1
|
Hinostroza F, Araya-Duran I, Piñeiro A, Lobos I, Pastenes L. Transcription factor roles in the local adaptation to temperature in the Andean Spiny Toad Rhinella spinulosa. Sci Rep 2024; 14:15158. [PMID: 38956427 PMCID: PMC11220030 DOI: 10.1038/s41598-024-66127-5] [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: 05/31/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Environmental temperature strongly influences the adaptation dynamics of amphibians, whose limited regulation capabilities render them susceptible to thermal oscillations. A central element of the adaptive strategies is the transcription factors (TFs), which act as master regulators that orchestrate stress responses, enabling species to navigate the fluctuations of their environment skillfully. Our study delves into the intricate relationship between TF expression and thermal adaptation mechanisms in the Rhinella spinulosa populations. We sought to elucidate the dynamic modulations of TF expression in prometamorphic and metamorphic tadpoles that inhabit two thermally contrasting environments (Catarpe and El Tatio Geyser, Chile) and which were exposed to two thermal treatments (25 °C vs. 20 °C). Our findings unravel an intriguing dichotomy in response strategies between these populations. First, results evidence the expression of 1374 transcription factors. Regarding the temperature shift, the Catarpe tadpoles show a multifaceted approach by up-regulating crucial TFs, including fosB, atf7, and the androgen receptor. These dynamic regulatory responses likely underpin the population's ability to navigate thermal fluctuations effectively. In stark contrast, the El Tatio tadpoles exhibit a more targeted response, primarily up-regulating foxc1. This differential expression suggests a distinct focus on specific TFs to mitigate the effects of temperature variations. Our study contributes to understanding the molecular mechanisms governing thermal adaptation responses and highlights the resilience and adaptability of amphibians in the face of ever-changing environmental conditions.
Collapse
Affiliation(s)
- Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
- Escuela de Química y Farmacia, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
- Centro Para la Investigación Traslacional en Neurofarmacología, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Araya-Duran
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alejandro Piñeiro
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Isabel Lobos
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile
| | - Luis Pastenes
- Laboratorio de Genética y Microevolución, Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca, Chile.
| |
Collapse
|
2
|
Zhu Y, Chen B, Zu Y. Identifying OGN as a Biomarker Covering Multiple Pathogenic Pathways for Diagnosing Heart Failure: From Machine Learning to Mechanism Interpretation. Biomolecules 2024; 14:179. [PMID: 38397416 PMCID: PMC10886937 DOI: 10.3390/biom14020179] [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: 11/21/2023] [Revised: 01/14/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The pathophysiologic heterogeneity of heart failure (HF) necessitates a more detailed identification of diagnostic biomarkers that can reflect its diverse pathogenic pathways. METHODS We conducted weighted gene and multiscale embedded gene co-expression network analysis on differentially expressed genes obtained from HF and non-HF specimens. We employed a machine learning integration framework and protein-protein interaction network to identify diagnostic biomarkers. Additionally, we integrated gene set variation analysis, gene set enrichment analysis (GSEA), and transcription factor (TF)-target analysis to unravel the biomarker-dominant pathways. Leveraging single-sample GSEA and molecular docking, we predicted immune cells and therapeutic drugs related to biomarkers. Quantitative polymerase chain reaction validated the expressions of biomarkers in the plasma of HF patients. A two-sample Mendelian randomization analysis was implemented to investigate the causal impact of biomarkers on HF. RESULTS We first identified COL14A1, OGN, MFAP4, and SFRP4 as candidate biomarkers with robust diagnostic performance. We revealed that regulating biomarkers in HF pathogenesis involves TFs (BNC2, MEOX2) and pathways (cell adhesion molecules, chemokine signaling pathway, cytokine-cytokine receptor interaction, oxidative phosphorylation). Moreover, we observed the elevated infiltration of effector memory CD4+ T cells in HF, which was highly related to biomarkers and could impact immune pathways. Captopril, aldosterone antagonist, cyclopenthiazide, estradiol, tolazoline, and genistein were predicted as therapeutic drugs alleviating HF via interactions with biomarkers. In vitro study confirmed the up-regulation of OGN as a plasma biomarker of HF. Mendelian randomization analysis suggested that genetic predisposition toward higher plasma OGN promoted the risk of HF. CONCLUSIONS We propose OGN as a diagnostic biomarker for HF, which may advance our understanding of the diagnosis and pathogenesis of HF.
