1
|
Zhao J, Liu S, Li K, Yang Y, Zhao Y, Zhu X. RBM3 Promotes Anti-inflammatory Responses in Microglia and Serves as a Neuroprotective Target of Ischemic Stroke. Mol Neurobiol 2024; 61:7384-7402. [PMID: 38386136 DOI: 10.1007/s12035-024-04052-4] [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: 06/27/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Ischemic stroke is a major cause of death and disability in adults. Hypothermic treatment is successful in treating neonatal cerebral ischemia, but its application is restricted in adult patients due to complex management strategies and severe adverse effects. Two homologous RNA-binding proteins, RBM3 and CIRP, are the only known cold-inducible proteins in vertebrates, and their expression levels are robustly elevated by mild to moderate hypothermia. In previous studies, we and others have demonstrated that both RBM3 and CIRP mediate the neuroprotective and neurogenic effects of hypothermia in cell and animal models. However, CIRP can also be detrimental to neurons by triggering neuroinflammatory responses, complicating its post-stroke functions. In this study, we compared the properties of the two cold-inducible RNA-binding proteins after ischemic stroke. Our results indicated that RBM3 expression was stimulated in the ischemic brain of stroke patients, while CIRP expression was not. In an experimental model, RBM3 can ameliorate ischemic-like insult by promoting neuronal survival and eliciting anti-inflammatory responses in activated microglia, while the impact of CIRP was intriguing. Collectively, our data supported the notion that RBM3 may be a more promising therapeutic target than CIRP for treating ischemic stroke. We further demonstrated that zr17-2, a small molecule initially identified to target CIRP, can specifically target RBM3 but not CIRP in microglia. zr17-2 demonstrated anti-inflammatory and neuroprotective effects after ischemic stroke both in vitro and in vivo, suggesting its potential therapeutic value.
Collapse
Affiliation(s)
- Junyi Zhao
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Siyu Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Kunyu Li
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
| | - Yulu Yang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yue Zhao
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China
| | - Xinzhou Zhu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, China.
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen, China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
2
|
Thibault E, Brandizzi F. Post-translational modifications: emerging directors of cell-fate decisions during endoplasmic reticulum stress in Arabidopsis thaliana. Biochem Soc Trans 2024; 52:831-848. [PMID: 38600022 PMCID: PMC11088923 DOI: 10.1042/bst20231025] [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: 01/19/2024] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Homeostasis of the endoplasmic reticulum (ER) is critical for growth, development, and stress responses. Perturbations causing an imbalance in ER proteostasis lead to a potentially lethal condition known as ER stress. In ER stress situations, cell-fate decisions either activate pro-life pathways that reestablish homeostasis or initiate pro-death pathways to prevent further damage to the organism. Understanding the mechanisms underpinning cell-fate decisions in ER stress is critical for crop development and has the potential to enable translation of conserved components to ER stress-related diseases in metazoans. Post-translational modifications (PTMs) of proteins are emerging as key players in cell-fate decisions in situations of imbalanced ER proteostasis. In this review, we address PTMs orchestrating cell-fate decisions in ER stress in plants and provide evidence-based perspectives for where future studies may focus to identify additional PTMs involved in ER stress management.
Collapse
Affiliation(s)
- Ethan Thibault
- Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, U.S.A
- Department of Plant Biology, Michigan State University, East Lansing, MI, U.S.A
| | - Federica Brandizzi
- Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, U.S.A
- Department of Plant Biology, Michigan State University, East Lansing, MI, U.S.A
- Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI, U.S.A
| |
Collapse
|
3
|
Gao H, Tian M, Geng X, Zhao J, Song Y, Wu B, Tian X, Yang Y, Ni W, Yang H. Cyfluthrin exposure during pregnancy causes neurotoxicity in offspring-Ca 2+ overload via IP3R-GRP75-VDAC1 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116218. [PMID: 38492481 DOI: 10.1016/j.ecoenv.2024.116218] [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: 10/31/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cyfluthrin (Cy) is a widely used pyrethroid insecticide. There is growing evidence that Cy can cause damage to the nervous, reproductive, and immune systems, but there is limited evidence on the potential effects of maternal Cy exposure on offspring. A model of maternal Cy exposure was used to assess its neurobehavioral effects on young-adult offspring. We found that gestational Cy exposure affected pregnancy outcomes and fetal development, and that offspring showed impairments in anxiety as well as learning and memory, accompanied by impairments in hippocampal synaptic ultrastructure and synaptic plasticity. In addition, the IP3R-GRP75-VDAC1 apoptogenic pathway was also upregulated, and in vitro models showed that inhibition of this pathway alleviated neuronal apoptosis as well as synaptic plasticity damage. In conclusion, maternal Cy exposure during pregnancy can cause neurobehavioral abnormalities and synaptic damage in offspring, which may be related to neuronal apoptosis induced by activation of the IP3R-GRP75-VDAC1 pathway in the hippocampus of offspring. Our findings provide clues to understand the neurotoxicity mechanism of maternal Cy exposure to offspring during pregnancy.
Collapse
Affiliation(s)
- Haoxuan Gao
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Mi Tian
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Xiaozhe Geng
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Ji Zhao
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Yanan Song
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Bing Wu
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Xueyan Tian
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Yong Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China
| | - Wensi Ni
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| | - Huifang Yang
- School of Public Health, Ningxia Medical University, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China; Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of National Key Laboratory, No.1160, Shengli Street, Xingqing District, Yinchuan, Ningxia, China.
| |
Collapse
|
4
|
Perez-Pouchoulen M, Jaiyesimi A, Bardhi K, Waddell J, Banerjee A. Hypothermia increases cold-inducible protein expression and improves cerebellar-dependent learning after hypoxia ischemia in the neonatal rat. Pediatr Res 2023; 94:539-546. [PMID: 36810641 PMCID: PMC10403381 DOI: 10.1038/s41390-023-02535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Hypoxic ischemic encephalopathy remains a significant cause of developmental disability.1,2 The standard of care for term infants is hypothermia, which has multifactorial effects.3-5 Therapeutic hypothermia upregulates the cold-inducible protein RNA binding motif 3 (RBM3) that is highly expressed in developing and proliferative regions of the brain.6,7 The neuroprotective effects of RBM3 in adults are mediated by its ability to promote the translation of mRNAs such as reticulon 3 (RTN3).8 METHODS: Hypoxia ischemia or control procedure was conducted in Sprague Dawley rat pups on postnatal day 10 (PND10). Pups were immediately assigned to normothermia or hypothermia at the end of the hypoxia. In adulthood, cerebellum-dependent learning was tested using the conditioned eyeblink reflex. The volume of the cerebellum and the magnitude of cerebral injury were measured. A second study quantified RBM3 and RTN3 protein levels in the cerebellum and hippocampus collected during hypothermia. RESULTS Hypothermia reduced cerebral tissue loss and protected cerebellar volume. Hypothermia also improved learning of the conditioned eyeblink response. RBM3 and RTN3 protein expression were increased in the cerebellum and hippocampus of rat pups subjected to hypothermia on PND10. CONCLUSIONS Hypothermia was neuroprotective in male and female pups and reversed subtle changes in the cerebellum after hypoxic ischemic. IMPACT Hypoxic ischemic produced tissue loss and a learning deficit in the cerebellum. Hypothermia reversed both the tissue loss and learning deficit. Hypothermia increased cold-responsive protein expression in the cerebellum and hippocampus. Our results confirm cerebellar volume loss contralateral to the carotid artery ligation and injured cerebral hemisphere, suggesting crossed-cerebellar diaschisis in this model. Understanding the endogenous response to hypothermia might improve adjuvant interventions and expand the clinical utility of this intervention.
Collapse
Affiliation(s)
| | - Ayodele Jaiyesimi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Keti Bardhi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Long F, Hu L, Chen Y, Duan X, Xie K, Feng J, Wang M. RBM3 is associated with acute lung injury in septic mice and patients via the NF-κB/NLRP3 pathway. Inflamm Res 2023; 72:731-744. [PMID: 36781430 DOI: 10.1007/s00011-023-01705-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/23/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Sepsis refers to host response disorders caused by infection, leading to life-threatening organ dysfunction. RNA-binding motif protein 3 (RBM3) is an important cold-shock protein that is upregulated in response to mild hypothermia or hypoxia. In this study, we aimed to investigate whether RBM3 is involved in sepsis-associated acute lung injury (ALI). Intraperitoneal injection of LPS (10 mg/kg) was performed in wild type (WT) and RBM3 knockout (KO, RBM3-/-) mice to establish an in vivo sepsis model. An NLRP3 inflammasome inhibitor, MCC950 (50 mg/kg), was injected intraperitoneally 30 min before LPS treatment. Serum, lung tissues, and BALF were collected 24 h later for further analysis. In addition, we also collected serum from sepsis patients and healthy volunteers to detect their RBM3 expression. The results showed that the expression of RBM3 in the lung tissues of LPS-induced sepsis mice and the serum of patients with sepsis was significantly increased and positively correlated with disease severity. In addition, RBM3 knockout (KO) mice had a low survival rate, and RBM3 KO mice had more severe lung damage, inflammation, lung cell apoptosis, and oxidative stress than WT mice. LPS treatment significantly increased the levels of nucleotide binding and oligomerization domain-like receptor family 3 (NLRP3) inflammasomes and mononuclear cell nuclear factor-κB (NF-κB) in the lung tissues of RBM3 KO mice. However, these levels were only slightly elevated in WT mice. Interestingly, MCC950 improved LPS-induced acute lung injury in WT and RBM3 KO mice but inhibited the expression of NLRP3, caspase-1, and IL-1β. In conclusion, RBM3 was overexpressed in sepsis patients and LPS-induced mice. RBM3 gene deficiency aggravated sepsis-associated ALI through the NF-κB/NLRP3 pathway.
Collapse
Affiliation(s)
- Feiyu Long
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Liren Hu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yingxu Chen
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Xiaoxia Duan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Maohua Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
6
|
Gao Y, Cao H, Huang D, Zheng L, Nie Z, Zhang S. RNA-Binding Proteins in Bladder Cancer. Cancers (Basel) 2023; 15:cancers15041150. [PMID: 36831493 PMCID: PMC9953953 DOI: 10.3390/cancers15041150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
RNA-binding proteins (RBPs) are key regulators of transcription and translation, with highly dynamic spatio-temporal regulation. They are usually involved in the regulation of RNA splicing, polyadenylation, and mRNA stability and mediate processes such as mRNA localization and translation, thereby affecting the RNA life cycle and causing the production of abnormal protein phenotypes that lead to tumorigenesis and development. Accumulating evidence supports that RBPs play critical roles in vital life processes, such as bladder cancer initiation, progression, metastasis, and drug resistance. Uncovering the regulatory mechanisms of RBPs in bladder cancer is aimed at addressing the occurrence and progression of bladder cancer and finding new therapies for cancer treatment. This article reviews the effects and mechanisms of several RBPs on bladder cancer and summarizes the different types of RBPs involved in the progression of bladder cancer and the potential molecular mechanisms by which they are regulated, with a view to providing information for basic and clinical researchers.
