1
|
Yu L, Shi H, Gao T, Xu W, Qian H, Jiang J, Yang X, Zhang X. Exomeres and supermeres: Current advances and perspectives. Bioact Mater 2025; 50:322-343. [PMID: 40276541 PMCID: PMC12020890 DOI: 10.1016/j.bioactmat.2025.04.012] [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: 01/26/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Recent studies have revealed a great diversity and complexity in extracellular vesicles and particles (EVPs). The developments in techniques and the growing awareness of the particle heterogeneity have spurred active research on new particle subsets. Latest discoveries highlighted unique features and roles of non-vesicular extracellular nanoparticles (NVEPs) as promising biomarkers and targets for diseases. These nanoparticles are distinct from extracellular vesicles (EVs) in terms of their smaller particle sizes and lack of a bilayer membrane structure and they are enriched with diverse bioactive molecules particularly proteins and RNAs, which are widely reported to be delivered and packaged in exosomes. This review is focused on the two recently identified membraneless NVEPs, exomeres and supermeres, to provide an overview of their biogenesis and contents, particularly those bioactive substances linked to their bio-properties. This review also explains the concepts and characteristics of these nanoparticles, to compare them with other EVPs, especially EVs, as well as to discuss their isolation and identification methods, research interests, potential clinical applications and open questions.
Collapse
Affiliation(s)
- Li Yu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Tingxin Gao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
2
|
Wang B, Pi Q, Mohsin A, Gao WQ, Guo M, Xu H. Exosomal miR-1246 of adipose stem cells attenuates obesity by polarizing M2 macrophages, reducing fat mass, and beiging of white adipose tissue. J Adv Res 2025:S2090-1232(25)00369-8. [PMID: 40419017 DOI: 10.1016/j.jare.2025.05.049] [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: 01/29/2025] [Revised: 03/28/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025] Open
Abstract
INTRODUCTION Adipose stem cells (ADSC) have demonstrated therapeutic potential in ameliorating obesity and metabolic disorders, with their exosomes showing comparable therapeutic effects. However, the underlying molecular mechanism remains incompletely understood. Furthermore, the limited availability and inherent heterogeneity of primary ADSC present substantial challenges for consistent therapeutic outcomes. OBJECTIVE This study aimed to investigate the molecular mechanism underlying the unique biological effects mediated by microRNAs (miRNAs) in exosomes derived from immortalized adipose stem cells (iADSC). METHODS AND RESULTS We first established stable iADSC and isolated their exosomes (iADSC-EXO), which exhibited both high yield and stability. In high fat diet (HFD)-fed obese mice, iADSC-EXO administration significantly attenuated obesity, reduced blood glucose and lipid levels, and alleviated hepatic steatosis. miRNA chips analysis revealed that miR-1246 is highly enriched in exosomes derived from iADSC and ADSC. Administration of miR-1246 to HFD-fed obese mice produced beneficial effects on obesity and metabolic disorders comparable to those with iADSC-EXO. Mechanistic studies revealed that miR-1246 exerts its beneficial effects through multiple pathways: First, reducing fat mass by downregulating of the expression of fat mass and obesity-associate protein (FTO) and subsequent inhibition of adipogenesis and lipogenesis. Second, enhancing white adipose tissue (WAT) beiging by suppressing Runt-related transcription factor 1 (RUNX1T1) and FTO expression. Third, promoting M2 macrophage polarization and suppressing WAT inflammation via inhibition of TNF receptor-associated factor 6 (TRAF6)-mediated inflammatory pathway. CONCLUSION Our study elucidates a novel molecular mechanism through which ADSC regulates adipose tissue homeostasis and metabolism and presents a potential therapeutic approach for treating obesity and related metabolic disorders via iADSC-EXO or miR-1246-based interventions.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qingmeng Pi
- Department of Plastic Surgery, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wei-Qiang Gao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Clinical Stem Cell Research Center, RenJi Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
3
|
Jiang B, Bai F, Hu Y, Ren Y, Su Y, Song W, Xie K, Wang D, Pan J, Liu Y, Feng Y, Li X, Zhang H, Zhu X, Bai H, Yang Q, Ben J, Chen Q. Endothelial major vault protein alleviates vascular remodeling via promoting Parkin-mediated mitophagy. Nat Commun 2025; 16:4365. [PMID: 40348769 PMCID: PMC12065841 DOI: 10.1038/s41467-025-59644-y] [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: 06/23/2024] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Many important vascular diseases including neointimal hyperplasia and atherosclerosis are characterized by the endothelial cell (EC) injury-initiated pathological vascular remodeling. However, the endogenous regulatory mechanisms underlying it are not fully understood. The present study investigates regulatory role of major vault protein (MVP) in the pathogenesis of vascular remodeling via controlling EC injury. By generating male murine vascular disease models, we find that ablation of endothelial MVP increases neointima formation and promotes atherosclerosis. Mechanistically, MVP directly binds with Parkin and inhibits the ubiquitination and proteasomal degradation of Parkin by dissociating the E3 ligase NEDD4L from Parkin, leading to activation of Parkin-mediated mitophagy pathway in the EC. Genetic modulation of endothelial MVP and Parkin influences the mitophagy, apoptosis, and neointima formation. These results demonstrate that MVP acts as an intracellular regulator promoting Parkin-mediated mitophagy. Our findings suggest that MVP/NEDD4L/Parkin axis may serve as the therapeutic target for treating intimal hyperplasia and atherosclerosis.
Collapse
Affiliation(s)
- Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Fan Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yunfu Hu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Ren
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Su
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Wanxuan Song
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Kunxin Xie
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Dongdong Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Junlu Pan
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuying Liu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuxin Feng
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China.
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Liu Q, Wang Y, Song M, Huang J, Shi J, Sun W, Ji X, Chang Y, Ma B, Zhang P, Yan Y, Zhang H. CCL20/CXCL5 Drives Crosstalk Between Anaplastic Thyroid Cancer Stem Cells and Tumor-Associated Macrophages to Promote Tumor Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405399. [PMID: 40091357 PMCID: PMC12061268 DOI: 10.1002/advs.202405399] [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] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
The dynamic interplay between tumor-associated macrophages (TAMs) and anaplastic thyroid cancer (ATC) shapes the tumor microenvironment and facilitates ATC progression. However, the mechanisms of communication between TAMs and anaplastic thyroid cancer stem cells (ATCSCs) remain largely unelucidated. Integrative analyses of single-cell RNA sequencing, cytokine/chemokine arrays, proteomics, and mRNA expression datasets are performed to reveal crosstalk between TAMs and ATCSCs and signaling pathways in ATCSCs. Subsequently, in vitro experiments are performed to validate the regulatory effects of key cytokines on ATCSC stemness. Last, xenogeneic orthotopic thyroid ATCSCs transplantation models are utilized to corroborate the regulatory effect of cytokines on stemness. CCL20 derived from THP-1-M2 activates the IRAK-1/NF-κB1/2 signaling pathway in ATCSCs, thereby positively regulating stemness characteristics and upregulating CXCL5 secretion. ATCSCs not only exhibit autocrine CXCL5 participation in the regulation of stemness but also demonstrate paracrine CXCL5 activity to recruit THP-1-Mφ and maintain the M2 phenotype. CCL20 and CXCL5 are involved in the crosstalk between TAMs and ATCSCs. The CCL20/CXCL5 axis plays a crucial role in the interaction between TAMs and ATCSCs, establishing a progressive tumor microenvironment.
Collapse
Affiliation(s)
- Qi Liu
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yan Wang
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Mingyuan Song
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jiapeng Huang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Jinyuan Shi
- Department of Thyroid SurgeryGeneral SurgeryQilu Hospital of Shandong UniversityJinan250012P. R. China
| | - Wei Sun
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Xiaoyu Ji
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuang Chang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Bing Ma
- Department of Clinical Epidemiology and Evidence‐based MedicineThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Ping Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| | - Yuanyuan Yan
- Department of PharmacologySchool of PharmacyChina Medical UniversityShenyang110122P. R. China
| | - Hao Zhang
- Department of Thyroid SurgeryThe First Hospital of China Medical UniversityShenyang110801P. R. China
| |
Collapse
|
5
|
Wang L, Zhang Y, Yue J, Zhou R. The Role of Ubiquitination on Macrophages in Cardiovascular Diseases and Targeted Treatment. Int J Mol Sci 2025; 26:4260. [PMID: 40362498 PMCID: PMC12072125 DOI: 10.3390/ijms26094260] [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: 01/31/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, with macrophage dysfunction playing a central role in its pathogenesis. Ubiquitination, a critical post-translational modification, regulates diverse macrophage functions, including lipoprotein metabolism, inflammation, oxidative stress, mitophagy, autophagy, efferocytosis, and programmed cell death (pyroptosis, necroptosis, ferroptosis, and apoptosis). This review highlights the regulatory roles of ubiquitination in macrophage-driven CVD progression, focusing on its effects on cholesterol metabolism, inflammation, activation, polarization, and the survival of macrophages. Targeting ubiquitination pathways has therapeutic potential by enhancing macrophage autophagy, reducing inflammation, and improving plaque stability. However, challenges, such as off-target effects, ubiquitination crosstalk, and macrophage heterogeneity, must be addressed. By integrating advances in ubiquitination biology, therapeutic strategies can be developed to mitigate CVD and other macrophage-driven inflammatory diseases. This review underscores the potential of ubiquitination-targeting therapies for mitigating CVD and highlights the key areas for further investigation.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Yue
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ronghua Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Xu J, Wang B, Liu Q, Guo S, Chen C, Wu J, Zhao X, Li M, Ma Z, Zhou S, Qian Y, Huang Y, Wang Z, Shu C, Xu Q, Ben J, Wang Q, Wang S. MVP-LCN2 axis triggers evasion of ferroptosis to drive hepatocarcinogenesis and sorafenib resistance. Drug Resist Updat 2025; 81:101246. [PMID: 40262414 DOI: 10.1016/j.drup.2025.101246] [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: 02/15/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
RNA-binding proteins (RBPs) are critical regulators in tumorigenesis and therapy resistance by modulating RNA metabolism. However, the role of RBPs in hepatocarcinogenesis and progression remains elusive. Here, RBPs screening and integrating analyses identify major vault protein (MVP) as an oncogenic RBP in the occurrence of hepatocellular carcinoma (HCC) and sorafenib resistance via suppressing ferroptosis. Mechanistically, reactive oxygen species (ROS) induces STAT3-mediated MVP transcription activation and high expression in HCC cells. Subsequently, phosphoglycerate mutase family member 5 (PGAM5) directly dephosphorylates MVP at S873, facilitating its binding to the mRNA of iron-sequestering cytokine LCN2 and maintains its stability, thereby attenuating ferroptosis by reducing lipid peroxidation and intracellular Fe2+ content following sorafenib treatment. Notably, tenapanor, a potent pharmacological inhibitor of MVP, effectively disrupts the interaction between MVP and LCN2 mRNA and enhances ferroptosis and sorafenib sensitivity. Collectively, these findings underscore the central role of MVP in hepatocarcinogenesis and offer promising avenues to improve HCC treatment.
Collapse
Affiliation(s)
- Jiawen Xu
- Nanjing University Medical School, Nanjing, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Qiaoyu Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | | | - Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Jun Wu
- Medical College, Yangzhou University, Yangzhou, China
| | - Xiaoya Zhao
- Nanjing University Medical School, Nanjing, China
| | - Mengmeng Li
- Nanjing University Medical School, Nanjing, China
| | - Zhuang Ma
- Nanjing University Medical School, Nanjing, China
| | - Shimeng Zhou
- Nanjing University Medical School, Nanjing, China
| | - Yun Qian
- Nanjing University Medical School, Nanjing, China
| | - Yijin Huang
- School of Medicine, University of Missouri, Columbia, USA
| | - Zhangding Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Chuanjun Shu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Qingxiang Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China.
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China.
| | - Shouyu Wang
- Nanjing University Medical School, Nanjing, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor Immunotherapy; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China; Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
7
|
Ibrahim RT, Moustafa YM, Alwaili MA, Alrebdi AN, Alharthi A, Noufal NR, Khodeer DM. Chromium and formoterol therapy for obesity-induced asthma in rats. Front Pharmacol 2025; 16:1537022. [PMID: 40242447 PMCID: PMC12000533 DOI: 10.3389/fphar.2025.1537022] [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: 11/29/2024] [Accepted: 02/06/2025] [Indexed: 04/18/2025] Open
Abstract
The development of asthma is impacted by fat. Asthma is more common in obese persons. The purpose of the experimental study is to determine how chromium, formoterol, and their combination can improve the quality of life for obese people with lung anomalies. Thirty-six male Wistar rats were divided into six groups: control (C), obesity (CO), obese-asthma (COA), and obese-asthma groups treated with formoterol (OAF), chromium (OACR), or both (OACRF). Except for group C, all groups received a high-fat diet for 4 weeks. Subsequently, ovalbumin (OVA) was administered subcutaneously (s.c.) to all groups except C and CO to induce sensitization. Asthma was triggered via 1% OVA aerosol challenges on days 26-28. Over 5 days, OAF and OACRF received daily formoterol inhalations (50 μg/kg), while OACR and OACRF were given chromium (400 μg/kg). Treatments were timed to align with asthma induction protocols. Lipid profile and inflammatory indicators were examined at the end of the trial-Immunohistochemical analysis of lung tissue, Histopathological and lung tissue stained with Hematoxylin and Eosin. The combination therapy (OACRF) significantly reduced body weight (p < 0.05), lowered LDL and triglycerides, increased HDL, and normalized lung tissue architecture compared to controls. Immunohistochemistry revealed reduced IL-1β and IL-17α expression. The (OACRF) group demonstrated superior asthma control by reducing body weight, improving inflammatory indicators, and restoring lung tissue to its normal state by administering chromium and formoterol therapy. The most effective strategy for treating both obesity and asthma is to address their two connected conditions. These findings demonstrate that combined chromium and formoterol therapy effectively addresses metabolic and inflammatory components of obesity-induced asthma, offering a promising dual-target therapeutic strategy.