Collapse
Affiliation(s)
- Yihao Zhu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Bin Chen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Lin-gang), Shanghai 201306, China
| | - Yao Zu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
| |
Collapse
|
3
|
Duan L, Tang H, Lan Y, Shi H, Pu P, He Q. Ring finger protein 10 improves pirarubicin-induced cardiac inflammation by regulating the AP-1/Meox2 signaling pathway. Toxicol Appl Pharmacol 2023; 462:116411. [PMID: 36740146 DOI: 10.1016/j.taap.2023.116411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Pirarubicin (THP) is widely used in clinical antitumor therapy, but its cardiotoxicity seriously affects the therapeutic effect in patients. In the study, we investigated the role of ring finger protein 10 (RNF10) in cardiotoxicity induced by THP. MATERIALS AND METHODS A cardiac toxicity model in Sprague-Dawley (SD) rats induced by THP was established. Changes in diet, weight, electrocardiogram (ECG), and echocardiography were observed. Serum levels of brain natriuretic peptide (BNP), creatine kinase MB (CK-MB), cardiac troponin T (cTnT), and lactate dehydrogenase (LDH) were measured. The expression of RNF10 in myocardium was observed by immunohistochemistry. The expressions of RNF10, activator protein-1 (AP-1), mesenchyme homeobox 2 (Meox2), total nuclear factor (NF)-κB p65 (T-P65), phosphorylated NF-κB p65 (PP65), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, interleukin (IL)-6, and mature IL-1β were detected by Western blot. A THP-induced H9c2 myocardial cell injury model was established. RNF10 was downregulated or overexpressed by RNF10 siRNA and a RNF10 lentiviral vector, respectively. Then, cell viability was measured. The expression of RNF10 in H9c2 cells was observed by immunofluorescence. All of the above signaling pathways were verified by Western blots. FINDINGS THP caused a series of cardiotoxic manifestations in SD rats. Our studies suggested that THP caused cardiac inflammation by inhibiting the expression of RNF10, while overexpression of RNF10 antagonized the cardiotoxicity induced by THP. SIGNIFICANCE Our study showed RNF10 improved THP-induced cardiac inflammation by regulating the AP-1/Meox2 signaling pathway. RNF10 may be a new target to treat THP-induced cardiotoxicity.
Collapse
Affiliation(s)
- Liang Duan
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Lan
- Department of Critical Care Medicine, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Hongwei Shi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quan He
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
RING Finger Protein 10 Regulates AP-1/Meox2 to Mediate Pirarubicin-Induced Cardiomyocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7872193. [PMID: 36713029 PMCID: PMC9883094 DOI: 10.1155/2023/7872193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 11/25/2022] [Indexed: 01/21/2023]
Abstract
Pirarubicin (THP) is one of the classic chemotherapy drugs for cancer treatment. It is often clinically limited because of its cardiotoxicity. The occurrence and development of THP-mediated chemotherapy-related cardiotoxicity (CRC) may be reversed by RING finger protein 10 (RNF10). This study was performed with the aim of evaluating the inhibitory effect of RNF10 on THP-mediated CRC and its molecular mechanism. In vivo, we found that the expression of RNF10 decreased in THP-induced CRC rats, accompanied by Meox2 inhibition and AP-1 activation, resulting in increased cardiomyocyte apoptosis. After small interfering RNA (siRNA) and lentivirus transfection (Lv) of RNF10 in vitro, the expression of RNF10, Meox2, and AP-1 proteins and the degree of cardiomyocyte apoptosis were detected. We found that overexpression of RNF10 in H9C2 cardiomyocytes significantly promoted Meox2 and inhibited AP-1, alleviated apoptosis, and showed further inhibitory activity on THP-induced cardiomyocyte toxicity. Silencing RNF10 showed the opposite result. Our study showed that RNF10 inhibited THP-induced CRC through the activity of Meox2 and AP-1 proteins. RNF10 may be the next drug target for the treatment of CRC and other related cardiovascular diseases.
