenues for KOA intervention.
Collapse
Affiliation(s)
- Shangbo Niu
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mengmeng Li
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jinling Wang
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peirui Zhong
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Wen
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fujin Huang
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linwei Yin
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yang Liao
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun Zhou
- Rehabilitation Medicine Center Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
7
|
Jin Z, Yamaguchi A, Takai H, Nakayama Y, Ogata Y. Interleukin-6 regulates human ODAM gene expression in gingival epithelial cells. J Periodontal Implant Sci 2025; 55:55.e12. [PMID: 40350767 DOI: 10.5051/jpis.2402980149] [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: 07/21/2024] [Revised: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 05/14/2025] Open
Abstract
PURPOSE Odontogenic ameloblast-associated protein (ODAM) is a small secretory protein produced by the junctional epithelium (JE) and mature ameloblasts. It plays a role in odontogenesis and mediates the adhesion of JE to enamel. We used human gingival epithelial cells to evaluate the mechanism of ODAM gene expression regulation in the JE by interleukin (IL)-6. METHODS Ca9-22, Sa3, and HSY cells were stimulated with IL-6 (10 ng/mL), after which total RNA and proteins were extracted. Real-time polymerase chain reaction and Western blot analyses were performed to assess the expression levels of ODAM mRNA and protein. Luciferase (LUC) assays were employed using LUC constructs with varying lengths of the ODAM gene promoter sequence. Gel mobility shift and chromatin immunoprecipitation (ChIP) analyses were conducted to investigate the binding of transcription factors to response elements within the gene promoter. RESULTS Treatment with IL-6 increased the expressions of ODAM mRNA and protein. Additionally, it induced promoter activity of the ODAM gene, while LUC activity was suppressed by inhibitors of protein kinase A, tyrosine kinase, MEK1/2, phosphatidylinositol 3-kinase, nuclear factor-κB, signal transducer and activator of transcription (STAT) 3, and glycoprotein 130. Gel mobility shift and ChIP analyses revealed that IL-6 induced the binding of yin yang 1 (YY1), CCAAT/enhancer-binding protein (C/EBP) β, GATA binding protein (GATA), and phospho-STAT3 to the YY1, C/EBP, GATA, and interferon-γ activated transcriptional element (GATE) 1-3 elements. CONCLUSIONS These findings indicate that IL-6 upregulates ODAM gene expression by targeting the YY1, C/EBP, GATA, and GATE1-3 elements in the promoter region of the human ODAM gene.
Collapse
Affiliation(s)
- Zhenyu Jin
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Arisa Yamaguchi
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Hideki Takai
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yohei Nakayama
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yorimasa Ogata
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.
| |
Collapse
|
8
|
Li C, Ling Y, Kuang H. Research progress on FSH-FSHR signaling in the pathogenesis of non-reproductive diseases. Front Cell Dev Biol 2024; 12:1506450. [PMID: 39633710 PMCID: PMC11615068 DOI: 10.3389/fcell.2024.1506450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Follicle-stimulating hormone (FSH), a glycoprotein hormone synthesized and secreted by the anterior pituitary gland, plays a critical role in reproductive development and regulation by binding to FSH receptor (FSHR). Beyond reproductive tissue, FSHRs have been identified in various non-reproductive tissues, indicating broader functions. FSH levels chronically rise during menopause and remain elevated in postmenopausal life. This increase in FSH level has been indicated to be associated with heightened risk of several non-reproductive diseases, including osteoporosis, hypercholesterolemia, type 2 diabetes mellitus, obesity, cardiovascular disease, Alzheimer's disease, and certain cancers. In this review, we will examine the role of FSH-FSHR signaling in the pathogenesis of these non-reproductive diseases and explore therapeutic strategies targeting FSH-FSHR signaling pathways.
Collapse
Affiliation(s)
- Chenhe Li
- Department of Clinical Medicine, School of Queen Mary, Nanchang University, Nanchang, Jiangxi, China
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yan Ling
- Department of Obstetrics and Gynecology, Jiangxi provincial People’s Hospital, Nanchang, Jiangxi, China
| | - Haibin Kuang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Rakshit K, Brown MR, Javeed N, Lee JH, Ordog T, Matveyenko AV. Core circadian transcription factor Bmal1 mediates β cell response and recovery from pro-inflammatory injury. iScience 2024; 27:111179. [PMID: 39524327 PMCID: PMC11550590 DOI: 10.1016/j.isci.2024.111179] [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: 02/05/2024] [Revised: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The circadian clock plays a vital role in modulating the cellular immune response. However, its role in mediating pro-inflammatory diabetogenic β cell injury remains largely unexplored. Our studies demonstrate that the exposure of β cells to IL-1β-mediated inflammation alters genome-wide DNA binding of core circadian transcription factors BMAL1:CLOCK enriched for genomic sites important for cellular response to inflammation. Correspondingly, conditional deletion of Bmal1 in mouse β cells was shown to impair the ability of β cells to recover from streptozotocin-mediated pro-inflammatory injury in vivo, leading to β cell failure and the development of diabetes. Further data integration analysis revealed that the β cell circadian clock orchestrates the recovery from pro-inflammatory injury by regulating transcriptional responses to oxidative stress, DNA damage, and nuclear factor κB(NF-κB)-driven inflammation. Our study suggests that the β cell circadian clock mediates β cell response and recovery from pro-inflammatory injury common to the pathogenesis of diabetes mellitus.
Collapse
Affiliation(s)
- Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Matthew R. Brown
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, MN, USA
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Jeong-Heon Lee
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Tamas Ordog
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, MN, USA
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Aleksey V. Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Rochester, MN, USA
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic School of Medicine, Rochester, MN, USA
| |
Collapse
|
10
|
Daga N, Servaas NH, Kisand K, Moonen D, Arnold C, Reyes-Palomares A, Kaleviste E, Kingo K, Kuuse R, Ulst K, Steinmetz L, Peterson P, Nakic N, Zaugg JB. Integration of genetic and chromatin modification data pinpoints autoimmune-specific remodeling of enhancer landscape in CD4 + T cells. Cell Rep 2024; 43:114810. [PMID: 39388354 DOI: 10.1016/j.celrep.2024.114810] [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: 12/23/2023] [Revised: 07/16/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
CD4+ T cells play a crucial role in adaptive immune responses and have been implicated in the pathogenesis of autoimmune diseases (ADs). Despite numerous studies, the molecular mechanisms underlying T cell dysregulation in ADs remain incompletely understood. Here, we used chromatin immunoprecipitation (ChIP)-sequencing of active chromatin and transcriptomic data from CD4+ T cells of healthy donors and patients with systemic lupus erythematosus (SLE), psoriasis, juvenile idiopathic arthritis (JIA), and Graves' disease to investigate the role of enhancers in AD pathogenesis. By generating enhancer-based gene regulatory networks (eGRNs), we identified disease-specific dysregulated pathways and potential downstream target genes of enhancers harboring AD-associated single-nucleotide polymorphisms (SNPs), which we also validated using chromatin-capture (HiC) data and CRISPR interference (CRISPRi) in primary CD4+ T cells. Our results suggest that alterations in the regulatory landscapes of CD4+ T cells, including enhancers, contribute to the development of ADs and provide a basis for developing new therapeutic approaches.
Collapse
Affiliation(s)
- Neha Daga
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Nila H Servaas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Dewi Moonen
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christian Arnold
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Armando Reyes-Palomares
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Epp Kaleviste
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venerology, Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia and Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Reet Kuuse
- Department of Internal Medicine, Tartu University Hospital, Tartu, Estonia
| | - Katrin Ulst
- Department of Internal Medicine, Tartu University Hospital, Tartu, Estonia
| | - Lars Steinmetz
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Department of Genetics, Stanford University, Stanford, CA, USA
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Nikolina Nakic
- Functional Genomics, Medicinal Science and Technology, GSK R&D, Stevenage, UK
| | - Judith B Zaugg
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
11
|
Lei WJ, Zhang F, Li MD, Pan F, Ling LJ, Lu JW, Myatt L, Sun K, Wang WS. C/EBPδ deficiency delays infection-induced preterm birth. BMC Med 2024; 22:432. [PMID: 39379940 PMCID: PMC11462803 DOI: 10.1186/s12916-024-03650-2] [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: 06/15/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Parturition is an inflammation process. Exaggerated inflammatory reactions in infection lead to preterm birth. Although nuclear factor kappa B (NF-κB) has been recognized as a classical transcription factor mediating inflammatory reactions, those mediated by NF-κB per se are relatively short-lived. Therefore, there may be other transcription factors involved to sustain NF-κB-initiated inflammatory reactions in gestational tissues in infection-induced preterm birth. METHODS Cebpd-deficient mice were generated to investigate the role of CCAAT enhancer-binding protein δ (C/EBPδ) in lipopolysaccharide (LPS)-induced preterm birth, and the contribution of fetal and maternal C/EBPδ was further dissected by transferring Cebpd-/- or WT embryos to Cebpd-/- or WT dams. The effects of C/EBPδ pertinent to parturition were investigated in mouse and human myometrial and amnion cells. The interplay between C/EBPδ and NF-κB was examined in cultured human amnion fibroblasts. RESULTS The mouse study showed that LPS-induced preterm birth was delayed by Cebpd deficiency in either the fetus or the dam, with further delay being observed in conceptions where both the dam and the fetus were deficient in Cebpd. Mouse and human studies showed that the abundance of C/EBPδ was significantly increased in the myometrium and fetal membranes in infection-induced preterm birth. Furthermore, C/EBPδ participated in LPS-induced upregulation of pro-inflammatory cytokines as well as genes pertinent to myometrial contractility and fetal membrane activation in the myometrium and amnion respectively. A mechanistic study in human amnion fibroblasts showed that C/EBPδ, upon induction by NF-κB, could serve as a supplementary transcription factor to NF-κB to sustain the expression of genes pertinent to parturition. CONCLUSIONS C/EBPδ is a transcription factor to sustain the expression of gene initiated by NF-κB in the myometrium and fetal membranes in infection-induced preterm birth. Targeting C/EBPδ may be of therapeutic value in the treatment of infection-induced preterm birth.
Collapse
Affiliation(s)
- Wen-Jia Lei
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Fan Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Meng-Die Li
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Fan Pan
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Li-Jun Ling
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China.
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China.
| |
Collapse
|
12
|
Secchia S, Beilinson V, Chen X, Yang ZF, Wayman JA, Dhaliwal J, Jurickova I, Angerman E, Denson LA, Miraldi ER, Weirauch MT, Ikegami K. Nutrient starvation activates ECM remodeling gene enhancers associated with inflammatory bowel disease risk in fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611754. [PMID: 39314475 PMCID: PMC11418948 DOI: 10.1101/2024.09.06.611754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Nutrient deprivation induces a reversible cell cycle arrest state termed quiescence, which often accompanies transcriptional silencing and chromatin compaction. Paradoxically, nutrient deprivation is associated with activated fibroblast states in pathological microenvironments in which fibroblasts drive extracellular matrix (ECM) remodeling to alter tissue environments. The relationship between nutrient deprivation and fibroblast activation remains unclear. Here, we report that serum deprivation extensively activates transcription of ECM remodeling genes in cultured fibroblasts, despite the induction of quiescence. Starvation-induced transcriptional activation accompanied large-scale histone acetylation of putative distal enhancers, but not promoters. The starvation-activated putative enhancers were enriched for non-coding genetic risk variants associated with inflammatory bowel disease (IBD), suggesting that the starvation-activated gene regulatory network may contribute to fibroblast activation in IBD. Indeed, the starvation-activated gene PLAU, encoding uPA serine protease for plasminogen and ECM, was upregulated in inflammatory fibroblasts in the intestines of IBD patients. Furthermore, the starvation-activated putative enhancer at PLAU, which harbors an IBD risk variant, gained chromatin accessibility in IBD patient fibroblasts. This study implicates nutrient deprivation in transcriptional activation of ECM remodeling genes in fibroblasts and suggests nutrient deprivation as a potential mechanism for pathological fibroblast activation in IBD.
