1
|
Snyder K, Dixon CE, Henchir J, Gorse K, Vagni VA, Janesko-Feldman K, Kochanek PM, Jackson TC. Gene knockout of RNA binding motif 5 in the brain alters RIMS2 protein homeostasis in the cerebellum and Hippocampus and exacerbates behavioral deficits after a TBI in mice. Exp Neurol 2024; 374:114690. [PMID: 38218585 PMCID: PMC11178365 DOI: 10.1016/j.expneurol.2024.114690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/28/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
RNA binding motif 5 (RBM5) is a tumor suppressor in cancer but its role in the brain is unclear. We used conditional gene knockout (KO) mice to test if RBM5 inhibition in the brain affects chronic cortical brain tissue survival or function after a controlled cortical impact (CCI) traumatic brain injury (TBI). RBM5 KO decreased baseline contralateral hemispheric volume (p < 0.0001) and exacerbated ipsilateral tissue loss at 21 d after CCI in male mice vs. wild type (WT) (p = 0.0019). CCI injury, but not RBM5 KO, impaired beam balance performance (0-5d post-injury) and swim speed on the Morris Water Maze (MWM) (19-20d) (p < 0.0001). RBM5 KO was associated with mild learning impairment in female mice (p = 0.0426), reflected as a modest increase in escape latency early in training (14-18d post-injury). However, KO did not affect spatial memory at 19d post-injury in male or in female mice but it was impaired by CCI in females (p = 0.0061). RBM5 KO was associated with impaired visual function in male mice on the visible platform test at 20d post-injury (p = 0.0256). To explore signaling disturbances in KOs related to behavior, we first cross-referenced known brain-specific RBM5-regulated gene targets with genes in the curated RetNet database that impact vision. We then performed a secondary literature search on RBM5-regulated genes with a putative role in hippocampal function. Regulating synaptic membrane exocytosis 2 (RIMS) 2 was identified as a gene of interest because it regulates both vision and hippocampal function. Immunoprecipitation and western blot confirmed protein expression of a novel ~170 kDa RIMS2 variant in the cerebellum, and in the hippocampus, it was significantly increased in KO vs WT (p < 0.0001), and in a sex-dependent manner (p = 0.0390). Furthermore, male KOs had decreased total canonical RIMS2 levels in the cerebellum (p = 0.0027) and hippocampus (p < 0.0001), whereas female KOs had increased total RIMS1 levels in the cerebellum (p = 0.0389). In summary, RBM5 modulates brain function in mammals. Future work is needed to test if RBM5 dependent regulation of RIMS2 splicing effects vision and cognition, and to verify potential sex differences on behavior in a larger cohort of mice.
Collapse
Affiliation(s)
- Kara Snyder
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| | - C Edward Dixon
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Jeremy Henchir
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6(th) floor, Pittsburgh, PA 15224, United States of America.
| | - Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL 33602, United States of America; University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, United States of America.
| |
Collapse
|
2
|
Snyder K, Gorse K, Kochanek PM, Jackson TC. Neuronal RBM5 modulates cell signaling responses to traumatic and hypoxic-ischemic injury in a sex-dependent manner. Cell Death Discov 2023; 9:379. [PMID: 37848418 PMCID: PMC10582027 DOI: 10.1038/s41420-023-01677-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023] Open
Abstract
It is not clear if inhibiting the pro-death gene RNA binding motif 5 (RBM5) is neuroprotective in isolated primary neurons or if it regulates cell survival in a sex-dependent manner. Here we established sex-dichotomized primary cortical neuron cultures from transgenic mice harboring a floxed RBM5 gene-trap. Lentivirus-mediated expression of CRE was used to silence RBM5 expression. Male and female neurons were maintained in next-generation Neurobasal-Plus media and subjected to a mechanical stretch-injury (to model traumatic brain injury) or oxygen-glucose deprivation/OGD (to model ischemia). RBM5 KO did not affect 24 h post-injury survival as determined by lactate dehydrogenase (LDH) release, in either paradigm. In contrast, female KO neurons had increased spectrin breakdown products post-insult (in both models). Furthermore, in OGD, RBM5 KO in male neurons exacerbated injury-induced downregulation of pro-survival AKT activation (pAKT473) but conversely led to pAKT473 sparing in female neurons. Moreover, global proteomics identified 19 differentially expressed (DE) proteins in OGD-injured male neurons, and 102 DE proteins in injured female neurons. Two novel RBM5-regulated proteins (PIGQ and EST1C) were identified in injured male KO neurons, and 8 novel proteins identified in injured female KO neurons (S35A5, DHTK1, STX3, IF3M, RN167, K1C14, DYHS, and MED13). In summary, RBM5 inhibition does not modify neuronal survival in primary mouse neurons in 2 clinically relevant models of excitotoxic insult, but RBM5 does regulate intracellular responses to injury in a sex-dependent manner.
