1
|
Jagodzinski N, Leichtle A, Depping R, Plötze-Martin K, Hakim SG, Bruchhage KL, Pries R. Differential influence of 1,8-Cineol on distinct hypoxia-related immune alterations in human monocytes. Sci Rep 2025; 15:12126. [PMID: 40204839 PMCID: PMC11982364 DOI: 10.1038/s41598-025-97314-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025] Open
Abstract
1,8-Cineol is a natural plant-based therapeutic agent and is commonly used to treat a broad range of acute and chronic airway inflammatory diseases. 1,8-Cineol has recently been shown to attenuate the checkpoint molecule PDL-1 in circulating monocytes in patients with chronic Otitis media (OM) and was associated with an improved clinical outcome. Hypoxia-inducible factor (HIF) is thought to play an essential role in the middle ear inflammatory process, mainly due to dysfunctions of the eustachian tube. However, the unambiguous impact of 1,8-Cineol on hypoxia-driven immune alterations of human monocytes and the related inflammatory microenvironment have not been investigated thus far. Therefore, we used the human monocytes to investigate the impact of 1,8-Cineol on the cellular hypoxia response with regards to expression levels of different adhesion molecules, chemokine receptors, and different cell stress-related proteins. Furthermore, the secretion patterns of a variety of chemokines and cytokines were evaluated. The study aimed to better understand the influence of the monoterpene 1,8-Cineol on hypoxia and normoxia-associated monocyte characteristics and related inflammatory processes, all of which are crucial for the development of various human diseases.
Collapse
Affiliation(s)
- Nele Jagodzinski
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Anke Leichtle
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Reinhard Depping
- Center for Structural and Cell Biology in Medicine, Institute of Physiology, Working Group Hypoxia, University of Lübeck, Lübeck, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Samer G Hakim
- Department of Oral and Maxillofacial Surgery and Plastic Reconstructive Head and Neck Surgery, Helios Medical Center, Schwerin, Germany
- Department of Maxillofacial Surgery, University of Lübeck, Lübeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| |
Collapse
|
2
|
Yatsushiro M, Katsuyama M, Nakamae T, Imahara K, Miyamoto M, Hayashi T. New molecular markers to differentiate carbon dioxide intoxication from asphyxia due to oxygen deficiency. Forensic Sci Med Pathol 2025:10.1007/s12024-025-00981-1. [PMID: 40167862 DOI: 10.1007/s12024-025-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE The lack of specific autopsy findings for carbon dioxide (CO2) intoxication hinders the determination of cause of death based on autopsy findings alone. In addition, when death occurs in a space is filled with CO2 or other gases, the cause of death must be distinguished between intoxication and asphyxia due to oxygen deficiency, which also has no specific autopsy findings. In this study, we aimed to identify diagnostic markers of mRNA expression in the brainstem that indicate cause of death in cases of suspected CO2 intoxication. METHODS Mouse models of CO2 intoxication (composition of ambient gases at 70% CO2, 20% O2, and 10% N2) and asphyxia due to oxygen deficiency (5% O2, 95% N2) were used to identify mRNA markers specific to intoxication or asphyxia. RESULTS Using RNA-Sequence analysis, we identified 7 candidate genes for qRT-PCR analysis: Acid-sensing ion channel 4 (Asic4), Early growth response protein 1 (Egr1), Neurogranin (Nrgn), Opioid receptor delta 1 (Oprd1), Semaphorin 3f (Sema3f), Transthyretin (Ttr), and Tryptophan hydroxylase 2 (Tph2). We observed a significant increase of Nrgn mRNA expression in the brainstem of CO2 intoxication and a significant increase of Ttr mRNA expression in the brainstem of asphyxia due to oxygen deficiency. CONCLUSION Assays for the expression of Nrgn and Ttr in the human brainstem may assist in the diagnosis/differential diagnosis of CO2 intoxication and asphyxia due to oxygen deficiency, respectively.