Collapse
Affiliation(s)
- Stefano Secchia
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, USA
- Department of Biology, Lund University, Lund, 22362, Sweden
- Present address: Institute of Human Biology, Basel, Switzerland
| | - Vera Beilinson
- Department of Pediatrics, The University of Chicago, Chicago, Illinois, USA
- Present address: California Institute of Technology, Pasadena, California, USA
| | - Xiaoting Chen
- Division of Allergy and Immunology, CCHMC Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Zi F Yang
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joseph A Wayman
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jasbir Dhaliwal
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ingrid Jurickova
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Elizabeth Angerman
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily R Miraldi
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Matthew T Weirauch
- Division of Allergy and Immunology, CCHMC Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kohta Ikegami
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
13
|
Fang X, Liu S, Muhammad B, Zheng M, Ge X, Xu Y, Kan S, Zhang Y, Yu Y, Zheng K, Geng D, Liu CF. Gut microbiota dysbiosis contributes to α-synuclein-related pathology associated with C/EBPβ/AEP signaling activation in a mouse model of Parkinson's disease. Neural Regen Res 2024; 19:2081-2088. [PMID: 38227539 DOI: 10.4103/1673-5374.391191] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00042/figure1/v/2024-01-16T170235Z/r/image-tiff Parkinson's disease is a neurodegenerative disease characterized by motor and gastrointestinal dysfunction. Gastrointestinal dysfunction can precede the onset of motor symptoms by several years. Gut microbiota dysbiosis is involved in the pathogenesis of Parkinson's disease, whether it plays a causal role in motor dysfunction, and the mechanism underlying this potential effect, remain unknown. CCAAT/enhancer binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling, activated by bacterial endotoxin, can promote α-synuclein transcription, thereby contributing to Parkinson's disease pathology. In this study, we aimed to investigate the role of the gut microbiota in C/EBPβ/AEP signaling, α-synuclein-related pathology, and motor symptoms using a rotenone-induced mouse model of Parkinson's disease combined with antibiotic-induced microbiome depletion and fecal microbiota transplantation. We found that rotenone administration resulted in gut microbiota dysbiosis and perturbation of the intestinal barrier, as well as activation of the C/EBP/AEP pathway, α-synuclein aggregation, and tyrosine hydroxylase-positive neuron loss in the substantia nigra in mice with motor deficits. However, treatment with rotenone did not have any of these adverse effects in mice whose gut microbiota was depleted by pretreatment with antibiotics. Importantly, we found that transplanting gut microbiota derived from mice treated with rotenone induced motor deficits, intestinal inflammation, and endotoxemia. Transplantation of fecal microbiota from healthy control mice alleviated rotenone-induced motor deficits, intestinal inflammation, endotoxemia, and intestinal barrier impairment. These results highlight the vital role that gut microbiota dysbiosis plays in inducing motor deficits, C/EBPβ/AEP signaling activation, and α-synuclein-related pathology in a rotenone-induced mouse model of Parkinson's disease. Additionally, our findings suggest that supplementing with healthy microbiota may be a safe and effective treatment that could help ameliorate the progression of motor deficits in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoli Fang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bilal Muhammad
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yan Xu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shu Kan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yang Zhang
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu Province, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Jiangsu Key Laboratory of Neuropsychiatric Disease and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
14
|
Liu Y, Qiao H, Zienkiewicz J, Hawiger J. Anti-inflammatory control of human skin keratinocytes by targeting nuclear transport checkpoint. SKIN HEALTH AND DISEASE 2024; 4:e356. [PMID: 38846687 PMCID: PMC11150741 DOI: 10.1002/ski2.356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/10/2024] [Accepted: 02/21/2024] [Indexed: 06/09/2024]
Abstract
Background In the two common inflammatory skin diseases, Atopic Dermatitis (AD) and Psoriasis (Ps), keratinocytes (KCs) respond to immune insults through activation of proinflammatory transcription factors (TFs) and their translocation to the cell's nucleus. Therein, the TFs induce expression of genes encoding mediators of skin inflammation. The Nuclear Transport Checkpoint Inhibitors (NTCIs) were developed to regulate nuclear translocation of activated TFs, the essential step of inflammatory response. This new class of cell-penetrating peptide therapeutics controls inflammation caused by allergic, autoimmune, metabolic, and microbial insults. In preclinical model of AD, the treatment with NTCI, cSN50.1 peptide, suppressed the expression of Thymic Stromal Lymphopoietin (TSLP), the key gene in the development of allergic inflammation, among the 15 genes silenced by the NTCI. Here, we report the mechanism of anti-inflammatory action of NTCI in human skin-derived KCs. Objectives We aimed to determine whether the NTCI treatment can protect human KCs from harmful inflammatory insults. Methods Human primary KCs were pretreated with NTCI and challenged with the mix of cytokines Tumour Necrosis Factor alpha (TNF-α) and Interleukin (IL)-17A, or with Phorbol 12-Myristate 13-Acetate (PMA), and analysed for nuclear content of TFs and the expression of genes encoding mediators of inflammation. Results The nuclear import of TFs, Nuclear Factor ĸB (NF-ĸB) and Signal Transduction and Activator of Transcription 3 (STAT3), was inhibited in cells treated with NTCI. The expression of TSLP, along with genes encoding the core mediators of inflammation (TNF, IL1B, and IL6) was suppressed by NTCI. Noteworthy, NTCI silenced genes encoding Granulocyte-Macrophage Colony-Stimulating Factor (CSF2), and chemokine IL-8 (CXCL8), responsible for skin infiltration by the eosinophils and other myelomonocytic cells. Conclusion The control of inflammatory response in human KCs by NTCI is attributed to the inhibition of nuclear import of proinflammatory TFs. The protection of human KCs by NTCI, adds new perspectives to the completed Phase two clinical trial of the NTCI (AMTX-100 CF) for AD (NCT04313400).
Collapse
Affiliation(s)
- Yan Liu
- Department of MedicineDivision of AllergyPulmonary and Critical Care MedicineVanderbilt University School of MedicineNashvilleTennesseeUSA
- Department of Veterans AffairsTennessee Valley Health Care SystemNashvilleTennesseeUSA
| | - Huan Qiao
- Department of MedicineDivision of AllergyPulmonary and Critical Care MedicineVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Jozef Zienkiewicz
- Department of MedicineDivision of AllergyPulmonary and Critical Care MedicineVanderbilt University School of MedicineNashvilleTennesseeUSA
- Department of Veterans AffairsTennessee Valley Health Care SystemNashvilleTennesseeUSA
| | - Jacek Hawiger
- Department of MedicineDivision of AllergyPulmonary and Critical Care MedicineVanderbilt University School of MedicineNashvilleTennesseeUSA
- Department of Veterans AffairsTennessee Valley Health Care SystemNashvilleTennesseeUSA
- Department of Molecular Physiology and BiophysicsVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
15
|
Silva RCMC. Mitochondria, Autophagy and Inflammation: Interconnected in Aging. Cell Biochem Biophys 2024; 82:411-426. [PMID: 38381268 DOI: 10.1007/s12013-024-01231-x] [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: 10/18/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
In this manuscript, I discuss the direct link between abnormalities in inflammatory responses, mitochondrial metabolism and autophagy during the process of aging. It is focused on the cytosolic receptors nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) and cyclic GMP-AMP synthase (cGAS); myeloid-derived suppressor cells (MDSCs) expansion and their associated immunosuppressive metabolite, methyl-glyoxal, all of them negatively regulated by mitochondrial autophagy, biogenesis, metabolic pathways and its distinct metabolites.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Chen J, Wu L, Li Y. FGL1 and FGL2: emerging regulators of liver health and disease. Biomark Res 2024; 12:53. [PMID: 38816776 PMCID: PMC11141035 DOI: 10.1186/s40364-024-00601-0] [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: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Liver disease is a complex group of diseases with high morbidity and mortality rates, emerging as a major global health concern. Recent studies have highlighted the involvement of fibrinogen-like proteins, specifically fibrinogen-like protein 1 (FGL1) and fibrinogen-like protein 2 (FGL2), in the regulation of various liver diseases. FGL1 plays a crucial role in promoting hepatocyte growth, regulating lipid metabolism, and influencing the tumor microenvironment (TME), contributing significantly to liver repair, non-alcoholic fatty liver disease (NAFLD), and liver cancer. On the other hand, FGL2 is a multifunctional protein known for its role in modulating prothrombin activity and inducing immune tolerance, impacting viral hepatitis, liver fibrosis, hepatocellular carcinoma (HCC), and liver transplantation. Understanding the functions and mechanisms of fibrinogen-like proteins is essential for the development of effective therapeutic approaches for liver diseases. Additionally, FGL1 has demonstrated potential as a disease biomarker in radiation and drug-induced liver injury as well as HCC, while FGL2 shows promise as a biomarker in viral hepatitis and liver transplantation. The expression levels of these molecules offer exciting prospects for disease assessment. This review provides an overview of the structure and roles of FGL1 and FGL2 in different liver conditions, emphasizing the intricate molecular regulatory processes and advancements in targeted therapies. Furthermore, it explores the potential benefits and challenges of targeting FGL1 and FGL2 for liver disease treatment and the prospects of fibrinogen-like proteins as biomarkers for liver disease, offering insights for future research in this field.
Collapse
Affiliation(s)
- Jiongming Chen
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Lei Wu
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Yongsheng Li
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
17
|
Luan X, Lei T, Fang J, Liu X, Fu H, Li Y, Chu W, Jiang P, Tong C, Qi H, Fu Y. Blockade of C5a receptor unleashes tumor-associated macrophage antitumor response and enhances CXCL9-dependent CD8 + T cell activity. Mol Ther 2024; 32:469-489. [PMID: 38098230 PMCID: PMC10861991 DOI: 10.1016/j.ymthe.2023.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 12/25/2023] Open
Abstract
Macrophages play a crucial role in shaping the immune state within the tumor microenvironment (TME) and are often influenced by tumors to hinder antitumor immunity. However, the underlying mechanisms are still elusive. Here, we observed abnormal expression of complement 5a receptor (C5aR) in human ovarian cancer (OC), and identified high levels of C5aR expression on tumor-associated macrophages (TAMs), which led to the polarization of TAMs toward an immunosuppressive phenotype. C5aR knockout or inhibitor treatment restored TAM antitumor response and attenuated tumor progression. Mechanistically, C5aR deficiency reprogrammed macrophages from a protumor state to an antitumor state, associating with the upregulation of immune response and stimulation pathways, which in turn resulted in the enhanced antitumor response of cytotoxic T cells in a manner dependent on chemokine (C-X-C motif) ligand 9 (CXCL9). The pharmacological inhibition of C5aR also improved the efficacy of immune checkpoint blockade therapy. In patients, C5aR expression associated with CXCL9 production and infiltration of CD8+ T cells, and a high C5aR level predicted poor clinical outcomes and worse benefits from anti-PD-1 therapy. Thus, our study sheds light on the mechanisms underlying the modulation of TAM antitumor immune response by the C5a-C5aR axis and highlights the potential of targeting C5aR for clinical applications.