Collapse
Affiliation(s)
- Rania T. Ibrahim
- Department of Scientific Research, Egypt Healthcare Authority, Ismailia, Egypt
| | - Yasser M. Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, Egypt, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Maha Abdullah Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amjad N. Alrebdi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Afaf Alharthi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Noha R. Noufal
- Basic Medical Science Department, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia
- Pathology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Dina M. Khodeer
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
8
|
Zahedi K, Morgan M, Prieto B, Brooks M, Howard TA, Barone S, Bissler JJ, Argyropoulos C, Soleimani M. Role of Extracellular Vesicles in TSC Renal Cystogenesis. Int J Mol Sci 2025; 26:3154. [PMID: 40243914 PMCID: PMC11989098 DOI: 10.3390/ijms26073154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in TSC1 or TSC2 genes and affects multiple organs. TSC proteins control cell growth by regulating the activity of the mechanistic target of rapamycin complex 1. Extracellular vesicles (EVs) are membrane-bound particles produced by cells that mediate cellular communication, function, and growth. Although extensive studies regarding the genetic basis of TSC exist, the exact mechanism contributing to its pathogenesis remains unresolved. It has been proposed that EVs generated by renal cyst epithelia of mice and cells with Tsc gene mutations contain factors that alter the function and proliferation of TSC-sufficient cells. To test this, EVs from the kidneys and kidney explants of wildtype and Tsc1KO mice were isolated and characterized by Western blotting, transmission electron microscopy, dynamic light scattering, and fluorescent nanoparticle tracking. Our results show an enrichment in EV-associated markers and particle sizes of similar ranges. RNA-seq and proteomic analyses identified EV shuttle factors. EV RNA and protein shuttle factors showed significant differences. Furthermore, EVs isolated from Tsc1KO mice inhibited the proliferation of M-1 cells. Understanding the role of EVs in cell proliferation and cystogenesis in TSC may lead to the development of new approaches for the treatment of this disease.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
- Research Services, Raymond J. Murphy Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - Mackenzie Morgan
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
| | - Brenda Prieto
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
| | - Marybeth Brooks
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
- Research Services, Raymond J. Murphy Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - Tamara A. Howard
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
| | - Sharon Barone
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
- Research Services, Raymond J. Murphy Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - John J. Bissler
- University of Tennessee Health Sciences Center, Le Bonheur Children’s Hospital, Memphis, TN 38163, USA;
- The Department of Pediatrics, St. Jude Children’s Hospital, Memphis, TN 38105, USA
| | - Christos Argyropoulos
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
| | - Manoocher Soleimani
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.M.); (B.P.); (M.B.); (T.A.H.); (S.B.); (C.A.)
- Research Services, Raymond J. Murphy Veterans Health Care Center, Albuquerque, NM 87108, USA
| |
Collapse
|
9
|
He F, Lang Z, Huang Y, Qiu Y, Xiong P, Li N, Zhao G, Peng Y. Exogenous L-Serine Alleviates Pasteurella multocida-Induced Inflammation by Reprogramming the Transcription and Metabolism of Macrophages. Vet Sci 2025; 12:254. [PMID: 40267013 PMCID: PMC11945856 DOI: 10.3390/vetsci12030254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/05/2025] [Accepted: 03/04/2025] [Indexed: 04/25/2025] Open
Abstract
P. multocida is notorious for inducing excessive inflammation with high lethality in multiple animals, such as cattle, pigs, and chickens. Our previous study revealed that L-serine was decreased in the lungs of mice infected with P. multocida capsular type A strain CQ2 (PmCQ2), and 2 mg/kg of L-serine could alleviate PmCQ2-induced lung inflammation in vivo, which may largely depend on macrophages. However, the underlying intrinsic alterations remain unknown. Here, we demonstrated that 10 mM of L-serine significantly inhibited the release of inflammatory cytokines (e.g., IL-1β and TNF-α) by blocking inflammasome activation (including NALP1, NLRP3, NLRC4, AIM2, and Caspase-1) in PmCQ2-infected macrophages. Furthermore, the results of RNA-seq and metabonomics revealed that exogenous L-serine supplementation substantially reprogrammed macrophage transcription and metabolism. Mechanically, L-serine reduced inflammatory responses via the inhibition of glycolysis in macrophages based on a seahorse assay. Together, these findings characterize the intrinsic molecular alterations in activated macrophages and provide new targets for modulating P. multocida infection-induced macrophage inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guangfu Zhao
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (F.H.); (Z.L.); (Y.H.); (Y.Q.); (P.X.); (N.L.)
| | - Yuanyi Peng
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (F.H.); (Z.L.); (Y.H.); (Y.Q.); (P.X.); (N.L.)
| |
Collapse
|
10
|
Su M, Zhao W, Jiang H, Zhao Y, Liao Z, Liu Z, Xu M, Jiang S, Wu L, Yang Y, Wang Z, Zeng Z, Fang Y, Tang C, Miller CL, Evans PC, Wang L, Banach M, Jo H, Berk BC, Offermanns S, Huang Y, Ge J, Xu S, Weng J. Endothelial IGFBP6 suppresses vascular inflammation and atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2025; 4:145-162. [PMID: 39794479 PMCID: PMC11825279 DOI: 10.1038/s44161-024-00591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 11/28/2024] [Indexed: 01/13/2025]
Abstract
Beyond dyslipidemia, inflammation contributes to the development of atherosclerosis. However, intrinsic factors that counteract vascular inflammation and atherosclerosis remain scarce. Here we identify insulin-like growth factor binding protein 6 (IGFBP6) as a homeostasis-associated molecule that restrains endothelial inflammation and atherosclerosis. IGFBP6 levels are significantly reduced in human atherosclerotic arteries and patient serum. Reduction of IGFBP6 in human endothelial cells by siRNA increases inflammatory molecule expression and monocyte adhesion. Conversely, pro-inflammatory effects mediated by disturbed flow (DF) and tumor necrosis factor (TNF) are reversed by IGFBP6 overexpression. Mechanistic investigations further reveal that IGFBP6 executes anti-inflammatory effects directly through the major vault protein (MVP)-c-Jun N-terminal kinase (JNK)/nuclear factor kappa B (NF-κB) signaling axis. Finally, IGFBP6-deficient mice show aggravated diet- and DF-induced atherosclerosis, whereas endothelial-cell-specific IGFBP6-overexpressing mice protect against atherosclerosis. Based on these findings, we propose that reduction of endothelial IGFBP6 is a predisposing factor in vascular inflammation and atherosclerosis, which can be therapeutically targeted.
Collapse
Affiliation(s)
- Meiming Su
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenqi Zhao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hui Jiang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yaping Zhao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaopeng Liao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenghong Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengyun Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shanshan Jiang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Lili Wu
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Yi Yang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhihua Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhutian Zeng
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yun Fang
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, China
| | - Clint L Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Paul C Evans
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Xuhui District, Shanghai, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Anhui Provincial Key Laboratory of Metabolic Health and Pan-vascular Diseases, Hefei, China.
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Anhui Provincial Key Laboratory of Metabolic Health and Pan-vascular Diseases, Hefei, China.
| |
Collapse
|
11
|
Wang C, Peng M, Gao Z, Fu F, Li G, Su D, Huang L, Guo J, Shan Y. Citrus aurantium 'Changshan-huyou' physiological premature fruit drop: A promising prebiotic to tackle obesity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156347. [PMID: 39765038 DOI: 10.1016/j.phymed.2024.156347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/22/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Presently, the mitigation and governance of obesity have surfaced as significant public health dilemmas on a global scale. A wealth of studies indicated that the host gut microbiota is instrumental in regulating the interplay between high-fat diet (HFD) intake and the pathogenesis of obesity. Physiological premature fruit drop, a major byproduct of citrus, is rich in a variety of bioactive constituents, yet its potential has remained underutilized for an extended period. PURPOSE The objective of this investigation is to examine the chemical constituents of Citrus aurantium'Changshan-huyou' premature fruit drop (HYFD) and investigate its anti-obesity effects, elucidating its potential pathways. METHODS Volatile compounds and flavonoids in HYFD were analyzed using chromatographic and mass spectrometric techniques. Furthermore, this study utilized biochemical assays and histopathological examinations to evaluate the effects of HYFD on HFD-fed mice. The impact of HYFD on the gut microbiota of the mice was examined through 16S rRNA gene sequencing, and fecal microbiota transplantation was employed to validate the role of the gut microbial community in host obesity prevention. Concurrently, transcriptome was employed to identify differentially expressed genes, providing further insights into the molecular mechanisms through which HYFD manifests its anti-obesity effects. RESULTS Our findings demonstrated that HYFD supplementation significantly alleviated adiposity and ameliorated the dysbiosis of gut microbiota in HFD-induced mice. HYFD rectified the HFD-induced gut microbiota dysregulation, enhanced the presence of beneficial microbial taxa linked to lipid metabolism, including Parabacteroides and Alistipes, and elevated concentrations of the anti-obesity short-chain fatty acids, comprising caproic acid and isocaproic acid. Additionally, transcriptomic analyses confirmed that HYFD prevented obesity in mice by enhancing fatty acid catabolism via the activation of the AMPK/PPARα/CPT1a signaling pathway. CONCLUSION Our results provided novel insights into the mechanism of citrus physiological premature fruit drop and its potential role in preventing obesity, while sparking greater interest in leveraging more biomass waste.
Collapse
Affiliation(s)
- Chao Wang
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Mingfang Peng
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Zhipeng Gao
- Fisheries College, Hunan Agricultural University, Changsha, Hunan Province 410128, China
| | - Fuhua Fu
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Gaoyang Li
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Donglin Su
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Lvhong Huang
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China
| | - Jiajing Guo
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China.
| | - Yang Shan
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha, Hunan Province 410125, China.
| |
Collapse
|
12
|
Han YH, Cui XW, Li YX, Chen X, Zhang H, Zhang Y, Wang SS, Li M. Bacterial cellulose is a desirable biological macromolecule that can prevent obesity via modulating lipid metabolism and gut microbiota. Int J Biol Macromol 2024; 283:137522. [PMID: 39537056 DOI: 10.1016/j.ijbiomac.2024.137522] [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: 12/12/2023] [Revised: 10/02/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Obesity has attracted great concern because of its undesirable effects on our life quality. Bacterial cellulose (BC) is a biological macromolecule that can improve gut homeostasis and lipid metabolism. However, its potential role in preventing obesity and associated mechanisms is still poorly understood. Herein, a supplement of BC was used to fully evaluate how it prevents obesity based on physio-biochemical and gut microbial analyses. Results showed that BC consumption helped decrease body and liver weight, and fat accumulation in kidney and epididymis. Correspondingly, glucose concentrations, total triglycerides, total cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol were reversed to the control levels. Consuming BC also improved liver fat metabolism and intestinal function, and alleviated ileum and epididymis inflammation. High-throughput sequencing suggested that a high-fat diet significantly decreased gut microbiota diversity, which could be reversed by consuming BC. A decreased Firmicutes and Proteobacteria and an increased Bacteroidetes following BC consumption were observed. The OTU-based analysis identified that Lachnospiraceae, Desulfovibrio, Lachnoclostridium, Blautia, Anaerotruncus, Bacteroides, Faecalibaculum, Bacteroidales S24-7 group, Prevotellaceae UCG-001 group, and Alloprevotella might be involved in obesity development or prevention. Our data suggest that BC is a good insoluble dietary fiber to prevent obesity via regulating lipid metabolism and gut microbiota.