Collapse
Affiliation(s)
- Masahiko Yatsushiro
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Midori Katsuyama
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takuma Nakamae
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kotomi Imahara
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Machiko Miyamoto
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takahito Hayashi
- Department of Legal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| |
Collapse
|
3
|
L'homme L, Sermikli BP, Haas JT, Fleury S, Quemener S, Guinot V, Barreby E, Esser N, Caiazzo R, Verkindt H, Legendre B, Raverdy V, Cheval L, Paquot N, Piette J, Legrand-Poels S, Aouadi M, Pattou F, Staels B, Dombrowicz D. Adipose tissue macrophage infiltration and hepatocyte stress increase GDF-15 throughout development of obesity to MASH. Nat Commun 2024; 15:7173. [PMID: 39169003 PMCID: PMC11339436 DOI: 10.1038/s41467-024-51078-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.
Collapse
Affiliation(s)
- Laurent L'homme
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Benan Pelin Sermikli
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sébastien Fleury
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sandrine Quemener
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Valentine Guinot
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Emelie Barreby
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nathalie Esser
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Robert Caiazzo
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Hélène Verkindt
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Benjamin Legendre
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Violeta Raverdy
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, Paris, France
| | - Nicolas Paquot
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Signal Transduction, University of Liège, Liège, Belgium
| | - Sylvie Legrand-Poels
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | - Myriam Aouadi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - François Pattou
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Bart Staels
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
4
|
Wang J, Ye X, Wang Y. Anshen Shumai Decoction inhibits post-infarction inflammation and myocardial remodeling through suppression of the p38 MAPK/c-FOS/EGR1 pathway. J Mol Histol 2024; 55:437-454. [PMID: 38874870 DOI: 10.1007/s10735-024-10214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Anshen Shumai Decoction (ASSMD) is traditionally employed to manage coronary artery disease arrhythmias. Its protective efficacy against myocardial infarction remains to be elucidated. This investigation employed a rat model of myocardial infarction, achieved through the ligation of the left anterior descending (LAD) coronary artery, followed by a 28-day administration of ASSMD. The study observed the decoction's mitigative impact on myocardial injury, with gene regulation effects discerned through transcriptomic analysis. Furthermore, ASSMD's influence on cardiomyocyte apoptosis and fibrotic protein secretion was assessed using an embryonic rat cardiomyocyte cell line (H9c2) under hypoxic conditions and rat cardiac fibroblasts subjected to normoxic culture conditions with TGF-β. A functional rescue assay involving overexpression of FOS and Early Growth Response Factor 1 (EGR1), combined with inhibition of the p38 Mitogen-activated Protein Kinase (MAPK) pathway, was conducted. Results indicated that ASSMD significantly curtailed cardiomyocyte apoptosis and myocardial fibrosis in infarcted rats, primarily by downregulating FOS and EGR1 gene expression and inhibiting the upstream p38 MAPK pathway. These actions of ASSMD culminated in reduced expression of pro-apoptotic, collagen, and fibrosis-associated proteins, conferring myocardial protection and anti-fibrotic effects on cardiac fibroblasts.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Cardiology, Chun'an County Traditional Chinese Medicine Hospital, No. 1 Xin'an West Road, Qiandaohu Town, Chun'an County, Hangzhou, 311700, P. R. China
| | - Xiaolei Ye
- School of Medicine, Ningbo University, Ningbo, 315211, P. R. China
| | - Yanqin Wang
- Department of Cardiology, Chun'an County Traditional Chinese Medicine Hospital, No. 1 Xin'an West Road, Qiandaohu Town, Chun'an County, Hangzhou, 311700, P. R. China.