Collapse
|
7
|
Ismail Abo El-Fadl HM, Mohamed MFA. Targeting endoplasmic reticulum stress, Nrf-2/HO-1, and NF-κB by myristicin and its role in attenuation of ulcerative colitis in rats. Life Sci 2022; 311:121187. [PMID: 36403646 DOI: 10.1016/j.lfs.2022.121187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/15/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
AIMS Ulcerative colitis (UC) is characterized by the up-regulation of pro-inflammatory mediators, apoptotic signals, and oxidative stress that can lead to an increased risk of colorectal cancer. The present study aims to investigate the possible role of myristicin in modulating endoplasmic reticulum stress (ERS) and risk-associated conditions in acetic acid (AA)-induced UC. MATERIALS AND METHODS Adult male rats were treated with 150 mg/kg body weight of myristicin or mesalazine orally either as pre/post treatment or post-treatment only. The gene expression of glucose-related protein 78 (GRP78), CCAAT/enhancer-binding protein homologous protein (CHOP), and nuclear factor kappa B (NF-κB), percentage of DNA fragmentation, and serum levels of some oxidative and inflammatory markers were measured. KEY FINDINGS The results indicated the potential upregulation of ERS, pro-apoptotic, lipid peroxidation, and pro-inflammatory cascades by induction of UC in rats. However, myristicin could effectively reverse the deteriorated effects of ulceration in colonic mucosa. It was mediated through downregulation of the ERS markers GRP78 and CHOP genes expression, reduction of NF-κB mRNA expression, DNA fragmentation, reduced lipid peroxidation, myeloperoxidase (MPO) activity and pro-inflammatory markers (Tumor necrosis factor-α (TNF-α), Interleukin-1β (IL-1β) and cyclo‑oxygenase (COX-2) activity). Accompanied by elevated levels of IL-10, colonic Nuclear erythroid factor (Nrf-2) and Heme oxygenase (HO-1) activity as well as blood antioxidant enzymes activity. Results of docking might confirm the biological results of our study. SIGNIFICANCE Myristicin could effectively modulate important stress, and inflammatory effectors and protect mucosal DNA from oxidative damage which can serve as a promising candidate for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Huda M Ismail Abo El-Fadl
- Biochemistry and Nutrition Department, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo, Egypt.
| | - Mamdouh F A Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, 82524 Sohag, Egypt
| |
Collapse
|
8
|
Liu Y, Shi H, Hu Y, Yao R, Liu P, Yang Y, Li S. RNA binding motif protein 3 (RBM3) promotes protein kinase B (AKT) activation to enhance glucose metabolism and reduce apoptosis in skeletal muscle of mice under acute cold exposure. Cell Stress Chaperones 2022; 27:603-618. [PMID: 36149580 PMCID: PMC9672220 DOI: 10.1007/s12192-022-01297-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/01/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023] Open
Abstract
The main danger of cold stress to animals in cold regions is systemic metabolic changes and protein synthesis inhibition. RBM3, an exceptional cold shock protein, is rapidly upregulated in response to hypothermia to resist the adverse effects of cold stress. However, the mechanism of the protective effect and the rapid upregulation of RBM3 remains unclear. O-GlcNAcylation, an atypical O-glycosylation, is precisely regulated only by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) and participates in the signal transduction of multiple cellular stress responses as a "stress and nutrition receptor." Therefore, our study aimed to explore the mechanism of RBM3 regulating glucose metabolism and promoting survival in skeletal muscle under acute cold exposure. Meanwhile, our study verifies whether O-GlcNAcylation mediated by OGT rapidly upregulates RBM3. The blood and skeletal muscle of mice were collected at the end of cold exposure treatment for 0, 2, and 4 h. Changes in levels of RBM3, AKT, glycolysis apoptosis, and OGT were measured. The results show that acute cold exposure upregulated RBM3, OGT, and AKT phosphorylation and increased energy consumption, which enhanced glycolysis and prevent apoptosis. In the 32 °C mild hypothermia model in vitro, overexpression of RBM3 enhanced AKT phosphorylation. Meanwhile, inactivation of AKT by wortmannin resulted in increased apoptosis and decreased glucose metabolism in skeletal muscle under acute cold exposure. In addition, OGT-mediated O-GlcNAcylation of p65 was confirmed in mouse myoblast cell line (C2C12) cells at mild hypothermia. O-GlcNAcylation level affected p65 activity and nuclear translocation. Compared with wild type (WT) mice, RBM3 and p65 phosphorylation were decreased in specific skeletal muscle Ogt (KO) mice, whereas AKT phosphorylation, glycolysis, and apoptosis were increased. Taken together, O-GlcNAcylation of p65 upregulates RBM3 to promote AKT phosphorylation, enhance glucose metabolism, and reduce apoptosis in skeletal muscle of mice under acute cold exposure.
Collapse
Affiliation(s)
- Yang Liu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Hongzhao Shi
- Department of Animal Engineering, Yangling Vocational & Technical College, Xianyang, 712199, People's Republic of China
| | - Yajie Hu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Ruizhi Yao
- College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, 028000, People's Republic of China
| | - Peng Liu
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Yuying Yang
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China
| | - Shize Li
- National Experimental Teaching Demonstration Center of Animal Medicine Foundation, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, People's Republic of China.
| |
Collapse
|
9
|
Hu Y, Liu Y, Quan X, Fan W, Xu B, Li S. RBM3 is an outstanding cold shock protein with multiple physiological functions beyond hypothermia. J Cell Physiol 2022; 237:3788-3802. [PMID: 35926117 DOI: 10.1002/jcp.30852] [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: 03/16/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022]
Abstract
RNA-binding motif protein 3 (RBM3), an outstanding cold shock protein, is rapidly upregulated to ensure homeostasis and survival in a cold environment, which is an important physiological mechanism in response to cold stress. Meanwhile, RBM3 has multiple physiological functions and participates in the regulation of various cellular physiological processes, such as antiapoptosis, circadian rhythm, cell cycle, reproduction, and tumogenesis. The structure, conservation, and tissue distribution of RBM3 in human are demonstrated in this review. Herein, the multiple physiological functions of RBM3 were summarized based on recent research advances. Meanwhile, the cytoprotective mechanism of RBM3 during stress under various adverse conditions and its regulation of transcription were discussed. In addition, the neuroprotection of RBM3 and its oncogenic role and controversy in various cancers were investigated in our review.
Collapse
Affiliation(s)
- Yajie Hu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| | - Yang Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| | - Xin Quan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| | - Wenxuan Fan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| | - Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| | - Shize Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, National Experimental Teaching Demonstration Center of Animal Medicine Foundation, Daqing, China
| |
Collapse
|
10
|
Witkamp D, Oudejans E, Hu‐A‐Ng GV, Hoogterp L, Krzywańska AM, Žnidaršič M, Marinus K, de Veij Mestdagh CF, Bartelink I, Bugiani M, van der Knaap MS, Abbink TEM. Guanabenz ameliorates disease in vanishing white matter mice in contrast to sephin1. Ann Clin Transl Neurol 2022; 9:1147-1162. [PMID: 35778832 PMCID: PMC9380178 DOI: 10.1002/acn3.51611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Vanishing white matter (VWM) is a leukodystrophy, characterized by stress-sensitive neurological deterioration and premature death. It is currently without curative treatment. It is caused by bi-allelic pathogenic variants in the genes encoding eukaryotic initiation factor 2B (eIF2B). eIF2B is essential for the regulation of the integrated stress response (ISR), a physiological response to cellular stress. Preclinical studies on VWM mouse models revealed that deregulated ISR is key in the pathophysiology of VWM and an effective treatment target. Guanabenz, an α2-adrenergic agonist, attenuates the ISR and has beneficial effects on VWM neuropathology. The current study aimed at elucidating guanabenz's disease-modifying potential and mechanism of action in VWM mice. Sephin1, an ISR-modulating guanabenz analog without α2-adrenergic agonistic properties, was included to separate effects on the ISR from α2-adrenergic effects. METHODS Wild-type and VWM mice were subjected to placebo, guanabenz or sephin1 treatments. Effects on clinical signs, neuropathology, and ISR deregulation were determined. Guanabenz's and sephin1's ISR-modifying effects were tested in cultured cells that expressed or lacked the α2-adrenergic receptor. RESULTS Guanabenz improved clinical signs, neuropathological hallmarks, and ISR regulation in VWM mice, but sephin1 did not. Guanabenz's effects on the ISR in VWM mice were not replicated in cell cultures and the contribution of α2-adrenergic effects on the deregulated ISR could therefore not be assessed. INTERPRETATION Guanabenz proved itself as a viable treatment option for VWM. The exact mechanism through which guanabenz exerts its ameliorating impact on VWM requires further studies. Sephin1 is not simply a guanabenz replacement without α2-adrenergic effects.
Collapse
Affiliation(s)
- Diede Witkamp
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Ellen Oudejans
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Gino V. Hu‐A‐Ng
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Leoni Hoogterp
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Aleksandra M. Krzywańska
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Milo Žnidaršič
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Kevin Marinus
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Christina F. de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
- Present address:
Alzheimer Center AmsterdamVU University Medical CenterAmsterdamThe Netherlands
| | - Imke Bartelink
- Department of Pharmacy and Clinical PharmacologyAmsterdam UMC, Location VUmcAmsterdamThe Netherlands
| | - Marianna Bugiani
- Department of PathologyAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Marjo S. van der Knaap
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| | - Truus E. M. Abbink
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy CenterAmsterdam University Medical Centers, Vrije Universiteit and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVU UniversityAmsterdamThe Netherlands
| |
Collapse
|
11
|
Fan L, Li Y, Zhang X, Wu Y, Song Y, Zhang F, Zhang J, Sun H. Time-resolved proteome and transcriptome of paraquat-induced pulmonary fibrosis. Pulm Pharmacol Ther 2022; 75:102145. [PMID: 35817254 DOI: 10.1016/j.pupt.2022.102145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/31/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUNDS Pulmonary fibrosis (PF) is a pathological state presenting at the progressive stage of heterogeneous interstitial lung disease (ILD). The current understanding of the molecular mechanisms involved is incomplete. This clinical toxicology study focused on the pulmonary fibrosis induced by paraquat (PQ), a widely-used herbicide. Using proteo-transcriptome analysis, we identified differentially expressed proteins (DEPs) derived from the initial development of fibrosis to the dissolved stage and provided further functional analysis. METHODS We established a mouse model of progressive lung fibrosis via intratracheal instillation of paraquat. To acquire a comprehensive and unbiased understanding of the onset of pulmonary fibrosis, we performed time-series proteomics profiling (iTRAQ) and RNA sequencing (RNA-Seq) on lung samples from paraquat-treated mice and saline control. The biological functions and pathways involved were evaluated through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway analysis. Correlation tests were conducted on comparable groups 7 days and 28 days post-exposure. Differentially expressed proteins and genes following the same trend on the protein and mRNA levels were selected for validation. The functions of the selected molecules were identified in vitro. The protein level was overexpressed by transfecting gene-containing plasmid or suppressed by transfecting specific siRNA in A549 cells. The levels of endothlial-mesenchymal transition (EMT) markers, including E-cadherin, vimentin, FN1, and α-SMA, were determined via western blot to evaluate the fibrotic process. RESULTS We quantified 1358 DEPs on day 7 and 426 DEPs on day 28 post exposure (Fold change >1.2; Q value < 0.05). The top 5 pathways - drug metabolism-cytochrome P450, metabolism of xenobiotics by cytochrome P450, complement and coagulation cascades, chemical carcinogenesis, protein digestion and absorption - were involved on both day 7 and day 28. Several pathways, including tight junction, focal adhesion, platelet activation, and ECM-receptor interaction, were more enriched on day 28 than on day 7. Integrative analysis of the proteome and transcriptome revealed a moderate correlation of quantitative protein abundance ratios with RNA abundance ratios (Spearman R = 0.3950 and 0.2477 on days 7 and 28, respectively), indicating that post-transcriptional regulation plays an important role in lung injury and repair. Western blot identified that the protein expressions of FN1, S100A4, and RBM3 were significantly upregulated while that of CYP1A1, FMO3, and PGDH were significantly downregulated on day 7. All proteins generally recovered to baseline on day 28. qPCR showed the mRNA levels of Fn1, S100a4, Rbm3, Cyp1a1, Fmo3, and Hpgd changed following the same trend as the levels of their respective proteins. Further, in vitro experiments showed that RBM3 was upregulated while PGDH was downregulated in an EMT model established in human lung epithelial A549 cells. RBM3 overexpression and PGDH knockout could both induce EMT in A549 cells. RBM3 knockout or PGDH overexpression had no reverse effect on EMT in A549 cells. CONCLUSIONS Our proteo-transcriptomic study determined the proteins responsible for fibrogenesis and uncovers their dynamic regulation from lung injury to repair, providing new insights for the development of biomarkers for diagnosis and treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Lu Fan
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China; Department of Emergency, Clinical Medical College, Yangzhou University, Yangzhou, PR China.