Collapse
Affiliation(s)
- Yong-He Han
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Xi-Wen Cui
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yi-Xi Li
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Xian Chen
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hong Zhang
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhang
- Fujian Key Laboratory of Pollution Control and Resource Reuse, College of Environmental and Resource Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Shan-Shan Wang
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; College of Life Science, Fujian Normal University, Fuzhou 350117, China.
| | - Min Li
- College of Life Science, Fujian Normal University, Fuzhou 350117, China.
| |
Collapse
|
13
|
Wang S, Li Z, Chen C, Guo T, Zhao S, Zhao J, Zhang W, Qi Y, Zhang J, Wang Y, Lv Y, Gu C. MACC1 enhances an oncogenic RNA splicing of IRAK1 through interacting with HNRNPH1 in lung adenocarcinoma. J Cell Physiol 2024; 239:e31426. [PMID: 39221900 DOI: 10.1002/jcp.31426] [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: 03/18/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Dysregulation of alternative pre-mRNA splicing plays a critical role in the progression of cancers, yet the underlying molecular mechanisms remain largely unknown. It is reported that metastasis-associated in colon cancer 1 (MACC1) is a novel prognostic and predictive marker in many types of cancers, including lung adenocarcinoma. Here, we reveal that the oncogene MACC1 specifically drives the progression of lung adenocarcinoma through its control over cancer-related splicing events. MACC1 depletion inhibits lung adenocarcinoma progression through triggering IRAK1 from its long isoform, IRAK1-L, to the shorter isoform, IRAK1-S. Mechanistically, MACC1 interacts with splicing factor HNRNPH1 to prevent the production of the short isoform of IRAK1 mRNA. Specifically, the interaction between MACC1 and HNRNPH1 relies on the involvement of MACC1's SH3 domain and HNRNPH1's GYR domain. Further, HNRNPH1 can interact with the pre-mRNA segment (comprising exon 11) of IRAK1, thereby bridging MACC1's regulation of IRAK1 splicing. Our research not only sheds light on the abnormal splicing regulation in cancer but also uncovers a hitherto unknown function of MACC1 in tumor progression, thereby presenting a novel potential therapeutic target for clinical treatment.
Collapse
Affiliation(s)
- Shiqing Wang
- Department of Thoracic Surgery & Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhuoshi Li
- Department of Thoracic Surgery & Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Chaoqun Chen
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Tao Guo
- Department of Thoracic Surgery & Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Shilei Zhao
- Department of Thoracic Surgery & Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jinyao Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wenjing Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yangfan Qi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinrui Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yang Wang
- Sino-US Research Center for Cancer Translational Medicine of the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuesheng Lv
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chundong Gu
- Department of Thoracic Surgery & Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Chen T, Wang Y, Yang JL, Ni J, You K, Li X, Song Y, Wang X, Li J, Shen X, Fan Y, You Y. Gentisic acid prevents the development of atherosclerotic lesions by inhibiting SNX10-mediated stabilization of LRP6. Pharmacol Res 2024; 210:107516. [PMID: 39603572 DOI: 10.1016/j.phrs.2024.107516] [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: 07/22/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerotic-related acute cardiovascular events remain a leading cause of mortality worldwide, yet there are currently no pharmacological interventions available to address plaque formation or plaque rupture (PR). Here we reported that gentisic acid (GA) exerted potent therapeutic effects on plaque formation and PR in a dose-dependent manner by inhibiting LRP6-mediated macrophage apoptosis. By using the CETSA assay and DARTS assay, we identified sorting nexin 10 (SNX10) as the direct target of GA. The binding of GA to SNX10 disrupts the interaction between SNX10 and LRP6, leading to the degradation of LRP6. The downregulation of LRP6 then significantly attenuated the activation of Wnt/β-catenin pathway to exert an inhibitory effect on apoptosis. Moreover, the specific depletion of SNX10 in macrophages significantly reduced LRP6 levels and subsequently macrophage apoptosis both in vivo and in vitro. In conclusion, our findings not only suggest that GA may serve as a potential therapeutic candidate for the prevention of atherosclerosis and acute cardiovascular events caused by PR, but also confirm the druggability of SNX10 as a promising therapeutic target for atherosclerotic rupture.
Collapse
Affiliation(s)
- Tongqing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yiming Wang
- Department of Cardiology, Worldwide Medical Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Lin Yang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiahui Ni
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuesong Li
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuping Song
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jian Li
- Department of Cardiology, Worldwide Medical Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China.
| | - Yujuan Fan
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China.
| |
Collapse
|
15
|
Li Z, Ngu R, Naik AA, Trinh K, Paharkova V, Liao H, Liu Y, Zhuang C, Le D, Pei H, Asante I, Mittelman SD, Louie S. Adipocyte maturation impacts daunorubicin disposition and metabolism. Eur J Clin Invest 2024; 54:e14307. [PMID: 39254480 DOI: 10.1111/eci.14307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Acute lymphoblastic leukaemia (ALL) is the most common type of childhood leukaemia with effective chemotherapeutic treatment. However, obesity has been associated with higher ALL chemoresistance rates and lower event-free survival rates. The molecular mechanism of how obesity promotes chemotherapy resistance is not well delineated. OBJECTIVES This study evaluated the effect of adipocyte maturation on sequestration and metabolism of chemotherapeutic drug daunorubicin (DNR). METHODS Using targeted LC-MS/MS multi-analyte assay, DNR sequestration and metabolism were studied in human preadipocyte and adipocyte cell lines, where expressions of DNR-metabolizing enzymes aldo-keto reductases (AKR) and carbonyl reductases (CBR) were also evaluated. In addition, to identify the most DNR-metabolizing AKR/CBR isoforms, recombinant human AKR and CBR enzymes were subject to DNR metabolism. The results were further validated by AKR-, CBR-specific inhibitors. RESULTS This report shows that adipocyte maturation upregulates expressions of AKR and CBR enzymes (by 4- to 60- folds, p < .05), which is positively associated with enhanced sequestration and metabolism of DNR in adipocytes compared to preadipocytes (by ~30%, p < .05). In particular, adipocyte maturation upregulates AKR1C3 and CBR1, which are the predominate metabolic enzyme isoforms responsible for DNR biotransformation to its metabolites. CONCLUSION Fat is an expandable tissue that can sequester and detoxify DNR when stimulated by obesity, likely through the upregulation of DNR-metabolizing enzymes AKR1C3 and CBR1. Our data partially explains why obese ALL patients may be more likely to become chemoresistant towards DNR, and provides evidence for potential clinical investigation targeting obesity to reduce DNR chemoresistance.
Collapse
Affiliation(s)
- Zeyang Li
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Rachael Ngu
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Aditya Anil Naik
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Kaitlyn Trinh
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Vladislava Paharkova
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Hanyue Liao
- College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Yulu Liu
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Cindy Zhuang
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Danh Le
- Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Hua Pei
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Isaac Asante
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, University of California Los Angeles (UCLA) Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA, Los Angeles, California, USA
| | - Stan Louie
- Alfred Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
16
|
Zhao J, Cao X, Li Q, Xie J, Wu H. Obesity Mediates the Association Between Serum Copper and Inflammation: A Cross-sectional and Mendelian Randomization Study. Biol Trace Elem Res 2024:10.1007/s12011-024-04405-z. [PMID: 39368047 DOI: 10.1007/s12011-024-04405-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Copper is an important biological trace element, but its overexposure can be harmful to the human body. Herein, we aimed to assess the association between serum copper levels and inflammation. A total of 5231 participants were analyzed from the National Health and Nutrition Examination Survey (NHANES) between 2011 and 2016. Participants with higher serum copper levels had higher values of systemic inflammation indexes. The concentration of high-sensitive C-reactive protein (hs-CRP) increased with serum copper concentration (β = 2.8, p < 0.001). Participants with high and very high copper levels had higher ORs (odds ratios) of having inflammation (high: OR 2.92 (0.77-11.04), p = 0.074; very high: OR 8.66 (3.18-23.54), p = 0.011), which were further exacerbated in people with diabetes and males. Body mass index (BMI) and body fat percentage are two main mediators in the association between serum copper and hs-CRP, accounting for 12.62% and 19.72%, respectively. The random-effects inverse variance-weighted (IVW) analysis revealed that there was a genetic causal relationship between serum copper and obesity (OR 1.15, p = 0.014). Our results suggest that serum copper is positively associated with inflammation, which may be mainly mediated by obesity.
Collapse
Affiliation(s)
- Jianfeng Zhao
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, People's Republic of China.
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, People's Republic of China.
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Xueer Cao
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Qingqi Li
- Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Junhao Xie
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, People's Republic of China
| | - Hongrong Wu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
17
|
Aroca-Esteban J, Souza-Neto FV, Aguilar-Latorre C, Tribaldo-Torralbo A, González-López P, Ruiz-Simón R, Álvarez-Villareal M, Ballesteros S, de Ceniga MV, Landete P, González-Rodríguez Á, Martín-Ventura JL, de Las Heras N, Escribano Ó, Gómez-Hernández A. Potential protective role of let-7d-5p in atherosclerosis progression reducing the inflammatory pathway regulated by NF-κB and vascular smooth muscle cells proliferation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167327. [PMID: 38945455 DOI: 10.1016/j.bbadis.2024.167327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
The prevalence of cardiovascular diseases (CVDs) is increasing in the last decades, even is the main cause of death in first world countries being atherosclerosis one of the principal triggers. Therefore, there is an urgent need to decipher the underlying mechanisms involved in atherosclerosis progression. In this respect, microRNAs dysregulation is frequently involved in the progression of multiple diseases including CVDs. Our aim was to demonstrate that let-7d-5p unbalance could contribute to the pathophysiology of atherosclerosis and serve as a potential diagnostic biomarker. We evaluated let-7d-5p levels in vascular biopsies and exosome-enriched extracellular vesicles (EVs) from patients with carotid atherosclerosis and healthy donors. Moreover, we overexpressed let-7d-5p in vitro in vascular smooth muscle cells (VSMCs) to decipher the targets and the underlying mechanisms regulated by let-7d-5p in atherosclerosis. Our results demonstrate that let-7d-5p was significantly upregulated in carotid plaques from overweight patients with carotid atherosclerosis. Moreover, in EVs isolated from plasma, we found that let-7d-5p levels were increased in carotid atherosclerosis patients compared to control subjects specially in overweight patients. Receiver Operating Characteristic (ROC) analyses confirmed its utility as a diagnostic biomarker for atherosclerosis. In VSMCs, we demonstrated that increased let-7d-5p levels impairs cell proliferation and could serve as a protective mechanism against inflammation by impairing NF-κB pathway without affecting insulin resistance. In summary, our results highlight the role of let-7d-5p as a potential therapeutic target for atherosclerosis since its overexpression induce a decrease in inflammation and VSMCs proliferation, and also, as a novel non-invasive diagnostic biomarker for atherosclerosis in overweight patients.
Collapse
Affiliation(s)
- Javier Aroca-Esteban
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Francisco V Souza-Neto
- Physiology Department, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Carlota Aguilar-Latorre
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Alba Tribaldo-Torralbo
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Paula González-López
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Rubén Ruiz-Simón
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Marta Álvarez-Villareal
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
| | - Sandra Ballesteros
- Physiology Department, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Melina Vega de Ceniga
- Department of Angiology and Vascular Surgery, Hospital of Galdakao-Usansolo, Galdakao, Bizkaia, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Pedro Landete
- Departmento de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa, Madrid, Spain; Faculty of Medicine, Autonoma University of Madrid, Madrid, Spain
| | - Águeda González-Rodríguez
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - José L Martín-Ventura
- IIS-Fundation Jimenez-Diaz, Autonoma University of Madrid and CIBERCV, Madrid, Spain
| | - Natalia de Las Heras
- Physiology Department, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Óscar Escribano
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| | - Almudena Gómez-Hernández
- Hepatic and Vascular Diseases Laboratory, Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
18
|
Masood A, Benabdelkamel H, Joy SS, Alhossan A, Alsuwayni B, Abdeen G, Aldhwayan M, Alfadda NA, Miras AD, Alfadda AA. Label-free quantitative proteomic profiling reveals differential plasma protein expression in patients with obesity after treatment with liraglutide. Front Mol Biosci 2024; 11:1458675. [PMID: 39324112 PMCID: PMC11422103 DOI: 10.3389/fmolb.2024.1458675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Treatment and management of obesity is clinically challenging. The inclusion of GLP-1 receptor agonists (GLP1RA) in the medical management of obesity has proven to be efficacious. However, mechanisms underlying the molecular changes arising from GLP1RA treatment in patients with obesity remain to be elucidated. Methods A single-center, prospective study was undertaken to evaluate the changes in the plasma proteins after liraglutide 3 mg therapy in twenty patients (M/F: 7/13) with obesity (mean BMI 40.65 ± 3.7 kg/m2). Anthropometric and laboratory parameters were measured, and blood samples were collected at two time points: baseline, before initiating treatment (pretreatment group, PT), and after three months of receiving the full dose liraglutide 3 mg (posttreatment group, PoT). An untargeted label-free LC MSMS mass spectrometric approach combined with bioinformatics and network pathway analysis was used to determine changes in the proteomic profiles. Results The mean age of the study participants was 36.0 ± 11.1 years. A statistically significant change was observed in weight, BMI and HbA1c levels between the PT and PoT groups (paired t-test, P < 0.001). A significant dysregulation was noted in the abundances of 151 proteins (31 up and 120 downregulated) between the two groups. The potential biomarkers were evaluated using receiver operating characteristic (ROC) curves. The top ten proteins (area under the curve (AUC) of 0.999 (95% CI)) were identified as potential biomarkers between PT and PoT groups and included Cystatin-B, major vault protein, and plastin-3, which were upregulated, whereas multimerin-2, large ribosomal P2, and proline-rich acidic protein 1 were downregulated in the PoT group compared with the PT group. The top network pathway identified using ingenuity pathway analysis (IPA), centered around dysregulation of MAPK, AKT, and PKc signaling pathways and related to cell-to-cell signaling and interaction, cellular assembly and organization, cellular compromise and a score of 46 with 25 focus proteins. Discussion Through label-free quantitative proteomic analysis, our study revealed significant dysregulation of plasma proteins after liraglutide 3 mg treatment in patients with obesity. The alterations in the proteomic profile between the PT and PoT groups demonstrated a decrease in levels of proteins involved in inflammation and oxidative stress pathways. On the other hand proteins involved in the glycolytic and lipolytic metabolic pathways as well as those participating in cytoskeletal and endothelial reorganization were observed to be increased. Understanding actions of liraglutide at a molecular and proteomic levels provides a holistic look into how liraglutide impacts metabolism, induces weight loss and improves overall metabolic health.