Collapse
Affiliation(s)
- Xiaojin Luan
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting Lei
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jie Fang
- Department of Gynecology, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Xue Liu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Huijia Fu
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yiran Li
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Chu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Jiang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chao Tong
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongbo Qi
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yong Fu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
18
|
Shao KM, Shao WH. Transcription Factors in the Pathogenesis of Lupus Nephritis and Their Targeted Therapy. Int J Mol Sci 2024; 25:1084. [PMID: 38256157 PMCID: PMC10816397 DOI: 10.3390/ijms25021084] [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] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototype inflammatory autoimmune disease, characterized by breakdown of immunotolerance to self-antigens. Renal involvement, known as lupus nephritis (LN), is one of the leading causes of morbidity and a significant contributor to mortality in SLE. Despite current pathophysiological advances, further studies are needed to fully understand complex mechanisms underlying the development and progression of LN. Transcription factors (TFs) are proteins that regulate the expression of genes and play a crucial role in the development and progression of LN. The mechanisms of TF promoting or inhibiting gene expression are complex, and studies have just begun to reveal the pathological roles of TFs in LN. Understanding TFs in the pathogenesis of LN can provide valuable insights into this disease's mechanisms and potentially lead to the development of targeted therapies for its management. This review will focus on recent findings on TFs in the pathogenesis of LN and newly developed TF-targeted therapy in renal inflammation.
Collapse
Affiliation(s)
- Kasey M. Shao
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Wen-Hai Shao
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
19
|
Ren Q, Liu Z, Wu L, Yin G, Xie X, Kong W, Zhou J, Liu S. C/EBPβ: The structure, regulation, and its roles in inflammation-related diseases. Biomed Pharmacother 2023; 169:115938. [PMID: 38000353 DOI: 10.1016/j.biopha.2023.115938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammation, a mechanism of the human body, has been implicated in many diseases. Inflammatory responses include the release of inflammatory mediators by activating various signaling pathways. CCAAT/enhancer binding protein β (C/EBPβ), a transcription factor in the C/EBP family, contains the leucine zipper (bZIP) domain. The expression of C/EBPβ is mediated at the transcriptional and post-translational levels, such as phosphorylation, acetylation, methylation, and SUMOylation. C/EBPβ has been involved in inflammatory responses by mediating several signaling pathways, such as MAPK/NF-κB and IL-6/JAK/STAT3 pathways. C/EBPβ plays an important role in the pathological development of inflammation-related diseases, such as osteoarthritis, pneumonia, hepatitis, inflammatory bowel diseases, and rheumatoid arthritis. Here, we comprehensively discuss the structure and biological effects of C/EBPβ and its role in inflammatory diseases.
Collapse
Affiliation(s)
- Qun Ren
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhaowen Liu
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Longhuo Wu
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou People's Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
20
|
Xiong J, Kang SS, Wang M, Wang Z, Xia Y, Liao J, Liu X, Yu SP, Zhang Z, Ryu V, Yuen T, Zaidi M, Ye K. FSH and ApoE4 contribute to Alzheimer's disease-like pathogenesis via C/EBPβ/δ-secretase in female mice. Nat Commun 2023; 14:6577. [PMID: 37852961 PMCID: PMC10584868 DOI: 10.1038/s41467-023-42282-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia. It is known that women with one ApoE4 allele display greater risk and earlier onset of AD compared with men. In mice, we previously showed that follicle-stimulating hormone (FSH), a gonadotropin that rises in post-menopausal females, activates its receptor FSHR in the hippocampus, to drive AD-like pathology and cognitive impairment. Here we show in mice that ApoE4 and FSH jointly trigger AD-like pathogenesis by activating C/EBPβ/δ-secretase signaling. ApoE4 and FSH additively activate C/EBPβ/δ-secretase pathway that mediates APP and Tau proteolytic fragmentation, stimulating Aβ and neurofibrillary tangles. Ovariectomy-provoked AD-like pathologies and cognitive defects in female ApoE4-TR mice are ameliorated by anti-FSH antibody treatment. FSH administration facilitates AD-like pathologies in both young male and female ApoE4-TR mice. Furthermore, FSH stimulates AD-like pathologies and cognitive defects in ApoE4-TR mice, but not ApoE3-TR mice. Our findings suggest that in mice, augmented FSH in females with ApoE4 but not ApoE3 genotype increases vulnerability to AD-like process by activating C/EBPβ/δ-secretase signalling.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mengmeng Wang
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Zhihao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan-Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Vitaly Ryu
- Mount Sinai Bone Program, Department of Medicine and Department of Orthopedics, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - Tony Yuen
- Mount Sinai Bone Program, Department of Medicine and Department of Orthopedics, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - Mone Zaidi
- Mount Sinai Bone Program, Department of Medicine and Department of Orthopedics, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
21
|
Han K, Dong H, Peng X, Sun J, Jiang H, Feng Y, Ding J, Xiao S. Transcriptome and the gut microbiome analysis of the impacts of Brucella abortus oral infection in BALB/c mice. Microb Pathog 2023; 183:106278. [PMID: 37532208 DOI: 10.1016/j.micpath.2023.106278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/22/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
Brucellosis is a zoonotic infectious disease caused by Brucella spp, which could cause serious economic losses to animal husbandry and threaten human public health. Ingestion of contaminated animal products is a common way to acquire Brucella infection in humans, while research on effect of oral Brucella infection on host gut microbiota and the gene expression in intestinal tissues is limited. In the present study, 16S rRNA sequencing and RNA sequencing were conducted to explore gut microbiota and expression profiles of mRNAs in the colon of BALB/c mice, which were infected by Brucella abortus 2308. The fecal samples were collected at 7 and 28 days post infection to observe changes in the gut microbiota during Brucella infection. In the alpha diversity analysis, significantly increased Chao 1 index was observed at 28 days after Brucella infection. The Bray-Curtis distancebased principal coordinate analysis indicated that the WT group showed a separation from the Brucella infection groups. In addition, analysis of composition of microbes revealed that Prevotellaceae_NK3B31_group were more abundant in 1 week and 4 week infection groups, while Turicibacter was only more abundant in 4 week infection group. Based on the RNA-seq assay, a total of 45 differentially expressed genes were detected between Brucella abortus infection group and control group. Furthermore, KEGG pathway enrichment analysis showed that protein processing in endoplasmic reticulum, Legionellosis, Spliceosome, Hippo signaling pathway and Influenza A were significantly enriched in response to Brucella abortus infection. Our finding will help to improve the knowledge of the mechanisms underlying Brucella infection and may provide novel targets for future treatment of this pathogen infection.
Collapse
Affiliation(s)
- Kun Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hao Dong
- National Institutes for Food and Drug Control, Beijing, 102600, China
| | - Xiaowei Peng
- China Institute of Veterinary Drug Control, Beijing, 102600, China
| | - Jiali Sun
- China Institute of Veterinary Drug Control, Beijing, 102600, China; College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Hui Jiang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yu Feng
- China Institute of Veterinary Drug Control, Beijing, 102600, China
| | - Jiabo Ding
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Sa Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
22
|
Lin Z, Huang L, Cao Q, Luo H, Yao W, Zhang JC. Inhibition of abnormal C/EBPβ/α-Syn signaling pathway through activation of Nrf2 ameliorates Parkinson's disease-like pathology. Aging Cell 2023; 22:e13958. [PMID: 37614147 PMCID: PMC10577548 DOI: 10.1111/acel.13958] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/24/2023] [Accepted: 07/30/2023] [Indexed: 08/25/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the formation of Lewy bodies (LBs) in the brain. These LBs are primarily composed of α-Synuclein (α-Syn), which has aggregated. A recent report proposes that CCAAT/enhancer-binding proteins β (C/EBPβ) may act as an age-dependent transcription factor for α-Syn, thereby initiating PD pathologies by regulating its transcription. Potential therapeutic approaches to address PD could involve targeting the regulation of α-Syn by C/EBPβ. This study has revealed that Nrf2, also known as nuclear factor (erythroid-derived 2)-like 2 (NFE2L2), suppresses the transcription of C/EBPβ in SH-SY5Y cells when treated with MPP+ . To activate Nrf2, sulforaphane, an Nrf2 activator, was administered. Additionally, C/EBPβ was silenced using C/EBPβ-DNA/RNA heteroduplex oligonucleotide (HDO). Both approaches successfully reduced abnormal α-Syn expression in primary neurons treated with MPP+ . Furthermore, sustained activation of Nrf2 via its activator or inhibition of C/EBPβ using C/EBPβ-HDO resulted in a reduction of aberrant α-Syn expression, thus leading to an improvement in the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc) in mouse models induced by 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP) and those treated with preformed fibrils (PFFs). The data presented in this study illustrate that the activation of Nrf2 may provide a potential therapeutic strategy for PD by inhibiting the abnormal C/EBPβ/α-Syn signaling pathway.
Collapse
Affiliation(s)
- Zefang Lin
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Lixuan Huang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Qianqian Cao
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Hanyue Luo
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
| | - Wei Yao
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ji-Chun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
23
|
Kim JY, Lee HY, Lee SY, Kim SY, Park JL, Lee SD. DNA methylome profiling of blood to identify individuals in a pair of monozygotic twins. Genes Genomics 2023; 45:1273-1279. [PMID: 37198375 PMCID: PMC10504115 DOI: 10.1007/s13258-023-01396-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/30/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Short tandem repeat (STR) markers cannot be used to distinguish between genetically identical monozygotic (MZ) twins, causing problems in a case with an MZ twin as a suspect. Many studies have shown that in older MZ twins, there are significant differences in overall content and genomic distribution of methylation. OBJECTIVE In this study, we analyzed the DNA methylome profile of blood to identify recurrent differentially methylated CpG sites (DMCs) to discriminate between MZ twins. METHODS Blood samples were collected from 47 paired MZ twins. We performed the DNA methylation profiling using the HumanMethylation EPIC BeadChip platform and identified recurrent DMCs between MZ twins. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and motif enrichment analyses were performed to reveal the biological functions of recurrent DMCs. We collected DNA methylome data from the Gene Expression Omnibus (GEO) public database to verify the recurrent DMCs between MZ twins. RESULTS We identified recurrent DMCs between MZ twin samples and observed that they were enriched in immune-related genes. In addition, we verified our DMCs in a public dataset. CONCLUSION Our results suggest that the methylation level at recurrent DMCs between MZ twins may serve as a valuable biomarker for identification of individuals in a pair of MZ twins.
Collapse
Affiliation(s)
- Jae-Yoon Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, 34141, Korea
| | - Hwan Young Lee
- Department of Forensic Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
- Institute of Forensic and Anthropological Science, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - So-Yeon Lee
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Korea
| | - Seon-Young Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, 34141, Korea
| | - Jong-Lyul Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, 34141, Korea.
- Aging Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, 34141, Korea.
| | - Soong Deok Lee
- Department of Forensic Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Institute of Forensic and Anthropological Science, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
24
|
Xia Y, Xiao Y, Wang ZH, Liu X, Alam AM, Haran JP, McCormick BA, Shu X, Wang X, Ye K. Bacteroides Fragilis in the gut microbiomes of Alzheimer's disease activates microglia and triggers pathogenesis in neuronal C/EBPβ transgenic mice. Nat Commun 2023; 14:5471. [PMID: 37673907 PMCID: PMC10482867 DOI: 10.1038/s41467-023-41283-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Gut dysbiosis contributes to Alzheimer's disease (AD) pathogenesis, and Bacteroides strains are selectively elevated in AD gut microbiota. However, it remains unknown which Bacteroides species and how their metabolites trigger AD pathologies. Here we show that Bacteroides fragilis and their metabolites 12-hydroxy-heptadecatrienoic acid (12-HHTrE) and Prostaglandin E2 (PGE2) activate microglia and induce AD pathogenesis in neuronal C/EBPβ transgenic mice. Recolonization of antibiotics cocktail-pretreated Thy1-C/EBPβ transgenic mice with AD patient fecal samples elicits AD pathologies, associated with C/EBPβ/Asparaginyl endopeptidase (AEP) pathway upregulation, microglia activation, and cognitive disorders compared to mice receiving healthy donors' fecal microbiota transplantation (FMT). Microbial 16S rRNA sequencing analysis shows higher abundance of proinflammatory Bacteroides fragilis in AD-FMT mice. Active components characterization from the sera and brains of the transplanted mice revealed that both 12-HHTrE and PGE2 activate primary microglia, fitting with poly-unsaturated fatty acid (PUFA) metabolites enrichment identified by metabolomics. Strikingly, recolonization with live but not dead Bacteroides fragilis elicited AD pathologies in Thy1-C/EBPβ transgenic mice, so did 12-HHTrE or PGE2 treatment alone. Collectively, our findings support a causal role for Bacteroides fragilis and the PUFA metabolites in activating microglia and inducing AD pathologies in Thy1- C/EBPβ transgenic mice.