Collapse
|
5
|
Vozzi F, Cecchettini A, Cabiati M, Mg F, Aretini P, Del Ry S, Rocchiccioli S, Pelosi G. Modulated molecular markers of restenosis and thrombosis by in-vitrovascular cells exposed to bioresorbable scaffolds. Biomed Mater 2021; 16. [PMID: 34020430 DOI: 10.1088/1748-605x/ac0401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/21/2021] [Indexed: 01/06/2023]
Abstract
Drug-eluting bioresorbable vascular scaffolds (BVSs) have emerged as a potential breakthrough for the treatment of coronary artery stenosis, providing mechanical support and drug delivery followed by complete resorption. Restenosis and thrombosis remain the primary limitations in clinical use. The study aimed to identify potential markers of restenosis and thrombosis analyzing the vascular wall cell transcriptomic profile modulation triggered by BVS at different values of shear stress (SS). Human coronary artery endothelial cells and smooth muscle cells were cultured under SS (1 and 20 dyne cm-2) for 6 h without and with application of BVS and everolimus 600 nM. Cell RNA-Seq and bioinformatics analysis identified modulated genes by direct comparison of SS conditions and Gene Ontology (GO). The results of different experimental conditions and GO analysis highlighted the modulation of specific genes as semaphorin 3E, mesenchyme homeobox 2, bone morphogenetic protein 4, (heme oxygenase 1) and selectin E, with different roles in pathological evolution of disease. Transcriptomic analysis of dynamic vascular cell cultures identifies candidate genes related to pro-restenotic and pro-thrombotic mechanisms in anin-vitrosetting of BVS, which are not adequately contrasted by everolimus addition.
Collapse
Affiliation(s)
- F Vozzi
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| | - A Cecchettini
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy.,Department of Clinical and Experimental Medicine, University of Pisa, Via Volta 4, Pisa, Italy
| | - M Cabiati
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| | - Fornaro Mg
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| | - P Aretini
- Fondazione Pisana per la Scienza ONLUS, Via Ferruccio Giovannini, 13, San Giuliano Terme, Italy
| | - S Del Ry
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| | - S Rocchiccioli
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| | - G Pelosi
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, Pisa, Italy
| |
Collapse
|
6
|
Cao X, Liu L, Zhang Y, Yang Y. Reduced RING finger protein 10 expression in macrophages is associated with aging-related inflammation. FEBS Open Bio 2021; 11:386-394. [PMID: 33249776 PMCID: PMC7876489 DOI: 10.1002/2211-5463.13049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/13/2020] [Accepted: 10/28/2020] [Indexed: 01/06/2023] Open
Abstract
Age‐associated decline of the immune system is referred to as immunosenescence. The E3 ligase RING finger 10 (RNF10) has long been associated with the innate immune response, although a potential role in immunosenescence has not previously been reported. In the present study, we identified that RNF10 expression is lower in aged mouse macrophages than in young cells. After lipopolysaccharide stimulation, RNF10 expression remained at a basal low level in aged mouse cells, but declined sharply in young mouse cells. Knockdown of RNF10 enhanced both the nuclear factor‐κB and interferon regulatory factor 3 signaling pathways and thus enhanced proinflammatory cytokines and type I interferons in macrophages, promoting clearance of Listeria monocytogenes. These findings indicate that dysregulated expression of RNF10 is associated with age‐associated immune dysfunction, and RNF10 may thus be a potential target for the treatment of age‐related inflammatory diseases.