Collapse
Affiliation(s)
- Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Salini Scaria Joy
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz Alhossan
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bashayr Alsuwayni
- Corporate of Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ghalia Abdeen
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Madhawi Aldhwayan
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Nora A. Alfadda
- Department of Community Health Sciences, Clinical Nutrition, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Alexander Dimitri Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolic Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
- School of Medicine, Ulster University, Derry, United Kingdom
| | - Assim A. Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Wang Y, Zhang W, Yang Y, Qin J, Wang R, Wang S, Fu W, Niu Q, Wang Y, Li C, Li H, Zhou Y, Liu M. Osteopontin deficiency promotes cartilaginous endplate degeneration by enhancing the NF-κB signaling to recruit macrophages and activate the NLRP3 inflammasome. Bone Res 2024; 12:53. [PMID: 39242551 PMCID: PMC11379908 DOI: 10.1038/s41413-024-00355-3] [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: 09/27/2023] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 09/09/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is a major cause of discogenic pain, and is attributed to the dysfunction of nucleus pulposus, annulus fibrosus, and cartilaginous endplate (CEP). Osteopontin (OPN), a glycoprotein, is highly expressed in the CEP. However, little is known on how OPN regulates CEP homeostasis and degeneration, contributing to the pathogenesis of IDD. Here, we investigate the roles of OPN in CEP degeneration in a mouse IDD model induced by lumbar spine instability and its impact on the degeneration of endplate chondrocytes (EPCs) under pathological conditions. OPN is mainly expressed in the CEP and decreases with degeneration in mice and human patients with severe IDD. Conditional Spp1 knockout in EPCs of adult mice enhances age-related CEP degeneration and accelerates CEP remodeling during IDD. Mechanistically, OPN deficiency increases CCL2 and CCL5 production in EPCs to recruit macrophages and enhances the activation of NLRP3 inflammasome and NF-κB signaling by facilitating assembly of IRAK1-TRAF6 complex, deteriorating CEP degeneration in a spatiotemporal pattern. More importantly, pharmacological inhibition of the NF-κB/NLRP3 axis attenuates CEP degeneration in OPN-deficient IDD mice. Overall, this study highlights the importance of OPN in maintaining CEP and disc homeostasis, and proposes a promising therapeutic strategy for IDD by targeting the NF-κB/NLRP3 axis.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wanqian Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Jinghao Qin
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ruoyu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Wenjuan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yanxia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hongli Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
20
|
Peshkova M, Korneev A, Revokatova D, Smirnova O, Klyucherev T, Shender V, Arapidi G, Kosheleva N, Timashev P. Four sides to the story: A proteomic comparison of liquid-phase and matrix-bound extracellular vesicles in 2D and 3D cell cultures. Proteomics 2024; 24:e2300375. [PMID: 38197488 DOI: 10.1002/pmic.202300375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024]
Abstract
Multipotent mesenchymal stromal cells (MSCs)-derived extracellular vesicles (EVs) play important roles in cellular communication and are extensively studied as promising therapeutic agents. While there is a substantial pool of studies on liquid-phase EVs, data on EVs bound to the extracellular matrix (ECM) is lacking. There is also an emerging trend of accumulating and comparing data on characteristics of EVs obtained in different culturing conditions. Aiming to reveal proteomic signatures of EVs obtained from conditioned media and ECM of MSCs cultured in 2D and 3D conditions, we performed liquid chromatography with tandem mass spectrometry. Bioinformatic analysis revealed common patterns in proteomic composition of liquid-phase EVs and matrix-bound vesicles (MBVs), namely extracellular environment organization, immune, and transport pathways enrichment. However, extracellular environmental organization pathways are more enriched in liquid-phase EVs than in MBVs, while MBVs proteins noticeably enrich enzymatic pathways. Furthermore, each type of EVs from 2D and 3D cultures has a unique differential abundance profile. We have also performed comparative functional assays, namely scratch assay to assess EVs effect on cell migration and tubulogenesis assay to evaluate EVs angiogenic potential. We found that both liquid-phase EVs and MBVs enhance cell migration, while angiogenic potential is higher in MBVs. Results of the present study suggest that while both liquid-phase EVs and MBVs have therapeutic potential, some unique features of each subgroup may determine optimal areas of their application.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
| | - Alexander Korneev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
- Laboratory of the Polymers Synthesis for Medical Applications, Sechenov University, Moscow, Russia
| | - Daria Revokatova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
| | - Olga Smirnova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
| | - Timofey Klyucherev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
| | - Victoria Shender
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Georgij Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Moscow Region, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
21
|
Routsias JG, Marinou D, Mavrouli M, Tsakris A, Pitiriga VC. Major Vault Protein/Lung Resistance-Related Protein: A Novel Biomarker for Inflammation and Acute Infections. Microorganisms 2024; 12:1762. [PMID: 39338437 PMCID: PMC11434279 DOI: 10.3390/microorganisms12091762] [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: 07/02/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION Vault particles are large cytoplasmic ribonucleoprotein particles that participate in inflammation. The aim of this study was to assess the diagnostic and prognostic value of major vault protein (MVP) in patients with inflammation, in order to determine whether MVP could be used as a biomarker for infection or inflammation. We also aimed to compare the diagnostic impact of MVP compared to other conventional measurements, such as CRP or white blood cell (WBC) counts. METHODS CRP and MVP levels were measured in 111 sera samples from 85 patients with inflammation admitted to a tertiary-care hospital and 26 healthy individuals during an 18-month period (2019-2020), using nephelometry and a custom MVP sandwich ELISA assay, respectively. In addition, WBC counts were measured using a commercial assay. RESULTS MVP levels were found to be elevated in patients with inflammation compared to healthy individuals (p < 0.0001). Moreover, MVP levels were higher in patients with inflammation due to an infectious etiology compared to those with non-infectious etiology (p = 0.0006). MVP levels significantly decreased during the first four days of infection in response to antibiotic treatment, while CRP levels showed a less-sensitive decline. An ROC curve analysis demonstrated that MVP and CRP have similarly high diagnostic accuracy, with AUCs of 0.955 and 0.995, respectively, followed by WBCs with an AUC of 0.805. CONCLUSIONS The ROC curves demonstrated that MVP has the potential to serve as a diagnostic biomarker for inflammation and infection. Additionally, MVP levels may reflect the efficacy of antibiotic treatment.
Collapse
Affiliation(s)
- John G. Routsias
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | | | | | | |
Collapse
|
22
|
Lodwick JE, Shen R, Erramilli S, Xie Y, Roganowicz K, Kossiakoff AA, Zhao M. Structural Insights into the Roles of PARP4 and NAD + in the Human Vault Cage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601040. [PMID: 38979142 PMCID: PMC11230398 DOI: 10.1101/2024.06.27.601040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Vault is a massive ribonucleoprotein complex found across Eukaryota. The major vault protein (MVP) oligomerizes into an ovular cage, which contains several minor vault components (MVCs) and is thought to transport transiently bound "cargo" molecules. Vertebrate vaults house a poly (ADP-ribose) polymerase (known as PARP4 in humans), which is the only MVC with known enzymatic activity. Despite being discovered decades ago, the molecular basis for PARP4's interaction with MVP remains unclear. In this study, we determined the structure of the human vault cage in complex with PARP4 and its enzymatic substrate NAD + . The structures reveal atomic-level details of the protein-binding interface, as well as unexpected NAD + -binding pockets within the interior of the vault cage. In addition, proteomics data show that human vaults purified from wild-type and PARP4-depleted cells interact with distinct subsets of proteins. Our results thereby support a model in which PARP4's specific incorporation into the vault cage helps to regulate vault's selection of cargo and its subcellular localization. Further, PARP4's proximity to MVP's NAD + -binding sites could support its enzymatic function within the vault.
Collapse
|
23
|
Zhang L, Kuang G, Gong X, Huang R, Zhao Z, Li Y, Wan J, Wang B. Piperine attenuates hepatic ischemia/reperfusion injury via suppressing the TLR4 signaling cascade in mice. Transpl Immunol 2024; 84:102033. [PMID: 38484898 DOI: 10.1016/j.trim.2024.102033] [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: 12/07/2022] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Piperine, the major active substance in black pepper, has been shown to have anti-inflammatory and antioxidant effects in several ischemic diseases. However, the role of piperine in hepatic ischemia/reperfusion injury (HIRI) and its underlying mechanisms remain unclear. In this study, the mice were administered piperine (30 mg/kg) intragastric administration before surgery. After 24 h of hepatic ischemia-reperfusion, liver histopathological evaluation, serum transaminase measurements, and TUNEL analysis were performed. The infiltration of inflammatory cells and production of inflammatory mediators in the liver tissue were determined by immunofluorescence and immunohistochemical staining. The protein levels of toll-like receptor 4 (TLR4) and related proteins such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), interleukin-1 receptor-associated kinase 1 (IRAK1), p65, and p38 were detected by western blotting. The results showed that plasma aminotransferase (ALT), aspartate aminotransferase (AST), hepatocyte apoptosis, oxidative stress, and inflammatory cell infiltration significantly increased in HIRI mice. Piperine pretreatment notably repaired liver function, improved the histopathology and apoptosis of liver cells, alleviated oxidative stress injury, and reduced inflammatory cell infiltration. Further analysis showed that piperine attenuated tumor necrosis factor-a (TNF-α) and interleukin 6 (IL-6) production and reduced TLR4 activation and phosphorylation of IRAK1, p38, and NF-κB in HIRI. Piperine has a protective effect against HIRI through the TLR4/IRAK1/NF-κB signaling pathway and may be a safer option for future clinical treatment and prevention of ischemia-related diseases.
Collapse
Affiliation(s)
- Lidan Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ge Kuang
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - Rui Huang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang 310058, China
| | - Zizuo Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yan Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jingyuan Wan
- Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, China.
| | - Bin Wang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
24
|
Bruck O, Pandit LM. Pulmonary Hypertension and Hyperglycemia-Not a Sweet Combination. Diagnostics (Basel) 2024; 14:1119. [PMID: 38893645 PMCID: PMC11171670 DOI: 10.3390/diagnostics14111119] [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: 04/02/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Hyperglycemia and pulmonary hypertension (PH) share common pathological pathways that lead to vascular dysfunction and resultant cardiovascular complications. These shared pathologic pathways involve endothelial dysfunction, inflammation, oxidative stress, and hormonal imbalances. Individuals with hyperglycemia or pulmonary hypertension also possess shared clinical factors that contribute to increased morbidity from both diseases. This review aims to explore the relationship between PH and hyperglycemia, highlighting the mechanisms underlying their association and discussing the clinical implications. Understanding these common pathologic and clinical factors will enable early detection for those at-risk for complications from both diseases, paving the way for improved research and targeted therapeutics.