| |
Collapse
|
5
|
Imamura I, Kiyama R. Potential involvement of KANK1 haploinsufficiency in centrosome aberrations. Biochim Biophys Acta Gen Subj 2024; 1868:130648. [PMID: 38830559 DOI: 10.1016/j.bbagen.2024.130648] [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: 12/21/2023] [Revised: 05/04/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
KANK1 was found as a tumor suppressor gene based on frequent deletions in renal cell carcinoma and the inhibitory activity of tumor cell proliferation. Previously, we reported that knockdown of KANK1 induced centrosomal amplification, leading to abnormal cell division, through the hyperactivation of RhoA small GTPase. Here, we investigated the loss of KANK1 function by performing CRISPR/Cas9-based genome editing to knockout the gene. After several rounds of genome editing, however, there were no cell lines with complete loss of KANK1, and the less the wild-type KANK1 dosage, the greater the number of cells with abnormal numbers of centrosomes and rates of cell-doubling and apoptosis, suggesting the involvement of KANK1 haploinsufficiency in centrosome aberrations. The rescue of KANK1-knockdown cells with a KANK1-expressing plasmid restored the rates of cells exhibiting centrosomal amplification to the control level. RNA-sequencing analysis of the cells with reduced dosages of functional KANK1 revealed potential involvement of other cell proliferation-related genes, such as EGR1, MDGA2, and BMP3, which have been reported to show haploinsufficiency when they function. When EGR1 protein expression was reduced by siRNA technology, the number of cells exhibiting centrosomal amplification increased, along with the reduction of KANK1 protein expression, suggesting their functional relationship. Thus, KANK1 haploinsufficiency may contribute to centrosome aberrations through the network of haploinsufficiency-related genes.
Collapse
Affiliation(s)
- Ikumi Imamura
- Faculty of Life Science, Kyushu Sangyo University, Japan
| | - Ryoiti Kiyama
- Faculty of Life Science, Kyushu Sangyo University, Japan.
| |
Collapse
|
6
|
Yan G, Wei T, Lan Y, Xu T, Qian P. Different parts of the mussel Gigantidas haimaensis holobiont responded differently to deep-sea sampling stress. Integr Zool 2024. [PMID: 39072987 DOI: 10.1111/1749-4877.12881] [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] [Indexed: 07/30/2024]
Abstract
Acute environmental changes cause stress during conventional deep-sea biological sampling without in situ fixation and affect gene expressions of samples collected. However, the degree of influence and underlying mechanisms are hardly investigated. Here, we conducted comparative transcriptomic analyses between in situ and onboard fixed gills and between in situ and onboard fixed mantles of deep-sea mussel Gigantidas haimaensis to assess the effects of incidental sampling stress. Results showed that transcription, translation, and energy metabolism were upregulated in onboard fixed gills and mantles, thereby mobilizing rapid gene expression to tackle the stress. Autophagy and phagocytosis that related to symbiotic interactions between the host and endosymbiont were downregulated in the onboard fixed gills. These findings demonstrated that symbiotic gill and nonsymbiotic mantle responded differently to sampling stress, and symbiosis in the gill was perturbed. Further comparative metatranscriptomic analysis between in situ and onboard fixed gills revealed that stress response genes, peptidoglycan biosynthesis, and methane fixation were upregulated in the onboard fixed endosymbiotic Gammaproteobacteria inside the gills, implying that energy metabolism of the endosymbiont was increased to cope with sampling stress. Furthermore, comparative analysis between the mussel G. haimaensis and the limpet Bathyacmaea lactea transcriptomes resultedidentified six transcription factor orthologs upregulated in both onboard fixed mussel mantles and limpets, including sharply increased early growth response protein 1 and Kruppel-like factor 5. They potentially play key roles in initiating the response of sampled deep-sea macrobenthos to sampling stress. Our results clearly show that in situ fixed biological samples are vital for studying deep-sea environmental adaptation.