Collapse
Affiliation(s)
- Xinyuan Cao
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Lidan Liu
- The Southern Medical District of Chinese PLA General Hospital, Beijing, China
| | - Yueyi Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yingyun Yang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|
7
|
Li S, Yu G, Huang W, Wang R, Pu P, Chen M. RING finger protein 10 is a potential drug target for diabetic vascular complications. Mol Med Rep 2019; 20:931-938. [PMID: 31173254 PMCID: PMC6625204 DOI: 10.3892/mmr.2019.10358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/28/2019] [Indexed: 12/23/2022] Open
Abstract
Vascular remodeling induced by long-term hyperglycaemia is the main pathological process in diabetic vascular complications. Thus, vascular remodeling may be a potential therapeutic target in diabetes mellitus (DM) with macrovascular disease. The present study aimed to investigate the effect of RING finger protein 10 (RNF10) on vascular remodeling under conditions of chronic hyperglycaemia stimulation. We found that overexpression of RNF10 clearly decreased intimal thickness and attenuated vascular remodeling in DM. TUNEL staining showed that apoptosis was clearly inhibited, an effect that may be mediated by decreases in Bcl-2 protein expression. Quantitative analysis demonstrated that overexpression of RNF10 could suppress inflammation by reducing the levels of TNF-α, and MCP-1 mRNA and NF-κB protein. Meanwhile, overexpression of RNF10 prevented vascular smooth muscle cell (VSMC) hyperproliferation through the downregulation of cyclin D1 and CDK4 proteins. Notably, short hairpin RNF10 (shRNF10) greatly aggravated the pathological responses of diabetic vascular remodeling. These outcomes revealed that the differential expression of RNF10 had a completely opposite effect on vascular damage under hyperglycaemia, further displaying the core function of RNF10 in regulating vascular remodeling induced by diabetes. Consequently, RNF10 could be a novel target for the treatment of diabetic vascular complications.
Collapse
Affiliation(s)
- Siyu Li
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Guiquan Yu
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wei Huang
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ruiyu Wang
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Peng Pu
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ming Chen
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
8
|
Carrano N, Samaddar T, Brunialti E, Franchini L, Marcello E, Ciana P, Mauceri D, Di Luca M, Gardoni F. The Synaptonuclear Messenger RNF10 Acts as an Architect of Neuronal Morphology. Mol Neurobiol 2019; 56:7583-7593. [DOI: 10.1007/s12035-019-1631-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
|
9
|
Li S, Yu G, Jing F, Chen H, Liu A, Luo M, Huang W, Pu P, Chen M. RING finger protein 10 attenuates vascular restenosis by inhibiting vascular smooth muscle cell hyperproliferation in vivo and vitro. IUBMB Life 2018; 71:632-642. [PMID: 30597731 DOI: 10.1002/iub.1995] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/28/2018] [Accepted: 12/02/2018] [Indexed: 01/24/2023]
Affiliation(s)
- Siyu Li
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Guiquan Yu
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Fuyu Jing
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Hui Chen
- Department of NeurosurgeryChildren's Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Aoyi Liu
- Department of CardiologyChildren's Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Minghao Luo
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Wei Huang
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Peng Pu
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Ming Chen
- Department of CardiologyFirst Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| |
Collapse
|
10
|
Yu G, Chen J, Li S, Pu P, Huang W, Zhao Y, Peng X, Wang R, Lei H. RING finger protein 10 prevents neointimal hyperplasia by promoting apoptosis in vitro and in vivo. Life Sci 2018; 208:325-332. [DOI: 10.1016/j.lfs.2018.04.055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 04/19/2018] [Accepted: 04/28/2018] [Indexed: 12/23/2022]
|
11
|
Hou L, Liu Y, Gao Q, Xu X, Ning M, Bi J, Liu H, Liu M, Gu W, Wang W, Meng Q. Spiroplasma eriocheiris Adhesin-Like Protein (ALP) Interacts with Epidermal Growth Factor (EGF) Domain Proteins to Facilitate Infection. Front Cell Infect Microbiol 2017; 7:13. [PMID: 28184355 PMCID: PMC5266718 DOI: 10.3389/fcimb.2017.00013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022] Open
Abstract
Spiroplasma eriocheiris is a novel pathogen found in recent years, causing the tremor disease (TD) of Chinese mitten crab Eriocheir sinensis. Like Spiroplasma mirum, S. eriocheiris infects the newborn mouse (adult mice are not infected) and can cause cataract. Adhesion-related protein is an important protein involved in the interaction between pathogen and host. In this study, the Adhesin-like Protein (ALP) of S. eriocheiris was detected on its outer membrane by using immune electron microscopy, and was found to be involved in the bacterium's infection of mouse embryo fibroblasts (3T6-Swiss albino). Yeast two-hybrid analysis demonstrated that ALP interacts with a diverse group of mouse proteins. The interactions between recombinant partial fibulin7 (FBLN7; including two epidermal growth factor [EGF] domains) and ALP were confirmed by Far-western blotting and colocalization. We synthetized the domains of FBLN7 [EGF domain: amino acids 136–172 and complement control protein (CCP) domain: 81–134 amino acids], and demonstrated that only EGF domain of FBLN7 can interact with ALP. Because the EGF domain has high degree of similarity to EGF, it can activate the downstream EGFR signaling pathway, in key site amino acids. The EGFR pathway in 3T6 cells was restrained after rALP stimulation resulting from competitive binding of ALP to EGF. The unborn mouse, newborn mouse, and the adult mouse with cataract have a small amount of expressed FBLN7; however, none was detected in the brain and very little expression was seen in the eye of normal adult mice. In short, ALP as a S. eriocheiris surface protein, is critical for infection and further supports the role of ALP in S. eriocheiris infection by competitive effection of the EGF/EGFR axis of the target cells.