Collapse
Affiliation(s)
- Or Bruck
- Section of Pulmonary, Critical Care, Sleep Medicine, Baylor College of Medicine, Houston, TX 77024, USA;
| | - L. M. Pandit
- Section of Pulmonary, Critical Care, Sleep Medicine, Baylor College of Medicine, Houston, TX 77024, USA;
- Michael E. DeBakey Veterans Affairs Medical Center, Center for Translational Research on Inflammatory Diseases (CTRID), Houston, TX 77030, USA
| |
Collapse
|
25
|
Wang D, Li Y, Yang H, Shen X, Shi X, Li C, Zhang Y, Liu X, Jiang B, Zhu X, Zhang H, Li X, Bai H, Yang Q, Gao W, Bai F, Ji Y, Chen Q, Ben J. Disruption of TIGAR-TAK1 alleviates immunopathology in a murine model of sepsis. Nat Commun 2024; 15:4340. [PMID: 38773142 PMCID: PMC11109194 DOI: 10.1038/s41467-024-48708-0] [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: 08/07/2023] [Accepted: 05/11/2024] [Indexed: 05/23/2024] Open
Abstract
Macrophage-orchestrated inflammation contributes to multiple diseases including sepsis. However, the underlying mechanisms remain to be defined clearly. Here, we show that macrophage TP53-induced glycolysis and apoptosis regulator (TIGAR) is up-regulated in murine sepsis models. When myeloid Tigar is ablated, sepsis induced by either lipopolysaccharide treatment or cecal ligation puncture in male mice is attenuated via inflammation inhibition. Mechanistic characterizations indicate that TIGAR directly binds to transforming growth factor β-activated kinase (TAK1) and promotes tumor necrosis factor receptor-associated factor 6-mediated ubiquitination and auto-phosphorylation of TAK1, in which residues 152-161 of TIGAR constitute crucial motif independent of its phosphatase activity. Interference with the binding of TIGAR to TAK1 by 5Z-7-oxozeaenol exhibits therapeutic effects in male murine model of sepsis. These findings demonstrate a non-canonical function of macrophage TIGAR in promoting inflammation, and confer a potential therapeutic target for sepsis by disruption of TIGAR-TAK1 interaction.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Yanxia Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Hao Yang
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiaoqi Shen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaolin Shi
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Chenyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yongjing Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Gao
- The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Fang Bai
- School of Life Science and Technology, and Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Yong Ji
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
26
|
Li C, Gao M, Zha N, Guo G. The prognostic value and immunological role of MVP in pan-cancer study. Aging (Albany NY) 2024; 16:8497-8510. [PMID: 38713157 PMCID: PMC11164508 DOI: 10.18632/aging.205802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/08/2024] [Indexed: 05/08/2024]
Abstract
Major Vault Protein (MVP) has emerged as a potential prognostic and immunological biomarker in various cancer types. This pan-cancer study aimed to investigate expression of MVP and its correlation with clinical outcomes and immune infiltration across diverse cancer types. We conducted an analysis of extensive transcriptomic and clinical data from publicly available databases. Our findings unveiled a significant association between MVP expression and cancer progression, with higher expression levels predicting poorer overall survival in multiple cancer types. Importantly, MVP expression demonstrated a close relationship with immune infiltration in the tumor microenvironment, showing that higher expression levels were associated with increased immune cell infiltration. We further validated expression of MVP and function in cancer cell lines A549 and AGS. These compelling results suggest that MVP holds promise as a valuable biomarker for prognostic assessment and the development of immunotherapeutic strategies across various cancer types. Consequently, targeting MVP may offer a compelling therapeutic approach in the treatment of human cancers.
Collapse
Affiliation(s)
- Chunlin Li
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
| | - Min Gao
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
- Inner Mongolia Medical University, Hohhot 010010, China
| | - Nashunbayaer Zha
- Department of Thoracic Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
| | - Gang Guo
- Department of Thyroid and Breast Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
| |
Collapse
|
27
|
Zhu X, Bai B, Ge X, Zheng B, Xiao Z, Tang Y, Fang L, Tang Y, Dai Y, Zhang B, Zhang Y. Costunolide attenuates LPS-induced inflammation and lung injury through inhibiting IKK/NF-κB signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1601-1610. [PMID: 37688623 DOI: 10.1007/s00210-023-02705-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
Inflammation is an important pathological process of many acute and chronic diseases, such as sepsis, arthritis, and cancer. Many factors can lead to an inflammatory state of the body, among which bacterial infection plays an important role. Bacterial infection often leads to sepsis, acute lung injury (ALI), or its more serious form of acute respiratory distress syndrome, which are the main fatal diseases in intensive care units. Costunolide has been reported to possess excellent anti-inflammatory activity; however, whether it can affect inflammation induced by gram-negative bacterial is still unclear. Lipopolysaccharide (LPS) stimulated mouse peritoneal macrophages (MPMs) to release proinflammatory cytokines was used as the cell model. The mouse model of sepsis and ALI was built through injecting intravenously and intratracheally of LPS. In the present study, costunolide inhibited LPS-induced inflammatory response through IKK/NF-κB signaling pathway in macrophages. In vivo, costunolide attenuated LPS-induced septic death in mice. Meanwhile, costunolide treatment alleviated LPS-induced lung injury and inflammation via inhibiting the infiltration of inflammatory cells and the expression of inflammatory cytokines. Taken together, these results demonstrated that costunolide could attenuate gram-negative bacterial induced inflammation and diseases and might be a potential candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xiaona Zhu
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Bin Bai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiangting Ge
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Bin Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zhongxiang Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Yue Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Letong Fang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yelin Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuanrong Dai
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Bing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
| | - Yali Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
28
|
Tan W, Zhang J, Dai F, Yang D, Gu R, Tang L, Liu H, Cheng YX. Insights on the NF-κB system in polycystic ovary syndrome, attractive therapeutic targets. Mol Cell Biochem 2024; 479:467-486. [PMID: 37097332 DOI: 10.1007/s11010-023-04736-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/07/2023] [Indexed: 04/26/2023]
Abstract
The nuclear factor κappa B (NF-κB) signaling plays a well-known function in inflammation and regulates a wide variety of biological processes. Low-grade chronic inflammation is gradually considered to be closely related to the pathogenesis of Polycystic ovary syndrome (PCOS). In this review, we provide an overview on the involvement of NF-κB in the progression of PCOS particularly, such as hyperandrogenemia, insulin resistance, cardiovascular diseases, and endometrial dysfunction. From a clinical perspective, progressive recognition of NF-κB pathway provides opportunities for therapeutic interventions aimed at inhibiting pathway-specific mechanisms. With the accumulation of basic experimental and clinical data, NF-κB signaling pathway was recognized as a therapeutic target. Although there have been no specific small molecule NF-κB inhibitors in PCOS, a plethora of natural and synthetic compound have emerged for the pharmacologic intervention of the pathway. The traditional herbs developed for NF-κB pathway have become increasingly popular in recent years. Abundant evidence elucidated that NF-κB inhibitors can significantly improve the symptoms of PCOS. Herein, we summarized evidence relating to how NF-κB pathway is involved in the development and progression of PCOS. Furthermore, we present an in-depth overview of NF-κB inhibitors for therapy interventions of PCOS. Taken together, the NF-κB signaling may be a futuristic treatment strategy for PCOS.
Collapse
Affiliation(s)
- Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Ran Gu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Lujia Tang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, 430060, Hubei, People's Republic of China.
| |
Collapse
|
29
|
Fuster-Martínez I, Català-Senent JF, Hidalgo MR, Roig FJ, Esplugues JV, Apostolova N, García-García F, Blas-García A. Integrated transcriptomic landscape of the effect of anti-steatotic treatments in high-fat diet mouse models of non-alcoholic fatty liver disease. J Pathol 2024; 262:377-389. [PMID: 38180387 DOI: 10.1002/path.6242] [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: 06/18/2023] [Revised: 10/20/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
High-fat diet (HFD) mouse models are widely used in research to develop medications to treat non-alcoholic fatty liver disease (NAFLD), as they mimic the steatosis, inflammation, and hepatic fibrosis typically found in this complex human disease. The aims of this study were to identify a complete transcriptomic signature of these mouse models and to characterize the transcriptional impact exerted by different experimental anti-steatotic treatments. For this reason, we conducted a systematic review and meta-analysis of liver transcriptomic studies performed in HFD-fed C57BL/6J mice, comparing them with control mice and HFD-fed mice receiving potential anti-steatotic treatments. Analyzing 21 studies broaching 24 different treatments, we obtained a robust HFD transcriptomic signature that included 2,670 differentially expressed genes and 2,567 modified gene ontology biological processes. Treated HFD mice generally showed a reversion of this HFD signature, although the extent varied depending on the treatment. The biological processes most frequently reversed were those related to lipid metabolism, response to stress, and immune system, whereas processes related to nitrogen compound metabolism were generally not reversed. When comparing this HFD signature with a signature of human NAFLD progression, we identified 62 genes that were common to both; 10 belonged to the group that were reversed by treatments. Altered expression of most of these 10 genes was confirmed in vitro in hepatocytes and hepatic stellate cells exposed to a lipotoxic or a profibrogenic stimulus, respectively. In conclusion, this study provides a vast amount of information about transcriptomic changes induced during the progression and regression of NAFLD and identifies some relevant targets. Our results may help in the assessment of treatment efficacy, the discovery of unmet therapeutic targets, and the search for novel biomarkers. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
| | - José F Català-Senent
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
| | - Marta R Hidalgo
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
| | - Francisco J Roig
- Computational Biomedicine Laboratory, Principe Felipe Research Center, Valencia, Spain
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario Villanueva de Gállego, Zaragoza, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
| | - Nadezda Apostolova
- Departamento de Farmacología, Universitat de València, Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
| | | | - Ana Blas-García
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
- CIBEREHD (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Madrid, Spain
- Departamento de Fisiología, Universitat de València, Valencia, Spain
| |
Collapse
|
30
|
Zhang X, Wang S, Chong N, Chen D, Shu J, Sun J, Sun Z, Wang R, Wang Q, Xu Y. GDF-15 alleviates diabetic nephropathy via inhibiting NEDD4L-mediated IKK/NF-κB signalling pathways. Int Immunopharmacol 2024; 128:111427. [PMID: 38181673 DOI: 10.1016/j.intimp.2023.111427] [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: 09/11/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024]
Abstract
Podocyte inflammatory injury has been indicated to play a pivotal role in the occurrence and development of diabetic nephropathy (DN). However, the pathogenesis of inflammation remains unclear. Recent researches have shown that GDF-15, a member of the transforming growth factor-β superfamily, were elevated under pathological conditions, such as myocardial ischemia, cancer, as well as inflammation. Here, we demonstrated that GDF-15 could alleviate podocyte inflammatory injury by modulating the NF-κB pathway. GDF-15 and other pro-inflammatory factors, such as TNF-α, IL-1β, and IL-6 were upregulated in the serum of HFD/STZ rat models. GDF-15 was also elevated in diabetic glomeruli and hyperglycemic stimuli treated-podocytes. The silence of GDF-15 in HG-stimulated podocytes further augmented inflammation and podocyte injury, while overexpression of GDF-15 significantly reduced the inflammatory response in podocytes. Mechanistically, we demonstrated that GDF-15 could inhibit the nuclear translocation of NF-κB through IKK and IκBα by interaction with ubiquitin ligase NEDD4L. Taken together, our data suggested a protective mechanism of elevated GDF-15 in DN through obstruction of ubiquitin degradation of IKK by inhibiting NEDD4L expression, thus decreasing the activation of NF-κB and relieving the inflammation. GDF-15 could serve as a potential therapeutic target for DN.
Collapse
Affiliation(s)
- Xinyu Zhang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Simeng Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Nannan Chong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dandan Chen
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jianqiang Shu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jingshu Sun
- Department of Nephrology, Weifang People's Hospital, Weifang, Shandong, China
| | - Zhikang Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Rong Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qinglian Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Ying Xu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
31
|
Lv JJ, Wang H, Zhang C, Zhang TJ, Wei HL, Liu ZK, Ma YH, Yang Z, He Q, Wang LJ, Duan LL, Chen ZN, Bian H. CD147 Sparks Atherosclerosis by Driving M1 Phenotype and Impairing Efferocytosis. Circ Res 2024; 134:165-185. [PMID: 38166463 DOI: 10.1161/circresaha.123.323223] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Atherosclerosis is a globally prevalent chronic inflammatory disease with high morbidity and mortality. The development of atherosclerotic lesions is determined by macrophages. This study aimed to investigate the specific role of myeloid-derived CD147 (cluster of differentiation 147) in atherosclerosis and its translational significance. METHODS AND RESULTS We generated mice with a myeloid-specific knockout of CD147 and mice with restricted CD147 overexpression, both in an apoE-deficient (ApoE-/-) background. Here, the myeloid-specific deletion of CD147 ameliorated atherosclerosis and inflammation. Consistent with our in vivo data, macrophages isolated from myeloid-specific CD147 knockout mice exhibited a phenotype shift from proinflammatory to anti-inflammatory macrophage polarization in response to lipopolysaccharide/IFN (interferon)-γ. These macrophages demonstrated a weakened proinflammatory macrophage phenotype, characterized by reduced production of NO and reactive nitrogen species derived from iNOS (inducible NO synthase). Mechanistically, the TRAF6 (tumor necrosis factor receptor-associated factor 6)-IKK (inhibitor of κB kinase)-IRF5 (IFN regulatory factor 5) signaling pathway was essential for the effect of CD147 on proinflammatory responses. Consistent with the reduced size of the necrotic core, myeloid-specific CD147 deficiency diminished the susceptibility of iNOS-mediated late apoptosis, accompanied by enhanced efferocytotic capacity mediated by increased secretion of GAS6 (growth arrest-specific 6) in proinflammatory macrophages. These findings were consistent in a mouse model with myeloid-restricted overexpression of CD147. Furthermore, we developed a new atherosclerosis model in ApoE-/- mice with humanized CD147 transgenic expression and demonstrated that the administration of an anti-human CD147 antibody effectively suppressed atherosclerosis by targeting inflammation and efferocytosis. CONCLUSIONS Myeloid CD147 plays a crucial role in the growth of plaques by promoting inflammation in a TRAF6-IKK-IRF5-dependent manner and inhibiting efferocytosis by suppressing GAS6 during proinflammatory conditions. Consequently, the use of anti-human CD147 antibodies presents a complementary therapeutic approach to the existing lipid-lowering strategies for treating atherosclerotic diseases.