Collapse
Affiliation(s)
- Kara Snyder
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA.
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
3
|
Farooq J, Snyder K, Janesko-Feldman K, Gorse K, Vagni V, Kochanek PM, Jackson TC. RNA Binding Motif 5 Gene Deletion Modulates Cell Signaling in a Sex-Dependent Manner but not Hippocampal Cell Death. J Neurotrauma 2022; 39:577-589. [PMID: 35152732 PMCID: PMC8978574 DOI: 10.1089/neu.2021.0362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
RNA-binding motif 5 (RBM5) is a pro-death tumor suppressor gene in cancer cells. It remains to be determined if it is neurotoxic in the brain or rather if it plays a fundamentally different role in the central nervous system (CNS). Brain-specific RBM5 knockout (KO) mice were given a controlled cortical impact (CCI) traumatic brain injury (TBI). Markers of acute cellular damage and repair were measured in hippocampal homogenates 48 h post-CCI. Hippocampal CA1/CA3 cell counts were assessed 7 days post-CCI to determine if early changes in injury markers were associated with histological outcome. No genotype-dependent differences were found in the levels of apoptotic markers (caspase 3, caspase 6, and caspase 9). However, KO females had a paradoxical increase in markers of pro-death calpain activation (145/150-spectrin and breakdown products [SBDP]) and in DNA repair/survival markers. (pH2A.x and pCREB). CCI-injured male KOs had a significant increase in phosphorylated calcium/calmodulin-dependent protein kinase II (pCaMKII). Despite sex/genotype-dependent differences in KOs in the levels of acute cell signaling targets involved in cell death pathways, 7 day hippocampal neuronal survival did not differ from that of wild types (WTs). Similarly, no differences in astrogliosis were observed. Finally, gene analysis revealed increased estrogen receptor α (ERα) levels in the KO hippocampus in females and may suggest a novel mechanism to explain sex-dimorphic effects on cell signaling. In summary, RBM5 inhibition did not affect hippocampal survival after a TBI in vivo but did modify targets involved in neural signal transduction/Ca2+ signaling pathways. Findings here support the view that RBM5 may serve a purpose in the CNS that is dissimilar from its traditional pro-death role in cancer.
Collapse
Affiliation(s)
- Jeffrey Farooq
- University of South Florida, 7831, Molecular Pharmacology and Physiology, Tampa, Florida, United States
- USF Health Morsani College of Medicine, 33697, USF Health Heart Institute, Tampa, Florida, United States
| | - Kara Snyder
- University of South Florida, 7831, Molecular Pharmacology and Physiology, Tampa, Florida, United States
- USF Health Morsani College of Medicine, 33697, USF Health Heart Institute, Tampa, Florida, United States
| | - Keri Janesko-Feldman
- University of Pittsburgh School of Medicine, Critical Care Medicine, Pittsburgh, Pennsylvania, United States,
| | - Kiersten Gorse
- University of South Florida, 7831, Molecular Pharmacology and Physiology, Tampa, Florida, United States
- USF Health Morsani College of Medicine, 33697, USF Health Heart Institute, Tampa, Florida, United States
| | - Vincent Vagni
- University of Pittsburgh School of Medicine, Critical Care Medicine, Pittsburgh, Pennsylvania, United States,
| | - Patrick M. Kochanek
- University of Pittsburgh School of Medicine, Critical Care Medicine, John G. Rangos Research Center, Safar Center for Resuscitation Research, 4401 Penn Avenue, Pittsburgh, Pennsylvania, United States, 15224
- United States
| | - Travis C. Jackson
- University of South Florida, 7831, Molecular Pharmacology and Physiology, 4202 E Fowler Ave, Tampa, Florida, United States, 33620-9951
- USF Health Morsani College of Medicine, 33697, USF Health Heart Institute, 560 Channelside Dr, Tampa, Florida, United States, 33602
| |
Collapse
|
4
|
Fitzgerald J, Fahey L, Holleran L, Ó Broin P, Donohoe G, Morris DW. Thirteen Independent Genetic Loci Associated with Preserved Processing Speed in a Study of Cognitive Resilience in 330,097 Individuals in the UK Biobank. Genes (Basel) 2022; 13:122. [PMID: 35052462 PMCID: PMC8774848 DOI: 10.3390/genes13010122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 02/04/2023] Open
Abstract