Collapse
Affiliation(s)
- Libo Hou
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Yuhan Liu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Qi Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Xuechuan Xu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Mingxiao Ning
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Jingxiu Bi
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Hui Liu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Min Liu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Wei Gu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal UniversityNanjing, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu ProvinceLianYungang, China
| | - Wen Wang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal University Nanjing, China
| | - Qingguo Meng
- Jiangsu Key Laboratory for Biodiversity and Biotechnology and Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Life Sciences, Nanjing Normal UniversityNanjing, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu ProvinceLianYungang, China
| |
Collapse
|
12
|
Dinamarca MC, Guzzetti F, Karpova A, Lim D, Mitro N, Musardo S, Mellone M, Marcello E, Stanic J, Samaddar T, Burguière A, Caldarelli A, Genazzani AA, Perroy J, Fagni L, Canonico PL, Kreutz MR, Gardoni F, Di Luca M. Ring finger protein 10 is a novel synaptonuclear messenger encoding activation of NMDA receptors in hippocampus. eLife 2016; 5:e12430. [PMID: 26977767 PMCID: PMC4805553 DOI: 10.7554/elife.12430] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/19/2016] [Indexed: 12/11/2022] Open
Abstract
Synapses and nuclei are connected by bidirectional communication mechanisms that enable information transfer encoded by macromolecules. Here, we identified RNF10 as a novel synaptonuclear protein messenger. RNF10 is activated by calcium signals at the postsynaptic compartment and elicits discrete changes at the transcriptional level. RNF10 is enriched at the excitatory synapse where it associates with the GluN2A subunit of NMDA receptors (NMDARs). Activation of synaptic GluN2A-containing NMDARs and induction of long term potentiation (LTP) lead to the translocation of RNF10 from dendritic segments and dendritic spines to the nucleus. In particular, we provide evidence for importin-dependent long-distance transport from synapto-dendritic compartments to the nucleus. Notably, RNF10 silencing prevents the maintenance of LTP as well as LTP-dependent structural modifications of dendritic spines. DOI:http://dx.doi.org/10.7554/eLife.12430.001 Brain activity depends on the communication between neurons. This process takes place at the junctions between neurons, which are known as synapses, and typically involves one of the cells releasing a chemical messenger that binds to receptors on the other cell. The binding triggers a cascade of events inside the recipient cell, including the production of new receptors and their insertion into the cell membrane. These changes strengthen the synapse and are thought to be one of the ways in which the brain establishes and maintains memories. However, in order to induce these changes at the synapse, neurons must be able to activate the genes that encode their component parts. These genes are present inside the cell nucleus, which is located some distance away from the synapse. Studies have shown that signals can be sent from the nucleus to the synapse and vice versa, enabling the two parts of the cell to exchange information. Synapses that communicate using a chemical called glutamate have been particularly well studied; but it still remains unclear how the activation of receptors at these “glutamatergic synapses” is linked to activation of genes inside the nucleus at the molecular level. Dinamarca, Guzzetti et al. have now discovered that this process at glutamatergic synapses involves the movement of a protein messenger to the nucleus. Specifically, activation at synapses of a particularly common subtype of receptor, called NMDA, causes a protein called Ring Finger protein 10 (or RNF10 for short) to move from the synapse to the nucleus. To leave the synapse, RNF10 first has to bind to proteins called importins, which transport RNF10 into the nucleus. Once inside the nucleus, RNF10 binds to another protein that interacts with the DNA to start the production of new synaptic proteins. Further work is required to identify the molecular mechanisms that trigger RNF10 to leave the synapse. In addition, future studies should evaluate the levels and activity of RNF10 in brain disorders in which synapses are known to function abnormally. DOI:http://dx.doi.org/10.7554/eLife.12430.002
Collapse
Affiliation(s)