Collapse
Affiliation(s)
- Jian-Jun Lv
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Cong Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Tian-Jiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Hao-Lin Wei
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Ze-Kun Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Yi-Hui Ma
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital (Z.Y.), Fourth Military Medical University, Xi'an, China
| | - Qian He
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Li-Juan Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Li-Li Duan
- Department of Gastrointestinal Surgery, State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases (L.-L.D.), Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Huijie Bian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| |
Collapse
|
32
|
Lv N, Zhang Y, Wang L, Suo Y, Zeng W, Yu Q, Yu B, Jiang X. LncRNA/CircRNA-miRNA-mRNA Axis in Atherosclerotic Inflammation: Research Progress. Curr Pharm Biotechnol 2024; 25:1021-1040. [PMID: 37842894 DOI: 10.2174/0113892010267577231005102901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 10/17/2023]
Abstract
Atherosclerosis is characterized by chronic inflammation of the arterial wall. However, the exact mechanism underlying atherosclerosis-related inflammation has not been fully elucidated. To gain insight into the mechanisms underlying the inflammatory process that leads to atherosclerosis, there is need to identify novel molecular markers. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-protein-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have gained prominence in recent years. LncRNAs/circRNAs act as competing endogenous RNAs (ceRNAs) that bind to miRNAs via microRNA response elements (MREs), thereby inhibiting the silencing of miRNA target mRNAs. Inflammatory mediators and inflammatory signaling pathways are closely regulated by ceRNA regulatory networks in atherosclerosis. In this review, we discuss the role of LncRNA/CircRNA-miRNA-mRNA axis in atherosclerotic inflammation and how it can be targeted for early clinical detection and treatment.
Collapse
Affiliation(s)
- Nuan Lv
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yilin Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luming Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanrong Suo
- Traditional Chinese Medicine Department, Ganzhou People's Hospital, Ganzhou, China
| | - Wenyun Zeng
- Oncology Department, Ganzhou People's Hospital, Ganzhou, China
| | - Qun Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
33
|
Møller AL, Vasan RS, Levy D, Andersson C, Lin H. Integrated omics analysis of coronary artery calcifications and myocardial infarction: the Framingham Heart Study. Sci Rep 2023; 13:21581. [PMID: 38062110 PMCID: PMC10703905 DOI: 10.1038/s41598-023-48848-1] [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: 02/08/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Gene function can be described using various measures. We integrated association studies of three types of omics data to provide insights into the pathophysiology of subclinical coronary disease and myocardial infarction (MI). Using multivariable regression models, we associated: (1) single nucleotide polymorphism, (2) DNA methylation, and (3) gene expression with coronary artery calcification (CAC) scores and MI. Among 3106 participants of the Framingham Heart Study, 65 (2.1%) had prevalent MI and 60 (1.9%) had incident MI, median CAC value was 67.8 [IQR 10.8, 274.9], and 1403 (45.2%) had CAC scores > 0 (prevalent CAC). Prevalent CAC was associated with AHRR (linked to smoking) and EXOC3 (affecting platelet function and promoting hemostasis). CAC score was associated with VWA1 (extracellular matrix protein associated with cartilage structure in endomysium). For prevalent MI we identified FYTTD1 (down-regulated in familial hypercholesterolemia) and PINK1 (linked to cardiac tissue homeostasis and ischemia-reperfusion injury). Incident MI was associated with IRX3 (enhancing browning of white adipose tissue) and STXBP3 (controlling trafficking of glucose transporter type 4 to plasma). Using an integrative trans-omics approach, we identified both putatively novel and known candidate genes associated with CAC and MI. Replication of findings is warranted.
Collapse
Affiliation(s)
- Amalie Lykkemark Møller
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
- Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark.
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
- University of Texas School of Public Health San Antonio, and Departments of Medicine and Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daniel Levy
- Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Institutes of Health, Bethesda, MD, USA
| | - Charlotte Andersson
- Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
34
|
Deng X, Liu Z, Han S. Cimifugin inhibits adipogenesis and TNF-α-induced insulin resistance in 3T3-L1 cells. Open Med (Wars) 2023; 18:20230855. [PMID: 38045856 PMCID: PMC10693008 DOI: 10.1515/med-2023-0855] [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: 08/08/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
To investigate the effects of cimifugin on adipogenesis and tumor necrosis factor (TNF-α)-induced insulin resistance (IR) and inflammation in 3T3-L1 adipocytes. 3T3-L1 adipocytes were treated with 3-isobutyl-1-methyl-xanthine, dexamethasone, and insulin or cimifugin and then Oil Red O staining and intracellular triglyceride content detection were performed to assess adipogenesis. Subsequently, after cimifugin treatment, TNF-α was used to induce IR and inflammation. The results showed that cimifugin reduced intracellular lipids accumulation of 3T3-L1 adipocytes. Cimifugin improved IR of 3T3-L1 adipocytes induced by TNF-α, as reflected in decreased adiponectin, GLUT-4, and IRS-1 mRNA and protein expression. Moreover, cimifugin reduced TNF-α-induced pro-inflammatory factors production and phospho-P65 expression, and MAPK pathway activation in the 3T3-L1 adipocytes. These findings suggested that cimifugin might be useful for the prevention and therapy of obesity-related IR and inflammation.
Collapse
Affiliation(s)
- Xiang Deng
- Department of Pediatrics, Chengdu Fifth People’s Hospital, No. 33, Mashi Street, Wenjiang District, Chengdu, Sichuan, 611130, China
| | - Zhenmin Liu
- Department of Pediatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| | - Siqi Han
- Department of Pediatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| |
Collapse
|
35
|
Zhou H, Yan L, Huang H, Li X, Xia Q, Zheng L, Shao B, Gao Q, Sun N, Shi J. Tat-NTS peptide protects neurons against cerebral ischemia-reperfusion injury via ANXA1 SUMOylation in microglia. Theranostics 2023; 13:5561-5583. [PMID: 37908731 PMCID: PMC10614677 DOI: 10.7150/thno.85390] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/26/2023] [Indexed: 11/02/2023] Open
Abstract
Rationale: Recent studies indicate that microglial activation and the resulting inflammatory response could be potential targets of adjuvant therapy for ischemic stroke. Many studies have emphasized a well-established function of Annexin-A1 (ANXA1) in the immune system, including the regulation of microglial activation. Nevertheless, few therapeutic interventions targeting ANXA1 in microglia for ischemic stroke have been conducted. In the present study, Tat-NTS, a small peptide developed to prevent ANXA1 from entering the nucleus, was utilized. We discovered the underlying mechanism that Tat-NTS peptide targets microglial ANXA1 to protect against ischemic brain injury. Methods: Preclinical studies of ischemic stroke were performed using an oxygen-glucose deprivation and reperfusion (OGD/R) cell model in vitro and the middle cerebral artery occlusion (MCAO) animal model of ischemic stroke in vivo. Confocal imaging and 3D reconstruction analyses for detecting the protein expression and subcellular localization of microglia in vivo. Co-immunoprecipitation (Co-IP), immunoblotting, ELISA, quantitative real-time PCR (qRT-PCR), Luciferase reporter assay for determining the precise molecular mechanism. Measurement on the cytotoxicity of Tat-NTS peptide for microglia was assessed by CCK-8 and LDH assay. TUNEL staining was used to detect the microglia conditioned medium-mediated neuronal apoptosis. Adeno-associated viruses (AAVs) were injected into the cerebral cortex, striatum and hippocampal CA1 region of adult male Cx3cr1-Cre mice, to further verify the neurofunctional outcome and mechanism of Tat-NTS peptide by TTC staining, the modified Neurological Severity Score (mNSS) test, the open field test (OFT), the novel object recognition task (NORT), the Morris water maze (MWM) test, the long-term potentiation (LTP) and the Transmission electron microscopy (TEM). Results: It was observed that administration of Tat-NTS led to a shift of subcellular localization of ANXA1 in microglia from the nucleus to the cytoplasm in response to ischemic injury. Notably, this shift was accompanied by an increase in ANXA1 SUMOylation in microglia and a transformation of microglia towards an anti-inflammatory phenotype. We confirmed that Tat-NTS-induced ANXA1 SUMOylation in microglia mediated IKKα degradation via NBR1-dependent selective autophagy, then blocking the activation of the NF-κB pathway. As a result, the expression and release of the pro-inflammatory factors IL-1β and TNF-α were reduced in both in vitro and in vivo experiments. Furthermore, we found that Tat-NTS peptide's protective effect on microglia relieved ischemic neuron apoptosis. Finally, we demonstrated that Tat-NTS peptide administration, through induction of ANXA1 SUMOylation in microglia, reduced infarct volume, improved neurological function and facilitated behavioral recovery in MCAO mice. Conclusions: Our study provides evidence for a novel mechanism of Tat-NTS peptide in regulating microglial ANXA1 function and its substantial neuroprotective effect on neurons with ischemic injuries. These findings suggest that Tat-NTS peptides have a high potential for clinical application and may be a promising therapeutic candidate for treating cerebral ischemia.
Collapse
Affiliation(s)
- Huijuan Zhou
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| | - Lulu Yan
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| | - Hezhou Huang
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xing Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Qian Xia
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lu Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Bin Shao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| | - Qian Gao
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| | - Ning Sun
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| | - Jing Shi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Neurological Diseases, Ministry of Education, Wuhan, Hubei 430030, China
| |
Collapse
|
36
|
Li H, Niu X, Zhang D, Qu MH, Yang K. The role of the canonical nf-κb signaling pathway in the development of acute liver failure. Biotechnol Genet Eng Rev 2023; 39:775-795. [PMID: 36578157 DOI: 10.1080/02648725.2022.2162999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
As a clinical emergency with a high mortality rate, the treatment of acute liver failure has been paid attention to by society. At present, liver transplantation is the most effective treatment for acute liver failure, but there is still an insufficient supply of liver sources and a poor prognosis. In view of the current therapeutic development of this disease, more researchers have turned their attention to the research of drugs related to the NF-κB pathway. The NF-κB canonical pathway has been proven to play a role in a variety of diseases, regulating inflammation, apoptosis, and other physiological processes. More and more evidence shows that the NF-κB canonical pathway regulates the pathogenesis of acute liver failure. In this review, we will summarize the regulation process of the NF-κB canonical pathway on acute liver failure, and develop a new way to treat acute liver failure by targeting the components of the pathway.
Collapse
Affiliation(s)
- Hanyue Li
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Xiao Niu
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Dajin Zhang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
| | - Mei-Hua Qu
- Biopharmaceutical Laboratory, Key Laboratory of Shandong Province Colleges and Universities, School of life science and Technology, Weifang Medical University, Weifang, China
| | - Kunning Yang
- Translational Medical Center, Weifang Second People's Hospital, Weifang Respiratory Disease Hospital, Weifang, China
| |
Collapse
|
37
|
Wang R, Yang Y, Zhang Z, Zhao N, Wiemer EAC, Ben J, Ma J, Yuan L. Major vault protein (MVP) suppresses aging- and estrogen deficiency-related bone loss through Fas-mediated apoptosis in osteoclasts. Cell Death Dis 2023; 14:604. [PMID: 37704623 PMCID: PMC10500014 DOI: 10.1038/s41419-023-05928-4] [Citation(s) in RCA: 4] [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: 11/07/2022] [Revised: 06/02/2023] [Accepted: 06/23/2023] [Indexed: 09/15/2023]
Abstract
Osteoclasts (OCs), derived from monocyte/macrophage lineage, are key orchestrators in bone remodeling. Targeting osteoclast apoptosis is a promising approach to cut down excessive osteoclast numbers, and thus slow down the rate of bone mass loss that inevitably occurs during aging. However, the therapeutic target of apoptosis in osteoclasts has not been fully studied. Our previous work generated Mvpf/fLyz2-Cre mice, conditionally depleting major vault protein (MVP) in monocyte lineage, and identified MVP as a bone protector for its negative role in osteoclastogenesis in vivo and in vitro. Here, we observed a notable decline of MVP in osteoclasts with aging in mice, encouraging us to further investigate the regulatory role of osteoclast MVP. Then, Mvpf/fLyz2-Cre mice were exploited in two osteoporosis contexts, aging and abrupt loss of estrogen, and we revealed that conditional knockout of MVP inhibited osteoclast apoptosis in vivo and in vitro. Moreover, we reported the interaction between MVP and death receptor Fas, and MVP-Fas signaling cascade was identified to positively regulate the apoptosis of osteoclasts, thus preventing osteoporosis. Collectively, our comprehensive discovery of MVP's regulatory role in osteoclasts provides new insight into osteoclast biology and therapeutic targets for osteoporosis.