Cognitive resilience is the ability to withstand the negative effects of stress on cognitive functioning and is important for maintaining quality of life while aging. The UK Biobank does not have measurements of the same cognitive phenotype at distal time points. Therefore, we used education years (EY) as a proxy phenotype for past cognitive performance and current cognitive performance was based on processing speed. This represented an average time span of 40 years between past and current cognitive performance in 330,097 individuals. A confounding factor was that EY is highly polygenic and masked the genetics of resilience. To overcome this, we employed Genomics Structural Equation Modelling (GenomicSEM) to perform a genome-wide association study (GWAS)-by-subtraction using two GWAS, one GWAS of EY and resilience and a second GWAS of EY but not resilience, to generate a GWAS of Resilience. Using independent discovery and replication samples, we found 13 independent genetic loci for Resilience. Functional analyses showed enrichment in several brain regions and specific cell types. Gene-set analyses implicated the biological process "neuron differentiation", the cellular component "synaptic part" and the "WNT signalosome". Mendelian randomisation analysis showed a causative effect of white matter volume on cognitive resilience. These results may contribute to the neurobiological understanding of resilience.
Collapse
Affiliation(s)
- Joan Fitzgerald
- Cognitive Genetics and Cognitive Therapy Group, Centre for Neuroimaging & Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, H91 TK33 Galway, Ireland; (J.F.); (L.F.); (L.H.); (G.D.)
| | - Laura Fahey
- Cognitive Genetics and Cognitive Therapy Group, Centre for Neuroimaging & Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, H91 TK33 Galway, Ireland; (J.F.); (L.F.); (L.H.); (G.D.)
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, H91 TK33 Galway, Ireland;
| | - Laurena Holleran
- Cognitive Genetics and Cognitive Therapy Group, Centre for Neuroimaging & Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, H91 TK33 Galway, Ireland; (J.F.); (L.F.); (L.H.); (G.D.)
| | - Pilib Ó Broin
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, H91 TK33 Galway, Ireland;
| | - Gary Donohoe
- Cognitive Genetics and Cognitive Therapy Group, Centre for Neuroimaging & Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, H91 TK33 Galway, Ireland; (J.F.); (L.F.); (L.H.); (G.D.)
| | - Derek W. Morris
- Cognitive Genetics and Cognitive Therapy Group, Centre for Neuroimaging & Cognitive Genomics, School of Psychology and Discipline of Biochemistry, National University of Ireland Galway, H91 TK33 Galway, Ireland; (J.F.); (L.F.); (L.H.); (G.D.)
| |
Collapse
|
5
|
Z-Guggulsterone alleviated oxidative stress and inflammation through inhibiting the TXNIP/NLRP3 axis in ischemic stroke. Int Immunopharmacol 2020; 89:107094. [PMID: 33129097 DOI: 10.1016/j.intimp.2020.107094] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 01/05/2023]
Abstract
Ischemic stroke is a serious and life-threatening cerebrovascular thrombotic disease; however, the therapeutic strategy is limited for the complicated mechanism and narrow therapeutic window. Our previous study suggested that Z-Guggulsterone (Z-GS), an active component derived from myrrh, is a good candidate for cerebral injury. The object of this study is to investigate the exact mechanisms of Z-GS in cerebral ischemic stroke. Rats were used to conduct middle cerebral artery occlusion (MCAO) model and were treated with different dosage of Z-GS. Morphological results showed that Z-GS significantly alleviated neurological deficits, infarct volume and histopathological damage in MCAO rats. A total of 8276 differentially expressed genes were identified based on microarray analysis. Oxidation-reduction process and inflammatory response were enriched as the significant gene ontology items. TXNIP and NLRP3 were screened as the potential target genes by Series Test of Cluster (STC) analysis. The results were validated by immunohistochemistry and immunofluorescence staining. Besides, Z-GS successfully inhibited oxidative stress and inflammatory response in oxygen-glucose deprivation (OGD) treated neurons. Knockdown of TXNIP significantly decreased the expression of NLRP3 in OGD-induced neurons. In addition, Z-GS treatment scarcely changed the expressions of NLRP3 in siRNA-TXNIP pretreated cells compared with the siRNA-TXNIP alone treatment group, suggesting that the neuroprotective effect of Z-GS was dependent on TXNIP-NLRP3 axis. Taken together, this study revealed that Z-GS exerted neuroprotective property through alleviated oxidative stress and inflammation via inhibiting the TXNIP/NLRP3 axis. Z-GS could be considered as a promising candidate for the treatment of ischemic stroke.
