1
|
Yao F, Cai SQ, Cheng HX, Ren LW, Hui KL, Liu QZ, Guo M, Chen LH, Qian B, Zeng Y, Li F, Duan ML. Therapeutic Hypothermia Increases the Expression of RNA-binding Protein Motif 3 and Attenuates Cognitive Deficits Following Cardiac Arrest in Rats. Neurochem Res 2025; 50:134. [PMID: 40257581 PMCID: PMC12011659 DOI: 10.1007/s11064-025-04383-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Cardiac arrest (CA) remains a leading cause of mortality and morbidity worldwide. Cognitive deficits are common neurological sequelae among CA survivors. Preclinical and clinical studies have confirmed that therapeutic hypothermia (TH) is an effective intervention for mitigating brain injury following CA. Hypothermia induces the expression of specific small proteins, including RNA-binding motif protein 3 (RBM3), which provides neuroprotection under stress conditions. However, the role of RBM3 in TH after CA has not been fully elucidated. In this study, we investigated the role of RBM3 in attenuating cognitive deficits following hypothermic brain resuscitation. We constructed a rat model of CA and resuscitation, and used shRNA transfection to interfere with RBM3 expression to explore the underlying mechanisms of TH's effects on cognitive alterations. Rats were randomly assigned to one of five groups: sham group (Sham), CA group (CA), TH group (TH), adeno-associated virus (AAV)-shRNA-RBM3 transfection group (shRNA-RBM3), and AAV-shRNA-negative control transfection group (shRNA-control). Key synaptic regulatory proteins, dendritic spines, and synaptic ultrastructures were examined. The rats exhibited spatial learning and memory impairments in the Morris water maze test and novel object recognition task. Hypothermia increased RBM3 expression in hippocampal neurons, mitigated early brain injury, preserved dendritic spine integrity and synaptic ultrastructure, upregulated key synaptic regulatory proteins, and ameliorated cognitive impairment following resuscitation. When RBM3 expression in the hippocampus was inhibited, the beneficial effects of therapeutic hypothermia were partially reversed. Overall, our findings provide new insights into the mechanisms of hypothermia-induced neuroprotection, demonstrating that neuroplasticity and rehabilitation can be achieved following global cerebral ischemia-reperfusion injury after CA. Therefore, the RBM3-mediated cold shock pathway represents a potential target for enhancing neuroprotection and neurorehabilitation through hypothermia.
Collapse
Affiliation(s)
- Fen Yao
- Department of Anesthesiology, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Anesthesiology, The First People's Hospital of Yancheng, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Shen-Quan Cai
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Hui-Xian Cheng
- Department of Anesthesiology, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Li-Wen Ren
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Kang-Li Hui
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Qing-Zhen Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Min Guo
- Department of Anesthesiology, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Li-Hui Chen
- Department of Anesthesiology, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Qian
- Department of Anesthesiology, The First People's Hospital of Yancheng, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Yang Zeng
- Department of Anesthesiology, The First People's Hospital of Yancheng, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Feng Li
- Department of Anesthesiology, The First People's Hospital of Yancheng, Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China.
| | - Man-Lin Duan
- Department of Anesthesiology, Nanjing Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
- Department of Anesthesiology, Nanjing Tianyishan Hospital, The First Affiliated Hospital of China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
2
|
Ávila-Gómez P, Vieites-Prado A, Correa-Paz C, Del Pozo-Filíu L, Palomar-Alonso N, Campos F, López-Arias E. Therapeutic modulation of protein RBM3 for ischemic stroke treatment. Front Pharmacol 2025; 16:1555115. [PMID: 40124786 PMCID: PMC11925906 DOI: 10.3389/fphar.2025.1555115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Several preclinical assays and clinical trials have found hypothermia as an efficient protective treatment for stroke. However, systemic hypothermia impairs several physiological functions being difficult to implement in acute critical patients. A deeper understanding of the mechanisms underlying the therapeutic effects of hypothermia could inspire new treatments based on the protective effects of cold. Furthermore, this could contribute to the reduction of the side effects associated with it. One of the metabolic landmarks of hypothermia is the overexpression of a small subset of shock proteins while global protein synthesis is reduced. Among these cold-shock proteins, RBM3 (RNA-binding motif protein 3) seems to play a central protective role. In physiological conditions, which is involved in the regulation of protein synthesis. In several models of cerebral diseases, in vitro and in vivo, RBM3 exhibited the ability to mitigate apoptosis or increase neural proliferation. In stroke models, RBM3 has shown specially promising effects attenuating neural damage and enhancing cell survival. Future prospects should be directed towards the design of efficient strategies to modulate RBM3 levels. This mini-review aims to summarize the progress made in understanding the role of RBM3 in cerebral tissue protection, while encouraging efforts to address research gaps, particularly in its modulation and clinical application.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Alba Vieites-Prado
- Brain Plasticity Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Brain Plasticity Laboratory, Centre for research in molecular medicine and chronic diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Neurological Sciences and Cerebrovascular Research Laboratory, Neurology and Cerebrovascular Disease Group, Department of Neurology and Stroke Centre, Neuroscience Area La Paz Institute for Health Research - idiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), Madrid, Spain
| | - Lucía Del Pozo-Filíu
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Nuria Palomar-Alonso
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
3
|
van de Wal MAE, Doornbos C, Bibbe JM, Homberg JR, van Karnebeek C, Huynen MA, Keijer J, van Schothorst EM, 't Hoen PAC, Janssen MCH, Adjobo-Hermans MJW, Wieckowski MR, Koopman WJH. Ndufs4 knockout mice with isolated complex I deficiency engage a futile adaptive brain response. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141055. [PMID: 39395749 DOI: 10.1016/j.bbapap.2024.141055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Paediatric Leigh syndrome (LS) is an early-onset and fatal neurodegenerative disorder lacking treatment options. LS is frequently caused by mutations in the NDUFS4 gene, encoding an accessory subunit of mitochondrial complex I (CI), the first complex of the oxidative phosphorylation (OXPHOS) system. Whole-body Ndufs4 knockout (KO) mice (WB-KO mice) are widely used to study isolated CI deficiency, LS pathology and interventions. These animals develop a brain-specific phenotype via an incompletely understood pathomechanism. Here we performed a quantitative analysis of the sub-brain proteome in six-weeks old WB-KO mice vs. wildtype (WT) mice. Brain regions comprised of a brain slice (BrSl), cerebellum (CB), cerebral cortex (CC), hippocampus (HC), inferior colliculus (IC), and superior colliculus (SC). Proteome analysis demonstrated similarities between CC/HC, and between IC/SC, whereas BrSl and CB differed from these two groups and each other. All brain regions displayed greatly reduced levels of two CI structural subunits (NDUFS4, NDUFA12) and an increased level of the CI assembly factor NDUFAF2. The level of CI-Q module subunits was significantly more reduced in IC/SC than in BrSl/CB/CC/HC, whereas other OXPHOS complex levels were not reduced. Gene ontology and pathway analysis demonstrated specific and common proteome changes between brain regions. Across brain regions, upregulation of cold-shock-associated proteins, mitochondrial fatty acid (FA) oxidation and synthesis (mtFAS) were the most prominent. FA-related pathways were predominantly upregulated in CB and HC. Based upon these results, we argue that stimulation of these pathways is futile and pro-pathological and discuss alternative strategies for therapeutic intervention in LS. SIGNIFICANCE: The Ndufs4 knockout mouse model is currently the most relevant and most widely used animal model to study the brain-linked pathophysiology of human Leigh Syndrome (LS) and intervention strategies. We demonstrate that the Ndufs4 knockout brain engages futile and pro-pathological responses. These responses explain both negative and positive outcomes of intervention studies in Leigh Syndrome mice and patients, thereby guiding novel intervention opportunities.
Collapse
Affiliation(s)
- Melissa A E van de Wal
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cenna Doornbos
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Janne M Bibbe
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clara van Karnebeek
- Departments of Pediatrics and Human Genetics, Emma Center for Personalized Medicine, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands; United for Metabolic Diseases, the Netherlands
| | - Martijn A Huynen
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | | | - Peter A C 't Hoen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mirian C H Janssen
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Merel J W Adjobo-Hermans
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Werner J H Koopman
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands.
| |
Collapse
|
4
|
Kline KE, Russell AL, Stezoski JP, Gober IG, Dimeo EG, Janesko-Feldman K, Drabek T, Kochanek PM, Wagner AK. Differential Effects of Targeted Temperature Management on Sex-Dependent Outcomes After Experimental Asphyxial Cardiac Arrest. Ther Hypothermia Temp Manag 2024; 14:299-309. [PMID: 38386544 PMCID: PMC11665272 DOI: 10.1089/ther.2023.0061] [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] [Indexed: 02/24/2024] Open
Abstract
Asphyxial cardiac arrest (ACA) survivors face lasting neurological disability from hypoxic ischemic brain injury. Sex differences in long-term outcomes after cardiac arrest (CA) are grossly understudied and underreported. We used rigorous targeted temperature management (TTM) to understand its influence on survival and lasting sex-specific neurological and neuropathological outcomes in a rodent ACA model. Adult male and female rats underwent either sham or 5-minute no-flow ACA with 18 hours TTM at either ∼37°C (normothermia) or ∼36°C (mild hypothermia). Survival, temperature, and body weight (BW) were recorded over the 14-day study duration. All rats underwent neurological deficit score (NDS) assessment on days 1-3 and day 14. Hippocampal pathology was assessed for cell death, degenerating neurons, and microglia on day 14. Although ACA females were less likely to achieve return of spontaneous circulation (ROSC), post-ROSC physiology and biochemical profiles were similar between sexes. ACA females had significantly greater 14-day survival, NDS, and BW recovery than ACA males at normothermia (56% vs. 29%). TTM at 36°C versus 37°C improved 14-day survival in males, producing similar survival in male (63%) versus female (50%). There were no sex or temperature effects on CA1 histopathology. We conclude that at normothermic conditions, sex differences favoring females were observed after ACA in survival, NDS, and BW recovery. We achieved a clinically relevant ACA model using TTM at 36°C to improve long-term survival. This model can be used to more fully characterize sex differences in long-term outcomes and test novel acute and chronic therapies.