Collapse
Affiliation(s)
- Guoyong Yan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Tong Wei
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yi Lan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ting Xu
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Peiyuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
7
|
Li X, Wu W, He H, Guan L, Chen G, Lin Z, Li H, Jiang J, Dong X, Guan Z, Chen P, Pan Z, Huang W, Yu R, Song W, Lu L, Yang Z, Chen Z, Wang L, Xian S, Chen J. Analysis and validation of hub genes in neutrophil extracellular traps for the long-term prognosis of myocardial infarction. Gene 2024; 914:148369. [PMID: 38485036 DOI: 10.1016/j.gene.2024.148369] [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: 01/12/2024] [Revised: 02/27/2024] [Accepted: 03/11/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION The study focuses on the long-term prognosis of myocardial infarction (MI) influenced by neutrophil extracellular traps (NETs). It also aims to analyze and validate relative hub genes in this process, in order to further explore new therapeutic targets that can improve the prognosis of MI. MATERIALS AND METHODS We established a MI model in mice by ligating the left anterior descending branch (LAD) and conducted an 8-week continuous observation to study the dynamic changes in the structure and function of the heart in these mice. Meanwhile, we administered Apocynin, an inhibitor of NADPH Oxidase, which has also been shown to inhibit the formation of NETs, to mice undergoing MI surgery in order to compare. This study employed hematoxylin-eosin (HE) staining, echocardiography, immunofluorescence, and real-time quantitative PCR (RT-qPCR) to examine the impact of NETs on the long-term prognosis of MI. Next, datasets related to MI and NETs were downloaded from the GEO database, respectively. The Limma package of R software was used to identify differentially expressed genes (DEGs). After analyzing the "Robust Rank Aggregation (RRA)" package, we conducted a screening for robust differentially expressed genes (DEGs) and performed pathway enrichment analysis using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to determine the functional roles of these robust DEGs. The protein-protein interaction (PPI) network was visualized and hub genes were filtered using Cytoscape. RESULTS Immunofluorescence and qPCR results showed an increase in the expression of Myeloperoxidase (MPO) at week 1 and week 8 in the hearts of mice after MI. HE staining reveals a series of pathological manifestations in the heart of the MI group during 8 weeks, including enlarged size, disordered arrangement of cardiomyocytes, infiltration of inflammatory cells, and excessive deposition of collagen fibers, among others. The utilization of Apocynin could significantly improve these poor performances. The echocardiography displayed the cardiac function of the heart in mice. The MI group has a reduced range of heart movement and decreased ejection ability. Moreover, the ventricular systolic movement was found to be abnormal, and its wall thickening rate decreased over time, indicating a progressive worsening of myocardial ischemia. The Apocynin group, on the contrary, showed fewer abnormal changes in the aforementioned aspects. A total of 81 DEGs and 4 hub genes (FOS, EGR1, PTGS2, and HIST1H4H) were obtained. The results of RT-qPCR demonstrated abnormal expression of these four genes in the MI group, which could be reversed by treatment of Apocynin. CONCLUSION The NETs formation could be highly related to MI and the long-term prognosis of MI can be significantly influenced by the NETs formation. Four hub genes, namely FOS, EGR1, PTGS2, and HIST1H4H, have the potential to be key genes related to this process. They could also serve as biomarkers for predicting MI prognosis and as targets for gene therapy.
Collapse
Affiliation(s)
- Xuan Li
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China.
| | - Wenyu Wu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Huan He
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Lin Guan
- Shandong Province Hospital of Traditional Chinese Medicine, Jinan 250011, China
| | - Guancheng Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Zhijun Lin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Huan Li
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Jialin Jiang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Xin Dong
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Zhuoji Guan
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Pinliang Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Zigang Pan
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Weiwei Huang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Runjia Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Wenxin Song
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Lu Lu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Zhongqi Yang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China
| | - Zixin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China.
| | - Lingjun Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China.
| | - Shaoxiang Xian
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China.
| | - Jie Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; National Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou 510405, China.