| | - Yuan Li
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Xiaomin Zhang
- Department of Emergency, The Second People's Hospital of Wuxi, Affiliated to Nanjing Medical University, Wuxi, PR China.
| | - Yuxuan Wu
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Yang Song
- Department of Emergency, Nanjing Jiangbei Hospital, Affiliated to Southeast University, Nanjing, PR China.
| | - Feng Zhang
- Department of Emergency, Jiangsu Province Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, PR China.
| | - Jinsong Zhang
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Hao Sun
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
12
|
Block H, Rossaint J, Zarbock A. The Fatal Circle of NETs and NET-Associated DAMPs Contributing to Organ Dysfunction. Cells 2022; 11:1919. [PMID: 35741047 PMCID: PMC9222025 DOI: 10.3390/cells11121919] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is the first line of defense against invading pathogens or sterile injuries. Pattern recognition receptors (PRR) sense molecules released from inflamed or damaged cells, or foreign molecules resulting from invading pathogens. PRRs can in turn induce inflammatory responses, comprising the generation of cytokines or chemokines, which further induce immune cell recruitment. Neutrophils represent an essential factor in the early immune response and fulfill numerous tasks to fight infection or heal injuries. The release of neutrophil extracellular traps (NETs) is part of it and was originally attributed to the capture and elimination of pathogens. In the last decade studies revealed a detrimental role of NETs during several diseases, often correlated with an exaggerated immune response. Overwhelming inflammation in single organs can induce remote organ damage, thereby further perpetuating release of inflammatory molecules. Here, we review recent findings regarding damage-associated molecular patterns (DAMPs) which are able to induce NET formation, as well as NET components known to act as DAMPs, generating a putative fatal circle of inflammation contributing to organ damage and sequentially occurring remote organ injury.
Collapse
Affiliation(s)
| | | | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, 48149 Muenster, Germany; (H.B.); (J.R.)
| |
Collapse
|
13
|
Pre-clinical and clinical studies on the role of RBM3 in muscle-invasive bladder cancer: longitudinal expression, transcriptome-level effects and modulation of chemosensitivity. BMC Cancer 2022; 22:131. [PMID: 35109796 PMCID: PMC8811987 DOI: 10.1186/s12885-021-09168-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 12/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background The response to neoadjuvant cisplatin-based chemotherapy (NAC) in muscle-invasive bladder cancer (MIBC) is impaired in up to 50% of patients due to chemoresistance, with no predictive biomarkers in clinical use. The proto-oncogene RNA-binding motif protein 3 (RBM3) has emerged as a putative modulator of chemotherapy response in several solid tumours but has a hitherto unrecognized role in MIBC. Methods RBM3 protein expression level in tumour cells was assessed via immunohistochemistry in paired transurethral resection of the bladder (TURB) specimens, cystectomy specimens and lymph node metastases from a consecutive cohort of 145 patients, 65 of whom were treated with NAC. Kaplan-Meier and Cox regression analyses were applied to estimate the impact of RBM3 expression on time to recurrence (TTR), cancer-specific survival (CSS), and overall survival (OS) in strata according to NAC treatment. The effect of siRNA-mediated silencing of RBM3 on chemosensitivity was examined in RT4 and T24 human bladder carcinoma cells in vitro. Cellular functions of RBM3 were assessed using RNA-sequencing and gene ontology analysis, followed by investigation of cell cycle distribution using flow cytometry. Results RBM3 protein expression was significantly higher in TURB compared to cystectomy specimens but showed consistency between primary tumours and lymph node metastases. Patients with high-tumour specific RBM3 expression treated with NAC had a significantly reduced risk of recurrence and a prolonged CSS and OS compared to NAC-untreated patients. In high-grade T24 carcinoma cells, which expressed higher RBM3 mRNA levels compared to RT4 cells, RBM3 silencing conferred a decreased sensitivity to cisplatin and gemcitabine. Transcriptomic analysis revealed potential involvement of RBM3 in facilitating cell cycle progression, in particular G1/S-phase transition, and initiation of DNA replication. Furthermore, siRBM3-transfected T24 cells displayed an accumulation of cells residing in the G1-phase as well as altered levels of recognised regulators of G1-phase progression, including Cyclin D1/CDK4 and CDK2. Conclusions The presented data highlight the potential value of RBM3 as a predictive biomarker of chemotherapy response in MIBC, which could, if prospectively validated, improve treatment stratification of patients with this aggressive disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09168-7.
Collapse
|
14
|
Roy S, Boral S, Maiti S, Kushwaha T, Basak AJ, Lee W, Basak A, Gholap SL, Inampudi KK, De S. Structural and dynamic studies of the human RNA binding protein RBM3 reveals the molecular basis of its oligomerization and RNA recognition. FEBS J 2021; 289:2847-2864. [PMID: 34837346 DOI: 10.1111/febs.16301] [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: 03/30/2021] [Revised: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 12/01/2022]
Abstract
Human RNA-binding motif 3 protein (RBM3) is a cold-shock protein which functions in various aspects of global protein synthesis, cell proliferation and apoptosis by interacting with the components of basal translational machinery. RBM3 plays important roles in tumour progression and cancer metastasis, and also has been shown to be involved in neuroprotection and endoplasmic reticulum stress response. Here, we have solved the solution NMR structure of the N-terminal 84 residue RNA recognition motif (RRM) of RBM3. The remaining residues are rich in RGG and YGG motifs and are disordered. The RRM domain adopts a βαββαβ topology, which is found in many RNA-binding proteins. NMR-monitored titration experiments and molecular dynamic simulations show that the beta-sheet and two loops form the RNA-binding interface. Hydrogen bond, pi-pi and pi-cation are the key interactions between the RNA and the RRM domain. NMR, size exclusion chromatography and chemical cross-linking experiments show that RBM3 forms oligomers in solution, which is favoured by decrease in temperature, thus, potentially linking it to its function as a cold-shock protein. Temperature-dependent NMR studies revealed that oligomerization of the RRM domain occurs via nonspecific interactions. Overall, this study provides the detailed structural analysis of RRM domain of RBM3, its interaction with RNA and the molecular basis of its temperature-dependent oligomerization.
Collapse
Affiliation(s)
- Sayantani Roy
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| | - Soumendu Boral
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| | - Snigdha Maiti
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| | - Tushar Kushwaha
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Aditya J Basak
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| | - Woonghee Lee
- Department of Chemistry, University of Colorado Denver, CO, USA
| | - Amit Basak
- School of Bioscience, Indian Institute of Technology Kharagpur, India.,Department of Chemistry, Indian Institute of Technology Kharagpur, India
| | - Shivajirao L Gholap
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India
| | - Krishna K Inampudi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Soumya De
- School of Bioscience, Indian Institute of Technology Kharagpur, India
| |
Collapse
|
15
|
Cooling and Sterile Inflammation in an Oxygen-Glucose-Deprivation/Reperfusion Injury Model in BV-2 Microglia. Mediators Inflamm 2021; 2021:8906561. [PMID: 34776788 PMCID: PMC8589512 DOI: 10.1155/2021/8906561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/02/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Cold-inducible RNA-binding protein (CIRBP) has been shown to be involved not only in cooling-induced cellular protection but also as a mediator of sterile inflammation, a critical mechanism of the innate immune response in ischemia/reperfusion (I/R) injury. The role of microglia and its activation in cerebral I/R injury warrants further investigation as both detrimental and regenerative properties have been described. Therefore, we investigated the effects of cooling, specifically viability, activation, and release of damage associated molecular patterns (DAMPs) on oxygen glucose deprivation/reperfusion- (OGD/R-) induced injury in murine BV-2 microglial cells. Methods Murine BV-2 microglial cells were exposed to 2 to 6 h OGD (0.2% O2 in glucose- and serum-free medium) followed by up to 19 h of reperfusion, simulated by restoration of oxygen (21% O2) and nutrients. Cells were maintained at either normothermia (37°C) or cooled to 33.5°C, 1 h after experimental start. Cultured supernatants were harvested after exposure to OGD for analysis of DAMP secretions, including high-mobility group box 1 (HMGB1), heat shock protein 70 (HSP70), and CIRBP, and cytotoxicity was assessed by lactate dehydrogenase releases after exposure to OGD and reperfusion. Intracellular cold-shock proteins CIRBP and RNA-binding motif 3 (RBM3) as well as caspases 9, 8, and 3 were also analyzed via Western blot analysis. Furthermore, inducible nitric oxide synthase (iNOS), ionized calcium-binding adaptor molecule 1 (Iba1), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β), interleukin-1α (IL-1α), monocyte chemotactic protein 1 (MCP-1), transforming growth factor β (TGFβ), CIRBP, and RBM3 gene expressions were assessed via reverse transcription polymerase chain reaction, and TNF-α, IL-6, and IL-1β releases into the cultured supernatants were assessed via enzyme-linked immunosorbent assays (ELISA). Results Prolonged exposure to OGD resulted in increased BV-2 necrotic cell death, which was attenuated by cooling. Cooling also significantly induced cold-shock proteins CIRBP and RBM3 gene expressions, with CIRBP expression more rapidly regulated than RBM3 and translatable to significantly increased protein expression. DAMPs including HMGB-1, HSP70, and CIRBP could be detected in cultured supernatants after 6 h of OGD with CIRBP release being significantly attenuated by cooling. Exposure to OGD suppressed cytokine gene expressions of IL-1β, TNF-α, MCP-1, and TGFβ independently of temperature management, whereas cooling led to a significant increase in IL-1α gene expression after 6 h of OGD. In the reperfusion phase, TNF-α and MCP-1 gene expressions were increased, and cooling was associated with significantly lower TGFβ gene expression. Interestingly, cooled Normoxia groups had significant upregulations of microglial activation marker, Iba1, IL-1β, and TNF-α gene expressions. Conclusion BV-2 microglial cells undergo necrotic cell death resulting in DAMP release due to OGD/R-induced injury. Cooling conveyed neuroprotection in OGD/R-injury as observable in increased cell viability as well as induced gene expressions of cold shock proteins. As cooling alone resulted in both upregulation of microglial activation, expression of proinflammatory cytokines, and cold shock protein transcript and protein expression, temperature management might have ambiguous effects in sterile inflammation. However, cooling resulted in a significant decrease of extracellular CIRBP, which has recently been characterized as a novel DAMP and a potent initiator and mediator of inflammation.