Collapse
Affiliation(s)
- Ruobing Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yan Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhongyin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Na Zhao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| | - Lichan Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
38
|
Voskamp AL, Tak T, Gerdes ML, Menafra R, Duijster E, Jochems SP, Kielbasa SM, Kormelink TG, Stam KA, van Hengel OR, de Jong NW, Hendriks RW, Kloet SL, Yazdanbakhsh M, de Jong EC, Gerth van Wijk R, Smits HH. Inflammatory and tolerogenic myeloid cells determine outcome following human allergen challenge. J Exp Med 2023; 220:e20221111. [PMID: 37428185 PMCID: PMC10333709 DOI: 10.1084/jem.20221111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/08/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Innate mononuclear phagocytic system (MPS) cells preserve mucosal immune homeostasis. We investigated their role at nasal mucosa following allergen challenge with house dust mite. We combined single-cell proteome and transcriptome profiling on nasal immune cells from nasal biopsies cells from 30 allergic rhinitis and 27 non-allergic subjects before and after repeated nasal allergen challenge. Biopsies of patients showed infiltrating inflammatory HLA-DRhi/CD14+ and CD16+ monocytes and proallergic transcriptional changes in resident CD1C+/CD1A+ conventional dendritic cells (cDC)2 following challenge. In contrast, non-allergic individuals displayed distinct innate MPS responses to allergen challenge: predominant infiltration of myeloid-derived suppressor cells (MDSC: HLA-DRlow/CD14+ monocytes) and cDC2 expressing inhibitory/tolerogenic transcripts. These divergent patterns were confirmed in ex vivo stimulated MPS nasal biopsy cells. Thus, we identified not only MPS cell clusters involved in airway allergic inflammation but also highlight novel roles for non-inflammatory innate MPS responses by MDSC to allergens in non-allergic individuals. Future therapies should address MDSC activity as treatment for inflammatory airway diseases.
Collapse
Affiliation(s)
- Astrid L. Voskamp
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Tamar Tak
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Maarten L. Gerdes
- Department of Ear, Nose and Throat, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Roberta Menafra
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Ellen Duijster
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Simon P. Jochems
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Szymon M. Kielbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Tom Groot Kormelink
- Department of Exp Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Koen A. Stam
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Nicolette W. de Jong
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Susan L. Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Esther C. de Jong
- Department of Exp Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Roy Gerth van Wijk
- Department of Internal Medicine, Section Allergology and Clinical Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Hallmann A, Leszczyńska D, Czumaj A, Świeżak J, Caban M, Michnowska A, Smolarz K. Oxytetracycline-induced inflammatory process without oxidative stress in blue mussels Mytilus trossulus. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:80462-80477. [PMID: 37301807 PMCID: PMC10345040 DOI: 10.1007/s11356-023-28057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Potentially harmful compounds including pharmaceuticals are commonly found in marine waters and sediments. Amongst those, antibiotics and their metabolites are detected worldwide in various abiotic (at concentrations as high as µg/L) and biotic matrices at ng/gram of tissue, posing a risk to non-target species exposed to them such as blue mussels. Amongst those, oxytetracycline (OTC) belongs to the most detected antibiotics in the marine environment. In this work, we concentrated on studying the potential induction of oxidative stress, activation of cellular detoxification processes (including Phase I and Phase II xenobiotic biotransformation enzymes) and multixenobiotic resistance pumps (Phase III) as well as changes in the aromatisation efficiency in Mytilus trossulus exposed to 100 μg/L OTC. Our results show that 100 µg/L OTC concentration did not provoke cellular oxidative stress and did not affect the expression of genes involved in detoxification processes in our model. Moreover, no effect of OTC on aromatisation efficiency was found. Instead, phenoloxidase activity measured in haemolymph was significantly higher in OTC exposed mussels than in those from the control (30.95 ± 3.33 U/L and 17.95 ± 2.75 U/L, respectively). OTC exposed mussels were also characterised by a tissue-dependant activation of major vault protein (MVP) gene expression (1.5 times higher in gills and 2.4 times higher in the digestive system) and a decreased expression of the nuclear factor kappa B-a (NF-κB) gene (3.4 times lower in the digestive system) when compared to those from the control. Additionally, an elevated number of regressive changes and inflammatory responses in tissues such as gills, digestive system and mantle (gonads) was observed underlining the worsening of bivalves' general health. Therefore, instead of a free-radical effect of OTC, we for the first time describe the occurrence of typical changes resulting from antibiotic therapy in non-target organisms like M. trossulus exposed to antibiotics such as OTC.
Collapse
Affiliation(s)
- Anna Hallmann
- Department of Pharmaceutical Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Dagmara Leszczyńska
- Department of Pharmaceutical Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Medical University of Gdańsk, Gdańsk, Poland
| | - Justyna Świeżak
- Department of Marine Ecosystem Functioning, University of Gdańsk, Gdynia, Poland
| | - Magda Caban
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Alicja Michnowska
- Department of Marine Ecosystem Functioning, University of Gdańsk, Gdynia, Poland
| | - Katarzyna Smolarz
- Department of Marine Ecosystem Functioning, University of Gdańsk, Gdynia, Poland.
| |
Collapse
|
40
|
Farina FM, Weber C, Santovito D. The emerging landscape of non-conventional RNA functions in atherosclerosis. Atherosclerosis 2023; 374:74-86. [PMID: 36725418 DOI: 10.1016/j.atherosclerosis.2023.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/15/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
Most of the human genome is transcribed into non-coding RNAs (ncRNAs), which encompass a heterogeneous family of transcripts including microRNAs (miRNAs), long ncRNAs (lncRNAs), circular RNAs (circRNAs), and others. Although the detailed modes of action of some classes are not fully elucidated, the common notion is that ncRNAs contribute to sculpting gene expression of eukaryotic cells at multiple levels. These range from the regulation of chromatin remodeling and transcriptional activity to post-transcriptional regulation of messenger RNA splicing, stability, and decay. Many of these functions ultimately govern the expression of coding and non-coding genes to affect diverse physiological and pathological mechanisms in vascular biology and beyond. As such, different classes of ncRNAs emerged as crucial regulators of vascular integrity as well as active players in the pathophysiology of atherosclerosis from the early stages of endothelial dysfunction to the clinically relevant complications. However, research in recent years revealed unexpected findings such as small ncRNAs being able to biophysically regulate protein function, the glycosylation of ncRNAs to be exposed on the cell surface, the release of ncRNAs in the extracellular space to act as ligands of receptors, and even the ability of non-coding portion of messenger RNAs to mediate structural functions. This evidence expanded the functional repertoire of ncRNAs far beyond gene regulation and highlighted an additional layer of biological control of cell function. In this Review, we will discuss these emerging aspects of ncRNA biology, highlight the implications for the mechanisms of vascular biology and atherosclerosis, and discuss possible translational implications.
Collapse
Affiliation(s)
- Floriana Maria Farina
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität (LMU), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany; Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy.
| |
Collapse
|
41
|
Yang Y, Zhao N, Wang R, Zhan Z, Guo S, Song H, Wiemer EAC, Ben J, Ma J. Macrophage MVP regulates fracture repair by promoting M2 polarization via JAK2-STAT6 pathway. Int Immunopharmacol 2023; 120:110313. [PMID: 37267856 DOI: 10.1016/j.intimp.2023.110313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 04/19/2023] [Accepted: 05/07/2023] [Indexed: 06/04/2023]
Abstract
OBJECTIVE Major vault protein (MVP) is vital in various macrophage-related inflammatory diseases. However, the effects of MVP on macrophage polarization during fracture repair are still unknown. METHODS We used Mvpflox/floxLyz2-Cre mice (myeloid-specific MVP gene knockout, abbreviated as MacKO) and Mvpflox/flox (abbreviated as MacWT) mice to compare their fracture healing phenotype. Next, we traced the changes in macrophage immune status in vivo and in vitro. We further explored the effects of MVP on osteogenesis and osteoclastogenesis. Finally, we re-expressed MVP in MacKO mice to confirm the role of MVP in fracture healing. RESULTS The lack of MVP in macrophages impaired their transition from a pro-inflammatory to an anti-inflammatory phenotype during fracture repair. The increased secretion of pro-inflammatory cytokines by macrophages promoted their osteoclastic differentiation and impaired BMSC osteogenic differentiation, ultimately leading to impaired fracture repair in MacKO mice. Last, adeno-associated virus (AAV)-Mvp tibial injection significantly promoted fracture repair in MacKO mice. CONCLUSIONS Our findings showed MVP has a previously unknown immunomodulatory role in macrophages during fracture repair. Targeting macrophage MVP may represent a novel therapeutic method for fracture treatment.
Collapse
Affiliation(s)
- Yan Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Na Zhao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Ruobing Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhuorong Zhan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Shuyu Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Haiyang Song
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
42
|
Yang H, Huang Z, Luo Y, Lei D, Yan P, Shen A, Liu W, Li D, Wu Z. TRIM37 exacerbates hepatic ischemia/reperfusion injury by facilitating IKKγ translocation. Mol Med 2023; 29:62. [PMID: 37158850 PMCID: PMC10165779 DOI: 10.1186/s10020-023-00653-2] [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: 01/26/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury is one of the major pathological processes associated with various liver surgeries. However, there is still a lack of strategies to protect against hepatic I/R injury because of the unknown underlying mechanism. The present study aimed to identify a potential strategy and provide a fundamental experimental basis for treating hepatic I/R injury. METHOD A classic 70% ischemia/reperfusion injury was established. Immunoprecipitation was used to identify direct interactions between proteins. The expression of proteins from different subcellular localizations was detected by Western blotting. Cell translocation was directly observed by immunofluorescence. HE, TUNEL and ELISA were performed for function tests. RESULT We report that tripartite motif containing 37 (TRIM37) aggravates hepatic I/R injury through the reinforcement of IKK-induced inflammation following dual patterns. Mechanistically, TRIM37 directly interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), inducing K63 ubiquitination and eventually leading to the phosphorylation of IKKβ. TRIM37 enhances the translocation of IKKγ, a regulatory subunit of the IKK complex, from the nucleus to the cytoplasm, thereby stabilizing the cytoplasmic IKK complex and prolonging the duration of inflammation. Inhibition of IKK rescued the function of TRIM37 in vivo and in vitro. CONCLUSION Collectively, the present study discloses some potential function of TRIM37 in hepatic I/R injury. Targeting TRIM37 might be potential for treatment against hepatic I/R injury.Targeting TRIM37 might be a potential treatment strategy against hepatic I/R injury.
Collapse
Affiliation(s)
- Hang Yang
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yunhai Luo
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Dengliang Lei
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
| | - Ping Yan
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ai Shen
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Wenbin Liu
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Dewei Li
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China.
| | - Zhongjun Wu
- The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
43
|
Wei D, Tian X, Zhai X, Sun C. Adipose Tissue Macrophage-Mediated Inflammation in Obesity: A Link to Posttranslational Modification. Immunol Invest 2023:1-25. [PMID: 37129471 DOI: 10.1080/08820139.2023.2205883] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Adipose tissue macrophages (ATM) are an essential type of immune cells in adipose tissue. Obesity induces the inflammation of adipose tissues, as expressed by ATM accumulation, that is more likely to become a source of systemic metabolic diseases, including insulin resistance. The process is characterized by the transcriptional regulation of inflammatory pathways by virtue of signaling molecules such as cytokines and free fatty acids. Notably, posttranslational modification (PTM) is a key link for these signaling molecules to trigger the proinflammatory or anti-inflammatory phenotype of ATMs. This review focuses on summarizing the functions and molecular mechanisms of ATMs regulating inflammation in obese adipose tissue. Furthermore, the role of PTM is elaborated, hoping to identify new horizons of treatment and prevention for obesity-mediated metabolic disease.