Collapse
Affiliation(s)
- Kelsey E. Kline
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ashley L. Russell
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason P. Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ian G. Gober
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Emma G. Dimeo
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
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
|
6
|
Rafnsdottir S, Jang K, Halldorsdottir ST, Vinod M, Tomasdottir A, Möller K, Halldorsdottir K, Reynisdottir T, Atladottir LH, Allison KE, Ostacolo K, He J, Zhang L, Northington FJ, Magnusdottir E, Chavez-Valdez R, Anderson KJ, Bjornsson HT. SMYD5 is a regulator of the mild hypothermia response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.11.540170. [PMID: 37333301 PMCID: PMC10274674 DOI: 10.1101/2023.05.11.540170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The mild hypothermia response (MHR) maintains organismal homeostasis during cold exposure and is thought to be critical for the neuroprotection documented with therapeutic hypothermia. To date, little is known about the transcriptional regulation of the MHR. We utilize a forward CRISPR-Cas9 mutagenesis screen to identify the histone lysine methyltransferase SMYD5 as a regulator of the MHR. SMYD5 represses the key MHR gene SP1 at euthermia. This repression correlates with temperature-dependent levels of H3K36me3 at the SP1-locus and globally, indicating that the mammalian MHR is regulated at the level of histone modifications. We have identified 37 additional SMYD5 regulated temperature-dependent genes, suggesting a broader MHR-related role for SMYD5. Our study provides an example of how histone modifications integrate environmental cues into the genetic circuitry of mammalian cells and provides insights that may yield therapeutic avenues for neuroprotection after catastrophic events.
Collapse
Affiliation(s)
- Salvor Rafnsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Kijin Jang
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Sara Tholl Halldorsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Meghna Vinod
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Arnhildur Tomasdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Katrin Möller
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Katrin Halldorsdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Tinna Reynisdottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | - Laufey Halla Atladottir
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
| | | | - Kevin Ostacolo
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
| | - Jin He
- Department of Biochemistry and Molecular Biology, College of Natural Science, Michigan State University; MI, USA
| | - Li Zhang
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, USA
| | - Frances J Northington
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University; Baltimore, MD, USA
| | - Erna Magnusdottir
- Department of Biomedical Science and Department of Anatomy, Faculty of Medicine, University of Iceland; Reykjavík, Iceland
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Neuroscience Intensive Care Nursery Program, Johns Hopkins University; Baltimore, MD, USA
| | - Kimberley Jade Anderson
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
| | - Hans Tomas Bjornsson
- Louma G. Laboratory of Epigenetic Research, Faculty of Medicine, University of Iceland; Reykjavik, Iceland
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University; Baltimore, MD, USA
- Department of Genetics and Molecular Medicine, Landspitali University Hospital; Reykjavik, Iceland
- Lead contact
| |
Collapse
|
7
|
Herrmann JR, Fink EL, Fabio A, Berger RP, Janesko-Feldman K, Gorse K, Clark RSB, Kochanek PM, Jackson TC. Characterization of Circulating Cold Shock Proteins FGF21 and RBM3 in a Multi-Center Study of Pediatric Cardiac Arrest. Ther Hypothermia Temp Manag 2024; 14:99-109. [PMID: 37669029 PMCID: PMC11391889 DOI: 10.1089/ther.2023.0035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
Fibroblast Growth Factor 21 (FGF21) is a neuroprotective hormone induced by cold exposure that targets the β-klotho co-receptor. β-klotho is abundant in the newborn brain but decreases rapidly with age. RNA-Binding Motif 3 (RBM3) is a potent neuroprotectant upregulated by FGF21 in hypothermic conditions. We characterized serum FGF21 and RBM3 levels in patients enrolled in a prospective multi-center study of pediatric cardiac arrest (CA) via a secondary analysis of samples collected to evaluate brain injury biomarkers. Patients (n = 111) with remnant serum samples available from at least two of three available timepoints (0-24, 24-48 or 48-72 hours post-resuscitation) were included. Serum samples from 20 healthy controls were used for comparison. FGF21 was measured by Luminex and internally validated enzyme-linked immunoassay (ELISA). RBM3 was measured by internally validated ELISA. Of postarrest patients, 98 were managed with normothermia, while 13 were treated with therapeutic hypothermia (TH). FGF21 increased >20-fold in the first 24 hours postarrest versus controls (681 pg/mL [200-1864] vs. 29 pg/mL [15-51], n = 99 vs. 19, respectively, p < 0.0001, median [interquartile range]) with no difference in RBM3. FGF21 did not differ by sex, while RBM3 was increased in females versus males at 48-72 hours postarrest (1866 pg/mL [873-5176] vs. 1045 pg/mL [535-2728], n = 40 vs. 54, respectively, p < 0.05). Patients requiring extracorporeal membrane oxygenation (ECMO) postresuscitation had increased FGF21 versus those who did not at 48-72 hours (6550 pg/mL [1455-66,781] vs. 1213 pg/mL [480-3117], n = 7 vs 74, respectively, p < 0.05). FGF21 and RBM3 did not correlate (Spearman's rho = 0.004, p = 0.97). We conclude that in a multi-center study of pediatric CA patients where normothermic targeted temperature management was largely used, FGF21 was markedly increased postarrest versus control and highest in patients requiring ECMO postresuscitation. RBM3 was sex-dependent. We provide a framework for future studies examining the effect of TH on FGF21 or use of FGF21 therapy after pediatric CA.
Collapse
Affiliation(s)
- Jeremy R Herrmann
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ericka L Fink
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony Fabio
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rachel P Berger
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kiersten Gorse
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Robert S B Clark
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Travis C Jackson
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
8
|
Lin P, Lin C, Diao L. RBM3 Ameliorates Acute Brain Injury-induced Inflammation and Oxidative Stress by Stabilizing GAS6 mRNA Through Nrf2 Signaling Pathway. Neuroscience 2024; 547:74-87. [PMID: 38555015 DOI: 10.1016/j.neuroscience.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
RNA-binding motif protein 3 (RBM3), as a cold-inducible protein, exhibits neuroprotective function in brain disorders. This study was conducted to investigate the effects of RBM3 on acute brain injury (ABI) and its underlying mechanism. The cerebral injury (CI) rat model and oxygen-glucose deprivation (OGD) cell model were established. The neurological severity score, wire-grip score, morris water maze test, and Y-maze test were used to detect the neurological damage, vestibular motor, learning, and memory functions. Cerebral injury, apoptosis, oxidative stress, and inflammatory level were evaluated by hematoxylin-eosin and TUNEL staining and specific kits. Flow cytometry was used to analyze the apoptosis rate. The relationship between RBM3 and growth arrest specific (GAS) 6 was analyzed by RNA immunoprecipitation assay. The results indicated that RBM3 recovered of neurological function and behaviour impairment of CI rats. Additionally, RBM3 reversed the increased oxidative stress, inflammatory level, and apoptosis induced by CI and OGD. RBM3 interacted with GAS6 to activate the Nrf2 signaling pathway, thus playing neuroprotection on ABI. Besides, the results of RBM3 treatment were similar to those of mild hypothermia treatment. In summary, RBM3 exerted neuroprotection and ameliorated inflammatory levels and oxidative stress by stabilizing GAS6 mRNA through the Nrf2 signaling pathway, suggesting that RBM3 might be a potential therapeutic candidate for treating ABI.
Collapse
Affiliation(s)
- Pingqing Lin
- Department Of Emergency, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China.
| | - Chengshi Lin
- Department Of Emergency, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China
| | - Liangbiao Diao
- Department Of Nephrology, Fuzhou Second General Hospital, Fuzhou City, Fujian Province 350007, China
| |
Collapse
|
9
|
Hoekstra MMB, Ness N, Badia-Soteras A, Brancaccio M. Bmal1 integrates circadian function and temperature sensing in the suprachiasmatic nucleus. Proc Natl Acad Sci U S A 2024; 121:e2316646121. [PMID: 38625943 PMCID: PMC11047078 DOI: 10.1073/pnas.2316646121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/22/2024] [Indexed: 04/18/2024] Open
Abstract
Circadian regulation and temperature dependency are important orchestrators of molecular pathways. How the integration between these two drivers is achieved, is not understood. We monitored circadian- and temperature-dependent effects on transcription dynamics of cold-response protein RNA Binding Motif 3 (Rbm3). Temperature changes in the mammalian master circadian pacemaker, the suprachiasmatic nucleus (SCN), induced Rbm3 transcription and regulated its circadian periodicity, whereas the core clock gene Per2 was unaffected. Rbm3 induction depended on a full Brain And Muscle ARNT-Like Protein 1 (Bmal1) complement: reduced Bmal1 erased Rbm3 responses and weakened SCN circuit resilience to temperature changes. By focusing on circadian and temperature dependency, we highlight weakened transmission between core clock and downstream pathways as a potential route for reduced circadian resilience.