Collapse
|
16
|
Inhibition of cardiac PERK signaling promotes peripartum cardiac dysfunction. Sci Rep 2021; 11:18687. [PMID: 34548576 PMCID: PMC8455649 DOI: 10.1038/s41598-021-98344-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a life-threatening heart failure occurring in the peripartum period. Although mal-angiogenesis, induced by the 16-kDa N-terminal prolactin fragment (16 K PRL), is involved in the pathogenesis, the effect of full-length prolactin (23 K PRL) is poorly understood. We transfected neonate rat cardiomyocytes with plasmids containing 23 K PRL or 16 K PRL in vitro and found that 23 K PRL, but not 16 K PRL, upregulated protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling, and hypoxia promoted this effect. During the perinatal period, cardiomyocyte-specific PERK homogenous knockout (CM-KO) mice showed PPCM phenotypes after consecutive deliveries. Downregulation of PERK or JAK/STAT signaling and upregulation of apoptosis were observed in CM-KO mouse hearts. Moreover, in bromocriptine-treated CM-KO mice, cardiac function did not improve and cardiomyocyte apoptosis was not suppressed during the peripartum period. These results demonstrate that interaction between 23 K PRL and PERK signaling is cardioprotective during the peripartum term.
Collapse
|
17
|
Research Progress of the Application of Hypothermia in the Eye. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3897168. [PMID: 33381263 PMCID: PMC7758138 DOI: 10.1155/2020/3897168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Hypothermia is widely used in the medical field to protect organs or tissues from damage. Different research fields have different explanations of the protection mechanism of hypothermia. Hypothermia is also widely used in the field of ophthalmology, for example, in the eye bank, the preservation of corneal tissue and the preservation of the eyeball. Low temperature can also be applied to some ophthalmic diseases, such as allergic conjunctivitis, retinal ischemia, and retinal hypoxia. It is used to relieve eye symptoms or reduce tissue damage. Hypothermic techniques have important applications in ophthalmic surgery, such as corneal refractive surgery, vitrectomy surgery, and ciliary body cryotherapy for end-stage glaucoma. Hypothermia can reduce the inflammation of the cornea and protect the retinal tissue. The eyeball is a complex organ, including collagen tissue of the eyeball wall and retinal nerve tissue and retinal blood vessels. The mechanism of low temperature protecting eye tissue is complicated. It is important to understand the mechanism of hypothermia and its applications in ophthalmology. This review introduces the mechanism of hypothermia and its application in the eye banks, eye diseases (allergic conjunctivitis, retinal ischemia, and hypoxia), and eye surgeries (corneal transplant surgery, corneal refractive surgery, and vitrectomy).
Collapse
|
18
|
Si W, Li Z, Huang Z, Ye S, Li X, Li Y, Kuang W, Chen D, Zhu M. RNA Binding Protein Motif 3 Inhibits Oxygen-Glucose Deprivation/Reoxygenation-Induced Apoptosis Through Promoting Stress Granules Formation in PC12 Cells and Rat Primary Cortical Neurons. Front Cell Neurosci 2020; 14:559384. [PMID: 32982696 PMCID: PMC7492797 DOI: 10.3389/fncel.2020.559384] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/13/2020] [Indexed: 11/17/2022] Open
Abstract
As a sensitive cold-shock protein, RNA binding protein motif 3 (RBM3) exhibits a neuroprotective function in the condition of brain injury. However, how RBM3 is involved in acute ischemic stroke by affecting stress granules (SGs) remains unclear. Here, we established an oxygen-glucose deprivation/reperfusion (OGD/R) model in rat primary cortical neurons and PC12 cells to explore the potential mechanism between RBM3 and SG formation in acute ischemic/reperfusion (I/R) condition. The immunofluorescence results showed that the SG formation significantly decreased in rat primary cortical neurons and PC12 cells during the reperfusion period after 6 h of OGD stimulation. The western blot results, flow cytometry analysis, and cell viability assessment showed that the RBM3 expression and ratio of cell viability significantly decreased, while the rate of apoptosis increased in PC12 cells during the reperfusion period after 6 h of OGD stimulation. Co-immunoprecipitation (Co-IP) and immunofluorescence indicated that RBM3 and GTPase-activating protein-binding protein 1 (G3BP1) colocalized cytoplasm of PC12 cells after 6 h of OGD stimulation when the SGs formation reached the highest level. Besides, overexpression and knockdown of the RBM3 were achieved via plasmid transfection and CRISPR-Cas9 technology, respectively. The results of overexpression and knockdown of RBM3 gene illustrated the pivotal role of RBM3 in affecting SG formation and apoptosis level in OGD-treated PC12 cells. In conclusion, RBM3 could combine with G3BP1 resulted in increasing stress granules generation in rat primary cortical neurons and PC12 cells after 6 h of oxygen-glucose deprivation (OGD) injury, which ultimately reduced the apoptosis in OGD-induced cells. Our study may enable a new promising target for alleviating ischemia-reperfusion injury in cells.
Collapse
Affiliation(s)
- Wenwen Si
- Shenzhen Bao'an Traditional Chinese Medicine Hospital (Group), Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhen Li
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zifeng Huang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shanyu Ye
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinrong Li
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yi Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Dongfeng Chen
- Department of Anatomy, The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meiling Zhu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
19
|
Shimizu T, Taguchi A, Higashijima Y, Takubo N, Kanki Y, Urade Y, Wada Y. PERK-Mediated Suppression of microRNAs by Sildenafil Improves Mitochondrial Dysfunction in Heart Failure. iScience 2020; 23:101410. [PMID: 32768667 PMCID: PMC7378464 DOI: 10.1016/j.isci.2020.101410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/03/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative/nitrosative stress is a major trigger of cardiac dysfunction, involving the unfolded protein response and mitochondrial dysfunction. Activation of nitric oxide-cyclic guanosine monophosphate-protein kinase G signaling by sildenafil improves cardiac mal-remodeling during pressure-overload-induced heart failure. Transcriptome analysis was conducted in failing hearts with or without sildenafil treatment. Protein kinase R-like endoplasmic reticulum (ER) kinase (PERK) downstream signaling pathways, EIF2 and NRF2, were significantly altered. Although EIF2 signaling was suppressed, NRF2 signaling was upregulated, inhibiting the maturation of miR 24-3p through EGFR-mediated Ago2 phosphorylation. To study the effect of sildenafil on these pathways, we generated cardiac-specific PERK knockout mice. In these mice, sildenafil could not inhibit the maturations, the nuclear translocation of NRF2 was suppressed, and mitochondrial dysfunction advanced. Altogether, these results show that PERK-mediated suppression of miRNAs by sildenafil is vital for maintaining mitochondrial homeostasis through NRF2-mediated oxidative stress response.
Collapse
Affiliation(s)
- Takashi Shimizu
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Akashi Taguchi
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yoshiki Higashijima
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; Department of Bioinformational Pharmacology, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Naoko Takubo
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yasuharu Kanki
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Yoshihiro Urade
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan
| |
Collapse
|
20
|
Liu B, Cao Y, Shi F, Wang L, Li N, Cheng X, Du J, Tian Q, Zhou X. The overexpression of RBM3 alleviates TBI-induced behaviour impairment and AD-like tauopathy in mice. J Cell Mol Med 2020; 24:9176-9188. [PMID: 32648620 PMCID: PMC7417709 DOI: 10.1111/jcmm.15555] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The therapeutic hypothermia is an effective tool for TBI‐associated brain impairment, but its side effects limit in clinical routine use. Hypothermia up‐regulates RNA‐binding motif protein 3 (RBM3), which is verified to protect synaptic plasticity. Here, we found that cognitive and LTP deficits, loss of spines, AD‐like tau pathologies are displayed one month after TBI in mice. In contrast, the deficits of LTP and cognitive, loss of spines and tau abnormal phosphorylation at several sites are obviously reversed in TBI mice combined with hypothermia pre‐treatment (HT). But, the neuroprotective role of HT disappears in TBI mouse models under condition of blocking RBM3 expression with RBM3 shRNA. In other hand, overexpressing RBM3 by AAV‐RBM3 plasmid can mimic HT‐like neuroprotection against TBI‐induced chronic brain injuries, such as improving LTP and cognitive, loss of spines and tau hyperphosphorylation in TBI mouse models. Taken together, hypothermia pre‐treatment reverses TBI‐induced chronic AD‐like pathology and behaviour deficits in RBM3 expression dependent manner, RBM3 may be a potential target for neurodegeneration diseases including Alzheimer disease.