Collapse
Affiliation(s)
- Dongqin Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
| | - Xin Tian
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
| | - Xiangyun Zhai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi, China
| |
Collapse
|
44
|
Li W, Huang Z, Luo Y, Cui Y, Xu M, Luo W, Wu G, Liang G. Tetrandrine alleviates atherosclerosis via inhibition of STING-TBK1 pathway and inflammation in macrophages. Int Immunopharmacol 2023; 119:110139. [PMID: 37099944 DOI: 10.1016/j.intimp.2023.110139] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/22/2023] [Accepted: 03/31/2023] [Indexed: 04/28/2023]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease. Recent studies have showed that stimulator of interferon genes (STING), an important protein in innate immunity, mediates pro-inflammatory activation of macrophages in the development of AS. Tetrandrine (TET) is a natural bisbenzylisoquinoline alkaloid isolated from Stepania tetrandra and possesses anti-inflammatory activities, with unknown effects and mechanisms in AS. In this study, we explored the anti-atherosclerotic effects of TET and investigated the underlying mechanisms. Mouse primary peritoneal macrophages (MPMs) are challenged with cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) or oxidized LDL (oxLDL). We found that pretreatment with TET dose-dependently inhibited cGAMP- or oxLDL-induced STING/ TANK-binding kinase 1 (TBK1) signaling, then suppressing nuclear factor kappa-B (NF-κB) activation and pro-inflammatory factor expression in MPMs. ApoE-/- mice were fed a high-fat diet (HFD) to develop an atherosclerotic phenotype. Administration of TET at 20 mg/kg/day significantly reduced HFD-induced atherosclerotic plaques, accompanied with decreased macrophage infiltration, inflammatory cytokine production, fibrosis, and STING/TBK1 activation in aortic plaque lesions. In summary, we demonstrate that TET inhibits STING/TBK1/NF-κB signaling pathway to reduce inflammation in oxLDL-challenged macrophages and alleviate atherosclerosis in HFD-fed ApoE-/- mice. These findings proved that TET could be a potential therapeutic candidate for the treatment of atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Weixin Li
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhuqi Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yue Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yaqian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mingjiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
45
|
Gao Y, Wang R, Li L, He Y, Yuan D, Zhang Y, Hu Y, Wang S, Yuan C. Total saponins from Panax japonicus reduce inflammation in adipocytes through the miR155/SOCS1/NFκB signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154827. [PMID: 37087792 DOI: 10.1016/j.phymed.2023.154827] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 04/07/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND The rising incidence of metabolic diseases due to chronic inflammation in the adipose tissue has been attributed to factors such as high fat diet (HFD). Previous studies have demonstrated that the total saponins from Panax japonicus (TSPJ) can reduce HFD-induced adipocyte inflammation, but the underlying mechanism remains unclear. In this work, we explored the molecular mechanism by which TSPJ reduces inflammation response in adipocytes. METHODS We first established C57BL/6 mouse and 3T3-L1 adipocyte models. Lentiviruses packaged with the plasmids were injected into mice through the tail vein or into adipocytes to generate the in vivo and in vitro models with miR155 knockdown and overexpression. The mice were fed with HFD to trigger inflammation and administered TSPJ (25 mg/kg∙d and 75 mg/kg∙d) by gavage. The adipocytes were treated with palmitic acid (PA) to trigger inflammation response, then treated with TSPJ (25 μg/ml and 50 μg/ml). Finally, the expression of miR155, inflammatory factors, SOCS1, and NFκB pathway-related proteins was explored. RESULTS TSPJ significantly inhibited the expression of inflammation-related genes and the miR155 expression in adipocytes both in vitro and in vivo. The dual luciferase reporter gene assay revealed that miR155 mediated the downregulation of SOCS1. TSPJ significantly inhibited and upregulated the phosphorylation of the NFκB protein and the SOCS1 proteins, respectively. CONCLUSION TSPJ inhibits miR155 to upregulate the SOCS1 expression, which subsequently inhibits the NFκB signaling pathway, thereby mitigating the inflammatory response in the adipocytes of HFD mice.
Collapse
Affiliation(s)
- Yan Gao
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Rui Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Luoying Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yumin He
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Ding Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Yifan Zhang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yaqi Hu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Shuwen Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| |
Collapse
|
46
|
Li Q, Wang C, Hu J, Jiao W, Tang Z, Song X, Wu Y, Dai J, Gao P, Du L, Jin Y. Cannabidiol-loaded biomimetic macrophage membrane vesicles against post-traumatic stress disorder assisted by ultrasound. Int J Pharm 2023; 637:122872. [PMID: 36958611 DOI: 10.1016/j.ijpharm.2023.122872] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
Post-traumatic stress disorder (PTSD), which normally follows psychological trauma, has been increasingly studied as a brain disease. However, the blood-brain barrier (BBB) prevents conventional drugs for PTSD from entering the brain. Our previous studies proved the effectiveness of cannabidiol (CBD) against PTSD, but low water solubility, low brain targeting efficiency and poor bioavailability restricted its applications. Here, a bionic delivery system, camouflage CBD-loaded macrophage-membrane nanovesicles (CMNVs), was constructed via co-extrusion of CBD with macrophage membranes, which had inflammatory and immune escape properties. In vitro anti-inflammatory, cellular uptake and pharmacokinetic experiments respectively verified the anti-inflammatory, inflammatory targeting and immune escape properties of CMNVs. Brain targeting and excellent anti-PTSD effects of CMNVs had been validated in vivo by imaging and pharmacodynamics studies. In our study, the potential of ultrasound to open BBBs and improve the brain-targeted delivery of CBD was evaluated. In conclusion, this cell membrane bionic delivery system assisted with ultrasound had good therapeutic effect against PTSD mice, which is expected to help convey CBD to inflammatory areas within the brain and alleviate the symptoms of PTSD.
Collapse
Affiliation(s)
- Qi Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chunqing Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jinglu Hu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Pharmaceutical College, Henan University, Kaifeng 475004, China
| | - Wencheng Jiao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Pharmaceutical College, Hebei University, Baoding 071000, China
| | - Ziyan Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xingshuang Song
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yanping Wu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Dai
- Information Department, General Hospital of Western Theater Command, Chengdu 610083, China
| | - Peng Gao
- R&D Institute, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Lina Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Pharmaceutical College, Henan University, Kaifeng 475004, China; Pharmaceutical College, Hebei University, Baoding 071000, China.
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Pharmaceutical College, Henan University, Kaifeng 475004, China
| |
Collapse
|
47
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Hakami ZH, Khamjan NA, Saad HM, Batiha GES, De Waard M. A Potential Link Between Visceral Obesity and Risk of Alzheimer's Disease. Neurochem Res 2023; 48:745-766. [PMID: 36409447 DOI: 10.1007/s11064-022-03817-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia characterized by the deposition of amyloid beta (Aβ) plaques and tau-neurofibrillary tangles in the brain. Visceral obesity (VO) is usually associated with low-grade inflammation due to higher expression of pro-inflammatory cytokines by adipose tissue. The objective of the present review was to evaluate the potential link between VO and the development of AD. Tissue hypoxia in obesity promotes tissue injury, production of adipocytokines, and release of pro-inflammatory cytokines leading to an oxidative-inflammatory loop with induction of insulin resistance. Importantly, brain insulin signaling is involved in the pathogenesis of AD and lower cognitive function. Obesity and enlargement of visceral adipose tissue are associated with the deposition of Aβ. All of this is consonant with VO increasing the risk of AD through the dysregulation of adipocytokines which affect the development of AD. The activated nuclear factor kappa B (NF-κB) pathway in VO might be a potential link in the development of AD. Likewise, the higher concentration of advanced glycation end-products in VO could be implicated in the pathogenesis of AD. Taken together, different inflammatory signaling pathways are activated in VO that all have a negative impact on the cognitive function and progression of AD except hypoxia-inducible factor 1 which has beneficial and neuroprotective effects in mitigating the progression of AD. In addition, VO-mediated hypoadiponectinemia and leptin resistance may promote the progression of Aβ formation and tau phosphorylation with the development of AD. In conclusion, VO-induced AD is mainly mediated through the induction of oxidative stress, inflammatory changes, leptin resistance, and hypoadiponectinemia that collectively trigger Aβ formation and neuroinflammation. Thus, early recognition of VO by visceral adiposity index with appropriate management could be a preventive measure against the development of AD in patients with VO.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120, Saint-Egrève, France.,L'institut du thorax, INSERM, CNRS, UNIV NANTES, 44007, Nantes, France.,LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
48
|
Xu M, Xue H, Kong L, Lin L, Zheng G. Smilax china L. Polyphenols Improves Insulin Resistance and Obesity in High-fat Diet-induced Mice Through IRS/AKT-AMPK and NF-κB Signaling Pathways. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2023:10.1007/s11130-023-01052-y. [PMID: 36826691 DOI: 10.1007/s11130-023-01052-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Smilax china L. is an important herb used in traditional Chinese medicine. In this study, the mechanism of Smilax china L. polyphenols (SCP) on insulin resistance and anti-obesity in mice induced by a high-fat diet (HFD) was investigated. Fifty female mice were randomly divided into five groups: control, HFD and low, medium, and high doses of SCP for 70 d. SCP significantly decreased intraperitoneal adipose tissue index, body weight gain, liver lipids, and serum inflammatory factor levels. Blood glucose and insulin concentrations, as well as insulin resistance index in SCP, were significantly lower than those in HFD. In addition, SCP markedly up-regulated the gene expression of glucose transporter 4 (GLUT4), insulin receptor substrate 1 (IRS1), insulin receptor substrate 2 (IRS2), serine-threonine kinase (AKT), Acyl-CoA oxidase (ACO), and protein kinase A (PKA), and down-regulated the expression of mammalian target of rapamycin complex 1 (mTORC1), sterol-responsive element-binding protein-1c (SREBP1c), fatty acid synthase (FAS), 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR), and forkhead box protein O1 (FOXO1). SCP significantly increased the protein expression of AKT, GLUT4, AMP-activated protein kinase (AMPK), phosphorylated-AMPK (p-AMPK), phosphorylated-AKT (p-AKT), and uncoupling protein 1 (UCP-1), and decreased the expression of SREBP1c, FAS, HMGCR, phosphorylation of IKBα (p-IKBα), and nuclear factor kappa B subunit p65 (P65) in the liver. Overall, SCP effectively reduced HFD-induced insulin resistance and obesity in mice, partly through NF-κB and IRS/AKT-AMPK signaling pathways to regulate inflammatory factors. Therefore, SCP may improve lifestyle diseases.
Collapse
Affiliation(s)
- Meng Xu
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Hui Xue
- Engineering Research Center of Biomass Conversion, Ministry of Education, Nanchang University, 330047, Nanchang, China
| | - Li Kong
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Lezhen Lin
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, School of Food Science and Engineering, Jiangxi Agricultural University, 330045, Nanchang, China.
| |
Collapse
|
49
|
Gan J, Guo L, Zhang X, Yu Q, Yang Q, Zhang Y, Zeng W, Jiang X, Guo M. Anti-inflammatory therapy of atherosclerosis: focusing on IKKβ. J Inflamm (Lond) 2023; 20:8. [PMID: 36823573 PMCID: PMC9951513 DOI: 10.1186/s12950-023-00330-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Chronic low-grade inflammation has been identified as a major contributor in the development of atherosclerosis. Nuclear Factor-κappa B (NF-κB) is a critical transcription factors family of the inflammatory pathway. As a major catalytic subunit of the IKK complex, IκB kinase β (IKKβ) drives canonical activation of NF-κB and is implicated in the link between inflammation and atherosclerosis, making it a promising therapeutic target. Various natural product derivatives, extracts, and synthetic, show anti-atherogenic potential by inhibiting IKKβ-mediated inflammation. This review focuses on the latest knowledge and current research landscape surrounding anti-atherosclerotic drugs that inhibit IKKβ. There will be more opportunities to fully understand the complex functions of IKKβ in atherogenesis and develop new effective therapies in the future.
Collapse
Affiliation(s)
- Jiali Gan
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Guo
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaolu Zhang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qun Yu
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuyue Yang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yilin Zhang
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- grid.459559.10000 0004 9344 2915Oncology department, Ganzhou People’s Hospital, Ganzhou, Jiangxi China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
50
|
Zhang B, Tian M, Zhu J, Zhu A. Global research trends in atherosclerosis-related NF-κB: a bibliometric analysis from 2000 to 2021 and suggestions for future research. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:57. [PMID: 36819535 PMCID: PMC9929801 DOI: 10.21037/atm-22-6145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
Background Atherosclerosis (AS) is closely related to stroke and cardiovascular diseases. Nuclear factor kappa-B (NF-κB) is the master regulator of inflammation, and thus, modulating the transcription of NF-κB can improve AS. Methods In this study, we conducted a bibliometric analysis to identify the frontiers, hotspots, and features of global research output on NF-κB in AS from 2000 to 2021. Papers published from 2000 to 2021 and the recorded information were retrieved from the Science Citation Index-Expanded of the Web of Science Core Collection. Bibliometric analysis and visualization were performed using VOSviewer and CiteSpace, including an analysis of the general distribution of annual output, highly productive countries, active journals, active institutions and authors, keywords, and co-cited references. Results A total of 5,439 original articles and reviews were retrieved and analyzed, and the results indicated that the annual number of publications on NF-κB in AS has been increasing in waves over the past 22 years. The majority of papers were published in China, while the USA had the highest number of citations and H-index. The most productive affiliation and journal were the University of California System and Arteriosclerosis Thrombosis and Vascular Biology, respectively. The papers of Chiu JJ. received the highest number of citations globally in 2011. The keywords, "nlrp3 inflammasome" and "microRNA", have recently attracted considerable attention, and very frequently occurring keywords included "NF kappa B", "atherosclerosis", "expression", "activation", "endogenous cell", and "oxidative stress". New keywords in 2021 included "muscle", "attenuates atherosclerosis", "mesenchymal transition", "metabolic disorder", and "palmitic acid". Conclusions AS and inflammation have become research hotspots lately. Over the past decade, most studies have focused on basic research, and pathways associated with the regulatory role of NF-κB in AS have become a particular focus in recent studies. Moreover, our study revealed that NF-κB plays a remarkable role in AS and may be a therapeutic target.
Collapse
Affiliation(s)
- Bingyue Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Mengyao Tian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Jinling Zhu
- Zhejiang Engineering Research Center for “Preventive Treatment” Smart Health of Traditional Chinese Medicine, Hangzhou, China
| | - Aisong Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China;,Zhejiang Engineering Research Center for “Preventive Treatment” Smart Health of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|