- Margarita C Dinamarca
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Francesca Guzzetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Anna Karpova
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Dmitry Lim
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Nico Mitro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Stefano Musardo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Elena Marcello
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Jennifer Stanic
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Tanmoy Samaddar
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | | | - Antonio Caldarelli
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Armando A Genazzani
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Julie Perroy
- CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Laurent Fagni
- CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Pier Luigi Canonico
- Dipartimento di Scienze del Farmaco, Università degli Studi del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Monica Di Luca
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
13
|
Ma Q, Yu H, Lin J, Sun Y, Shen X, Ren L. Screening for cardiac HERG potassium channel interacting proteins using the yeast two-hybrid technique. Cell Biol Int 2013; 38:239-45. [PMID: 24154981 DOI: 10.1002/cbin.10196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/25/2013] [Indexed: 12/12/2022]
Abstract
The human ERG protein (HERG or Kv 11.1) encoded by the human ether-a-go-go-related gene (herg) is the pore-forming subunit of the cardiac delayed rectifier potassium current (IKr) responsible for action potential (AP) repolarization. Mutations in HERG lead to long-QT syndrome, a major cause of arrhythmias. Protein-protein interactions are fundamental for ion channel trafficking, membrane localization, and functional modulation. To identify proteins involved in the regulation of the HERG channel, we conducted a yeast two-hybrid screen of a human heart cDNA library using the C-terminus or N-terminus of HERG as bait. Fifteen proteins were identified as HERG amino terminal (HERG-NT)-interacting proteins, including Caveolin-1 (a membrane scaffold protein with multiple interacting partners, including G-proteins, kinases and NOS), the zinc finger protein, FHL2 and PTPN12 (a non-receptor tyrosine phosphatase). Eight HERG carboxylic terminal (HERG-CT)-interacting proteins were also identified, including the NF-κB-interacting protein myotrophin, We have identified multiple potential interacting proteins that may regulate cardiac IKr through cytoskeletal interactions, G-protein modulation, phosphorylation and downstream second messenger and transcription cascades. These findings provide further insight into dynamic modulation of HERG under physiological conditions and arrhythmogenesis.
Collapse
Affiliation(s)
- Qingyan Ma
- Department of Cardiology, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | | | | | | | | | | |
Collapse
|
14
|
Malik YS, Sheikh MA, Lai M, Cao R, Zhu X. RING finger protein 10 regulates retinoic acid-induced neuronal differentiation and the cell cycle exit of P19 embryonic carcinoma cells. J Cell Biochem 2013; 114:2007-15. [DOI: 10.1002/jcb.24544] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 03/06/2013] [Indexed: 02/04/2023]
Affiliation(s)
- Yousra S. Malik
- Key Laboratory of Molecular Epigenetics of Ministry of Education; Institute of Cytology and Genetics, Northeast Normal University; Changchun; 130024; China
| | - Muhammad A. Sheikh
- Key Laboratory of Molecular Epigenetics of Ministry of Education; Institute of Cytology and Genetics, Northeast Normal University; Changchun; 130024; China
| | - Mingming Lai
- Key Laboratory of Molecular Epigenetics of Ministry of Education; Institute of Cytology and Genetics, Northeast Normal University; Changchun; 130024; China
| | - Rangjuan Cao
- Key Laboratory of Molecular Epigenetics of Ministry of Education; Institute of Cytology and Genetics, Northeast Normal University; Changchun; 130024; China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education; Institute of Cytology and Genetics, Northeast Normal University; Changchun; 130024; China
| |
Collapse
|
15
|
Host cell targets of tegument protein VP22 of herpes simplex virus 1. Arch Virol 2011; 156:1079-84. [DOI: 10.1007/s00705-011-0960-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 02/21/2011] [Indexed: 10/18/2022]
|
16
|
Liu L, Li Y, Lin J, Liang Q, Sheng X, Wu J, Huang R, Liu S, Li Y. Connexin43 interacts with Caveolin-3 in the heart. Mol Biol Rep 2009; 37:1685-91. [PMID: 19544087 DOI: 10.1007/s11033-009-9584-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 06/08/2009] [Indexed: 02/05/2023]