Collapse
Affiliation(s)
- Bingjin Liu
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou, China
| | - Yun Cao
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangxiao Shi
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Li
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangshu Cheng
- Department of Neurology, Center for Translational Medicine, Huaihe Hospital of Henan University, Kaifeng, China
| | - Jin Du
- Department of Neurology, Center for Translational Medicine, Huaihe Hospital of Henan University, Kaifeng, China
| | - Qing Tian
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwen Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Ávila-Gómez P, Vieites-Prado A, Dopico-López A, Bashir S, Fernández-Susavila H, Gubern C, Pérez-Mato M, Correa-Paz C, Iglesias-Rey R, Sobrino T, Bustamante A, Wellmann S, Montaner J, Serena J, Castillo J, Hervella P, Campos F. Cold stress protein RBM3 responds to hypothermia and is associated with good stroke outcome. Brain Commun 2020; 2:fcaa078. [PMID: 33585816 PMCID: PMC7869850 DOI: 10.1093/braincomms/fcaa078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
RNA-binding motif protein 3 is a molecular marker of hypothermia that has proved neuroprotective in neurodegenerative disease models. However, its relationship to the well-recognized therapeutic effect of hypothermia in ischaemic stroke had not been studied. In this work, the expression of RNA-binding motif protein 3 was investigated in ischaemic animal models subjected to systemic and focal brain hypothermia, specifically the effects of RNA-binding motif protein 3 silencing and overexpression on ischaemic lesions. Moreover, the association of RNA-binding motif protein 3 levels with body temperature and clinical outcome was evaluated in two independent cohorts of acute ischaemic stroke patients (n = 215); these levels were also determined in a third cohort of 31 patients derived from the phase III EuroHYP-1 trial of therapeutic cooling in ischaemic stroke. The preclinical data confirmed the increase of brain RNA-binding motif protein 3 levels in ischaemic animals subjected to systemic and focal hypothermia; this increase was selectively higher in the cooled hemisphere of animals undergoing focal brain hypothermia, thus confirming the direct effect of hypothermia on RNA-binding motif protein 3 expression, while RNA-binding motif protein 3 up-regulation in ischaemic brain regions led to functional recovery. Clinically, patients with body temperature <37.5°C in the first two cohorts had higher RNA-binding motif protein 3 values at 24 h and good outcome at 3 months post-ischaemic stroke, while RNA-binding motif protein 3 levels in the cooled third cohort tended to exceed those in placebo-treated patients. These results make RNA-binding motif protein 3 a molecular marker associated with the effect of hypothermia in ischaemic stroke and suggest its potential application as a promising protective target.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alba Vieites-Prado
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Saima Bashir
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - Héctor Fernández-Susavila
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Carme Gubern
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - María Pérez-Mato
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain.,Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Hospital La Paz Institute for Health Research (IdiPAZ), Autonomous University of Madrid, 28046, Madrid, Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Regensburg (KUNO), University of Regensburg, 93049 Regensburg, Germany
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Joaquín Serena
- Stroke Unit, Department of Neurology, Hospital Universitari Dr. Josep Trueta de Girona, IDIBGI, Girona 17007, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Santiago de Compostela 15706, Spain
| |
Collapse
|
22
|
Rosenthal LM, Leithner C, Tong G, Streitberger KJ, Krech J, Storm C, Schmitt KRL. RBM3 and CIRP expressions in targeted temperature management treated cardiac arrest patients-A prospective single center study. PLoS One 2019; 14:e0226005. [PMID: 31821351 PMCID: PMC6903712 DOI: 10.1371/journal.pone.0226005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/17/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Management of cardiac arrest patients includes active body temperature control and strict prevention of fever to avoid further neurological damage. Cold-shock proteins RNA-binding motif 3 (RBM3) and cold inducible RNA-binding protein (CIRP) expressions are induced in vitro in response to hypothermia and play a key role in hypothermia-induced neuroprotection. OBJECTIVE To measure gene expressions of RBM3, CIRP, and inflammatory biomarkers in whole blood samples from targeted temperature management (TTM)-treated post-cardiac arrest patients for the potential application as clinical biomarkers for the efficacy of TTM treatment. METHODS A prospective single center trial with the inclusion of 22 cardiac arrest patients who were treated with TTM (33°C for 24 hours) after ROSC was performed. RBM3, CIRP, interleukin 6 (IL-6), monocyte chemotactic protein 1 (MCP-1), and inducible nitric oxide synthase (iNOS) mRNA expressions were quantified by RT-qPCR. Serum RBM3 protein concentration was quantified using an enzyme-linked immunosorbent assay (ELISA). RESULTS RBM3 mRNA expression was significantly induced in post-cardiac arrest patients in response to TTM. RBM3 mRNA was increased 2.2-fold compared to before TTM. A similar expression kinetic of 1.4-fold increase was observed for CIRP mRNA, but did not reached significancy. Serum RBM3 protein was not increased in response to TTM. IL-6 and MCP-1 expression peaked after ROSC and then significantly decreased. iNOS expression was significantly increased 24h after return of spontaneous circulation (ROSC) and TTM. CONCLUSIONS RBM3 is temperature regulated in patients treated with TTM after CA and ROSC. RBM3 is a possible biomarker candidate to ensure the efficacy of TTM treatment in post-cardiac arrest patients and its pharmacological induction could be a potential future intervention strategy that warrants further research.
Collapse
Affiliation(s)
- Lisa-Maria Rosenthal
- Dept. for Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Christoph Leithner
- Dept. of Neurology, Charité Universtitätsmedizin Berlin, Berlin, Germany
| | - Giang Tong
- Dept. for Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Kaspar Josche Streitberger
- Berlin Institute of Health, Berlin, Germany
- Dept. of Neurology, Charité Universtitätsmedizin Berlin, Berlin, Germany
| | - Jana Krech
- Dept. for Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Christian Storm
- Dept. of Internal Medicine, Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Rose Luise Schmitt
- Dept. for Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
- Dept. for Pediatric Cardiology, Charité Universitätsmedizin Berlin, Berlin, Germany
- DHZK (German Centre for Cardiovascular Research), Berlin, Germany
- * E-mail:
| |
Collapse
|
23
|
RBM3 promotes neurogenesis in a niche-dependent manner via IMP2-IGF2 signaling pathway after hypoxic-ischemic brain injury. Nat Commun 2019; 10:3983. [PMID: 31484925 PMCID: PMC6726629 DOI: 10.1038/s41467-019-11870-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Hypoxic ischemia (HI) is an acute brain threat across all age groups. Therapeutic hypothermia ameliorates resulting injury in neonates but its side effects prevent routine use in adults. Hypothermia up-regulates a small protein subset that includes RNA-binding motif protein 3 (RBM3), which is neuroprotective under stressful conditions. Here we show how RBM3 stimulates neuronal differentiation and inhibits HI-induced apoptosis in the two areas of persistent adult neurogenesis, the subventricular zone (SVZ) and the subgranular zone (SGZ), while promoting neural stem/progenitor cell (NSPC) proliferation after HI injury only in the SGZ. RBM3 interacts with IGF2 mRNA binding protein 2 (IMP2), elevates its expression and thereby stimulates IGF2 release in SGZ but not SVZ-NSPCs. In summary, we describe niche-dependent regulation of neurogenesis after adult HI injury via the novel RBM3-IMP2-IGF2 signaling pathway. Therapeutic hypothermia is a potent tool in the treatment of neonatal hypoxic-ischemic (HI) injury, yet the underlying mechanisms remain unclear. Here, authors demonstrate how the RNA-binding motif protein RBM3, which is induced by mild cooling while global translation rate is slowed down, contributes substantially to neuroregeneration after adult HI injury, specifically in the subventricular zone and subgranular zone.
Collapse
|
24
|
Sun YJ, Zhang ZY, Fan B, Li GY. Neuroprotection by Therapeutic Hypothermia. Front Neurosci 2019; 13:586. [PMID: 31244597 PMCID: PMC6579927 DOI: 10.3389/fnins.2019.00586] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
Hypothermia therapy is an old and important method of neuroprotection. Until now, many neurological diseases such as stroke, traumatic brain injury, intracranial pressure elevation, subarachnoid hemorrhage, spinal cord injury, hepatic encephalopathy, and neonatal peripartum encephalopathy have proven to be suppressed by therapeutic hypothermia. Beneficial effects of therapeutic hypothermia have also been discovered, and progress has been made toward improving the benefits of therapeutic hypothermia further through combination with other neuroprotective treatments and by probing the mechanism of hypothermia neuroprotection. In this review, we compare different hypothermia induction methods and provide a summarized account of the synergistic effect of hypothermia therapy with other neuroprotective treatments, along with an overview of hypothermia neuroprotection mechanisms and cold/hypothermia-induced proteins.
Collapse
Affiliation(s)
- Ying-Jian Sun
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Zi-Yuan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Bin Fan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Guang-Yu Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Shi H, Yao R, Lian S, Liu P, Liu Y, Yang YY, Yang H, Li S. Regulating glycolysis, the TLR4 signal pathway and expression of RBM3 in mouse liver in response to acute cold exposure. Stress 2019; 22:366-376. [PMID: 30821572 DOI: 10.1080/10253890.2019.1568987] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
At low temperatures, the liver increases glucose utilization and expresses RNA-binding motif 3 (RBM3) to cope with cold exposure. In this study, the expression of heat shock protein 70 (HSP70), Toll-like receptor 4 (TLR4), bone marrow differentiation factor 88 (MYD88), and phosphorylated nuclear factor-κB (NF-κB) was consistent with fluctuations in insulin in fasted cold-exposed mice. We also found up-regulation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in acute cold exposure with a decrease in core body temperature. RBM3 transcription and translation were activated 2 h after cold exposure. The anti-apoptotic factor Bcl-2/Bax ratio also increased, while expression of apoptosis factors: cleaved caspase-3, cleaved poly(ADP-ribose)polymerase 1 (PARP-1) and cytochrome-c (Cyt-c) was unchanged. Liver glycogen was depleted after 2 h of cold exposure, and blood glucose decreased after 4 h. Glycogen synthase kinase 3β (GSK3β) phosphorylation continued to increase to promote hepatic glycogen synthesis. We found a high level of protein kinase B (AKT) phosphorylation after 6 h of cold exposure. In addition, we demonstrated that after cold exposure for 2 h, in the liver, continued phosphorylation of fructose-2,6-diphosphate (PFKFB2) and decreased accumulation of glycogen intermediates fructose-1,6-diphosphate (FDP) and pyruvic acid (PA). In summary, the liver responds to cold exposure through a number of different pathways, including activation of HSP70/TLR4 signaling pathways, up-regulation of RBM3 expression, and increased glycolysis and glycogen synthesis. We propose a possible signaling pathway in which regulation of RBM3 expression by the liver affects the AKT metabolic signaling pathway. Lay summary In response to changes in ambient temperature, mice regulate global metabolism and gene expression through hormones. This study focused on the effects of environmental hypothermia on molecular pathways of glucose metabolism in the liver, which is the important metabolic organ in mice. This provides a basis for further study of mice against cold exposure damage.
Collapse
Affiliation(s)
- Hongzhao Shi
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Ruizhi Yao
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Shuai Lian
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Peng Liu
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Yang Liu
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Yu Ying Yang
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Huanmin Yang
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| | - Shize Li
- a College of Animal Science and Veterinary Medicine , Heilongjiang Bayi Agricultural University , Daqing , PR China
| |
Collapse
|
26
|
Nie M, Tan X, Lu Y, Wu Z, Li J, Xu D, Zhang P, You F. Network of microRNA-transcriptional factor-mRNA in cold response of turbot Scophthalmus maximus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:583-597. [PMID: 30790148 DOI: 10.1007/s10695-019-00611-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 01/21/2019] [Indexed: 05/19/2023]
Abstract
The aim of this study is to understand fish cold-tolerant mechanism. We analyzed the transcriptional reactions to the cold condition in turbot Scophthalmus maximus by using RNA-seq and microRNA (miRNA)-seq. Meio-gynogenetic diploid turbots were treated at 0 °C to distinguish the cold-tolerant (CT) and cold-sensitive (CS) groups. The results showed that there were quite different responses at both mRNA and miRNA levels, with more up-regulated mRNAs (1069 vs. 194) and less down-regulated miRNAs (4 vs. 1) in CT versus CS relative to the control group. The network of miRNA-transcription factor-mRNA, regulating turbot different response to cold stress, was constructed, which involved in cell cycle, component of cell membrane, signal transduction, and circadian rhythm pathways. The above information demonstrates mechanisms by which cold tolerance is increased in fish.