Collapse
Affiliation(s)
- Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China
| | - Yifan Xiao
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China
| | - Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ashfaqul M Alam
- University of Kentucky, Microbiology, Immunology & Molecular Genetics Office - MN 376, Medical Science Building, 800 Rose Street, Lexington, KY, USA
| | - John P Haran
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Beth A McCormick
- Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Xiji Shu
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China.
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neurodegeneration, Nantong University, Nantong, Jiangsu, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
25
|
Liu L, Yao D, Chen Z, Duan S. A comprehensive signature based on endoplasmic reticulum stress-related genes in predicting prognosis and immunotherapy response in melanoma. Sci Rep 2023; 13:8232. [PMID: 37217516 DOI: 10.1038/s41598-023-35031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
Melanoma is considered as one of the most invasion types of skin cancer with high mortality rates. Although combination of immune checkpoint therapy with local surgical excision provide a novel promising therapeutic strategies, the overall prognosis of melanoma patients remains unsatisfactory. Endoplasmic reticulum (ER) stress, a process of protein misfolding and undue accumulation, has been proven to play an indispensable regulatory role in tumor progression and tumor immunity. However, whether the signature based ER genes has predictive value for the prognosis and immunotherapy of melanoma has not been systematically manifested. In this study, the LASSO regression and multivariate Cox regression were applied to construct a novel signature for predicting melanoma prognosis both in the training and testing set. Intriguingly, we found that patients endowed with high- and low-risk scores displayed differences in clinicopathologic classification, immune cell infiltration level, tumor microenvironment, and immune checkpoint treatment response. Subsequently, based on molecular biology experiments, we validated that silencing the expression of RAC1, an ERG composed of the risk signature, could restrain the proliferation and migration, promote apoptosis, as well as increase the expression of PD-1/PD-L1 and CTLA4 in melanoma cells. Taken together, the risk signature was regarded as promising predictors for melanoma prognosis and might provide prospective strategies to ameliorate patients' response to immunotherapy.
Collapse
Affiliation(s)
- Longqing Liu
- Department of Otolaryngology Head and Neck Surgery, Enshi Prefecture Ethnic Hospital, 178 Hangkong Avenue, Enshi, Hubei Province, China
| | - Dilang Yao
- Department of Otolaryngology Head and Neck Surgery, Enshi Prefecture Ethnic Hospital, 178 Hangkong Avenue, Enshi, Hubei Province, China
| | - Zhiwei Chen
- Department of Otolaryngology Head and Neck Surgery, Enshi Prefecture Ethnic Hospital, 178 Hangkong Avenue, Enshi, Hubei Province, China.
| | - Shidong Duan
- Department of Otolaryngology Head and Neck Surgery, Enshi Prefecture Ethnic Hospital, 178 Hangkong Avenue, Enshi, Hubei Province, China.
| |
Collapse
|
26
|
Jeon KB, Kim J, Lim CM, Park JY, Kim NY, Lee J, Oh DK, Yoon DY. Unsaturated oxidated fatty acid 12(S)-HETE attenuates TNF-α expression in TNF-α/IFN-γ-stimulated human keratinocytes. Int Immunopharmacol 2023; 120:110298. [PMID: 37207444 DOI: 10.1016/j.intimp.2023.110298] [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: 01/22/2023] [Revised: 04/06/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Chronic skin inflammatory diseases are associated with abnormal immune responses characterized by skin barrier dysfunction. Keratinocytes participate in immune homeostasis regulated by immune cells. Immune homeostasis dysfunction contributes to the pathogenesis of skin diseases mediated by pro-inflammatory cytokines and chemokines, such as tumor necrosis factor (TNF)-α, which are produced by activated keratinocytes. 12(S)-Hydroxy eicosatetraenoic acid [12(S)-HETE], an arachidonic acid metabolite, has anti-inflammatory properties. However, the role of 12(S)-HETE in chronic skin inflammatory diseases has not been elucidated yet. In this study, we investigated the effect of 12(S)-HETE on TNF-α/interferon (IFN)-γ-induced pro-inflammatory cytokine and chemokine expression. Our data showed that 12(S)-HETE modulates TNF-α mRNA and protein expression in TNF-α-/IFN-γ-treated human keratinocytes. Molecular docking analyses demonstrated that 12(S)-HETE bound to extracellular signal-regulated kinase (ERK)1/2, thus preventing ERK activation and downregulating phosphorylated ERK expression. We also demonstrated that 12(S)-HETE treatment inhibited IκB and ERK phosphorylation and nuclear factor (NF)-κB, p65/p50, and CCAAT/enhancerbindingproteinβ (C/EBPβ) translocation. Overall, our results showed that 12(S)-HETE attenuated TNF-α expression and secretion by inhibiting the mitogen-activated protein kinase ERK/NF-κB and C/EBPβ signaling pathways. Overall, these results suggest that 12(S)-HETE effectively resolved TNF-α-induced inflammation.
Collapse
Affiliation(s)
- Kyeong-Bae Jeon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jinju Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Chae-Min Lim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Deok-Kun Oh
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
27
|
Jagoda E, Marnetto D, Senevirathne G, Gonzalez V, Baid K, Montinaro F, Richard D, Falzarano D, LeBlanc EV, Colpitts CC, Banerjee A, Pagani L, Capellini TD. Regulatory dissection of the severe COVID-19 risk locus introgressed by Neanderthals. eLife 2023; 12:e71235. [PMID: 36763080 PMCID: PMC9917435 DOI: 10.7554/elife.71235] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Individuals infected with the SARS-CoV-2 virus present with a wide variety of symptoms ranging from asymptomatic to severe and even lethal outcomes. Past research has revealed a genetic haplotype on chromosome 3 that entered the human population via introgression from Neanderthals as the strongest genetic risk factor for the severe response to COVID-19. However, the specific variants along this introgressed haplotype that contribute to this risk and the biological mechanisms that are involved remain unclear. Here, we assess the variants present on the risk haplotype for their likelihood of driving the genetic predisposition to severe COVID-19 outcomes. We do this by first exploring their impact on the regulation of genes involved in COVID-19 infection using a variety of population genetics and functional genomics tools. We then perform a locus-specific massively parallel reporter assay to individually assess the regulatory potential of each allele on the haplotype in a multipotent immune-related cell line. We ultimately reduce the set of over 600 linked genetic variants to identify four introgressed alleles that are strong functional candidates for driving the association between this locus and severe COVID-19. Using reporter assays in the presence/absence of SARS-CoV-2, we find evidence that these variants respond to viral infection. These variants likely drive the locus' impact on severity by modulating the regulation of two critical chemokine receptor genes: CCR1 and CCR5. These alleles are ideal targets for future functional investigations into the interaction between host genomics and COVID-19 outcomes.
Collapse
Affiliation(s)
- Evelyn Jagoda
- Department of Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Davide Marnetto
- Estonian Biocentre, Institute of Genomics, University of TartuTartuEstonia
| | - Gayani Senevirathne
- Department of Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Victoria Gonzalez
- Department of Veterinary Microbiology, University of SaskatchewanSaskatoonCanada
- Vaccine and Infectious Disease Organization, University of SaskatchewanSaskatoonCanada
| | - Kaushal Baid
- Vaccine and Infectious Disease Organization, University of SaskatchewanSaskatoonCanada
| | - Francesco Montinaro
- Estonian Biocentre, Institute of Genomics, University of TartuTartuEstonia
- Department of Biology, University of BariBariItaly
| | - Daniel Richard
- Department of Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Darryl Falzarano
- Department of Veterinary Microbiology, University of SaskatchewanSaskatoonCanada
- Vaccine and Infectious Disease Organization, University of SaskatchewanSaskatoonCanada
| | - Emmanuelle V LeBlanc
- Department of Biomedical and Molecular Sciences, Queen’s UniversityKingstonCanada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s UniversityKingstonCanada
| | - Arinjay Banerjee
- Department of Veterinary Microbiology, University of SaskatchewanSaskatoonCanada
- Vaccine and Infectious Disease Organization, University of SaskatchewanSaskatoonCanada
- Department of Biology, University of WaterlooWaterlooCanada
- Department of Laboratory Medicine and Pathobiology, University of TorontoTorontoCanada
| | - Luca Pagani
- Estonian Biocentre, Institute of Genomics, University of TartuTartuEstonia
- Department of Biology, University of PadovaPadovaItaly
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Broad Institute of MIT and HarvardCambridgeUnited States
| |
Collapse
|
28
|
Suzuki K, Shibato J, Rakwal R, Takaura M, Hotta R, Masuo Y. Biomarkers in the Rat Hippocampus and Peripheral Blood for an Early Stage of Mental Disorders Induced by Water Immersion Stress. Int J Mol Sci 2023; 24:ijms24043153. [PMID: 36834565 PMCID: PMC9960135 DOI: 10.3390/ijms24043153] [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: 12/19/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
It is difficult to evaluate the pre-symptomatic state of mental disorders and prevent its onset. Since stress could be a trigger of mental disorders, it may be helpful to identify stress-responsive biomarkers (stress markers) for the evaluation of stress levels. We have so far performed omics analyses of the rat brain and peripheral blood after various kinds of stress and have found numerous factors that respond to stress. In this study, we investigated the effects of relatively moderate stress on these factors in the rat to identify stress marker candidates. Adult male Wistar rats underwent water immersion stress for 12 h, 24 h, or 48 h. Stress caused weight loss and elevated serum corticosterone levels, and alterations regarded as anxiety and/or fear-like behaviors. Reverse-transcription PCR and Western blot analyses revealed significant alterations in the expressions of hippocampal genes and proteins by the stress for no longer than 24 h, such as mitogen-activated protein kinase phosphatase 1 (MKP-1), CCAAT/enhancer-binding protein delta (CEBPD), small ubiquitin-like modifier proteins 1/sentrin-specific peptidase 5 (SENP5), matrix metalloproteinase-8 (MMP-8), kinase suppressor of Ras 1 (KSR1), and MKP-1, MMP-8, nerve growth factor receptor (NGFR). Similar alterations were observed in three genes (MKP-1, CEBPD, MMP-8) in the peripheral blood. The present results strongly suggest that these factors may serve as stress markers. The correlation of these factors in the blood and brain may enable the evaluation of stress-induced changes in the brain by blood analysis, which will contribute to preventing the onset of mental disorders.