Collapse
Affiliation(s)
- Marieke M. B. Hoekstra
- Department of Brain Science, Imperial College London, LondonW12 0NN, United Kingdom
- Department of Brain Sciences, United Kingdom Dementia Research Institute at Imperial College London, LondonW12 0NN, United Kingdom
| | - Natalie Ness
- Department of Brain Science, Imperial College London, LondonW12 0NN, United Kingdom
- Department of Brain Sciences, United Kingdom Dementia Research Institute at Imperial College London, LondonW12 0NN, United Kingdom
| | - Aina Badia-Soteras
- Department of Brain Science, Imperial College London, LondonW12 0NN, United Kingdom
- Department of Brain Sciences, United Kingdom Dementia Research Institute at Imperial College London, LondonW12 0NN, United Kingdom
| | - Marco Brancaccio
- Department of Brain Science, Imperial College London, LondonW12 0NN, United Kingdom
- Department of Brain Sciences, United Kingdom Dementia Research Institute at Imperial College London, LondonW12 0NN, United Kingdom
| |
Collapse
|
10
|
Wolf-Johnston A, Ikeda Y, Zabbarova I, Kanai AJ, Bastacky S, Moldwin R, Stern JN, Jackson EK, Birder LA. Purine nucleoside phosphorylase inhibition is an effective approach for the treatment of chemical hemorrhagic cystitis. JCI Insight 2024; 9:e176103. [PMID: 38271096 PMCID: PMC10972598 DOI: 10.1172/jci.insight.176103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024] Open
Abstract
Hemorrhagic cystitis may be induced by infection, radiation therapy, or medications or may be idiopathic. Along with hemorrhagic features, symptoms include urinary urgency and frequency, dysuria (painful urination), and visceral pain. Cystitis-induced visceral pain is one of the most challenging types of pain to treat, and an effective treatment would address a major unmet medical need. We assessed the efficacy of a purine nucleoside phosphorylase inhibitor, 8-aminoguanine (8-AG), for the treatment of hemorrhagic/ulcerative cystitis. Lower urinary tract (LUT) function and structure were assessed in adult Sprague-Dawley rats, treated chronically with cyclophosphamide (CYP; sacrificed day 8) and randomized to daily oral treatment with 8-AG (begun 14 days prior to CYP induction) or its vehicle. CYP-treated rats exhibited multiple abnormalities, including increased urinary frequency and neural mechanosensitivity, reduced bladder levels of inosine, urothelial inflammation/damage, and activation of spinal cord microglia, which is associated with pain hypersensitivity. 8-AG treatment of CYP-treated rats normalized all observed histological, structural, biochemical, and physiological abnormalities. In cystitis 8-AG improved function and reduced both pain and inflammation likely by increasing inosine, a tissue-protective purine metabolite. These findings demonstrate that 8-AG has translational potential for reducing pain and preventing bladder damage in cystitis-associated LUT dysfunctions.
Collapse
Affiliation(s)
| | - Youko Ikeda
- Renal-Electrolyte Division, Department of Medicine
| | | | - Anthony J Kanai
- Renal-Electrolyte Division, Department of Medicine
- Department of Pharmacology and Chemical Biology; and
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert Moldwin
- Arthur Smith Institute for Urology, Northwell Health, Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, USA
| | - Joel Nh Stern
- Arthur Smith Institute for Urology, Northwell Health, Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York, USA
| | | | - Lori A Birder
- Renal-Electrolyte Division, Department of Medicine
- Department of Pharmacology and Chemical Biology; and
| |
Collapse
|
11
|
Jackson TC, Herrmann JR, Fink EL, Au AK, Kochanek PM. Harnessing the Promise of the Cold Stress Response for Acute Brain Injury and Critical Illness in Infants and Children. Pediatr Crit Care Med 2024; 25:259-270. [PMID: 38085024 PMCID: PMC10932834 DOI: 10.1097/pcc.0000000000003424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Affiliation(s)
- Travis C. Jackson
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Jeremy R. Herrmann
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Children’s Hospital of Philadelphia, Philadelphia, PA
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Alicia K. Au
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
12
|
Herrmann JR, Kochanek PM, Vagni VA, Janesko-Feldman K, Stezoski J, Gorse K, Jackson TC. FGF21 modulates hippocampal cold-shock proteins and CA2-subregion proteins in neonatal mice with hypoxia-ischemia. Pediatr Res 2023; 94:1355-1364. [PMID: 37193753 PMCID: PMC10690493 DOI: 10.1038/s41390-023-02652-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/30/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) is a neuroprotectant with cognitive enhancing effects but with poorly characterized mechanism(s) of action, particularly in females. Prior studies suggest that FGF21 may regulate cold-shock proteins (CSPs) and CA2-marker proteins in the hippocampus but empirical evidence is lacking. METHODS We assessed in normothermic postnatal day (PND) 10 female mice, if hypoxic-ischemic (HI) brain injury (25 min 8% O2/92% N2) altered endogenous levels of FGF21 in serum or in the hippocampus, or its receptor β-klotho. We also tested if systemic administration of FGF21 (1.5 mg/kg) modulated hippocampal CSPs or CA2 proteins. Finally, we measured if FGF21 therapy altered markers of acute hippocampal injury. RESULTS HI increased endogenous serum FGF21 (24 h), hippocampal tissue FGF21 (4d), and decreased hippocampal β-klotho levels (4d). Exogenous FGF21 therapy modulated hippocampal CSP levels, and dynamically altered hippocampal CA2 marker expression (24 h and 4d). Finally, FGF21 ameliorated neuronal damage markers at 24 h but did not affect GFAP (astrogliosis) or Iba1 (microgliosis) levels at 4d. CONCLUSIONS FGF21 therapy modulates CSP and CA2 protein levels in the injured hippocampus. These proteins serve different biological functions, but our findings suggest that FGF21 administration modulates them in a homeostatic manner after HI. IMPACT Hypoxic-ischemic (HI) injury in female post-natal day (PND) 10 mice decreases hippocampal RNA binding motif 3 (RBM3) levels in the normothermic newborn brain. HI injury in normothermic newborn female mice alters serum and hippocampal fibroblast growth factor 21 (FGF21) levels 24 h post-injury. HI injury in normothermic newborn female mice alters hippocampal levels of N-terminal EF-hand calcium binding protein 2 (NECAB2) in a time-dependent manner. Exogenous FGF21 therapy ameliorates the HI-mediated loss of hippocampal cold-induced RNA-binding protein (CIRBP). Exogenous FGF21 therapy modulates hippocampal levels of CA2-marker proteins after HI.
Collapse
Affiliation(s)
- Jeremy R Herrmann
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Vincent A Vagni
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Keri Janesko-Feldman
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Jason Stezoski
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Kiersten Gorse
- USF Health Heart Institute, University of South Florida Morsani College of Medicine, MDD 0630, 560 Channelside Drive, Tampa, FL, 33602, USA
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Boulevard, Tampa, FL, 33612-4799, USA
| | - Travis C Jackson
- USF Health Heart Institute, University of South Florida Morsani College of Medicine, MDD 0630, 560 Channelside Drive, Tampa, FL, 33602, USA.
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Boulevard, Tampa, FL, 33612-4799, USA.
| |
Collapse
|
13
|
Anatychuk L, Zadorozhnyy O, Naumenko V, Maltsev E, Kobylianskyi R, Nazaretyan R, Umanets M, Kustryn T, Nasinnyk I, Korol A, Pasyechnikova N. Vitreoretinal Surgery with Temperature Management: A Preliminary Study in Rabbits. Ther Hypothermia Temp Manag 2023; 13:126-133. [PMID: 36827431 DOI: 10.1089/ther.2022.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
The present study aimed to evaluate the structure of the rabbit retina after vitreoretinal surgery using prolonged irrigation with solutions of different temperatures. Thirty-six rabbits (72 eyes) were included in this study and randomly divided into 3 equal groups according to the temperature of the intraocular irrigating fluid they received during vitrectomy. Vitreoretinal surgery was performed with a 5°C irrigation solution in group 1 (12 rabbits, 24 eyes), a 22°C irrigation solution in group 2 (12 rabbits, 24 eyes), and a 36°C irrigation solution in group 3 (12 rabbits, 24 eyes). In each group of animals, the mean irrigation/aspiration time was 30 minutes for left eyes and 60 minutes for right eyes. Histological examination of the retina was performed 1, 7, and 30 days after surgery. During surgery, the temperature in the vitreous cavity of the eyes of rabbits of groups 1, 2, and 3 dropped by 26.0°C, 11.2°C (deep hypothermia), and 1.0°C (mild hypothermia), respectively. The highest rewarming rate was detected in group 1 (0.9°C/min) compared with group 2 (0.7°C/min) and group 3 (0.2°C/min). After 60 minutes of irrigation, retinal structural changes were detected in the animals of groups 1 and 2 (in contrast to the animals of group 3). After surgery with irrigation lasting 30 minutes, no retinal structural changes were observed. This study showed that temperature management, avoidance of intraoperative deep hypothermia, and prevention of rapid uncontrolled rewarming may protect the retinal morphology and increase the safety of prolonged vitreoretinal surgery.
Collapse
Affiliation(s)
- Lukyan Anatychuk
- Medical Department, Institute of Thermoelectricity of the National Academy of Sciences of Ukraine and the Ministry of Education and Science of Ukraine, Chernivtsi, Ukraine
- Department of Thermoelectricity, Yuriy Fedkovych Chernivtsi National University, Chernivtsi, Ukraine
| | - Oleg Zadorozhnyy
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Volodymyr Naumenko
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Eduard Maltsev
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Roman Kobylianskyi
- Medical Department, Institute of Thermoelectricity of the National Academy of Sciences of Ukraine and the Ministry of Education and Science of Ukraine, Chernivtsi, Ukraine
- Department of Thermoelectricity, Yuriy Fedkovych Chernivtsi National University, Chernivtsi, Ukraine
| | - Rudolph Nazaretyan
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Mykola Umanets
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Taras Kustryn
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Illia Nasinnyk
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Andrii Korol
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| | - Nataliya Pasyechnikova
- Department of Laser Microsurgery of Eye Diseases, State Institution "The Filatov Institute of Eye Diseases and Tissue Therapy of the National Academy of Medical Sciences of Ukraine," Odesa, Ukraine
| |
Collapse
|
14
|
Li L, Ye W, Li Y, Chen Y, Zeng J. Intraoperative accidental hypothermia as a probable cause of malignant ventricular arrhythmias in an elderly patient undergoing transurethral resection of prostate: A case report. Heliyon 2023; 9:e19006. [PMID: 37600405 PMCID: PMC10432956 DOI: 10.1016/j.heliyon.2023.e19006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/23/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
Background Intraoperative hypothermia is a common but severe condition that is defined as a core body temperature below 36 °C. Accidental hypothermia can produce coagulopathy, immunosuppression and peripheral hypoperfusion that can ultimately lead to life-threatening ventricular arrhythmias and vital organ injury, and it is significantly associated with perioperative complications and mortality. Case description We report the case of an 82-year-old man who presented with persistent ventricular tachycardia intraoperatively due to accidental hypothermia. The patient was diagnosed with benign prostatic hypertrophy and scheduled for transurethral resection of the prostate. Laboratory tests showed moderate anemia, and echocardiography indicated mild tricuspid and mitral regurgitation. The patient received general anesthesia with endotracheal intubation. Four hours after the start of surgery, the patient developed sudden ventricular tachycardia with severe hypotension. Arterial blood gas sampling indicated that there was no disturbance of electrolytes, acid-base balance or excessive bleeding. The rectal temperature was measured immediately, and the core temperature was 32 °C. The patient received antiarrhythmic therapy and rewarming measures. No additional ventricular arrhythmias appeared after the core temperature rose to 35 °C and the blood pressure returned to normal. The patient was transferred to the intensive care unit after surgery for further observation and was moved to the general ward the next day. He was discharged 4 days later without significant organ damage. Conclusions Intraoperative hypothermia may increase ventricular arrhythmia risk, especially in elderly patients. Surgeons and anesthesiologists should pay more attention to preventing and reversing accidental hypothermia, necessitating aggressive efforts to maintain normothermia during surgery.