Abstract
Gap junctions (GJs), collections of multiple intercellular channels between neighboring cells, are specialized channels facilitating intercellular electrical and chemical communication. GJs are important for synchronizing coupling and coordinated contraction in the heart, and are crucial regulators of heart gene transcription, cardiac development, and protection of ischemic cardiomyocytes through second messenger communication. Identification of proteins that interact with Connexin43 (Cx43), the predominant protein in cardiac GJs, may contribute to the understanding of GJ functional regulation. Using a yeast two-hybrid system, we identified Caveolin-3 (Cav3) as a new Cx43-interacting protein. This interaction was confirmed by co-immunoprecipitation and co-localization experiments. CX43 interacts with Cav3, suggesting that Cav3 may participate in the functional regulation of GJs.
Collapse
Affiliation(s)
- Limei Liu
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, 57 Changping Road, 515041, Shantou, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lin J, Lin S, Choy PC, Shen X, Deng C, Kuang S, Wu J, Xu W. The regulation of the cardiac potassium channel (HERG) by caveolin-1. Biochem Cell Biol 2008; 86:405-15. [PMID: 18923542 DOI: 10.1139/o08-118] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Protein-protein interaction plays a key role in the regulation of biological processes. The human potassium (HERG) channel is encoded by the ether-à-go-go-related gene (herg), and its activity may be regulated by association with other cellular proteins. To identify cellular proteins that might play a role in the regulation of the HERG channel, we screened a human heart cDNA library with the N terminus of HERG using a yeast 2-hybrid system, and identified caveolin-1 as a potential HERG partner. The interaction between these 2 proteins was confirmed by coimmunoprecipitation assay, and their overlapping subcellular localization was demonstrated by fluorescence immunocytochemistry. The physiologic implication of the protein-protein interaction was studied in whole-cell patch-clamp electrophysiology experiments. A significant increase in HERG current amplitude and a faster deactivation of tail current were observed in HEK293/HERG cells in a membrane lipid rafts disruption model and caveolin-1 knocked down cells by RNA interference. Alternatively, when caveolin-1 was overexpressed, the HERG current amplitude was significantly reduced and the tail current was deactivated more slowly. Taken together, these data indicate that HERG channels interact with caveolin-1 and are negatively regulated by this interaction. The finding from this study clearly demonstrates the regulatory role of caveolin-1 on HERG channels, and may help to understand biochemical events leading to arrhythmogenesis in the long QT syndrome in cardiac patients.
Collapse
Affiliation(s)
- Jijin Lin
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Provincial Cardiovascular Institute, 96 Dongchuan Road, Guangzhou 510080, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Hoshikawa S, Ogata T, Fujiwara S, Nakamura K, Tanaka S. A novel function of RING finger protein 10 in transcriptional regulation of the myelin-associated glycoprotein gene and myelin formation in Schwann cells. PLoS One 2008; 3:e3464. [PMID: 18941509 PMCID: PMC2565066 DOI: 10.1371/journal.pone.0003464] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 09/22/2008] [Indexed: 11/23/2022] Open
Abstract
Myelin-associated glycoprotein (MAG) has been detected in Schwann cells prior to the onset of myelination, suggesting its functions in the initiation of myelination. However, transcriptional regulatory mechanisms of MAG remain to be elucidated. Here, we analyzed the promoter of the MAG gene by using luciferase reporter systems in the primary rat Schwann cells. We identified a novel cis-acting element located 160 bp upstream from the MAG transcription initiation site. Using the identified cis-element as a bait, we performed yeast one-hybrid screening and isolated a cDNA encoding a RNF10 as a putative trans-acting protein. When overexpressed in Schwann cells, RNF10 enhanced the activity of the MAG promoter. When RNF10 expression in Schwann cells was knocked down by siRNA, endogenous MAG mRNA and protein expression decreased. Furthermore, we evaluated myelin synthesis using Schwann cell-DRG neuron cocultures. When Schwann cells were infected with retrovirus expressing RNF10 siRNA, myelin formation was inhibited. These data suggest that RNF10 regulates MAG expression and is required for myelin formation.