Collapse
Affiliation(s)
- Keisuke Suzuki
- Laboratory of Neuroscience, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi 274-8510, Japan
| | - Junko Shibato
- Department of Functional Morphology, Shonan University of Medical Sciences, 16-48 Kamishinano, Totsuka-ku, Yokohama 244-0806, Japan
| | - Randeep Rakwal
- Institute of Health and Sport Sciences and Tsukuba International Academy for Sport Studies (TIAS2.0), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8574, Japan
| | - Masahiko Takaura
- Laboratory of Neuroscience, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi 274-8510, Japan
| | - Ryotaro Hotta
- Laboratory of Neuroscience, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi 274-8510, Japan
| | - Yoshinori Masuo
- Laboratory of Neuroscience, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi 274-8510, Japan
- Correspondence:
| |
Collapse
|
29
|
Wang H, Chen G, Ahn EH, Xia Y, Kang SS, Liu X, Liu C, Han MH, Chen S, Ye K. C/EBPβ/AEP is age-dependently activated in Parkinson's disease and mediates α-synuclein in the gut and brain. NPJ Parkinsons Dis 2023; 9:1. [PMID: 36609384 PMCID: PMC9822984 DOI: 10.1038/s41531-022-00430-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/11/2022] [Indexed: 01/09/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder, and its pathologic hallmarks include extensive dopaminergic neuronal degeneration in the Substantia nigra associated with Lewy bodies, predominantly consisting of phosphorylated and truncated α-Synuclein (α-Syn). Asparagine endopeptidase (AEP) cleaves human α-Syn at N103 residue and promotes its aggregation, contributing to PD pathogenesis. However, how AEP mediates Lewy body pathologies during aging and elicits PD onset remains incompletely understood. Knockout of AEP or C/EBPβ from α-SNCA mice, and their chronic rotenone exposure models were used, and the mechanism of α-Syn from the gut that spread to the brain was observed. Here we report that C/EBPβ/AEP pathway, aggravated by oxidative stress, is age-dependently activated and cleaves α-Syn N103 and regulates Lewy body-like pathologies spreading from the gut into the brain in human α-SNCA transgenic mice. Deletion of C/EBPβ or AEP substantially diminished the oxidative stress, neuro-inflammation, and PD pathologies, attenuating motor dysfunctions in aged α-SNCA mice. Noticeably, PD pathologies initiate in the gut and progressively spread into the brain. Chronic gastric exposure to a low dose of rotenone initiates Lewy body-like pathologies in the gut that propagate into the brain in a C/EBPβ/AEP-dependent manner. Hence, our studies demonstrate that C/EBPβ/AEP pathway is critical for mediating Lewy body pathology progression in PD.
Collapse
Affiliation(s)
- Hualong Wang
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China ,grid.452458.aDepartment of Neurology, The First Hospital of Hebei Medical University (Department of Neurology, Hebei Hospital of Xuanwu Hospital Capital Medical University), Brain Aging and Cognitive Neuroscience Laboratory of Hebei Province, Neuromedical Technology Innovation Center of Hebei Province, Shijiazhuang, 050031 Hebei P. R. China ,grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Guiqin Chen
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.412632.00000 0004 1758 2270Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060 China
| | - Eun Hee Ahn
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.256753.00000 0004 0470 5964Department of Physiology, College of Medicine, Hallym University, Hallymdaehak-gil, Chuncheon-si, Gangwon-Do, 24252, South Korea
| | - Yiyuan Xia
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Seong Su Kang
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Xia Liu
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA
| | - Chang Liu
- grid.458489.c0000 0001 0483 7922CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China ,grid.458489.c0000 0001 0483 7922Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
| | - Ming-Hu Han
- grid.458489.c0000 0001 0483 7922Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China
| | - Shengdi Chen
- grid.16821.3c0000 0004 0368 8293Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Keqiang Ye
- grid.189967.80000 0001 0941 6502Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322 USA ,grid.458489.c0000 0001 0483 7922Department of Biology, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China
| |
Collapse
|
30
|
House JS, Gray S, Owen JR, Jima DD, Smart RC, Hall JR. C/EBPβ deficiency enhances the keratinocyte innate immune response to direct activators of cytosolic pattern recognition receptors. Innate Immun 2023; 29:14-24. [PMID: 37094088 PMCID: PMC10164275 DOI: 10.1177/17534259231162192] [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: 10/27/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 04/26/2023] Open
Abstract
The skin is the first line of defense to cutaneous microbes and viruses, and epidermal keratinocytes play a critical role in preventing infection by viruses and pathogens through activation of the type I interferon (IFN) response. Using RNAseq analysis, here we report that the conditional deletion of C/EBPβ transcription factor in mouse epidermis (CKOβ mice) resulted in the upregulation of IFNβ and numerous keratinocyte interferon-stimulated genes (ISGs). The expression of cytosolic pattern recognition receptors (cPRRs), that recognize viral RNA and DNA, were significantly increased, and enriched in the RNAseq data set. cPRRs stimulate a type I IFN response that can trigger cell death to eliminate infected cells. To determine if the observed increases in cPRRs had functional consequences, we transfected CKOβ primary keratinocytes with the pathogen and viral mimics poly(I:C) (dsRNA) or poly(dA:dT) (synthetic B-DNA) that directly activate PRRs. Transfected CKOβ primary keratinocytes displayed an amplified type I IFN response which was accompanied by increased activation of IRF3, enhanced ISG expression, enhanced activation of caspase-8, caspase-3 and increased apoptosis. Our results identify C/EBPβ as a critical repressor of the keratinocyte type I IFN response, and demonstrates that the loss of C/EBPβ primes keratinocytes to the activation of cytosolic PRRs by pathogen RNA and DNA to induce cell death mediated by caspase-8 and caspase-3.
Collapse
Affiliation(s)
- John S. House
- Center of Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
- Toxicology Graduate Program, North Carolina State University, Raleigh, NC, 27695, USA
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC 27709, USA
| | - Sophia Gray
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Jennifer R. Owen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Dereje D. Jima
- Center of Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA
| | - Robert C. Smart
- Center of Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
- Toxicology Graduate Program, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Jonathan R. Hall
- Center of Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
- Toxicology Graduate Program, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| |
Collapse
|
31
|
Xiao X, Cai W, Ding Z, Mao Z, Shi Y, Zhang Q. LincRNA00612 inhibits apoptosis and inflammation in LPS-induced BEAS-2B cells via enhancing interaction between p-STAT3 and A2M promoter. PeerJ 2023; 11:e14986. [PMID: 36883061 PMCID: PMC9985899 DOI: 10.7717/peerj.14986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been reported as key regulators of chronic obstructive pulmonary disease (COPD). This study aimed to figure out the regulatory mechanism as well as the effects of lncRNA00612 (LINC00612) in lipopolysaccharide (LPS)-induced inflammation and apoptosis in BEAS-2B cells. LINC00612 and its co-expressed gene alpha-2-macroglobulin (A2M) were strikingly downregulated in the peripheral venous blood of COPD patients. Overexpressed LINC00612 enhances BEAS-2B cells against apoptosis and inflammatory reactions mediated by LPS, however, an A2M knockdown can attenuate the degree of the enhancement. Bioinformatics analysis revealed putative binding sites between LINC00612, signal transducer and activator of transcription 3 (STAT3) and the A2M promoter, while RNA antisense purification and Chromatin immunoprecipitation were performed to confirm the prediction. Knockdown of LINC00612 impaired the binding of p-STAT3 to the promoter of A2M, which meant that LINC00612 was critical for the binding of STAT3 with the A2M promoter. Therefore, it can be concluded that LINC00612 ameliorates LPS-induced cell apoptosis and inflammation via recruiting STAT3 to bind to A2M. This conclusion will serve as a theoretical foundation for the treatment of COPD.
Collapse
Affiliation(s)
- Xinru Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China.,Department of the Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Wei Cai
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Zhengdao Mao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
32
|
Guindi C, Khan FU, Cloutier A, Khongorzul P, Raki AA, Gaudreau S, McDonald PP, Gris D, Amrani A. Inhibition of PI3K/C/EBPβ axis in tolerogenic bone marrow-derived dendritic cells of NOD mice promotes Th17 differentiation and diabetes development. Transl Res 2022; 255:37-49. [PMID: 36400308 DOI: 10.1016/j.trsl.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Dendritic cells (DCs) are key regulators of the adaptive immune response. Tolerogenic dendritic cells play a crucial role in inducing and maintaining immune tolerance in autoimmune diseases such as type 1 diabetes in humans as well as in the NOD mouse model. We previously reported that bone marrow-derived DCs (BM.DCs) from NOD mice, generated with a low dose of GM-CSF (GM/DCs), induce Treg differentiation and are able to protect NOD mice from diabetes. We had also found that the p38 MAPK/C/EBPβ axis is involved in regulating the phenotype, as well as the production of IL-10 and IL-12p70, by tolerogenic GM/DCs. Here, we report that the inhibition of the PI3K signaling switched the cytokine profile of GM/DCs toward Th17-promoting cytokines without affecting their phenotype. PI3K inhibition abrogated the production of IL-10 by GM/DCs, whereas it enhanced their production of IL-23 and TGFβ. Inhibition of PI3K signaling in tolerogenic GM/DCs also induced naive CD4+ T cells differentiation toward Th17 cells. Mechanistically, PI3K inhibition increased the DNA-binding activity of C/EBPβ through a GSK3-dependent pathway, which is important to maintain the semimature phenotype of tolerogenic GM/DCs. Furthermore, analysis of C/EBPβ-/- GM/DCs demonstrated that C/EBPβ is required for IL-23 production. Of physiological relevance, the level of protection from diabetes following transfusion of GM/DCs into young NOD mice was significantly reduced when NOD mice were transfused with GM/DCs pretreated with a PI3K inhibitor. Our data suggest that PI3K/C/EBPβ signaling is important in controlling tolerogenic function of GM/DCs by limiting their Th17-promoting cytokines.
Collapse
Affiliation(s)
- Chantal Guindi
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Farhan Ullah Khan
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Alexandre Cloutier
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Puregmaa Khongorzul
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Ahmed Aziz Raki
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Simon Gaudreau
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Patrick P McDonald
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Denis Gris
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Abdelaziz Amrani
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada.
| |
Collapse
|
33
|
Liu Y, Zienkiewicz J, Qiao H, Gibson-Corley KN, Boyd KL, Veach RA, Hawiger J. Genomic control of inflammation in experimental atopic dermatitis. Sci Rep 2022; 12:18891. [PMID: 36344555 PMCID: PMC9640569 DOI: 10.1038/s41598-022-23042-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Atopic Dermatitis (AD) or eczema, a recurrent allergic inflammation of the skin, afflicts 10-20% of children and 5% adults of all racial and ethnic groups globally. We report a new topical treatment of AD by a Nuclear Transport Checkpoint Inhibitor (NTCI), which targets two nuclear transport shuttles, importin α5 and importin β1. In the preclinical model of AD, induced by the active vitamin D3 analog MC903 (calcipotriol), NTCI suppressed the expression of keratinocyte-derived cytokine, Thymic Stromal Lymphopoietin (TSLP), the key gene in AD development. Moreover, the genes encoding mediators of TH2 response, IL-4 and its receptor IL-4Rα were also silenced together with the genes encoding cytokines IL-1β, IL-6, IL-13, IL-23α, IL-33, IFN-γ, GM-CSF, VEGF A, the chemokines RANTES and IL-8, and intracellular signal transducers COX-2 and iNOS. Consequently, NTCI suppressed skin infiltration by inflammatory cells (eosinophils, macrophages, and CD4 + T lymphocytes), and reduced MC903-evoked proliferation of Ki-67-positive cells. Thus, we highlight the mechanism of action and the potential utility of topical NTCI for treatment of AD undergoing Phase 1/2 clinical trial (AMTX-100 CF, NCT04313400).
Collapse
Affiliation(s)
- Yan Liu
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, 21St Avenue South, T-1218, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| | - Jozef Zienkiewicz
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, 21St Avenue South, T-1218, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| | - Huan Qiao
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, 21St Avenue South, T-1218, MCN, Nashville, TN, 37232, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ruth Ann Veach
- Department of Medicine, Division of Nephrology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jacek Hawiger
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, 21St Avenue South, T-1218, MCN, Nashville, TN, 37232, USA.
- Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
34
|
Darvishi E, Ghamsari L, Leong SF, Ramirez R, Koester M, Gallagher E, Yu M, Mason JM, Merutka G, Kappel BJ, Rotolo JA. Anticancer Activity of ST101, A Novel Antagonist of CCAAT/Enhancer Binding Protein β. Mol Cancer Ther 2022; 21:1632-1644. [PMID: 36121385 PMCID: PMC9630826 DOI: 10.1158/1535-7163.mct-21-0962] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/29/2022] [Accepted: 09/14/2022] [Indexed: 12/15/2022]
Abstract
CCAAT/enhancer binding protein β (C/EBPβ) is a basic leucine zipper (bZIP) family transcription factor, which is upregulated or overactivated in many cancers, resulting in a gene expression profile that drives oncogenesis. C/EBPβ dimerization regulates binding to DNA at the canonical TTGCGCAA motif and subsequent transcriptional activity, suggesting that disruption of dimerization represents a powerful approach to inhibit this previously "undruggable" oncogenic target. Here we describe the mechanism of action and antitumor activity of ST101, a novel and selective peptide antagonist of C/EBPβ that is currently in clinical evaluation in patients with advanced solid tumors. ST101 binds the leucine zipper domain of C/EBPβ, preventing its dimerization and enhancing ubiquitin-proteasome dependent C/EBPβ degradation. ST101 exposure attenuates transcription of C/EBPβ target genes, including a significant decrease in expression of survival, transcription factors, and cell-cycle-related proteins. The result of ST101 exposure is potent, tumor-specific in vitro cytotoxic activity in cancer cell lines including glioblastoma, breast, melanoma, prostate, and lung cancer, whereas normal human immune and epithelial cells are not impacted. Further, in mouse xenograft models ST101 exposure results in potent tumor growth inhibition or regression, both as a single agent and in combination studies. These data provide the First Disclosure of ST101, and support continued clinical development of ST101 as a novel strategy for targeting C/EBPβ-dependent cancers.
Collapse
Affiliation(s)
- Emad Darvishi
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Lila Ghamsari
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Siok F. Leong
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Ricardo Ramirez
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Mark Koester
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Erin Gallagher
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Miao Yu
- Department of Biology & Biochemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Jody M. Mason
- Department of Biology & Biochemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Gene Merutka
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Barry J. Kappel
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528
| | - Jim A. Rotolo
- Sapience Therapeutics, Inc. 500 Mamaroneck Ave. Suite 320, Harrison, NY 10528, Corresponding Author (, telephone: 914-607-6935)
| |
Collapse
|
35
|
Dickson MJ, Sheldon IM, Bromfield JJ. Lipopolysaccharide alters CEBPβ signaling and reduces estradiol production in bovine granulosa cells. CABI AGRICULTURE AND BIOSCIENCE 2022; 3:66. [PMID: 37576606 PMCID: PMC10419969 DOI: 10.1186/s43170-022-00133-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/27/2022] [Indexed: 08/15/2023]
Abstract
Background Bacterial infection of the uterus in postpartum dairy cows limits ovarian follicle growth, reduces blood estradiol concentrations, and leads to accumulation of bacterial lipopolysaccharide (LPS) in ovarian follicular fluid. Although treating granulosa cells with LPS in vitro decreases the expression of the estradiol synthesis enzyme CYP19A1 and reduces estradiol secretion, the molecular mechanisms are unclear. The transcription factor CCAAT enhancer binding protein beta (CEBPβ) not only facilitates the transcription of LPS regulated cytokines, but also binds to the promoter region of CYP19A1 in humans, mice, and buffalo. We hypothesized that LPS alters CEBPβ signaling to reduce CYP19A1 expression, resulting in decreased estradiol secretion. Methods Bovine granulosa cells were isolated from small/medium or large follicles and treated with LPS in the presence of FSH and androstenedione for up to 24 h. Results Treatment with LPS increased CXCL8 and IL6 gene expression and reduced estradiol secretion in granulosa cells from both small/medium and large follicles. However, LPS only reduced CYP19A1 expression in granulosa cells from large follicles. Treatment with LPS increased CEBPB expression and reduced CEBPβ nuclear localization in granulosa cells from small/medium follicles, but not granulosa cells from large follicles. Conclusions Although LPS reduces estradiol synthesis in bovine granulosa cells, the effects of LPS on CYP19A1 and CEBPβ are dependent on follicle size.
Collapse
Affiliation(s)
| | - I. Martin Sheldon
- Swansea University Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | |
Collapse
|
36
|
Xiong J, Liao J, Liu X, Zhang Z, Adams J, Pacifici R, Ye K. A TrkB agonist prodrug prevents bone loss via inhibiting asparagine endopeptidase and increasing osteoprotegerin. Nat Commun 2022; 13:4820. [PMID: 35973996 PMCID: PMC9381595 DOI: 10.1038/s41467-022-32435-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/26/2022] [Indexed: 11/12/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its tropomyosin-related kinase B receptor (TrkB) are expressed in human osteoblasts and mediate fracture healing. BDNF/TrkB signaling activates Akt that phosphorylates and inhibits asparagine endopeptidase (AEP), which regulates the differentiation fate of human bone marrow stromal cells (hBMSC) and is altered in postmenopausal osteoporosis. Here we show that R13, a small molecular TrkB receptor agonist prodrug, inhibits AEP and promotes bone formation. Though both receptor activator of nuclear factor kappa-Β ligand (RANK-L) and osteoprotegerin (OPG) induced by ovariectomy (OVX) remain comparable between WT and BDNF+/− mice, R13 treatment significantly elevates OPG in both mice without altering RANKL, blocking trabecular bone loss. Strikingly, both R13 and anti-RANK-L exhibit equivalent therapeutic efficacy. Moreover, OVX increases RANK-L and OPG in WT and AEP KO mice with RANK-L/OPG ratio lower in the latter than the former, attenuating bone turnover. 7,8-DHF, released from R13, activates TrkB and its downstream effector CREB, which is critical for OPG augmentation. Consequently, 7,8-DHF represses C/EBPβ/AEP pathway, inhibiting RANK-L-induced RAW264.7 osteoclastogenesis. Therefore, our findings support that R13 exerts its therapeutic efficacy toward osteoporosis via inhibiting AEP and escalating OPG. BDNS and TrkB are involved in bone fracture healing by inhibiting AEP. Here the authors show that a TrkB agonist prodrug can inhibit AEP and promote bone formation in osteoporotic mice.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology (SIAT) Shenzhen, Guangdong, PR China.
| |
Collapse
|
37
|
Kan M, Sun M, Jiang X, Diwadkar AR, Parikh V, Cao G, Gebski E, Jester W, Lan B, Panettieri RA, Koziol-White C, Lu Q, Himes BE. CEBPD modulates the airway smooth muscle transcriptomic response to glucocorticoids. Respir Res 2022; 23:193. [PMID: 35902923 PMCID: PMC9331514 DOI: 10.1186/s12931-022-02119-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CCAAT/Enhancer Binding Protein D (CEBPD), a pleiotropic glucocorticoid-responsive transcription factor, modulates inflammatory responses. Of relevance to asthma, expression of CEBPD in airway smooth muscle (ASM) increases with glucocorticoid exposure. We sought to characterize CEBPD-mediated transcriptomic responses to glucocorticoid exposure in ASM by measuring changes observed after knockdown of CEBPD and its impact on asthma-related ASM function. METHODS Primary ASM cells derived from four donors were transfected with CEBPD or non-targeting (NT) siRNA and exposed to vehicle control, budesonide (100 nM, 18 h), TNFα (10 ng/ml, 18 h), or both budesonide and TNFα. Subsequently, RNA-Seq was used to measure gene expression levels, and pairwise differential expression results were obtained for exposures versus vehicle and knockdown versus control conditions. Weighted gene co-expression analysis was performed to identify groups of genes with similar expression patterns across the various experimental conditions (i.e., CEBPD knockdown status, exposures). RESULTS CEBPD knockdown altered expression of 3037 genes under at least one exposure (q-value < 0.05). Co-expression analysis identified sets of 197, 152 and 290 genes that were correlated with CEBPD knockdown status, TNFα exposure status, and both, respectively. JAK-STAT signaling pathway genes, including IL6R and SOCS3, were among those influenced by both TNFα and CEBPD knockdown. Immunoblot assays revealed that budesonide-induced IL-6R protein expression and augmented IL-6-induced STAT3 phosphorylation levels were attenuated by CEBPD knockdown in ASM. CONCLUSIONS CEBPD modulates glucocorticoid responses in ASM, in part via modulation of IL-6 receptor signaling.
Collapse
Affiliation(s)
- Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Maoyun Sun
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xiaofeng Jiang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Avantika R Diwadkar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Vishal Parikh
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - William Jester
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Bo Lan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA.
| |
Collapse
|
38
|
Liao J, Chen G, Liu X, Wei ZZ, Yu SP, Chen Q, Ye K. C/EBPβ/AEP signaling couples atherosclerosis to the pathogenesis of Alzheimer's disease. Mol Psychiatry 2022; 27:3034-3046. [PMID: 35422468 PMCID: PMC9912845 DOI: 10.1038/s41380-022-01556-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/08/2022]
Abstract
Atherosclerosis (ATH) and Alzheimer's disease (AD) are both age-dependent inflammatory diseases, associated with infiltrated macrophages and vascular pathology and overlapping molecules. C/EBPβ, an Aβ or inflammatory cytokine-activated transcription factor, and AEP (asparagine endopeptidase) are intimately implicated in both ATH and AD; however, whether C/EBPβ/AEP signaling couples ATH to AD pathogenesis remains incompletely understood. Here we show that C/EBPβ/AEP pathway mediates ATH pathology and couples ATH to AD. Deletion of C/EBPβ or AEP from primary macrophages diminishes cholesterol load, and inactivation of this pathway reduces foam cell formation and lesions in aorta in ApoE-/- mice, fed with HFD (high-fat-diet). Knockout of ApoE from 3xTg AD mouse model augments serum LDL and increases lesion areas in the aorta. Depletion of C/EBPβ or AEP from 3xTg/ApoE-/- mice substantially attenuates these effects and elevates cerebral blood flow and vessel length, improving cognitive functions. Strikingly, knockdown of ApoE from the hippocampus of 3xTg mice decreases the cerebral blood flow and vessel length and aggravates AD pathologies, leading to cognitive deficits. Inactivation of C/EBPβ/AEP pathway alleviates these events and restores cognitive functions. Hence, our findings demonstrate that C/EBPβ/AEP signaling couples ATH to AD via mediating vascular pathology.
Collapse
Affiliation(s)
- Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Guiqin Chen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
39
|
Feng QL, Gu JJ, Chen JY, Zheng WY, Pan HH, Xu XY, Deng CC, Yang B. TSP1 promotes fibroblast proliferation and extracellular matrix deposition via the IL6/JAK2/STAT3 signalling pathway in keloids. Exp Dermatol 2022; 31:1533-1542. [PMID: 35661430 DOI: 10.1111/exd.14623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
Keloids are benign fibroproliferative diseases with abnormally proliferated bulges beyond the edge of the skin lesions, and they are characterized by uncontrolled fibroblast proliferation and excessive extracellular matrix deposition in the dermis. However, the definite mechanisms that increase fibroblast proliferation and collagen deposition in keloids remain unclear. Thrombospondin 1 (TSP1) has been suggested to play an important role in wound healing and fibrotic disorders, but its role in keloids is unknown. In this study, we aimed to clarify the specific role of TSP1 in keloids and explore the potential mechanism. Our results demonstrated that TSP1 was highly expressed in keloid lesions compared to normal skin. Knockdown of TSP1 in keloid fibroblasts decreased cell proliferation and collagen I deposition. Exogenous TSP1 treatment increased cell proliferation and collagen I deposition in normal fibroblasts. We further investigated the underlying mechanism and found that TSP1 promoted fibroblast proliferation and extracellular matrix deposition by upregulating the IL6/JAK2/STAT3 pathway. Moreover, we verified that TSP1 expression was positively correlated with IL6/STAT3 signalling activity in keloids. Taken together, our findings indicate that TSP1 promotes keloid development via the IL6/JAK2/STAT3 signalling pathway and blocking TSP1 may represent a potential strategy for keloid therapy.