Collapse
Affiliation(s)
| | | | - Yongxing Li
- Department of Anesthesiology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yingzhen Chen
- Department of Anesthesiology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianfeng Zeng
- Department of Anesthesiology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Preußner M, Smith HL, Hughes D, Zhang M, Emmerichs A, Scalzitti S, Peretti D, Swinden D, Neumann A, Haltenhof T, Mallucci GR, Heyd F. ASO targeting RBM3 temperature-controlled poison exon splicing prevents neurodegeneration in vivo. EMBO Mol Med 2023; 15:e17157. [PMID: 36946385 PMCID: PMC10165353 DOI: 10.15252/emmm.202217157] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 03/23/2023] Open
Abstract
Neurodegenerative diseases are increasingly prevalent in the aging population, yet no disease-modifying treatments are currently available. Increasing the expression of the cold-shock protein RBM3 through therapeutic hypothermia is remarkably neuroprotective. However, systemic cooling poses a health risk, strongly limiting its clinical application. Selective upregulation of RBM3 at normothermia thus holds immense therapeutic potential. Here we identify a poison exon within the RBM3 gene that is solely responsible for its cold-induced expression. Genetic removal or antisense oligonucleotide (ASO)-mediated manipulation of this exon yields high RBM3 levels independent of cooling. Notably, a single administration of ASO to exclude the poison exon, using FDA-approved chemistry, results in long-lasting increased RBM3 expression in mouse brains. In prion-diseased mice, this treatment leads to remarkable neuroprotection, with prevention of neuronal loss and spongiosis despite high levels of disease-associated prion protein. Our promising results in mice support the possibility that RBM3-inducing ASOs might also deliver neuroprotection in humans in conditions ranging from acute brain injury to Alzheimer's disease.
Collapse
Affiliation(s)
- Marco Preußner
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Heather L Smith
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Daniel Hughes
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Min Zhang
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Ann‐Kathrin Emmerichs
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Silvia Scalzitti
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| | - Diego Peretti
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Dean Swinden
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Alexander Neumann
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
- Omiqa BioinformaticsBerlinGermany
| | - Tom Haltenhof
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
- Omiqa BioinformaticsBerlinGermany
| | - Giovanna R Mallucci
- UK Dementia Research Institute and Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Altos LabsCambridge Institute of ScienceCambridgeUK
| | - Florian Heyd
- Institut für Chemie und Biochemie, RNA BiochemieFreie Universität BerlinBerlinGermany
| |
Collapse
|
16
|
Shevelev OA, Petrova MV, Mengistu EM, Yuriev MY, Kostenkova IZ, Vesnin SG, Kanarskii MM, Zhdanova MA, Goryanin I. Correction of Local Brain Temperature after Severe Brain Injury Using Hypothermia and Medical Microwave Radiometry (MWR) as Companion Diagnostics. Diagnostics (Basel) 2023; 13:diagnostics13061159. [PMID: 36980467 PMCID: PMC10047658 DOI: 10.3390/diagnostics13061159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/30/2023] Open
Abstract
The temperature of the brain can reflect the activity of its different regions, allowing us to evaluate the connections between them. A study involving 111 patients in a vegetative state or minimally conscious state used microwave radiometry to measure their cortical temperature. The patients were divided into a main group receiving a 10-day selective craniocerebral hypothermia (SCCH) procedure, and a control group receiving basic therapy and rehabilitation. The main group showed a significant improvement in consciousness level as measured by CRS-R assessment on day 14 compared to the control group. Temperature heterogeneity increased in patients who received SCCH, while remaining stable in the control group. The use of microwave radiometry to assess rehabilitation effectiveness and the inclusion of SCCH in rehabilitation programs appears to be a promising approach.
Collapse
Affiliation(s)
- Oleg A Shevelev
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
- Department of Anaesthesiology and Intensive Care, Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia, 117198 Moscow, Russia
| | - Marina V Petrova
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
- Department of Anaesthesiology and Intensive Care, Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia, 117198 Moscow, Russia
| | - Elias M Mengistu
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
- Department of Anaesthesiology and Intensive Care, Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia, 117198 Moscow, Russia
| | - Mikhail Y Yuriev
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
| | - Inna Z Kostenkova
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
| | - Sergey G Vesnin
- Medical Microwave Radiometry (MMWR) LTD, Edinburgh EH10 5LZ, UK
| | - Michael M Kanarskii
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
| | - Maria A Zhdanova
- Federal Research and Clinical Centre for Resuscitation and Rehabilitology, 107031 Moscow, Russia
| | - Igor Goryanin
- School of Informatics, University of Edinburgh, Edinburgh EH8 9YL, UK
- Biological Systems Unit, Okinawa Institute Science and Technology, Onna 904-0495, Japan
| |
Collapse
|
17
|
King BMN, Mintz S, Lin X, Morley GE, Schlamp F, Khodadadi-Jamayran A, Fishman GI. Chronic Kidney Disease Induces Proarrhythmic Remodeling. Circ Arrhythm Electrophysiol 2023; 16:e011466. [PMID: 36595632 PMCID: PMC9852080 DOI: 10.1161/circep.122.011466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) are at increased risk of developing cardiac arrhythmogenesis and sudden cardiac death; however, the basis for this association is incompletely known. METHODS Here, using murine models of CKD, we examined interactions between kidney disease progression and structural, electrophysiological, and molecular cardiac remodeling. RESULTS C57BL/6 mice with adenine supplemented in their diet developed progressive CKD. Electrocardiographically, CKD mice developed significant QT prolongation and episodes of bradycardia. Optical mapping of isolated-perfused hearts using voltage-sensitive dyes revealed significant prolongation of action potential duration with no change in epicardial conduction velocity. Patch-clamp studies of isolated ventricular cardiomyocytes revealed changes in sodium and potassium currents consistent with action potential duration prolongation. Global transcriptional profiling identified dysregulated expression of cellular stress response proteins RBM3 (RNA-binding motif protein 3) and CIRP (cold-inducible RNA-binding protein) that may underlay the ion channel remodeling. Unexpectedly, we found that female sex is a protective factor in the progression of CKD and its cardiac sequelae. CONCLUSIONS Our data provide novel insights into the association between CKD and pathologic proarrhythmic cardiac remodeling. Cardiac cellular stress response pathways represent potential targets for pharmacologic intervention for CKD-induced heart rhythm disorders.
Collapse
Affiliation(s)
- Benjamin M N King
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Shana Mintz
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Xianming Lin
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Gregory E Morley
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | - Florencia Schlamp
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| | | | - Glenn I Fishman
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY
| |
Collapse
|
18
|
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
|
19
|
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: 10] [Impact Index Per Article: 3.3] [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
|
20
|
Ávila-Gómez P, Pérez-Mato M, Hervella P, Dopico-López A, da Silva-Candal A, Bugallo-Casal A, López-Amoedo S, Candamo-Lourido M, Sobrino T, Iglesias-Rey R, Castillo J, Campos F. Associations between RNA-Binding Motif Protein 3, Fibroblast Growth Factor 21, and Clinical Outcome in Patients with Stroke. J Clin Med 2022; 11:jcm11040949. [PMID: 35207221 PMCID: PMC8875775 DOI: 10.3390/jcm11040949] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023] Open
Abstract
Background: RNA-binding motif protein 3 (RBM3) is a cold-induced marker of good functional outcome of ischemic stroke that is promising as a protective target. Fibroblast growth factor 21 (FGF21) is an obesity- and temperature-related hormone that upregulates the expression of RBM3, which is beneficial as a recombinant treatment and has been tested under different experimental pathological conditions, including stroke. However, the interaction between RBM3 and FGF21 has not yet been tested for clinical stroke conditions. Methods: In a sample of 66 stroke patients, we analyzed the associations between the FGF21 and RBM3 serum concentrations on admission and at 72 h, body weight, maximum temperature during the first 24 h, and the outcome of patients at 3 months. We also analyzed their association with biomarkers of obesity (adiponectin and leptin) and inflammation (interleukin-6 (IL-6) and interleukin (IL-10)). Results: Higher concentrations of FGF21 on admission and RBM3 at 72 h were associated with good outcomes. Serum FGF21 and RBM3 were directly related to body mass index and inversely related to the maximum temperature during the first 24 h. We found a positive association between the FGF21 concentrations in obese patients with leptin and a negative correlation with adiponectin in non-obese participants. Conclusions: This clinical study demonstrates the association between RBM3 and FGF21 levels and the outcome of stroke patients. Although further investigations are required, these data support the pharmacological induction of RBM3 as a promising protective therapy.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - María Pérez-Mato
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Paseo de la Castellana 261, 28046 Madrid, Spain;
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Andrés da Silva-Candal
- Neurovascular Diseases Laboratory, Neurology Service, Biomedical Research Institute (INIBIC), University Hospital Complex of A Coruña, 15006 A Coruña, Spain;
| | - Ana Bugallo-Casal
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Sonia López-Amoedo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - María Candamo-Lourido
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
- Correspondence: ; Tel./Fax: +34-981951097
| |
Collapse
|
21
|
Dalton HJ, Berg RA, Nadkarni VM, Kochanek PM, Tisherman SA, Thiagarajan R, Alexander P, Bartlett RH. Cardiopulmonary Resuscitation and Rescue Therapies. Crit Care Med 2021; 49:1375-1388. [PMID: 34259654 DOI: 10.1097/ccm.0000000000005106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The history of cardiopulmonary resuscitation and the Society of Critical Care Medicine have much in common, as many of the founders of the Society of Critical Care Medicine focused on understanding and improving outcomes from cardiac arrest. We review the history, the current, and future state of cardiopulmonary resuscitation.