Collapse
Affiliation(s)
- Miaomiao Nie
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 10049, People's Republic of China
| | - Xungang Tan
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
| | - Yunliang Lu
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
| | - Zhihao Wu
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
| | - Jun Li
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
| | - Dongdong Xu
- Key Lab of Mariculture and Enhancement of Zhejiang Province, Marine Fishery Institute of Zhejiang Province, Zhoushan, 316100, Zhejiang Province, People's Republic of China
| | - Peijun Zhang
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China
| | - Feng You
- CAS Key Laboratory of Experimental Marine Biology, National and Local Joint Engineering Laboratory of Ecological Mariculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, People's Republic of China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, People's Republic of China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
27
|
Jackson TC, Kochanek PM. A New Vision for Therapeutic Hypothermia in the Era of Targeted Temperature Management: A Speculative Synthesis. Ther Hypothermia Temp Manag 2019; 9:13-47. [PMID: 30802174 PMCID: PMC6434603 DOI: 10.1089/ther.2019.0001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Three decades of animal studies have reproducibly shown that hypothermia is profoundly cerebroprotective during or after a central nervous system (CNS) insult. The success of hypothermia in preclinical acute brain injury has not only fostered continued interest in research on the classic secondary injury mechanisms that are prevented or blunted by hypothermia but has also sparked a surge of new interest in elucidating beneficial signaling molecules that are increased by cooling. Ironically, while research into cold-induced neuroprotection is enjoying newfound interest in chronic neurodegenerative disease, conversely, the scope of the utility of therapeutic hypothermia (TH) across the field of acute brain injury is somewhat controversial and remains to be fully defined. This has led to the era of Targeted Temperature Management, which emphasizes a wider range of temperatures (33–36°C) showing benefit in acute brain injury. In this comprehensive review, we focus on our current understandings of the novel neuroprotective mechanisms activated by TH, and discuss the critical importance of developmental age germane to its clinical efficacy. We review emerging data on four cold stress hormones and three cold shock proteins that have generated new interest in hypothermia in the field of CNS injury, to create a framework for new frontiers in TH research. We make the case that further elucidation of novel cold responsive pathways might lead to major breakthroughs in the treatment of acute brain injury, chronic neurological diseases, and have broad potential implications for medicines of the distant future, including scenarios such as the prevention of adverse effects of long-duration spaceflight, among others. Finally, we introduce several new phrases that readily summarize the essence of the major concepts outlined by this review—namely, Ultramild Hypothermia, the “Responsivity of Cold Stress Pathways,” and “Hypothermia in a Syringe.”
Collapse
Affiliation(s)
- Travis C Jackson
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 1 John G. Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Safar Center for Resuscitation Research, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Thorne JL, Battaglia S, Baxter DE, Hayes JL, Hutchinson SA, Jana S, Millican-Slater RA, Smith L, Teske MC, Wastall LM, Hughes TA. MiR-19b non-canonical binding is directed by HuR and confers chemosensitivity through regulation of P-glycoprotein in breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:996-1006. [DOI: 10.1016/j.bbagrm.2018.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/09/2018] [Accepted: 08/23/2018] [Indexed: 12/25/2022]
|
29
|
Van Pelt DW, Confides AL, Judge AR, Vanderklish PW, Dupont-Versteegden EE. Cold shock protein RBM3 attenuates atrophy and induces hypertrophy in skeletal muscle. J Muscle Res Cell Motil 2018; 39:35-40. [PMID: 30051360 DOI: 10.1007/s10974-018-9496-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/23/2018] [Indexed: 12/22/2022]
Abstract
RNA-binding motif protein 3 (RBM3), a stress-inducible RNA-binding protein that increases protein synthesis and confers cell protection in multiple cell types, has been identified as a possible regulator of skeletal muscle mass. Therefore, the primary aim of this study was to examine the impact of elevated RBM3 on skeletal muscle hypertrophy and resistance to atrophy. Plasmid-mediated overexpression of RBM3 in vitro and in vivo was used to assess the role of RBM3 in muscle. C2C12 myotubes overexpressing RBM3 were approximately 1.6 times larger than non-transfected myotubes, suggesting a role for RBM3 in hypertrophy. In addition, elevated RBM3 attenuated atrophy in myotubes exposed to dexamethasone. In agreement with in vitro results, overexpression of RBM3 in soleus muscle of F344/BN rats using electroporation techniques increased the cross sectional area of muscle fibers. Overexpression of RBM3 also attenuated muscle atrophy in rat soleus muscle undergoing disuse atrophy. These findings provide direct evidence for a novel role of RBM3 in inducing hypertrophy as well as attenuating atrophy.
Collapse
Affiliation(s)
- Douglas W Van Pelt
- Department Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S. Limestone, RM 210E, Lexington, KY, 40536-0200, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, 40536-0200, USA
| | - Amy L Confides
- Department Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S. Limestone, RM 210E, Lexington, KY, 40536-0200, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, 40536-0200, USA
| | - Andrew R Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610-0154, USA
| | - Peter W Vanderklish
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Esther E Dupont-Versteegden
- Department Rehabilitation Sciences, College of Health Sciences, University of Kentucky, 900 S. Limestone, RM 210E, Lexington, KY, 40536-0200, USA. .,Center for Muscle Biology, University of Kentucky, Lexington, KY, 40536-0200, USA.
| |
Collapse
|
30
|
Zhao L, Zou T, Gomez NA, Wang B, Zhu MJ, Du M. Raspberry alleviates obesity-induced inflammation and insulin resistance in skeletal muscle through activation of AMP-activated protein kinase (AMPK) α1. Nutr Diabetes 2018; 8:39. [PMID: 29961765 PMCID: PMC6026595 DOI: 10.1038/s41387-018-0049-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/23/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Through dynamic means, etiological factors, including chronic inflammation and insulin resistance have the potential to perpetuate metabolic incidences such as type 2 diabetes and obesity. Abatement of such syndromes can be achieved by complex mechanisms initiated through bioactive compounds such as polyphenols derived from fruits. Using a whole-fruit approach, the effects of dietary red raspberry, which is rich in polyphenols, on inflammatory responses and insulin resistance in the skeletal muscles of Mus musculus were studied along with the potential role of AMP-activated protein kinase (AMPK) to act as a key mediator. SUBJECTS Wild-type (WT) mice and mice deficient in the catalytic subunit (α1) of AMPK (AMPKα1-/-) were fed with a high-fat diet (HFD) or HFD supplemented with raspberry (5% dry weight) for 10 weeks. Factors involved in inflammatory responses, insulin signaling transduction, and mitochondrial biogenesis were evaluated. RESULTS Dietary raspberry reduced ectopic lipid storage, alleviated inflammation responses, improved whole-body insulin sensitivity, and promoted mitochondrial biogenesis in the skeletal muscle of WT mice, but not AMPKα1-/- mice. CONCLUSIONS AMPKα1 is an important mediator for the beneficial effects of raspberry through alleviating inflammatory responses and sensitizing insulin signaling in skeletal muscle of HFD-fed mice.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Animal Sciences, Nutrigenomics and Growth Biology laboratory, Washington State University, Pullman, WA, 99164, USA
| | - Tiande Zou
- Department of Animal Sciences, Nutrigenomics and Growth Biology laboratory, Washington State University, Pullman, WA, 99164, USA
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, 330045, China
| | - Noe Alberto Gomez
- Department of Animal Sciences, Nutrigenomics and Growth Biology laboratory, Washington State University, Pullman, WA, 99164, USA
| | - Bo Wang
- Department of Animal Sciences, Nutrigenomics and Growth Biology laboratory, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Min Du
- Department of Animal Sciences, Nutrigenomics and Growth Biology laboratory, Washington State University, Pullman, WA, 99164, USA.
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, 100194, China.
| |
Collapse
|
31
|
Lian S, Wang D, Xu B, Guo W, Wang L, Li W, Ji H, Wang J, Kong F, Zhen L, Li S, Zhang L, Guo J, Yang H. Prenatal cold stress: Effect on maternal hippocampus and offspring behavior in rats. Behav Brain Res 2018; 346:1-10. [DOI: 10.1016/j.bbr.2018.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 01/08/2023]
|
32
|
Morphoregulatory functions of the RNA-binding motif protein 3 in cell spreading, polarity and migration. Sci Rep 2018; 8:7367. [PMID: 29743635 PMCID: PMC5943363 DOI: 10.1038/s41598-018-25668-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/27/2018] [Indexed: 02/01/2023] Open
Abstract
RNA-binding proteins are emerging as key regulators of transitions in cell morphology. The RNA-binding motif protein 3 (RBM3) is a cold-inducible RNA-binding protein with broadly relevant roles in cellular protection, and putative functions in cancer and development. Several findings suggest that RBM3 has morphoregulatory functions germane to its roles in these contexts. For example, RBM3 helps maintain the morphological integrity of cell protrusions during cell stress and disease. Moreover, it is highly expressed in migrating neurons of the developing brain and in cancer invadopodia, suggesting roles in migration. We here show that RBM3 regulates cell polarity, spreading and migration. RBM3 was present in spreading initiation centers, filopodia and blebs that formed during cell spreading in cell lines and primary myoblasts. Reducing RBM3 triggered exaggerated spreading, increased RhoA expression, and a loss of polarity that was rescued by Rho kinase inhibition and overexpression of CRMP2. High RBM3 expression enhanced the motility of cells migrating by a mesenchymal mode involving extension of long protrusions, whereas RBM3 knockdown slowed migration, greatly reducing the ability of cells to extend protrusions and impairing multiple processes that require directional migration. These data establish novel functions of RBM3 of potential significance to tissue repair, metastasis and development.
Collapse
|
33
|
Blagden S, Abdel Mouti M, Chettle J. Ancient and modern: hints of a core post-transcriptional network driving chemotherapy resistance in ovarian cancer. WILEY INTERDISCIPLINARY REVIEWS. RNA 2018; 9:e1432. [PMID: 28762650 PMCID: PMC5763387 DOI: 10.1002/wrna.1432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/12/2017] [Accepted: 06/12/2017] [Indexed: 01/04/2023]
Abstract
RNA-binding proteins (RBPs) and noncoding (nc)RNAs (such as microRNAs, long ncRNAs, and others) cooperate within a post-transcriptional network to regulate the expression of genes required for many aspects of cancer behavior including its sensitivity to chemotherapy. Here, using an RBP-centric approach, we explore the current knowledge surrounding contributers to post-transcriptional gene regulation (PTGR) in ovarian cancer and identify commonalities that hint at the existence of an evolutionarily conserved core PTGR network. This network regulates survival and chemotherapy resistance in the contemporary context of the cancer cell. There is emerging evidence that cancers become dependent on PTGR factors for their survival. Further understanding of this network may identify innovative therapeutic targets as well as yield crucial insights into the hard-wiring of many malignancies, including ovarian cancer. WIREs RNA 2018, 9:e1432. doi: 10.1002/wrna.1432 This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications Translation > Translation Mechanisms RNA in Disease and Development > RNA in Disease.
Collapse
|
34
|
Hetz C, Papa FR. The Unfolded Protein Response and Cell Fate Control. Mol Cell 2018; 69:169-181. [DOI: 10.1016/j.molcel.2017.06.017] [Citation(s) in RCA: 744] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/08/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
|
35
|
Zhou RB, Lu XL, Zhang CY, Yin DC. RNA binding motif protein 3: a potential biomarker in cancer and therapeutic target in neuroprotection. Oncotarget 2017; 8:22235-22250. [PMID: 28118608 PMCID: PMC5400660 DOI: 10.18632/oncotarget.14755] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022] Open
Abstract
RNA binding motif 3 (RBM3) is a highly conserved cold-induced RNA binding protein that is transcriptionally up-regulated in response to harsh stresses. Featured as RNA binding protein, RBM3 is involved in mRNA biogenesis as well as stimulating protein synthesis, promoting proliferation and exerting anti-apoptotic functions. Nowadays, accumulating immunohistochemically studies have suggested RBM3 function as a proto-oncogene that is associated with tumor progression and metastasis in various cancers. Moreover, emerging evidences have also indicated that RBM3 is equally effective in neuroprotection. In the present review, we provide an overview of current knowledge concerning the role of RBM3 in various cancers and neuroprotection. Additionally, its potential roles as a promising diagnostic marker for cancer and a possible therapeutic target for neuro-related diseases are discussed.