Collapse
Affiliation(s)
- Shinya Hoshikawa
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
19
|
Lin J, Lin S, Yu X, Choy PC, Shen X, Deng C, Kuang S, Wu J. The four and a half LIM domain protein 2 interacts with and regulates the HERG channel. FEBS J 2008; 275:4531-9. [DOI: 10.1111/j.1742-4658.2008.06596.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Douville JM, Wigle JT. Regulation and function of homeodomain proteins in the embryonic and adult vascular systems. Can J Physiol Pharmacol 2007; 85:55-65. [PMID: 17487245 DOI: 10.1139/y06-091] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
During embryonic development, the cardiovascular system first forms and then gives rise to the lymphatic vascular system. Homeobox genes are essential for both the development of the blood and lymphatic vascular systems, as well as for their maintenance in the adult. These genes all encode proteins that are transcription factors that contain a well conserved DNA binding motif, the homeodomain. It is through the homeodomain that these transcription factors bind to the promoters of target genes and regulate their expression. Although many homeodomain proteins have been found to be expressed within the vascular systems, little is known about their downstream target genes. This review highlights recent advances made in the identification of novel genes downstream of the homeodomain proteins that are necessary for regulating vascular cellular processes such as proliferation, migration, and endothelial tube formation. Factors known to regulate the functions of vascular cells via modulating the expression of homeobox genes will be discussed. We will also review current methods used to identify and characterize downstream target genes of homeodomain proteins.
Collapse
Affiliation(s)
- Josette M Douville
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre and Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | | |
Collapse
|
21
|
Lallier TE, Spencer A. Use of microarrays to find novel regulators of periodontal ligament fibroblast differentiation. Cell Tissue Res 2006; 327:93-109. [PMID: 17024420 DOI: 10.1007/s00441-006-0282-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 06/13/2006] [Indexed: 01/07/2023]
Abstract
Periodontal regeneration requires the coordinated movement and differentiation of several cell types in order to re-establish the cementum, periodontal ligament (PDL), and alveolar bone. Cells in culture are often used as model systems for mature tissues, although they may represent expanded progenitor cell populations. Comparison of transcript expression between fresh PDL tissue and PDL cell isolates by MicroArray analysis has revealed numerous molecular differences. Several transcripts (including alkaline phosphatase, bone sialoprotein, periostin, and fibromodulin) are expressed at higher levels in fresh PDL than in cultured PDL cells. In contrast, PDL cells in culture selectively express a variety of growth factors. Several of these growth factors alter PDL fibroblast behavior. Two members of the transforming growth factor beta family of growth factors, namely, bone morphogenic protein-7 (BMP7) and growth differentiation factor-5 (GDF5), reduce cell proliferation and Stro-1 expression (a bone marrow stromal stem cell marker), whereas only BMP7 induces alkaline phosphatase activity. In contrast, fibroblast growth factor-5 induces enhanced cell proliferation and Stro-1 expression, while repressing alkaline phosphatase activity. The stimulation of PDL cells to differentiate (either by BMP7 or GDF5) inhibits cell motility. Thus, PDL cells in culture are regulated by several factors that differentially stimulate a mineralized (cementoblast-like) fate, a non-mineralized fate (mature fibroblasts), or the propagation of a more naive phenotype (potential progenitors).
Collapse
Affiliation(s)
- Thomas E Lallier
- Department of Cell Biology and Anatomy, Center of Excellence in Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Science Center, 1100 Florida Avenue, New Orleans, LA 70119, USA.
| | | |
Collapse
|