Collapse
Affiliation(s)
- Heidi J Dalton
- Heart and Vascular Institute and Department of Pediatrics, INOVA Fairfax Medical Center, Falls Church, VA. Department of Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA. Department of Anesthesiology/Critical Care Medicine, Peter Safer Resuscitation Center, Pittsburgh, PA. Department of Surgery, R Adams Cowley Shock Trauma Center, Baltimore, MD. Department of Cardiology, Division of Cardiovascular Critical Care, Boston Children's Hospital, Boston, MA. Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Arrich J, Herkner H, Müllner D, Behringer W. Targeted temperature management after cardiac arrest. A systematic review and meta-analysis of animal studies. Resuscitation 2021; 162:47-55. [PMID: 33582259 DOI: 10.1016/j.resuscitation.2021.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
AIM Animal studies are an important knowledge base when information from clinical trials is missing or conflicting. The goal of this systematic review and meta-analysis was to investigate the effect of conventional targeted temperature management (TTM) between 32-36 °C in animal cardiac arrest models, and to estimate the influence of effect modifiers on the pooled effect of TTM. DATA SOURCES We searched Medline and Scopus from inception to May 2020 for randomised controlled animal trials assessing the effect of conventional TTM versus normothermia on neurologic outcome after cardiac arrest. We extracted data on study characteristics, study quality data, neurologic outcome, mortality, and potential effect modifiers. RESULTS We retrieved 1635 studies, 45 studies comprising data of 981 animals met the inclusion criteria. Risk of bias was high in 17 studies and moderate in 28 studies. We undertook random-effects meta-analyses and meta-regression analyses to calculate the pooled effect and the influence of effect modifiers. There was a strong beneficial effect of TTM as compared to normothermia on neurologic outcome (standardised mean difference of 1.4 [95% CI -1.7 to -1.1; I2 = 75%]). Faster cooling rates, lower target temperature of TTM within the range of 32-36 °C, and shorter duration of cooling were independently associated with an increasing effect size of TTM. CONCLUSIONS This systematic review of animal cardiac arrest studies showed a consistent favourable effect of postresuscitation TTM as compared to normothermia on neurologic outcome that increased with lower target temperatures.
Collapse
Affiliation(s)
- Jasmin Arrich
- Department of Emergency Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Wien, Austria; Department of Emergency Medicine, Jena University Hospital, Friedrich Schiller University Jena, Faculty of Medicine, Am Klinikum 1, 07747 Jena, Germany.
| | - Harald Herkner
- Department of Emergency Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Wien, Austria
| | - David Müllner
- Department of Emergency Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Wien, Austria
| | - Wilhelm Behringer
- Department of Emergency Medicine, Jena University Hospital, Friedrich Schiller University Jena, Faculty of Medicine, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
23
|
Peretti D, Smith HL, Verity N, Humoud I, de Weerd L, Swinden DP, Hayes J, Mallucci GR. TrkB signaling regulates the cold-shock protein RBM3-mediated neuroprotection. Life Sci Alliance 2021; 4:4/4/e202000884. [PMID: 33563652 PMCID: PMC7893816 DOI: 10.26508/lsa.202000884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 02/02/2023] Open
Abstract
Increasing levels of the cold-shock protein, RNA-binding motif 3 (RBM3), either through cooling or by ectopic over-expression, prevents synapse and neuronal loss in mouse models of neurodegeneration. To exploit this process therapeutically requires an understanding of mechanisms controlling cold-induced RBM3 expression. Here, we show that cooling increases RBM3 through activation of TrkB via PLCγ1 and pCREB signaling. RBM3, in turn, has a hitherto unrecognized negative feedback on TrkB-induced ERK activation through induction of its specific phosphatase, DUSP6. Thus, RBM3 mediates structural plasticity through a distinct, non-canonical activation of TrkB signaling, which is abolished in RBM3-null neurons. Both genetic reduction and pharmacological antagonism of TrkB and its downstream mediators abrogate cooling-induced RBM3 induction and prevent structural plasticity, whereas TrkB inhibition similarly prevents RBM3 induction and the neuroprotective effects of cooling in prion-diseased mice. Conversely, TrkB agonism induces RBM3 without cooling, preventing synapse loss and neurodegeneration. TrkB signaling is, therefore, necessary for the induction of RBM3 and related neuroprotective effects and provides a target by which RBM3-mediated synapse-regenerative therapies in neurodegenerative disorders can be used therapeutically without the need for inducing hypothermia.
Collapse
Affiliation(s)
- Diego Peretti
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Heather L Smith
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas Verity
- MRC Toxicology Unit at the University of Cambridge, Leicester, UK
| | - Ibrahim Humoud
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Lis de Weerd
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Dean P Swinden
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Joseph Hayes
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Giovanna R Mallucci
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
24
|
Jackson TC, Gorse K, Herrmann JR, Kochanek PM. Hippocampal and Prefrontal Cortical Brain Tissue Levels of Irisin and GDF15 Receptor Subunits in Children. Mol Neurobiol 2021; 58:2145-2157. [PMID: 33411243 PMCID: PMC7788542 DOI: 10.1007/s12035-020-02250-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
Cold-stress hormones (CSHs) stimulate thermogenesis and have direct neuroprotective effects on the brain. The obligatory receptor components of two new CSHs (irisin and growth differentiation factor-15 [GDF15]) were recently discovered. Irisin binds integrin-αV/β5 heterodimers while GDF-15 binds to the orphan receptor glial cell-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL). In addition, integrin-αV/β5 was just identified as the key receptor mediating Zika virus infection in the CNS. We measured integrin-αV, integrin-β5, and GFRAL protein levels across 78 high-quality human male/female brain tissues in infants, toddlers, preschoolers, adolescent, and adults-providing the most robust analysis to date on their levels in the human cortex and hippocampus. We report that integrin-αV was detected at all ages in the prefrontal cortex with levels greatest in adults. Integrin-αV was also detected in the hippocampus in all age groups. In contrast, integrin-β5 was detected in cortex and hippocampus largely restricted to infants. Co-expression of integrin-αV/β5 in the human infant hippocampus and cortex suggests the possibility that irisin has a more robust effect on the developing vs. the adult brain and may have implications for Zika virus infection in infants and young children.
Collapse
Affiliation(s)
- Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0742, 560 Channelside Dr, Tampa, FL, 33602, USA.
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, 12901 Bruce B Downs BLVD, MDC 2532, Tampa, FL, 33612-4799, USA.
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0742, 560 Channelside Dr, Tampa, FL, 33602, USA
- Department of Molecular Pharmacology & Physiology, University of South Florida, Morsani College of Medicine, 12901 Bruce B Downs BLVD, MDC 2532, Tampa, FL, 33612-4799, USA
| | - Jeremy R Herrmann
- School of Medicine, Children's Hospital of Pittsburgh of UPMC, Safar Center for Resuscitation Research, University of Pittsburgh, John G. Rangos Research Center - 6th Floor, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Scaife Hall 3550 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Patrick M Kochanek
- School of Medicine, Children's Hospital of Pittsburgh of UPMC, Safar Center for Resuscitation Research, University of Pittsburgh, John G. Rangos Research Center - 6th Floor, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Scaife Hall 3550 Terrace Street, Pittsburgh, PA, 15213, USA
| |
Collapse
|
25
|
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
|
26
|
Rosenthal LM, Tong G, Wowro S, Walker C, Pfitzer C, Böttcher W, Miera O, Berger F, Schmitt KRL. A Prospective Clinical Trial Measuring the Effects of Cardiopulmonary Bypass Under Mild Hypothermia on the Inflammatory Response and Regulation of Cold-Shock Protein RNA-Binding Motif 3. Ther Hypothermia Temp Manag 2020; 10:60-70. [DOI: 10.1089/ther.2018.0038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lisa-Maria Rosenthal
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Giang Tong
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
| | - Sylvia Wowro
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
| | - Christoph Walker
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
| | - Constanze Pfitzer
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- German Centre for Cardiovascular Research (DHZK), Partner Site Berlin, Berlin, Germany
| | - Wolfgang Böttcher
- Department for Cardiovascular Perfusion, German Heart Institute Berlin, Berlin, Germany
| | - Oliver Miera
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
| | - Felix Berger
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DHZK), Partner Site Berlin, Berlin, Germany
- Department of Pediatric Cardiology, Charité–Universitaetsmedizin Berlin, Berlin, Germany
| | - Katharina Rose Luise Schmitt
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Institute Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DHZK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
27
|
Olai H, Thornéus G, Watson H, Macleod M, Rhodes J, Friberg H, Nielsen N, Cronberg T, Deierborg T. Meta-analysis of targeted temperature management in animal models of cardiac arrest. Intensive Care Med Exp 2020; 8:3. [PMID: 31953652 PMCID: PMC6969098 DOI: 10.1186/s40635-019-0291-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/29/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Targeted temperature management (TTM) of 32 to 34 °C has been the standard treatment for out-of-hospital cardiac arrest since clinical trials in 2002 indicated benefit on survival and neurological outcome. In 2013, a clinical trial showed no difference in outcome between TTM of 33 °C and TTM of 36 °C. In this meta-analysis, we investigate the evidence for TTM in animal models of cardiac arrest. METHODS We searched PubMed and EMBASE for adult animal studies using TTM as a treatment in different models of cardiac arrest or global brain ischemia which reported neurobehavioural outcome, brain histology or mortality. We used a random effects model to calculate estimates of efficacy and assessed risk of bias using an adapted eight-item version of the Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES) quality checklist. We also used a scoring system based on the recommendations of the Stroke Treatment Academic Industry Roundtable (STAIR), to assess the scope of testing in the field. Included studies which investigated a post-ischemic induction of TTM had their treatment regimens characterized with regard to depth, duration and time to treatment and scored against the modified STAIR criteria. RESULTS The initial and updated search generated 17809 studies after duplicate removal. One hundred eighty-one studies met the inclusion criteria, including data from 1,787, 6,495 and 2,945 animals for neurobehavioural, histological and mortality outcomes, respectively. TTM was favoured compared to control for all outcomes. TTM was beneficial using short and prolonged cooling, deep and moderate temperature reduction, and early and delayed time to treatment. Median [IQR] study quality was 4 [3 to 6]. Eighteen studies checked seven or more of the eight CAMARADES quality items. There was no clear correlation between study quality and efficacy for any outcome. STAIR analysis identified 102 studies investigating post-ischemic induction of TTM, comprising 147 different treatment regimens of TTM. Only 2 and 8 out of 147 regimens investigated comorbid and gyrencephalic animals, respectively. CONCLUSIONS TTM is beneficial under most experimental conditions in animal models of cardiac arrest or global brain ischemia. However, research on gyrencephalic species and especially comorbid animals is uncommon and a possible translational gap. Also, low study quality suggests risk of bias within studies. Future animal research should focus on mimicking the clinical scenario and employ similar rigour in trial design to that of modern clinical trials.