Collapse
|
6
|
Jackson TC, Kochanek PM. RNA Binding Motif 5 (RBM5) in the CNS-Moving Beyond Cancer to Harness RNA Splicing to Mitigate the Consequences of Brain Injury. Front Mol Neurosci 2020; 13:126. [PMID: 32765218 PMCID: PMC7381114 DOI: 10.3389/fnmol.2020.00126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Gene splicing modulates the potency of cell death effectors, alters neuropathological disease processes, influences neuronal recovery, but may also direct distinct mechanisms of secondary brain injury. Therapeutic targeting of RNA splicing is a promising avenue for next-generation CNS treatments. RNA-binding proteins (RBPs) regulate a variety of RNA species and are prime candidates in the hunt for druggable targets to manipulate and tailor gene-splicing responses in the brain. RBPs preferentially recognize unique consensus sequences in targeted mRNAs. Also, RBPs often contain multiple RNA-binding domains (RBDs)—each having a unique consensus sequence—suggesting the possibility that drugs could be developed to block individual functional domains, increasing the precision of RBP-targeting therapies. Empirical characterization of most RBPs is lacking and represents a major barrier to advance this emerging therapeutic area. There is a paucity of data on the role of RBPs in the brain including, identification of their unique mRNA targets, defining how CNS insults affect their levels and elucidating which RBPs (and individual domains within) to target to improve neurological outcomes. This review focuses on the state-of-the-art of the RBP tumor suppressor RNA binding motif 5 (RBM5) in the CNS. We discuss its potent pro-death roles in cancer, which motivated our interest to study it in the brain. We review recent studies showing that RBM5 levels are increased after CNS trauma and that it promotes neuronal death in vitro. Finally, we conclude with recent reports on the first set of RBM5 regulated genes identified in the intact brain, and discuss how those findings provide new clues germane to its potential function(s) in the CNS, and pose new questions on its therapeutic utility to mitigate CNS injury.
Collapse
Affiliation(s)
- Travis C Jackson
- Morsani College of Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, United States.,Morsani College of Medicine, Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Identification of Novel Targets of RBM5 in the Healthy and Injured Brain. Neuroscience 2020; 440:299-315. [PMID: 32335213 DOI: 10.1016/j.neuroscience.2020.04.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 11/20/2022]
Abstract
The tumor suppressor RNA-binding motif 5 (RBM5) regulates the expression levels and cassette exon-definition (i.e. splicing) of a select set of mRNAs in a tissue-specific manner. Most RBM5-regulated targets were identified in oncological investigations and frequently involve genes which mediate apoptotic cell death. Little is known about the role of RBM5 in the brain. Also, it is unclear if a brain injury may be required to detect RBM5 mediated effects on pro-apoptotic genes due to their low expression levels in the healthy adult CNS at baseline. Conditional/floxed (brain-specific) gene deleter mice were generated to elucidate CNS-specific RBM5 mRNA targets. Male/female mice were subjected to a severe controlled cortical impact (CCI) traumatic brain injury (TBI) in order to increase the background expression of pro-death mRNAs and facilitate testing of the hypothesis that RBM5 inhibition decreases post-injury upregulation of caspases/FAS in the CNS. As expected, a CCI increased caspases/FAS mRNA in the injured cortex. RBM5 KO did not affect their levels or splicing. Surprisingly, KO increased the mRNA levels of novel targets including casein kinase 2 alpha prime interacting protein (Csnka2ip/CKT2) - a gene not thought to be expressed in the brain, contrary to findings here. Twenty-two unique splicing events were also detected in KOs including increased block-inclusion of cassette exons 20-22 in regulating synaptic membrane exocytosis 2 (Rims2). In conclusion, here we used genome-wide transcriptomic analysis on healthy and injured RBM5 KO mouse brain tissue to elucidate the first known gene targets of this enigmatic RBP in this CNS.