| |
Collapse
|
8
|
Yang Q, Murata K, Ikeda T, Minatoya K, Masumoto H. miR-124-3p downregulates EGR1 to suppress ischemia-hypoxia reperfusion injury in human iPS cell-derived cardiomyocytes. Sci Rep 2024; 14:14811. [PMID: 38926457 PMCID: PMC11208498 DOI: 10.1038/s41598-024-65373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Ischemic heart diseases are a major global cause of death, and despite timely revascularization, heart failure due to ischemia-hypoxia reperfusion (IH/R) injury remains a concern. The study focused on the role of Early Growth Response 1 (EGR1) in IH/R-induced apoptosis in human cardiomyocytes (CMs). Human induced pluripotent stem cell (hiPSC)-derived CMs were cultured under IH/R conditions, revealing higher EGR1 expression in the IH/R group through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB). Immunofluorescence analysis (IFA) showed an increased ratio of cleaved Caspase-3-positive apoptotic cells in the IH/R group. Using siRNA for EGR1 successfully downregulated EGR1, suppressing cleaved Caspase-3-positive apoptotic cell ratio. Bioinformatic analysis indicated that EGR1 is a plausible target of miR-124-3p under IH/R conditions. The miR-124-3p mimic, predicted to antagonize EGR1 mRNA, downregulated EGR1 under IH/R conditions in qRT-PCR and WB, as confirmed by IFA. The suppression of EGR1 by the miR-124-3p mimic subsequently reduced CM apoptosis. The study suggests that treatment with miR-124-3p targeting EGR1 could be a potential novel therapeutic approach for cardioprotection in ischemic heart diseases in the future.
Collapse
Affiliation(s)
- Qiaoke Yang
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kozue Murata
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Tadashi Ikeda
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
9
|
Tian Q, Chen JH, Ding Y, Wang XY, Qiu JY, Cao Q, Zhuang LL, Jin R, Zhou GP. EGR1 transcriptionally regulates SVEP1 to promote proliferation and migration in human coronary artery smooth muscle cells. Mol Biol Rep 2024; 51:365. [PMID: 38409611 DOI: 10.1007/s11033-024-09322-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
A low-frequency variant of sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SVEP1) is associated with the risk of coronary artery disease, as determined by a genome-wide association study. SVEP1 induces vascular smooth muscle cell proliferation and an inflammatory phenotype to promote atherosclerosis. In the present study, qRT‒PCR demonstrated that the mRNA expression of SVEP1 was significantly increased in atherosclerotic plaques compared to normal tissues. Bioinformatics revealed that EGR1 was a transcription factor for SVEP1. The results of the luciferase reporter assay, siRNA interference or overexpression assay, mutational analysis and ChIP confirmed that EGR1 positively regulated the transcriptional activity of SVEP1 by directly binding to its promoter. EGR1 promoted human coronary artery smooth muscle cell (HCASMC) proliferation and migration via SVEP1 in response to oxidized low-density lipoprotein (ox-LDL) treatment. Moreover, the expression level of EGR1 was increased in atherosclerotic plaques and showed a strong linear correlation with the expression of SVEP1. Our findings indicated that EGR1 binding to the promoter region drive SVEP1 transcription to promote HCASMC proliferation and migration.