Collapse
Affiliation(s)
- Hilmer Olai
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Gustav Thornéus
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Hannah Watson
- Department of Anaesthesia, Western General Hospital, NHS Lothian, Edinburgh, UK
- Department of Critical Care, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Jonathan Rhodes
- Department of Anaesthesia, Critical care and Pain Medicine/NHS Lothian, University of Edinburgh, Edinburgh, UK
| | - Hans Friberg
- Department of Clinical Sciences, Anesthesia & Intensive care, Skåne University Hospital, Lund University, Lund, Sweden
| | - Niklas Nielsen
- Department of Clinical Sciences Lund, Anesthesia & Intensive care, Helsingborg Hospital, Lund University, Lund, Sweden
| | - Tobias Cronberg
- Department of Clinical Sciences Lund, Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden.
| |
Collapse
|
28
|
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
|
29
|
Specific BK Channel Activator NS11021 Protects Rat Renal Proximal Tubular Cells from Cold Storage-Induced Mitochondrial Injury In Vitro. Biomolecules 2019; 9:biom9120825. [PMID: 31817165 PMCID: PMC6995623 DOI: 10.3390/biom9120825] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
Kidneys from deceased donors used for transplantation are placed in cold storage (CS) solution during the search for a matched recipient. However, CS causes mitochondrial injury, which may exacerbate renal graft dysfunction. Here, we explored whether adding NS11021, an activator of the mitochondrial big-conductance calcium-activated K+ (mitoBK) channel, to CS solution can mitigate CS-induced mitochondrial injury. We used normal rat kidney proximal tubular epithelial (NRK) cells as an in vitro model of renal cold storage (18 h) and rewarming (2 h) (CS + RW). Western blots detected the pore-forming α subunit of the BK channel in mitochondrial fractions from NRK cells. The fluorescent K+-binding probe, PBFI-AM, revealed that isolated mitochondria from NRK cells exhibited mitoBK-mediated K+ uptake, which was impaired ~70% in NRK cells subjected to CS + RW compared to control NRK cells maintained at 37 °C. Importantly, the addition of 1 μM NS11021 to CS solution prevented CS + RW-induced impairment of mitoBK-mediated K+ uptake. The NS11021–treated NRK cells also exhibited less cell death and mitochondrial injury after CS + RW, including mitigated mitochondrial respiratory dysfunction, depolarization, and superoxide production. In summary, these new data show for the first time that mitoBK channels may represent a therapeutic target to prevent renal CS-induced injury.
Collapse
|
30
|
Jackson TC, Janesko-Feldman K, Carlson SW, Kotermanski SE, Kochanek PM. Robust RBM3 and β-klotho expression in developing neurons in the human brain. J Cereb Blood Flow Metab 2019; 39:2355-2367. [PMID: 31566073 PMCID: PMC6890998 DOI: 10.1177/0271678x19878889] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RNA binding motif 3 (RBM3) is a powerful neuroprotectant that inhibits neurodegenerative cell death in vivo and is a promising therapeutic target in brain ischemia. RBM3 is increased by the hormone fibroblast growth factor 21 (FGF21) in an age- and temperature-dependent manner in rat cortical neurons. FGF21 receptor binding is controlled by the transmembrane protein β-klotho, which is mostly absent in the adult brain. We discovered that RBM3/β-klotho is unexpectedly high in the human infant vs. adult brain (hippocampus/prefrontal cortex). The use of tissue homogenates in that study precluded a comparison of RBM3/β-klotho expression among different CNS cell-types, thus, omitted key evidence (i.e. confirmation of neuronal expression) that would otherwise provide a critical link to support their possible direct neuroprotective effects in humans. This report addresses that knowledge gap. High-quality fixed human hippocampus, cortex, and hypothalamic tissues were acquired from the NIH Neurobiobank (<1 yr (premature born) infants, 1 yr, 4 yr, and 34 yr). Dual labeling of cell-type markers vs. RBM3/β-klotho revealed enriched staining of targets in neurons in the developing brain. Identifying that RBM3/β-klotho is abundant in neurons in the immature brain is fundamentally important to guide protocol design and conceptual frameworks germane to future testing of these neuroprotective pathways in humans.
Collapse
Affiliation(s)
- Travis C Jackson
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, School of Medicine Children's Hospital of Pittsburgh of UPMC John G. Rangos Research Center, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shawn E Kotermanski
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, School of Medicine Children's Hospital of Pittsburgh of UPMC John G. Rangos Research Center, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Yan J, Goerne T, Zelmer A, Guzman R, Kapfhammer JP, Wellmann S, Zhu X. The RNA-Binding Protein RBM3 Promotes Neural Stem Cell (NSC) Proliferation Under Hypoxia. Front Cell Dev Biol 2019; 7:288. [PMID: 31824945 PMCID: PMC6881237 DOI: 10.3389/fcell.2019.00288] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Neural stem cells (NSCs) reside physiologically in a hypoxic niche to maintain self-renewal and multipotency. Whereas mild hypoxia is known to promote NSC proliferation, severe hypoxia in pathological conditions exerts the reverse effect. The multi-functional RNA-binding protein RBM3 is abundant in NSCs and can be regulated by hypoxic exposure. Although RBM3 has been shown to accelerate cell growth in many cell types, whether and how it affects NSC proliferation in hypoxic environment remains largely unknown. In this study, we tested how RBM3 regulates cell proliferation under hypoxia in C17.2 mouse NSC cell line and in primary mouse NSCs from both the forebrain of postnatal day 0 (P0) mice and the subgranular zone (SGZ) of adult mice. Our results demonstrated that RBM3 expression was highly sensitive to hypoxia, and NSCs were arrested in G0/G1 phase by 5, 2.5, and 1% O2 treatment. When we overexpressed RBM3, hypoxia-induced cell cycle arrest in G0/G1 phase was relieved and more cell transit into S phase was observed. Furthermore, cell viability under hypoxia was also increased by RBM3. In contrast, in RBM3-depleted primary NSCs, less BrdU-incorporated cells were detected, indicating exacerbated cell cycle arrest in G1 to S phase transition. Instead, overexpressed RBM3 significantly increased proliferation ratio in primary NSCs. Our findings indicate RBM3 as a potential target to maintain the proliferation capacity of NSCs under hypoxia, which can be important in NSC-based therapies of acute brain injury and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingyi Yan
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Tessa Goerne
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Andrea Zelmer
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland.,Department of Neonatology, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Xinzhou Zhu
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| |
Collapse
|
32
|
Yang H, Zhuang R, Li Y, Li T, Yuan X, Lei B, Xie Y, Wang M. Cold-inducible protein RBM3 mediates hypothermic neuroprotection against neurotoxin rotenone via inhibition on MAPK signalling. J Cell Mol Med 2019; 23:7010-7020. [PMID: 31436914 PMCID: PMC6787511 DOI: 10.1111/jcmm.14588] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/11/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Mild hypothermia and its key product, cold-inducible protein RBM3, possess robust neuroprotective effects against various neurotoxins. However, we previously showed that mild hypothermia fails to attenuate the neurotoxicity from MPP+ , one of typical neurotoxins related to the increasing risk of Parkinson disease (PD). To better understand the role of mild hypothermia and RBM3 in PD progression, another known PD-related neurotoxin, rotenone (ROT) was utilized in this study. Using immunoblotting, cell viability assays and TUNEL staining, we revealed that mild hypothermia (32°C) significantly reduced the apoptosis induced by ROT in human neuroblastoma SH-SY5Y cells, when compared to normothermia (37°C). Meanwhile, the overexpression of RBM3 in SH-SY5Y cells mimicked the neuroprotective effects of mild hypothermia on ROT-induced cytotoxicity. Upon ROT stimulation, MAPK signalling like p38, JNK and ERK, and AMPK and GSK-3β signalling were activated. When RBM3 was overexpressed, only the activation of p38, JNK and ERK signalling was inhibited, leaving AMPK and GSK-3β signalling unaffected. Similarly, mild hypothermia also inhibited the activation of MAPKs induced by ROT. Lastly, it was demonstrated that the MAPK (especially p38 and ERK) inhibition by their individual inhibitors significantly decreased the neurotoxicity of ROT in SH-SY5Y cells. In conclusion, these data demonstrate that RBM3 mediates mild hypothermia-related neuroprotection against ROT by inhibiting the MAPK signalling of p38, JNK and ERK.