Collapse
Affiliation(s)
- Qing-Lan Feng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Jing Gu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Yi Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Yue Zheng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Hui-Hui Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xue-Yan Xu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Cheng-Cheng Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Gao X, Zhao D, Han J, Zhang Z, Wang Z. Identification of microRNA-mRNA-TF regulatory networks in periodontitis by bioinformatics analysis. BMC Oral Health 2022; 22:118. [PMID: 35397550 PMCID: PMC8994180 DOI: 10.1186/s12903-022-02150-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/24/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Periodontitis is a complex infectious disease with various causes and contributing factors. The aim of this study was to identify key genes, microRNAs (miRNAs) and transcription factors (TFs) and construct a miRNA-mRNA-TF regulatory networks to investigate the underlying molecular mechanism in periodontitis. METHODS The GSE54710 miRNA microarray dataset and the gene expression microarray dataset GSE16134 were downloaded from the Gene Expression Omnibus database. The differentially expressed miRNAs (DEMis) and mRNAs (DEMs) were screened using the "limma" package in R. The intersection of the target genes of candidate DEMis and DEMs were considered significant DEMs in the regulatory network. Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted. Subsequently, DEMs were uploaded to the STRING database, a protein-protein interaction (PPI) network was established, and the cytoHubba and MCODE plugins were used to screen out key hub mRNAs and significant modules. Ultimately, to investigate the regulatory network underlying periodontitis, a global triple network including miRNAs, mRNAs, and TFs was constructed using Cytoscape software. RESULTS 8 DEMis and 121 DEMs were found between the periodontal and control groups. GO analysis showed that mRNAs were most significantly enriched in positive regulation of the cell cycle, and KEGG pathway analysis showed that mRNAs in the regulatory network were mainly involved in the IL-17 signalling pathway. A PPI network was constructed including 81 nodes and 414 edges. Furthermore, 12 hub genes ranked by the top 10% genes with high degree connectivity and five TFs, including SRF, CNOT4, SIX6, SRRM3, NELFA, and ONECUT3, were identified and might play crucial roles in the molecular pathogenesis of periodontitis. Additionally, a miRNA-mRNA-TF coregulatory network was established. CONCLUSION In this study, we performed an integrated analysis based on public databases to identify specific TFs, miRNAs, and mRNAs that may play a pivotal role in periodontitis. On this basis, a TF-miRNA-mRNA network was established to provide a comprehensive perspective of the regulatory mechanism networks of periodontitis.
Collapse
Affiliation(s)
- Xiaoli Gao
- Department of Stomatology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gongti Nan Lu, Chaoyang District, Beijing, 100020 China
| | - Dong Zhao
- Department of Stomatology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gongti Nan Lu, Chaoyang District, Beijing, 100020 China
| | - Jing Han
- Department of Stomatology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gongti Nan Lu, Chaoyang District, Beijing, 100020 China
| | - Zheng Zhang
- Department of Periodontology, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, 75 Dagu Bei Lu, Heping District, Tianjin, 300041 China
| | - Zuomin Wang
- Department of Stomatology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gongti Nan Lu, Chaoyang District, Beijing, 100020 China
| |
Collapse
|
41
|
Gilbert S, Péant B, Malaquin N, Tu V, Fleury H, Leclerc-Desaulniers K, Rodier F, Mes-Masson AM, Saad F. Targeting IKKε in Androgen-Independent Prostate Cancer Causes Phenotypic Senescence and Genomic Instability. Mol Cancer Ther 2022; 21:407-418. [PMID: 34965959 PMCID: PMC9377745 DOI: 10.1158/1535-7163.mct-21-0519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/12/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023]
Abstract
Advanced prostate cancer will often progress to a lethal, castration-resistant state. We previously demonstrated that IKKε expression correlated with the aggressiveness of prostate cancer disease. Here, we address the potential of IKKε as a therapeutic target in prostate cancer. We examined cell fate decisions (proliferation, cell death, and senescence) in IKKε-depleted PC-3 cells, which exhibited delayed cell proliferation and a senescent phenotype, but did not undergo cell death. Using IKKε/TBK1 inhibitors, BX795 and Amlexanox, we measured their effects on cell fate decisions in androgen-sensitive prostate cancer and androgen-independent prostate cancer cell lines. Cell-cycle analyses revealed a G2-M cell-cycle arrest and a higher proportion of cells with 8N DNA content in androgen-independent prostate cancer cells only. Androgen-independent prostate cancer cells also displayed increased senescence-associated (SA)-β-galactosidase activity; increased γH2AX foci; genomic instability; and altered p15, p16, and p21 expression. In our mouse model, IKKε inhibitors also decreased tumor growth of androgen-independent prostate cancer xenografts but not 22Rv1 androgen-sensitive prostate cancer xenografts. Our study suggests that targeting IKKε with BX795 or Amlexanox in androgen-independent prostate cancer cells induces a senescence phenotype and demonstrates in vivo antitumor activity. These results strengthen the potential of exploiting IKKε as a therapeutic target.
Collapse
Affiliation(s)
- Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Véronique Tu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada.,Département de Radiologie, Radio-oncologie et Médicine Nucléaire, Université de Montréal, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada.,Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Corresponding Author: Anne-Marie Mes-Masson, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada. Phone: 514-890-8000, ext. 25496; E-mail:
| | - Fred Saad
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM) et Institut du cancer de Montréal, Montréal, Quebec, Canada.,Department of Surgery, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Tsuruya Y, Yamaguchi A, Yamazaki-Takai M, Zhenyu J, Takai H, Nakayama Y, Ogata Y. Interleukin-1β regulates odontogenic ameloblast-associated protein gene transcription in human gingival epithelial cells. Odontology 2022; 110:557-568. [PMID: 35179670 DOI: 10.1007/s10266-022-00689-6] [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: 09/20/2021] [Accepted: 01/30/2022] [Indexed: 11/30/2022]
Abstract
Junction epithelium (JE) is located apical to the bottom of the gingival sulcus and binds enamel to hemidesmosomes to protect the periodontal tissue from bacterial infection. Function of odontogenic ameloblast-associated protein (ODAM) is suggested by its expression sites (JE and maturation stage ameloblasts) to be involved in the adhesion between the JE and enamel, and odontogenesis. To analyze the changes in ODAM gene and protein expressions in inflamed gingiva, Ca9-22 gingival epithelial cells were stimulated with 1 ng/ml interleukin-1β (IL-1β) for 3-24 h, and ODAM mRNA and protein levels were analyzed by real-time PCR and Western blotting. Luciferase (LUC) constructs were made ligating various lengths of human ODAM gene promoters and performed LUC analyses in Ca9-22 cells. Gel shift and chromatin immunoprecipitation (ChIP) assays were performed. IL-1β induced ODAM mRNA and protein levels at 6-24 h. IL-1β increased LUC activities of the ODAM gene promoter constructs from - 85 to - 950. These activities were blocked by protein kinase A, tyrosine kinase, mitogen-activated protein (MAP) kinase kinase and phosphoinositide 3-kinase inhibitors. Gel shift and ChIP assays showed that IL-1β induced CCAAT/enhancer-binding protein (C/EBP) β and Yin Yang1 (YY1) binding to C/EBP1, 2, 3, and YY1 elements. These data indicate that IL-1β stimulates ODAM gene transcription mediated through C/EBP1, C/EBP2, C/EBP3, and YY1 elements in the human ODAM gene promoter.
Collapse
Affiliation(s)
- Yuto Tsuruya
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Arisa Yamaguchi
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Mizuho Yamazaki-Takai
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Jin Zhenyu
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Hideki Takai
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Yohei Nakayama
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
| | - Yorimasa Ogata
- Departments of Periodontology, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan. .,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan.
| |
Collapse
|
43
|
Okazaki K, Anzawa H, Katsuoka F, Kinoshita K, Sekine H, Motohashi H. CEBPB is Required for NRF2-Mediated Drug Resistance in NRF2-Activated Non-Small Cell Lung Cancer Cells. J Biochem 2022; 171:567-578. [PMID: 35137113 DOI: 10.1093/jb/mvac013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/03/2022] [Indexed: 11/12/2022] Open
Abstract
NRF2 is a transcription activator that plays a key role in cytoprotection against oxidative stress. While increased NRF2 activity is principally beneficial for our health, NRF2 activation in cancer cells is detrimental, as it drives their malignant progression. We previously found that CEBPB cooperates with NRF2 in NRF2-activated lung cancer and enhances tumor-initiating activity by promoting NOTCH3 expression. However, the general contribution of CEBPB in lung cancer is rather controversial, probably because the role of CEBPB depends on cooperating transcription factors in each cellular context. To understand how NRF2 shapes the function of CEBPB in NRF2-activated lung cancers and its biological consequence, we comprehensively explored NRF2-CEBPB-coregulated genes and found that genes involved in drug metabolism and detoxification were characteristically enriched. Indeed, CEBPB and NRF2 cooperatively contribute to the drug resistance. We also found that CEBPB is directly regulated by NRF2, which is likely to be advantageous for the coexpression and cooperative function of NRF2 and CEBPB. These results suggest that drug resistance of NRF2-activated lung cancers is achieved by the cooperative function of NRF2 and CEBPB.
Collapse
Affiliation(s)
- Keito Okazaki
- Department of Gene Expression Regulation and 6Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Hayato Anzawa
- Department of System Bioinformatics, Graduate School of Information Sciences, Tohoku University Sendai 980-8579, Japan
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization Tohoku University, Sendai 980-8573, Japan
| | - Kengo Kinoshita
- Department of System Bioinformatics, Graduate School of Information Sciences, Tohoku University Sendai 980-8579, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank Organization Tohoku University, Sendai 980-8573, Japan
| | - Hiroki Sekine
- Department of Gene Expression Regulation and 6Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation and 6Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
44
|
Fabian P, Tseng KC, Thiruppathy M, Arata C, Chen HJ, Smeeton J, Nelson N, Crump JG. Lifelong single-cell profiling of cranial neural crest diversification in zebrafish. Nat Commun 2022; 13:13. [PMID: 35013168 PMCID: PMC8748784 DOI: 10.1038/s41467-021-27594-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/30/2021] [Indexed: 01/13/2023] Open
Abstract
The cranial neural crest generates a huge diversity of derivatives, including the bulk of connective and skeletal tissues of the vertebrate head. How neural crest cells acquire such extraordinary lineage potential remains unresolved. By integrating single-cell transcriptome and chromatin accessibility profiles of cranial neural crest-derived cells across the zebrafish lifetime, we observe progressive and region-specific establishment of enhancer accessibility for distinct fates. Neural crest-derived cells rapidly diversify into specialized progenitors, including multipotent skeletal progenitors, stromal cells with a regenerative signature, fibroblasts with a unique metabolic signature linked to skeletal integrity, and gill-specific progenitors generating cell types for respiration. By retrogradely mapping the emergence of lineage-specific chromatin accessibility, we identify a wealth of candidate lineage-priming factors, including a Gata3 regulatory circuit for respiratory cell fates. Rather than multilineage potential being established during cranial neural crest specification, our findings support progressive and region-specific chromatin remodeling underlying acquisition of diverse potential.