Collapse
Affiliation(s)
- Ren-Bin Zhou
- Key Laboratory for Space Bioscience & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| | - Xiao-Li Lu
- Key Laboratory for Space Bioscience & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| | - Chen-Yan Zhang
- Key Laboratory for Space Bioscience & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience & Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| |
Collapse
|
36
|
Mammalian ECD Protein Is a Novel Negative Regulator of the PERK Arm of the Unfolded Protein Response. Mol Cell Biol 2017; 37:MCB.00030-17. [PMID: 28652267 PMCID: PMC5574048 DOI: 10.1128/mcb.00030-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/17/2017] [Indexed: 01/01/2023] Open
Abstract
Mammalian Ecdysoneless (ECD) is a highly conserved ortholog of the DrosophilaEcd gene product whose mutations impair the synthesis of Ecdysone and produce cell-autonomous survival defects, but the mechanisms by which ECD functions are largely unknown. Here we present evidence that ECD regulates the endoplasmic reticulum (ER) stress response. ER stress induction led to a reduced ECD protein level, but this effect was not seen in PKR-like ER kinase knockout (PERK-KO) or phosphodeficient eukaryotic translation initiation factor 2α (eIF2α) mouse embryonic fibroblasts (MEFs); moreover, ECD mRNA levels were increased, suggesting impaired ECD translation as the mechanism for reduced protein levels. ECD colocalizes and coimmunoprecipitates with PERK and GRP78. ECD depletion increased the levels of both phospho-PERK (p-PERK) and p-eIF2α, and these effects were enhanced upon ER stress induction. Reciprocally, overexpression of ECD led to marked decreases in p-PERK, p-eIF2α, and ATF4 levels but robust increases in GRP78 protein levels. However, GRP78 mRNA levels were unchanged, suggesting a posttranscriptional event. Knockdown of GRP78 reversed the attenuating effect of ECD overexpression on PERK signaling. Significantly, overexpression of ECD provided a survival advantage to cells upon ER stress induction. Taken together, our data demonstrate that ECD promotes survival upon ER stress by increasing GRP78 protein levels to enhance the adaptive folding protein in the ER to attenuate PERK signaling.
Collapse
|
37
|
Cold-Inducible Protein RBM3 Protects UV Irradiation-Induced Apoptosis in Neuroblastoma Cells by Affecting p38 and JNK Pathways and Bcl2 Family Proteins. J Mol Neurosci 2017; 63:142-151. [DOI: 10.1007/s12031-017-0964-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/10/2017] [Indexed: 12/24/2022]
|
38
|
Papadima EM, Niola P, Melis C, Pisanu C, Congiu D, Cruceanu C, Lopez JP, Turecki G, Ardau R, Severino G, Chillotti C, Del Zompo M, Squassina A. Evidence towards RNA Binding Motif (RNP1, RRM) Protein 3 (RBM3) as a Potential Biomarker of Lithium Response in Bipolar Disorder Patients. J Mol Neurosci 2017; 62:304-308. [PMID: 28616776 DOI: 10.1007/s12031-017-0938-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/06/2017] [Indexed: 01/05/2023]
Abstract
Lithium has been used for more than six decades for the management of bipolar disorder (BD). In a previous transcriptomic study, we showed that patients affected by either BD or cluster headache, both disorders characterized by circadian disturbances and response to lithium in a subgroup of patients, have higher expression of the RNA binding motif (RNP1, RRM) protein 3 (RBM3) gene compared to controls. To investigate whether RBM3 could represent a biomarker of lithium response, we screened raw microarray expression data from lymphoblastoid cell lines (LCLs) derived from 20 BD patients, responders or non-responders to lithium. RBM3 was the most significantly differentially expressed gene in the list, being overexpressed in responders compared to non-responders (fold change = 2.0; p = 1.5 × 10-16). We therefore sought to validate the microarray finding by quantitative reverse transcription polymerase chain reaction (RT-qPCR) and explore whether RBM3 expression was modulated by lithium treatment in vitro in LCLs as well as in human-derived neural progenitor cells (NPCs). Our findings confirmed the higher expression of RBM3 in responders compared to non-responders (fold change = 3.78; p = 0.0002). Lithium did not change RBM3 expression in LCLs in any of the groups, but it increased its expression in NPCs. While preliminary, our data suggest that higher levels of RBM3 might be required for better lithium response and that the expression of this gene could be modulated by lithium in a tissue-specific manner.
Collapse
Affiliation(s)
- Eleni Merkouri Papadima
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Paola Niola
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Carla Melis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Cristiana Cruceanu
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Juan Pablo Lopez
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Raffaella Ardau
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Maria Del Zompo
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy.,Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, School of Medicine, University of Cagliari, sp 6, 09042, Cagliari, Italy.
| |
Collapse
|
39
|
Zhao Q, Liu C, Shen X, Xiao L, Wang H, Liu P, Wang L, Xu H. Cytoprotective effects of myristicin against hypoxia‑induced apoptosis and endoplasmic reticulum stress in rat dorsal root ganglion neurons. Mol Med Rep 2017; 15:2280-2288. [PMID: 28260107 DOI: 10.3892/mmr.2017.6258] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/25/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the role of myristicin (Myr; 1‑allyl‑5‑methoxy‑3,4‑methylenedioxybenzene), an active aromatic compound isolated from nutmeg, carrot, basil, cinnamon and parsley, in hypoxia‑induced apoptosis in rat dorsal root ganglion (DRG) neurons. It was observed that Myr significantly enhanced cell viability in hypoxia‑induced DRG neurons in a dose‑dependent manner; the optimal concentration of Myr was 50 µM. Furthermore, Myr reduced the percentage of deoxynucleotidyl transferase‑mediated dUTP nick end‑labeling‑positive neuronal cells and influenced the expression of the pro‑apoptotic gene B‑cell lymphoma 2 (Bcl‑2) associated X protein, the apoptosis protease cleaved caspase‑3 and the anti‑apoptotic gene Bcl‑2, in the hypoxia‑induced group. In addition, Myr protected against hypoxic injury in DRG neurons by inhibiting malondialdehyde and lactate dehydrogenase, however upregulating superoxide dismutase and glutathione peroxidase. Myr reduced the expression of endoplasmic reticulum stress (ERS) markers, including CCAAT/enhancer‑binding protein‑homologous protein, glucose‑related protein 78 and cleaved caspase‑12 in the hypoxia‑induced group. To the best of our knowledge, this is the first demonstration of the activity of Myr against hypoxia‑induced apoptosis in rat DRG neurons via inhibition of the ERS pathway.
Collapse
Affiliation(s)
- Quanlai Zhao
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Chen Liu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Xiang Shen
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Liang Xiao
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Hong Wang
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Ping Liu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Lingting Wang
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Hongguang Xu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| |
Collapse
|
40
|
Zhu X, Zelmer A, Wellmann S. Visualization of Protein-protein Interaction in Nuclear and Cytoplasmic Fractions by Co-immunoprecipitation and In Situ Proximity Ligation Assay. J Vis Exp 2017. [PMID: 28117799 DOI: 10.3791/55218] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein-protein interactions are involved in thousands of cellular processes and occur in distinct spatial context. Traditionally, co-immunoprecipitation is a popular technique to detect protein-protein interactions. Subsequent Western blot analysis is the most common method to visualize co-immunoprecipitated proteins. Recently, the proximity ligation assay has become a powerful tool to visualize protein-protein interactions in situ and provides the possibility to quantify protein-protein interactions by this method. Similar to conventional immunocytochemistry, the proximity ligation assay technique is also based on the accessibility of primary antibodies to the antigens, but in contrast, proximity ligation assay detects protein-protein interactions with a unique technique involving rolling-circle PCR, while conventional immunocytochemistry only shows co-localization of proteins. Nuclear factor 90 (NF90) and RNA-binding motif protein 3 (RBM3) have been previously demonstrated as interacting partners. They are predominantly localized in the nucleus, but also migrate into the cytoplasm and regulate signaling pathways in the cytoplasmic compartment. Here, we compared NF90-RBM3 interaction in both the nucleus and the cytoplasm by co-immunoprecipitation and proximity ligation assay. In addition, we discussed the advantages and limitations of these two techniques in visualizing protein-protein interactions in respect to spatial distribution and the properties of protein-protein interactions.
Collapse
Affiliation(s)
- Xinzhou Zhu
- University Children's Hospital Basel (UKBB), University of Basel;
| | - Andrea Zelmer
- University Children's Hospital Basel (UKBB), University of Basel
| | - Sven Wellmann
- University Children's Hospital Basel (UKBB), University of Basel; Department of Clinical Research, University of Basel
| |
Collapse
|
41
|
Zhu X, Bührer C, Wellmann S. Cold-inducible proteins CIRP and RBM3, a unique couple with activities far beyond the cold. Cell Mol Life Sci 2016; 73:3839-59. [PMID: 27147467 PMCID: PMC5021741 DOI: 10.1007/s00018-016-2253-7] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/22/2016] [Accepted: 04/26/2016] [Indexed: 12/21/2022]
Abstract
Cold-inducible RNA-binding protein (CIRP) and RNA-binding motif protein 3 (RBM3) are two evolutionarily conserved RNA-binding proteins that are transcriptionally upregulated in response to low temperature. Featuring an RNA-recognition motif (RRM) and an arginine-glycine-rich (RGG) domain, these proteins display many similarities and specific disparities in the regulation of numerous molecular and cellular events. The resistance to serum withdrawal, endoplasmic reticulum stress, or other harsh conditions conferred by RBM3 has led to its reputation as a survival gene. Once CIRP protein is released from cells, it appears to bolster inflammation, contributing to poor prognosis in septic patients. A variety of human tumor specimens have been analyzed for CIRP and RBM3 expression. Surprisingly, RBM3 expression was primarily found to be positively associated with the survival of chemotherapy-treated patients, while CIRP expression was inversely linked to patient survival. In this comprehensive review, we summarize the evolutionary conservation of CIRP and RBM3 across species as well as their molecular interactions, cellular functions, and roles in diverse physiological and pathological processes, including circadian rhythm, inflammation, neural plasticity, stem cell properties, and cancer development.