Collapse
Affiliation(s)
- Hai‐Jie Yang
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological PsychiatrySecond Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Rui‐Juan Zhuang
- School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Yuan‐Bo Li
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Tian Li
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Xin Yuan
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Bing‐Bing Lei
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Yun‐Fei Xie
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Mian Wang
- School of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
33
|
Aziz M, Brenner M, Wang P. Extracellular CIRP (eCIRP) and inflammation. J Leukoc Biol 2019; 106:133-146. [PMID: 30645013 PMCID: PMC6597266 DOI: 10.1002/jlb.3mir1118-443r] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/27/2018] [Accepted: 12/30/2018] [Indexed: 12/22/2022] Open
Abstract
Cold-inducible RNA-binding protein (CIRP) was discovered 2 decades ago while studying the mechanism of cold stress adaptation in mammals. Since then, the role of intracellular CIRP (iCIRP) as a stress-response protein has been extensively studied. Recently, extracellular CIRP (eCIRP) was discovered to also have an important role, acting as a damage-associated molecular pattern, raising critical implications for the pathobiology of inflammatory diseases. During hemorrhagic shock and sepsis, inflammation triggers the translocation of CIRP from the nucleus to the cytosol and its release to the extracellular space. eCIRP then induces inflammatory responses in macrophages, neutrophils, lymphocytes, and dendritic cells. eCIRP also induces endoplasmic reticulum stress and pyroptosis in endothelial cells by activating the NF-κB and inflammasome pathways, and necroptosis in macrophages via mitochondrial DNA damage. eCIRP works through the TLR4-MD2 receptors. Studies with CIRP-/- mice reveal protection against inflammation, implicating eCIRP to be a novel drug target. Anti-CIRP Ab or CIRP-derived small peptide may have effective therapeutic potentials in sepsis, acute lung injury, and organ ischemia/reperfusion injuries. The current review focuses on the pathobiology of eCIRP by emphasizing on signal transduction machineries, leading to discovering novel therapeutic interventions targeting eCIRP in various inflammatory diseases.
Collapse
Affiliation(s)
- Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset,
NY
| |
Collapse
|
34
|
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
|
35
|
A "Metamorphosis" in Our Approach to Treatment Is Not Likely to Result From a Meta-Analysis of the Use of Therapeutic Hypothermia in Severe Traumatic Brain Injury. Crit Care Med 2019; 45:744-745. [PMID: 28291098 DOI: 10.1097/ccm.0000000000002240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
|
37
|
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
|
38
|
Downie Ruiz Velasco A, Welten SMJ, Goossens EAC, Quax PHA, Rappsilber J, Michlewski G, Nossent AY. Posttranscriptional Regulation of 14q32 MicroRNAs by the CIRBP and HADHB during Vascular Regeneration after Ischemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:329-338. [PMID: 30665182 PMCID: PMC6350214 DOI: 10.1016/j.omtn.2018.11.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022]
Abstract
After induction of ischemia in mice, 14q32 microRNAs are regulated in three distinct temporal patterns. These expression patterns, as well as basal expression levels, are independent of the microRNA genes’ order in the 14q32 locus. This implies that posttranscriptional processing is a major determinant of 14q32 microRNA expression. Therefore, we hypothesized that RNA binding proteins (RBPs) regulate posttranscriptional processing of 14q32, and we aimed to identify these RBPs. To identify proteins responsible for this posttranscriptional regulation, we used RNA pull-down SILAC mass spectrometry (RP-SMS) on selected precursor microRNAs. We observed differential binding of cold-inducible RBP (CIRBP) and hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit beta (HADHB) to the precursors of late-upregulated miR-329-3p and unaffected miR-495-3p. Immunohistochemical staining confirmed expression of both CIRBP and HADHB in the adductor muscle of mice. Expression of both CIRBP and HADHB was upregulated after hindlimb ischemia in mice. Using RBP immunoprecipitation experiments, we showed specific binding of CIRBP to pre-miR-329 but not to pri-miR-329. Finally, using CRISPR/Cas9, we generated HADHB−/− 3T3 cells, which display reduced expression of miR-329 and miR-495 but not their precursors. These data suggest a novel role for CIRBP and HADHB in posttranscriptional regulation of 14q32 microRNAs.
Collapse
Affiliation(s)
- Angela Downie Ruiz Velasco
- Division of Infection and Pathway Medicine, University of Edinburgh, The Chancellor's Building, Edinburgh, UK; The Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Sabine M J Welten
- Department of Surgery, Leiden University Medical, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical, Leiden, the Netherlands
| | - Eveline A C Goossens
- Department of Surgery, Leiden University Medical, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical, Leiden, the Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical, Leiden, the Netherlands
| | - Juri Rappsilber
- The Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK; Department of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Gracjan Michlewski
- Division of Infection and Pathway Medicine, University of Edinburgh, The Chancellor's Building, Edinburgh, UK; The Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK; Zhejiang University - University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, Zhejiang, P.R. China.
| | - A Yaël Nossent
- Department of Surgery, Leiden University Medical, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical, Leiden, the Netherlands; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.
| |
Collapse
|
39
|
Jha RM, Kochanek PM. A Precision Medicine Approach to Cerebral Edema and Intracranial Hypertension after Severe Traumatic Brain Injury: Quo Vadis? Curr Neurol Neurosci Rep 2018; 18:105. [PMID: 30406315 PMCID: PMC6589108 DOI: 10.1007/s11910-018-0912-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Standard clinical protocols for treating cerebral edema and intracranial hypertension after severe TBI have remained remarkably similar over decades. Cerebral edema and intracranial hypertension are treated interchangeably when in fact intracranial pressure (ICP) is a proxy for cerebral edema but also other processes such as extent of mass lesions, hydrocephalus, or cerebral blood volume. A complex interplay of multiple molecular mechanisms results in cerebral edema after severe TBI, and these are not measured or targeted by current clinically available tools. Addressing these underpinnings may be key to preventing or treating cerebral edema and improving outcome after severe TBI. RECENT FINDINGS This review begins by outlining basic principles underlying the relationship between edema and ICP including the Monro-Kellie doctrine and concepts of intracranial compliance/elastance. There is a subsequent brief discussion of current guidelines for ICP monitoring/management. We then focus most of the review on an evolving precision medicine approach towards cerebral edema and intracranial hypertension after TBI. Personalization of invasive neuromonitoring parameters including ICP waveform analysis, pulse amplitude, pressure reactivity, and longitudinal trajectories are presented. This is followed by a discussion of cerebral edema subtypes (continuum of ionic/cytotoxic/vasogenic edema and progressive secondary hemorrhage). Mechanisms of potential molecular contributors to cerebral edema after TBI are reviewed. For each target, we present findings from preclinical models, and evaluate their clinical utility as biomarkers and therapeutic targets for cerebral edema reduction. This selection represents promising candidates with evidence from different research groups, overlap/inter-relatedness with other pathways, and clinical/translational potential. We outline an evolving precision medicine and translational approach towards cerebral edema and intracranial hypertension after severe TBI.
Collapse
Affiliation(s)
- Ruchira M Jha
- Department of Critical Care Medicine, Room 646A, Scaife Hall, 3550 Terrace Street, Pittsburgh, 15261, PA, USA.
- Safar Center for Resuscitation Research John G. Rangos Research Center, 6th Floor; 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Patrick M Kochanek
- Department of Critical Care Medicine, Room 646A, Scaife Hall, 3550 Terrace Street, Pittsburgh, 15261, PA, USA
- Safar Center for Resuscitation Research John G. Rangos Research Center, 6th Floor; 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh John G. Rangos Research Center, 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| |
Collapse
|
40
|
Jackson TC, Kotermanski SE, Kochanek PM. Infants Uniquely Express High Levels of RBM3 and Other Cold-Adaptive Neuroprotectant Proteins in the Human Brain. Dev Neurosci 2018; 40:325-336. [PMID: 30399610 DOI: 10.1159/000493637] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022] Open
Abstract
Neuroprotective cold-shock proteins (CSPs) are abundant in the normothermic neonatal rodent brain but decrease with advancing neurodevelopmental age and are low or absent in the adult brain. It has not been established if neurodevelopmental age alters the baseline expression of CSPs in the human brain. Here, we tested the hypothesis that protein levels of RNA-binding motif 3 (RBM3), reticulon-3 (RTN3), and cold-induced RNA-binding protein (CIRBP) are abundant in the normothermic developing human brain but low-to-absent in adults. We also tested if β-klotho (KLB) is expressed in the developing brain; KLB functions as a coreceptor that controls tissue-specific binding and activity of the systemically circulating thermogenic hormone fibroblast growth factor 21 (FGF21), and is predominantly expressed in the liver, pancreas, and in adipose cells. Methods: Hippocampi and anterior prefrontal cortices (aPFCs/BA10) from a total of 20 male and 20 female subjects were obtained from the NIH NeuroBioBank. CSP and KLB levels were measured in: infants < 1 year old (n = 8), toddlers aged 1-2 years (n = 8), children aged 3-5 years (n = 7), 18-year-old adolescents (n = 8), and adults aged 31-34 years (n = 8). An equal number of male and female (n = 4 each) samples were pooled into each age group, except in the 3- to 5-year-olds which comprised 3 male and 4 female specimens due to sample availability. In total, 78 whole-brain tissues were dissociated using a bead-based Precellys homogenizer to generate equivalent homogenates, and levels of protein targets subsequently analyzed by Western blotting. Results: Infants had the highest levels of RBM3 and other CSPs in the brain compared to all other ages. In the hippocampus, CSPs were detected predominantly in infants. In the aPFC, CSP levels were highest in infants, moderate-to-low in toddlers/children, and below assay detection limits in adolescents/adults. Germane to the thermogenic FGF21/KLB signaling axis, our results confirm that KLB is absent in the adult hippocampus/aPFC as reported by others. In contrast, we report for the first time that KLB is abundant in the early developing human brain; KLB levels were highest in the infant hippocampus/aPFC and moderately expressed in toddlers. RBM3 is a potent neuroprotective CSP. Thus, the impact of these findings on the observed efficacy of therapeutic hypothermia in neonatal brain injury merits further investigation.