Collapse
|
8
|
Jing J, Wang B, Liu P. The Functional Role of SEC23 in Vesicle Transportation, Autophagy and Cancer. Int J Biol Sci 2019; 15:2419-2426. [PMID: 31595159 PMCID: PMC6775307 DOI: 10.7150/ijbs.37008] [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: 05/26/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
SEC23, the core component of the coat protein complex II (COPII), functions to transport newly synthesized proteins and lipids from the endoplasmic reticulum (ER) to the Golgi apparatus in cells for secretion. SEC23 protein has two isoforms (SEC23A and SEC23B) and their aberrant expression and mutations were reported to cause human diseases and oncogenesis, whereas SEC23A and SEC23B may have the opposite activity in human cancer, for a reason that remains unclear. This review summarizes recent research in SEC23, COPII-vesicle transportation, autophagy, and cancer.
Collapse
Affiliation(s)
- Jingchen Jing
- Center for Translational Medicine, The First Affiliated Hospital, Xi'an Jiaotong University.,The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bo Wang
- Center for Translational Medicine, The First Affiliated Hospital, Xi'an Jiaotong University.,The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital, Xi'an Jiaotong University.,The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
9
|
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
|
10
|
Jackson TC, Dixon CE, Janesko-Feldman K, Vagni V, Kotermanski SE, Jackson EK, Kochanek PM. Acute Physiology and Neurologic Outcomes after Brain Injury in SCOP/PHLPP1 KO Mice. Sci Rep 2018; 8:7158. [PMID: 29739983 PMCID: PMC5940799 DOI: 10.1038/s41598-018-25371-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/20/2018] [Indexed: 11/12/2022] Open
Abstract
Suprachiasmatic nucleus circadian oscillatory protein (SCOP) (a.k.a. PHLPP1) regulates long-term memory consolidation in the brain. Using a mouse model of controlled cortical impact (CCI) we tested if (1) brain tissue levels of SCOP/PHLPP1 increase after a traumatic brain injury (TBI), and (2) if SCOP/PHLPP1 gene knockout (KO) mice have improved (or worse) neurologic outcomes. Blood chemistry (pH, pCO2, pO2, pSO2, base excess, sodium bicarbonate, and osmolarity) and arterial pressure (MAP) differed in isoflurane anesthetized WT vs. KOs at baseline and up to 1 h post-injury. CCI injury increased cortical/hippocampal SCOP/PHLPP1 levels in WTs 7d and 14d post-injury. Injured KOs had higher brain tissue levels of phosphorylated AKT (pAKT) in cortex (14d post-injury), and higher levels of phosphorylated MEK (pMEK) in hippocampus (7d and 14d post-injury) and in cortex (7d post-injury). Consistent with an important role of SCOP/PHLPP1 on memory function, injured-KOs had near normal performance on the probe trial of the Morris water maze, whereas injured-WTs were impaired. CA1/CA3 hippocampal survival was lower in KOs vs. WTs 24 h post-injury but equivalent by 7d. No difference in 21d cortical lesion volume was detected. SCOP/PHLPP1 overexpression in cultured rat cortical neurons had no effect on 24 h cell death after a mechanical stretch-injury.
Collapse
Affiliation(s)
- Travis C Jackson
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA. .,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA.
| | - C Edward Dixon
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Neurology, 811 Kaufmann Medical Building, 3471 Fifth Avenue, Pittsburgh, USA
| | - Keri Janesko-Feldman
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| | - Vincent Vagni
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| | - Shawn E Kotermanski
- University of Pittsburgh School of Medicine, Department of Pharmacology and Chemical Biology, Bridgeside Point Building 1, 100 Technology Drive, Pittsburgh, USA
| | - Edwin K Jackson
- University of Pittsburgh School of Medicine, Department of Pharmacology and Chemical Biology, Bridgeside Point Building 1, 100 Technology Drive, Pittsburgh, USA
| | - Patrick M Kochanek
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| |
Collapse
|