Collapse
Affiliation(s)
- Xinzhou Zhu
- University Children's Hospital Basel (UKBB), Spitalstrasse 33, 4056, Basel, Switzerland
| | - Christoph Bührer
- Department of Neonatology, Charité University Medical Center, Berlin, Germany
| | - Sven Wellmann
- University Children's Hospital Basel (UKBB), Spitalstrasse 33, 4056, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
42
|
Mollereau B, Rzechorzek NM, Roussel BD, Sedru M, Van den Brink DM, Bailly-Maitre B, Palladino F, Medinas DB, Domingos PM, Hunot S, Chandran S, Birman S, Baron T, Vivien D, Duarte CB, Ryoo HD, Steller H, Urano F, Chevet E, Kroemer G, Ciechanover A, Calabrese EJ, Kaufman RJ, Hetz C. Adaptive preconditioning in neurological diseases - therapeutic insights from proteostatic perturbations. Brain Res 2016; 1648:603-616. [PMID: 26923166 PMCID: PMC5010532 DOI: 10.1016/j.brainres.2016.02.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 02/06/2023]
Abstract
In neurological disorders, both acute and chronic neural stress can disrupt cellular proteostasis, resulting in the generation of pathological protein. However in most cases, neurons adapt to these proteostatic perturbations by activating a range of cellular protective and repair responses, thus maintaining cell function. These interconnected adaptive mechanisms comprise a 'proteostasis network' and include the unfolded protein response, the ubiquitin proteasome system and autophagy. Interestingly, several recent studies have shown that these adaptive responses can be stimulated by preconditioning treatments, which confer resistance to a subsequent toxic challenge - the phenomenon known as hormesis. In this review we discuss the impact of adaptive stress responses stimulated in diverse human neuropathologies including Parkinson׳s disease, Wolfram syndrome, brain ischemia, and brain cancer. Further, we examine how these responses and the molecular pathways they recruit might be exploited for therapeutic gain. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- B Mollereau
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France.
| | - N M Rzechorzek
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, United Kingdom
| | - B D Roussel
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - M Sedru
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D M Van den Brink
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - B Bailly-Maitre
- INSERM U1065, C3M, Team 8 (Hepatic Complications in Obesity), Nice, France
| | - F Palladino
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile
| | - P M Domingos
- ITQB-UNL, Av. da Republica, EAN, 2780-157 Oeiras, Portugal
| | - S Hunot
- Inserm, U 1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - S Chandran
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - S Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS UMR 8249, ESPCI ParisTech, PSL Research University, 75005 Paris, France
| | - T Baron
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Neurodegenerative Diseases Unit, 31, avenue Tony Garnier, 69364 Lyon Cedex 07, France
| | - D Vivien
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - C B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Faculty of Medicine, Rua Larga, and Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - H D Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - H Steller
- Howard Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - F Urano
- Washington University School of Medicine, Department of Internal Medicine, St. Louis, MO 63110 USA
| | - E Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - G Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women׳s and Children׳s Health, Karolinska University Hospital, Stockholm, Sweden
| | - A Ciechanover
- The Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 30196, Israel
| | - E J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA 01003, USA
| | - R J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - C Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
43
|
Lee JK, Wang B, Reyes M, Armstrong JS, Kulikowicz E, Santos PT, Lee JH, Koehler RC, Martin LJ. Hypothermia and Rewarming Activate a Macroglial Unfolded Protein Response Independent of Hypoxic-Ischemic Brain Injury in Neonatal Piglets. Dev Neurosci 2016; 38:277-294. [PMID: 27622292 DOI: 10.1159/000448585] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/20/2016] [Indexed: 12/22/2022] Open
Abstract
Therapeutic hypothermia provides incomplete neuroprotection after hypoxia-ischemia (HI)-induced brain injury in neonates. We previously showed that cortical neuron and white matter apoptosis are promoted by hypothermia and early rewarming in a piglet model of HI. The unfolded protein response (UPR) may be one of the potential mediators of this cell death. Here, neonatal piglets underwent HI or sham surgery followed by 29 h of normothermia, 2 h of normothermia + 27 h of hypothermia or 18 h of hypothermia + rewarming. Piglets recovered for 29 h. Immunohistochemistry for endoplasmic reticulum to nucleus signaling-1 protein (ERN1), a marker of UPR activation, was used to determine the ratios of ERN1+ macroglia and neurons in the motor subcortical white matter and cerebral cortex. The ERN1+ macroglia were immunophenotyped as oligodendrocytes and astrocytes by immunofluorescent colabeling. Temperature (p = 0.046) and HI (p < 0.001) independently affected the ratio of ERN1+ macroglia. In sham piglets, sustained hypothermia (p = 0.011) and rewarming (p = 0.004) increased the ERN1+ macroglia ratio above that in normothermia. HI prior to hypothermia diminished the UPR. Ratios of ERN1+ macroglia correlated with white matter apoptotic profile counts in shams (r = 0.472; p = 0.026), thereby associating UPR activation with white matter apoptosis during hypothermia and rewarming. Accordingly, macroglial cell counts decreased in shams that received sustained hypothermia (p = 0.009) or rewarming (p = 0.007) compared to those in normothermic shams. HI prior to hypothermia neutralized the macroglial cell loss. Neither HI nor temperature affected ERN1+ neuron ratios. In summary, delayed hypothermia and rewarming activate the macroglial UPR, which is associated with white matter apoptosis. HI may decrease the macroglial endoplasmic reticulum stress response after hypothermia and rewarming.
Collapse
Affiliation(s)
- Jennifer K Lee
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Md., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Borowiec AS, Sion B, Chalmel F, D Rolland A, Lemonnier L, De Clerck T, Bokhobza A, Derouiche S, Dewailly E, Slomianny C, Mauduit C, Benahmed M, Roudbaraki M, Jégou B, Prevarskaya N, Bidaux G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation. FASEB J 2016; 30:3155-70. [PMID: 27317670 PMCID: PMC5001517 DOI: 10.1096/fj.201600257r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/23/2016] [Indexed: 12/21/2022]
Abstract
Testes of most male mammals present the particularity of being externalized from the body and are consequently slightly cooler than core body temperature (4-8°C below). Although, hypothermia of the testis is known to increase germ cells apoptosis, little is known about the underlying molecular mechanisms, including cold sensors, transduction pathways, and apoptosis triggers. In this study, using a functional knockout mouse model of the cold and menthol receptors, dubbed transient receptor potential melastatine 8 (TRPM8) channels, we found that TRPM8 initiated the cold-shock response by differentially modulating cold- and heat-shock proteins. Besides, apoptosis of germ cells increased in proportion to the cooling level in control mice but was independent of temperature in knockout mice. We also observed that the rate of germ cell death correlated positively with the reactive oxygen species level and negatively with the expression of the detoxifying enzymes. This result suggests that the TRPM8 sensor is a key determinant of germ cell fate under hypothermic stimulation.-Borowiec, A.-S., Sion, B., Chalmel, F., Rolland, A. D., Lemonnier, L., De Clerck, T., Bokhobza, A., Derouiche, S., Dewailly, E., Slomianny, C., Mauduit, C., Benahmed, M., Roudbaraki, M., Jégou, B., Prevarskaya, N., Bidaux, G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation.
Collapse
Affiliation(s)
| | - Benoit Sion
- Pharmacologie Fondamentale et Clinique de la Douleur, INSERM, U1107, Neuro-Dol, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | | | | | - Loïc Lemonnier
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Tatiana De Clerck
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Alexandre Bokhobza
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Sandra Derouiche
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Etienne Dewailly
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Christian Slomianny
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Claire Mauduit
- Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, INSERM, U1065, Nice, France; and
| | - Mohamed Benahmed
- Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, INSERM, U1065, Nice, France; and
| | - Morad Roudbaraki
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Bernard Jégou
- INSERM, U1085-Irset, Campus de Beaulieu, Rennes, France
| | - Natalia Prevarskaya
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Gabriel Bidaux
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France; Laboratoire de Physique des Lasers, Atomes et Molécules (PhLAM), UMR8523, Biophotonic Team, Villeneuve d'Ascq, France
| |
Collapse
|
45
|
Neuroprotective hypothermia - Why keep your head cool during ischemia and reperfusion. Biochim Biophys Acta Gen Subj 2016; 1860:2521-2528. [PMID: 27475000 DOI: 10.1016/j.bbagen.2016.07.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/28/2016] [Accepted: 07/25/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Targeted temperature management (TTM) is the induced cooling of the entire body or specific organs to help prevent ischemia and reperfusion (I/R) injury, as may occur during major surgery, cardiac resuscitation, traumatic brain injury and stroke. Ischemia and reperfusion induce neuronal damage by mitochondrial dysfunction and oxidative injury, ER stress, neuronal excitotoxicity, and a neuroinflammatory response, which may lead to activation of apoptosis pathways. SCOPE OF REVIEW The aim of the current review is to discuss TTM targets that convey neuroprotection and to identify potential novel pharmacological intervention strategies for the prevention of cerebral ischemia and reperfusion injury. MAJOR CONCLUSIONS TTM precludes I/R injury by reducing glutamate release and oxidative stress and inhibiting release of pro-inflammatory factors and thereby counteracts mitochondrial induced apoptosis, neuronal excitotoxicity, and neuroinflammation. Moreover, TTM promotes regulation of the unfolded protein response and induces SUMOylation and the production of cold shock proteins. These advantageous effects of TTM seem to depend on the clinical setting, as well as type and extent of the injury. Therefore, future aims should be to refine hypothermia management in order to optimize TTM utilization and to search for pharmacological agents mimicking the cellular effects of TTM. GENERAL SIGNIFICANCE Bundling knowledge about TTM in the experimental, translational and clinical setting may result in better approaches for diminishing I/R damage. While application of TTM in the clinical setting has some disadvantages, targeting its putative protective pathways may be useful to prevent I/R injury and reduce neurological complications.
Collapse
|
46
|
Rzechorzek NM, Connick P, Livesey MR, Borooah S, Patani R, Burr K, Story D, Wyllie DJA, Hardingham GE, Chandran S. Hypothermic Preconditioning Reverses Tau Ontogenesis in Human Cortical Neurons and is Mimicked by Protein Phosphatase 2A Inhibition. EBioMedicine 2016; 3:141-154. [PMID: 26870825 PMCID: PMC4739435 DOI: 10.1016/j.ebiom.2015.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 11/18/2022] Open
Abstract
Hypothermia is potently neuroprotective, but the molecular basis of this effect remains obscure. Changes in neuronal tau protein are of interest, since tau becomes hyperphosphorylated in injury-resistant, hypothermic brains. Noting inter-species differences in tau isoforms, we have used functional cortical neurons differentiated from human pluripotent stem cells (hCNs) to interrogate tau modulation during hypothermic preconditioning at clinically-relevant temperatures. Key tau developmental transitions (phosphorylation status and splicing shift) are recapitulated during hCN differentiation and subsequently reversed by mild (32 °C) to moderate (28 °C) cooling--conditions which reduce oxidative and excitotoxic stress-mediated injury in hCNs. Blocking a major tau kinase decreases hCN tau phosphorylation and abrogates hypothermic neuroprotection, whilst inhibition of protein phosphatase 2A mimics cooling-induced tau hyperphosphorylation and protects normothermic hCNs from oxidative stress. These findings indicate a possible role for phospho-tau in hypothermic preconditioning, and suggest that cooling drives human tau towards an earlier ontogenic phenotype whilst increasing neuronal resilience to common neurotoxic insults. This work provides a critical step forward in understanding how we might exploit the neuroprotective benefits of cooling without cooling patients.
Collapse
Affiliation(s)
- Nina M Rzechorzek
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Midlothian EH16 4UU, United Kingdom; Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom.
| | - Peter Connick
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom
| | - Matthew R Livesey
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Midlothian EH8 9XD, United Kingdom
| | - Shyamanga Borooah
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Midlothian EH16 4UU, United Kingdom; Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom
| | - Rickie Patani
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Karen Burr
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Midlothian EH16 4UU, United Kingdom; Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom
| | - David Story
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Midlothian EH16 4UU, United Kingdom; Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom
| | - David J A Wyllie
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Midlothian EH8 9XD, United Kingdom
| | - Giles E Hardingham
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Midlothian EH8 9XD, United Kingdom
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, Midlothian EH16 4UU, United Kingdom; Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom; The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian EH16 4SB, United Kingdom.
| |
Collapse
|