Collapse
Affiliation(s)
- Peter Fabian
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Kuo-Chang Tseng
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Mathi Thiruppathy
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Claire Arata
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Hung-Jhen Chen
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Joanna Smeeton
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Nellie Nelson
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - J Gage Crump
- Eli and Edythe Broad California Institute for Regenerative Medicine Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA.
| |
Collapse
|
45
|
Liu L, Zhong J, Chen B, Wang W, Xi H, Su X. CCAAT/enhancer binding protein (C/EBP) delta promotes the expression of PTX3 and macrophage phagocytosis during A. fumigatus infection. J Leukoc Biol 2021; 111:1225-1234. [PMID: 34939225 DOI: 10.1002/jlb.4ma1121-451rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 12/22/2022] Open
Abstract
Given the increasing incidence of pulmonary aspergillosis, it is important to understand the natural defense mechanisms by which the body can kill Aspergillus fumigatus conidia. Pentraxin 3 (PTX3) plays a nonredundant role in resistance to A. fumigatus. Here, we found that the key predicted PTX3 transcription factor, CCAAT/enhancer-binding protein δ (CEBPD), was up-regulated during A. fumigatus conidia infection. Functionally, CEBPD significantly promoted the expression of PTX3 and the phagocytic ability of macrophages. Mechanistically, CEBPD activated the PTX3 by directly binding to the promoter region of the PTX3 gene. We also showed that the RNA-binding protein human antigen R promoted CEBPD expression. These findings provide new insights into the crucial role of CEBPD in the phagocytosis of A. fumigatus conidia by macrophages and highlight this protein as a potential therapeutic target for invasive pulmonary aspergillosis.
Collapse
Affiliation(s)
- Lulu Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jinjin Zhong
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bilin Chen
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiping Wang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing, China
| | - Haiyan Xi
- Department of Clinical Laboratory, Jinling Hospital, Nanjing, China
| | - Xin Su
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Clinical Laboratory, Jinling Hospital, Nanjing, China
| |
Collapse
|
46
|
Mitochondrial dysfunction triggers the pathogenesis of Parkinson's disease in neuronal C/EBPβ transgenic mice. Mol Psychiatry 2021; 26:7838-7850. [PMID: 34489530 DOI: 10.1038/s41380-021-01284-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/03/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
Respiratory chain complex I deficiency elicits mitochondrial dysfunction and reactive oxidative species (ROS), which plays a crucial role in Parkinson's disease (PD) pathogenesis. However, it remains unclear whether the impairment in other complexes in the mitochondrial oxidative phosphorylation chain is also sufficient to trigger PD onset. Here we show that inhibition of Complex II or III in the electron transport chain (ETC) induces the motor disorder and PD pathologies in neuronal Thy1-C/EBPβ transgenic mice. Through a cell-based screening of mitochondrial respiratory chain inhibitors, we identified TTFA (complex II inhibitor) and Atovaquone (complex III inhibitor), which robustly block the oxidative phosphorylation functions, strongly escalate ROS, and activate C/EBPβ/AEP pathway that triggers dopaminergic neuronal cell death. Oral administration of these inhibitors to Thy1-C/EBPβ mice elicits constipation and motor defects, associated with Lewy body-like inclusions. Deletion of SDHD (Succinate dehydrogenase) gene from the complex II in the Substantia Nigra of Thy1-C/EBPβ mice triggers ROS and PD pathologies, resulting in motor disorders. Hence, our findings demonstrate that mitochondrial ETC inactivation triggers PD pathogenesis via activating C/EBPβ/AEP pathway.
Collapse
|
47
|
Chu S, Ma L, Wu Y, Zhao X, Xiao B, Pan Q. C-EBPβ mediates in cigarette/IL-17A-induced bronchial epithelial-mesenchymal transition in COPD mice. BMC Pulm Med 2021; 21:376. [PMID: 34794427 PMCID: PMC8603568 DOI: 10.1186/s12890-021-01738-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/05/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cigarettes smoking and IL-17A contribute to chronic obstructive pulmonary disease (COPD), and have synergistical effect on bronchial epithelial cell proliferation. CCAAT/enhancer-binding protein β (C-EBPβ) could be induced by IL-17A and is up-regulated in COPD. We explored the effect of cigarettes and IL-17 on bronchial epithelial-mesenchymal transition (EMT) in COPD mice and potential mechanism involved with C-EBPβ in this study. METHODS COPD model was established with mice by exposing to cigarettes. E-Cadherin, Vimentin, IL-17A and C-EBPβ distributions were detected in lung tissues. Primary bronchial epithelial cells were separated from health mice and cocultured with cigarette smoke extract (CSE) or/and IL-17A. E-Cadherin, Vimentin and IL-17 receptor (IL-17R) expressions in vitro were assessed. When C-EBPβ were silenced by siRNA in cells, E-Cadherin, Vimentin and C-EBPβ expressions were detected. RESULTS E-Cadherin distribution was less and Vimentin distribution was more in bronchus of COPD mice than controls. IL-17A and C-EBPβ expressions were higher in lung tissues of COPD mice than controls. In vitro, C-EBPβ protein expression was highest in CSE + IL-17A group, followed by CSE and IL-17A groups. E-cadherin expression in vitro was lowest and Vimentin expression was highest in CSE + IL-17A group, followed by CSE or IL-17A group. Those could be inhibited by C-EBPβ silenced. CONCLUSIONS C-EBPβ mediates in cigarette/IL-17A-induced bronchial EMT in COPD mice. Our findings contribute to a better understanding on the progress from COPD to lung cancers, which will provide novel avenues in preventing tumorigenesis of airway in the context of cigarette smoking.
Collapse
Affiliation(s)
- Shuyuan Chu
- Laboratory of Respiratory Disease, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Yashan Wu
- Guangxi University of Chinese Medicine, Nanning, 530222, Guangxi, China
| | - Xiaoli Zhao
- Laboratory of Respiratory Disease, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Bo Xiao
- Laboratory of Respiratory Disease, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qilu Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| |
Collapse
|
48
|
Tsuruya Y, Yamaguchi A, Yamazaki-Takai M, Mezawa M, Takai H, Nakayama Y, Ogata Y. Transcriptional regulation of human odontogenic ameloblast-associated protein gene by tumor necrosis factor-α. Inflamm Res 2021; 71:119-129. [PMID: 34787682 DOI: 10.1007/s00011-021-01523-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/23/2021] [Accepted: 11/03/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Odontogenic ameloblast-associated protein (ODAM) is produced by maturation stage ameloblasts and junctional epithelium (JE). The function of ODAM is thought to be involved in the attachment of teeth and JE. To elucidate transcriptional regulation of human ODAM gene in inflamed gingiva, we have analyzed the effects of TNF-α on the expression of ODAM gene in Ca9-22 and Sa3 gingival epithelial cells. MATERIALS AND METHODS Total RNAs were extracted from Ca9-22 and Sa3 cells after stimulation by TNF-α (10 ng/ml). ODAM mRNA and protein levels were analyzed by qPCR and Western blotting. Luciferase (LUC) analyses were performed using LUC constructs inserted in various lengths of ODAM gene promoter. Gel shift and chromatin immunoprecipitation (ChIP) assays were carried out. RESULTS TNF-α increased ODAM mRNA and protein levels at 3 to 24 h. TNF-α induced LUC activities of the ODAM gene promoter constructs, and the activities were inhibited by protein kinase A, tyrosine kinase, MEK1/2, PI3-kinase and NF-κB inhibitors. Gel shift and ChIP assays revealed that TNF-α increased CCAAT/enhancer-binding protein (C/EBP) β and Yin Yang1 (YY1) binding to three kinds of C/EBPs and YY1 elements. CONCLUSION These results demonstrate that TNF-α stimulates ODAM gene transcription via C/EBPs and YY1 elements in the human ODAM gene promoter.
Collapse
Affiliation(s)
- Yuto Tsuruya
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Arisa Yamaguchi
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Mizuho Yamazaki-Takai
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Masaru Mezawa
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Hideki Takai
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yohei Nakayama
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yorimasa Ogata
- Department of Periodontology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan. .,Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo, Japan.
| |
Collapse
|
49
|
Chera JS, Kumar S, Vats A, Kushwaha P, Behera M, De S. PU.1 is involved in the transcriptional up-regulation of RNA and DNA sensing pathway genes in buffalo fibroblasts. Vet Immunol Immunopathol 2021; 242:110349. [PMID: 34695651 DOI: 10.1016/j.vetimm.2021.110349] [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: 06/29/2021] [Revised: 09/26/2021] [Accepted: 10/17/2021] [Indexed: 11/30/2022]
Abstract
PU.1, CEBPA, and CEBPB are Lineage Determining Transcription Factors (LDTFs) that play roles in biological processes such as cell differentiation and the immune system regulation including the innate immune pathways. The roles of these LDTFs in the innate RNA and DNA sensing pathways have received little attention. We show that in buffalo fibroblasts, PU.1 causes the mRNA up-regulation of the RNA and DNA sensors such as RIG-I (65.1 fold), MDA5 (20.4 fold), IFI16-l (8.0 fold), and cGAS (60.5 fold) while CEBPA does the same but to a lesser extent (RIG-I-26.4 fold, MDA5-10.8 fold, IFI16-l- 3.3 fold and cGAS-8.6 fold). CEBPB does not appear to have a role in the up-regulation of these genes. PU.1 expression also primes the cells to develop a strong immune response against the dsRNA virus mimic polyinosinic:polycytidylic acid (poly I:C) by significantly up-regulating Interferon-β (14.9 fold change with p-value <0.0001). CEBPA up-regulates Interferon-β to a lower level than PU.1 (4.7 fold change with p-value 0.0024), whereas CEBPB exhibits non-significant up-regulation (2.1 fold with p-value of 0.1449). As PU.1 robustly up-regulates the nucleic acid sensing pathways, it can prove to be useful in improving the defence against viruses that can cause losses to animal husbandry.
Collapse
Affiliation(s)
- Jatinder Singh Chera
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Sushil Kumar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Ashutosh Vats
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Parmanand Kushwaha
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Manisha Behera
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Sachinandan De
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana, India.
| |
Collapse
|
50
|
Spek CA, Aberson HL, Butler JM, de Vos AF, Duitman J. CEBPD Potentiates the Macrophage Inflammatory Response but CEBPD Knock-Out Macrophages Fail to Identify CEBPD-Dependent Pro-Inflammatory Transcriptional Programs. Cells 2021; 10:cells10092233. [PMID: 34571881 PMCID: PMC8470509 DOI: 10.3390/cells10092233] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 12/25/2022] Open
Abstract
CCAAT/enhancer-binding protein delta (C/EBPδ) is a member of the C/EBP family of transcription factors. According to the current paradigm, C/EBPδ potentiates cytokine production and modulates macrophage function thereby enhancing the inflammatory response. Remarkably, however, C/EBPδ deficiency does not consistently lead to a reduction in Lipopolysaccharide (LPS)-induced cytokine production by macrophages. Here, we address this apparent discrepancy and show that the effect of C/EBPδ on cytokine production and macrophage function depends on both the macrophage subtype and the LPS concentration used. Using CRISPR-Cas generated macrophages in which the transactivation domain of C/EBPδ was deleted from the endogenous locus (ΔTAD macrophages), we next show that the context-dependent role of C/EBPδ in macrophage biology relies on compensatory transcriptional activity in the absence of C/EBPδ. We extend these findings by revealing a large discrepancy between transcriptional programs in C/EBPδ knock-out and C/EBPδ transactivation dead (ΔTAD) macrophages implying that compensatory mechanisms do not specifically modify C/EBPδ-dependent inflammatory responses but affect overall macrophage biology. Overall, these data imply that knock-out approaches are not suited for identifying the genuine transcriptional program regulated by C/EBPδ, and we suggest that this phenomenon applies for transcription factor families in general.
Collapse
Affiliation(s)
- C. Arnold Spek
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (H.L.A.); (J.M.B.); (A.F.d.V.); (J.D.)
- Correspondence:
| | - Hella L. Aberson
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (H.L.A.); (J.M.B.); (A.F.d.V.); (J.D.)
| | - Joe M. Butler
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (H.L.A.); (J.M.B.); (A.F.d.V.); (J.D.)
| | - Alex F. de Vos
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (H.L.A.); (J.M.B.); (A.F.d.V.); (J.D.)
| | - JanWillem Duitman
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (H.L.A.); (J.M.B.); (A.F.d.V.); (J.D.)
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|