Collapse
Affiliation(s)
- Travis C Jackson
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, John G. Rangos Research Center, Pittsburgh, Pennsylvania, USA, .,Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Scaife Hall, Pittsburgh, Pennsylvania, USA,
| | - Shawn E Kotermanski
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, School of Medicine, Bridgeside Point Building 1, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, John G. Rangos Research Center, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Scaife Hall, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
41
|
Duration of therapeutic hypothermia or targeted temperature management in pediatric cardiac arrest: Seeing through the ice. Resuscitation 2018; 133:A3-A4. [PMID: 30278203 DOI: 10.1016/j.resuscitation.2018.09.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/27/2018] [Indexed: 11/23/2022]
|
42
|
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
|
43
|
Atkins CM, Bramlett HM, Dietrich WD. Is temperature an important variable in recovery after mild traumatic brain injury? F1000Res 2017; 6:2031. [PMID: 29188026 PMCID: PMC5698917 DOI: 10.12688/f1000research.12025.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2017] [Indexed: 12/03/2022] Open
Abstract
With nearly 42 million mild traumatic brain injuries (mTBIs) occurring worldwide every year, understanding the factors that may adversely influence recovery after mTBI is important for developing guidelines in mTBI management. Extensive clinical evidence exists documenting the detrimental effects of elevated temperature levels on recovery after moderate to severe TBI. However, whether elevated temperature alters recovery after mTBI or concussion is an active area of investigation. Individuals engaged in exercise and competitive sports regularly experience body and brain temperature increases to hyperthermic levels and these temperature increases are prolonged in hot and humid ambient environments. Thus, there is a strong potential for hyperthermia to alter recovery after mTBI in a subset of individuals at risk for mTBI. Preclinical mTBI studies have found that elevating brain temperature to 39°C before mTBI significantly increases neuronal death within the cortex and hippocampus and also worsens cognitive deficits. This review summarizes the pathology and behavioral problems of mTBI that are exacerbated by hyperthermia and discusses whether hyperthermia is a variable that should be considered after concussion and mTBI. Finally, underlying pathophysiological mechanisms responsible for hyperthermia-induced altered responses to mTBI and potential gender considerations are discussed.
Collapse
Affiliation(s)
- Coleen M Atkins
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Lois Pope LIFE Center, 1095 NW 14th Terrace (R-48), Miami, FL, 33136-1060, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Lois Pope LIFE Center, 1095 NW 14th Terrace (R-48), Miami, FL, 33136-1060, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Lois Pope LIFE Center, 1095 NW 14th Terrace (R-48), Miami, FL, 33136-1060, USA
| |
Collapse
|
44
|
Kochanek PM, Jackson TC. Therapeutic Hypothermia and Targeted Temperature Management With or Without the "Cold Stress" Response. Ther Hypothermia Temp Manag 2017; 7:134-136. [PMID: 28800291 PMCID: PMC5610381 DOI: 10.1089/ther.2017.0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Patrick M Kochanek
- Department of Critical Care Medicine/Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Travis C Jackson
- Department of Critical Care Medicine/Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
45
|
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]
|
46
|
Seebacher F, Little AG. Plasticity of Performance Curves Can Buffer Reaction Rates from Body Temperature Variation in Active Endotherms. Front Physiol 2017; 8:575. [PMID: 28824463 PMCID: PMC5543086 DOI: 10.3389/fphys.2017.00575] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022] Open
Abstract
Endotherms regulate their core body temperature by adjusting metabolic heat production and insulation. Endothermic body temperatures are therefore relatively stable compared to external temperatures. The thermal sensitivity of biochemical reaction rates is thought to have co-evolved with body temperature regulation so that optimal reaction rates occur at the regulated body temperature. However, recent data show that core body temperatures even of non-torpid endotherms fluctuate considerably. Additionally, peripheral temperatures can be considerably lower and more variable than core body temperatures. Here we discuss whether published data support the hypothesis that thermal performance curves of physiological reaction rates are plastic so that performance is maintained despite variable body temperatures within active (non-torpid) endotherms, and we explore mechanisms that confer plasticity. There is evidence that thermal performance curves in tissues that experience thermal fluctuations can be plastic, although this question remains relatively unexplored for endotherms. Mechanisms that alter thermal responses locally at the tissue level include transient potential receptor ion channels (TRPV and TRPM) and the AMP-activated protein kinase (AMPK) both of which can influence metabolism and energy expenditure. Additionally, the thermal sensitivity of processes that cause post-transcriptional RNA degradation can promote the relative expression of cold-responsive genes. Endotherms can respond to environmental fluctuations similarly to ectotherms, and thermal plasticity complements core body temperature regulation to increase whole-organism performance. Thermal plasticity is ancestral to endothermic thermoregulation, but it has not lost its selective advantage so that modern endotherms are a physiological composite of ancestral ectothermic and derived endothermic traits.
Collapse
Affiliation(s)
- Frank Seebacher
- School of Life and Environmental Sciences, University of SydneySydney, NSW, Australia
| | - Alexander G Little
- Rosenstiel School of Marine and Atmospheric Science, The University of MiamiMiami, FL, United States
| |
Collapse
|
47
|
Kapinos G, Becker LB. The American Academy of Neurology affirms the revival of cooling for the revived. Neurology 2017; 88:2076-2077. [DOI: 10.1212/wnl.0000000000003968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
48
|
Jackson EK, Kotermanski SE, Menshikova EV, Dubey RK, Jackson TC, Kochanek PM. Adenosine production by brain cells. J Neurochem 2017; 141:676-693. [PMID: 28294336 DOI: 10.1111/jnc.14018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 02/06/2023]
Abstract
The early release of adenosine following traumatic brain injury (TBI) suppresses seizures and brain inflammation; thus, it is important to elucidate the cellular sources of adenosine following injurious stimuli triggered by TBI so that therapeutics for enhancing the early adenosine-release response can be optimized. Using mass spectrometry with 13 C-labeled standards, we investigated in cultured rat neurons, astrocytes, and microglia the effects of oxygen-glucose deprivation (OGD; models energy failure), H2 O2 (produces oxidative stress), and glutamate (induces excitotoxicity) on intracellular and extracellular levels of 5'-AMP (adenosine precursor), adenosine, and inosine and hypoxanthine (adenosine metabolites). In neurons, OGD triggered increases in intracellular 5'-AMP (2.8-fold), adenosine (2.6-fold), inosine (2.2-fold), and hypoxanthine (5.3-fold) and extracellular 5'-AMP (2.2-fold), adenosine (2.4-fold), and hypoxanthine (2.5-fold). In neurons, H2 O2 did not affect intracellular or extracellular purines; yet, glutamate increased intracellular adenosine, inosine, and hypoxanthine (1.7-fold, 1.7-fold, and 1.6-fold, respectively) and extracellular adenosine, inosine, and hypoxanthine (2.9-fold, 2.1-fold, and 1.6-fold, respectively). In astrocytes, neither H2 O2 nor glutamate affected intracellular or extracellular purines, and OGD only slightly increased intracellular and extracellular hypoxanthine. Microglia were unresponsive to OGD and glutamate, but were remarkably responsive to H2 O2 , which increased intracellular 5'-AMP (1.6-fold), adenosine (1.6-fold), inosine (2.1-fold), and hypoxanthine (1.6-fold) and extracellular 5'-AMP (5.9-fold), adenosine (4.0-fold), inosine (4.3-fold), and hypoxanthine (1.9-fold). CONCLUSION Under these particular experimental conditions, cultured neurons are the main contributors to adenosine production/release in response to OGD and glutamate, whereas cultured microglia are the main contributors upon oxidative stress. Developing therapeutics that recruit astrocytes to produce/release adenosine could have beneficial effects in TBI.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shawn E Kotermanski
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elizabeth V Menshikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Raghvendra K Dubey
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Reproductive Endocrinology, University Hospital Zurich and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Travis C Jackson
- Department of Critical Care Medicine and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
49
|
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: 157] [Impact Index Per Article: 17.4] [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
|
50
|
Anderson KB, Poloyac SM, Kochanek PM, Empey PE. Effect of Hypothermia and Targeted Temperature Management on Drug Disposition and Response Following Cardiac Arrest: A Comprehensive Review of Preclinical and Clinical Investigations. Ther Hypothermia Temp Manag 2016; 6:169-179. [PMID: 27622966 DOI: 10.1089/ther.2016.0003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Targeted temperature management (TTM) has been shown to reduce mortality and improve neurological outcomes in out-of-hospital cardiac arrest (CA) patients and in neonates with hypoxic-ischemic encephalopathy (HIE). TTM has also been associated with adverse drug events in the critically ill patient due to its effect on drug pharmacokinetics (PKs) and pharmacodynamics (PDs). We aim to evaluate the current literature on the effect of TTM on drug PKs and PDs following CA. MEDLINE/PubMed databases were searched for publications, which include the MeSH terms hypothermia, drug metabolism, drug transport, P450, critical care, cardiac arrest, hypoxic-ischemic encephalopathy, pharmacokinetics, and pharmacodynamics between July 2006 and October 2015. Twenty-three studies were included in this review. The studies demonstrate that hypothermia impacts PK parameters and increases concentrations of cytochrome-P450-metabolized drugs in the cooling and rewarming phase. Furthermore, the current data demonstrate a combined effect of CA and hypothermia on drug PK. Importantly, these effects can last greater than 4-5 days post-treatment. Limited evidence suggests hypothermia-mediated changes in the Phase II metabolism and the Phase III transport of drugs. Hypothermia also has been shown to potentially decrease the effect of specific drugs at the receptor level. Therapeutic hypothermia, as commonly deployed/applied during TTM, alters PK, and elevates concentrations of several commonly used medications. Hypothermia-mediated effects are an important factor when dosing and monitoring patients undergoing TTM treatment.
Collapse
Affiliation(s)
- Kacey B Anderson
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Samuel M Poloyac
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 2 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Philip E Empey
- 3 Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|