Collapse
Affiliation(s)
- Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-He Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Ding
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Yu Wang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Yun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Cao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Awaad A, Olama ZA, El-Subruiti GM, Ali SM. The dual activity of CaONPs as a cancer treatment substance and at the same time resistance to harmful microbes. Sci Rep 2023; 13:22940. [PMID: 38135693 PMCID: PMC10746744 DOI: 10.1038/s41598-023-49637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Nanotechnology holds significant promise for the development of novel and necessary products that enhance human health. Pharmacology and nanotechnology have contributed to developing advanced and highly effective drugs for cancer treatment and combating microbial infections. The microbiological effectiveness against the variety of examined microorganisms was assessed using the time killer curve, scanning electron microscopy (SEM), MIC techniques, and the agar well diffusion method. SEM was utilized to enhance the analysis of the mechanisms underlying the bio-interface interaction and intracellular localization of calcium oxide nanoparticles (CaONPs). The MTT test was used to examine the cytotoxicity of CaONP anticancer activity in various cancer cells, including colon, breast, and hepatic cells. The efficacy of CaONPs as an anticancer medication was elucidated by analyzing the gene expression of both treated and untreated cancer cells. MIC and MBC of CaONPs against Escherichia coli and Staphylococcus epidermidis were 150, 150, 150, and 200 µg/ml, respectively. The MIC and MFC of CaONPs against Candida albicans were 200 µg/ml and 250 µg/ml, respectively. The IC50 values of various CaONPs vary depending on the type of cancer cells. The gene expression analysis of breast cancer cells undergoing treatment revealed the identification of several cancer-controlling genes, namely BAX, BCL2, P53, TERT, KRAS1, KRAS2, and RB1. The study demonstrated the notable antibacterial efficacy of CaONPs, highlighting their potential as cancer therapies.
Collapse
Affiliation(s)
- Amr Awaad
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Zakia A Olama
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Gehan M El-Subruiti
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Safaa M Ali
- Nucleic Acid Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg El-Arab City, 21934, Alexandria, Egypt.
| |
Collapse
|
11
|
Saint Paraskeva Medical Center, Lviv, Ukraine, Chooklin S, Chuklin S, Saint Paraskeva Medical Center, Lviv, Ukraine. PATHOPHYSIOLOGICAL MECHANISMS OF DEEP VEIN THROMBOSIS. FIZIOLOHICHNYĬ ZHURNAL 2023; 69:133-144. [DOI: 10.15407/fz69.06.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Deep venous thrombosis is a frequent multifactorial disease and most of the time is triggered by the interaction between acquired risk factors, particularly immobility, and hereditary risk factors such as thrombophilias. The mechanisms underlying deep venous thrombosis are not fully elucidated; however, in recent years the role of venous flow, endothelium, platelets, leukocytes, and the interaction between inflammation and hemostasis has been determined. Alteration of venous blood flow produces endothelial activation, favoring the adhesion of platelets and leukocytes, which, through tissue factor expression and neutrophil extracellular traps formation, contribute to the activation of coagulation, trapping more cells, such as red blood cells, monocytes, eosinophils, lymphocytes. The coagulation factor XI-driven propagation phase of blood coagulation plays a major role in venous thrombus growth, but a minor role in hemostasis. In this work, the main mechanisms involved in the pathophysiology of deep vein thrombosis are described.
Collapse
|
12
|
Pries R, Jeschke S, Leichtle A, Bruchhage KL. Modes of Action of 1,8-Cineol in Infections and Inflammation. Metabolites 2023; 13:751. [PMID: 37367909 DOI: 10.3390/metabo13060751] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
The monoterpene 1,8-Cineol is a natural plant-based therapeutic agent that is commonly applied to treat different inflammatory diseases due to its mucolytic, anti-microbial and anti-inflammatory properties. It has become increasingly clear in the recent years that 1,8-Cineol spreads almost everywhere in the human body after its oral administration, from the gut to the blood to the brain. Its anti-microbial potential and even its anti-viral effects have been observed to include numerous bacteria and fungi species. Many recent studies help to better understand the cellular and molecular immunological consequences of 1,8-Cineol treatment in inflammatory diseases and further provide information concerning the mechanistic modes of action in the regulation of distinct inflammatory biosynthetic pathways. This review aims to present a holistic and understandable overview of the different aspects of 1,8-Cineol in infections and inflammation.
Collapse
Affiliation(s)
- Ralph Pries
- Department of Otorhinolaryngology, University of Luebeck, 23538 Luebeck, Germany
| | - Stephanie Jeschke
- Department of Otorhinolaryngology, University of Luebeck, 23538 Luebeck, Germany
| | - Anke Leichtle
- Department of Otorhinolaryngology, University of Luebeck, 23538 Luebeck, Germany
| | | |
Collapse
|