Next Article in Journal
Support Vector Machine as a Supervised Learning for the Prioritization of Novel Potential SARS-CoV-2 Main Protease Inhibitors
Next Article in Special Issue
The Next Frontier: Translational Development of Ubiquitination, SUMOylation, and NEDDylation in Cancer
Previous Article in Journal
Chronic and Intermittent Hyperglycemia Modulates Expression of Key Molecules of PI3K/AKT Pathway in Differentiating Human Visceral Adipocytes
Previous Article in Special Issue
The Role of Autophagy in Chemical Proteasome Inhibition Model of Retinal Degeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Space Radiation-Induced Alterations in the Hippocampal Ubiquitin-Proteome System

by
Alyssa Tidmore
1,2,3,4,
Sucharita M. Dutta
2,3,
Arriyam S. Fesshaye
1,2,3,4,
William K. Russell
5,
Vania D. Duncan
1 and
Richard A. Britten
1,2,3,4,*
1
Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA 23507, USA
2
Department of Microbiology and Molecular Cell Biology; Eastern Virginia Medical School, Norfolk, VA 23507, USA
3
Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
4
Center for Integrative Neuroinflammatory and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
5
Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(14), 7713; https://doi.org/10.3390/ijms22147713
Submission received: 3 June 2021 / Revised: 8 July 2021 / Accepted: 15 July 2021 / Published: 19 July 2021
(This article belongs to the Special Issue Ubiquitination in Health and Diseases: Volume II)

Abstract

:
Exposure of rodents to <20 cGy Space Radiation (SR) impairs performance in several hippocampus-dependent cognitive tasks, including spatial memory. However, there is considerable inter-individual susceptibility to develop SR-induced spatial memory impairment. In this study, a robust label-free mass spectrometry (MS)-based unbiased proteomic profiling approach was used to characterize the composition of the hippocampal proteome in adult male Wistar rats exposed to 15 cGy of 1 GeV/n 48Ti and their sham counterparts. Unique protein signatures were identified in the hippocampal proteome of: (1) sham rats, (2) Ti-exposed rats, (3) Ti-exposed rats that had sham-like spatial memory performance, and (4) Ti-exposed rats that impaired spatial memory performance. Approximately 14% (159) of the proteins detected in hippocampal proteome of sham rats were not detected in the Ti-exposed rats. We explored the possibility that the loss of the Sham-only proteins may arise as a result of SR-induced changes in protein homeostasis. SR-exposure was associated with a switch towards increased pro-ubiquitination proteins from that seen in Sham. These data suggest that the role of the ubiquitin-proteome system as a determinant of SR-induced neurocognitive deficits needs to be more thoroughly investigated.

1. Introduction

NASA’s planned mission to Mars will take about three years to complete, during which time astronauts will be exposed to ~13 cGy Space Radiation (SR) each year [1]. SR is composed of highly energetic charged ions: protons, helium nuclei, and high-mass and energy (HZE) (Z ≤ 28, nuclei [2]). It has been calculated that roughly a quarter of the estimated 43 million hippocampal neurons within the brain will be hit directly hit by one or more particles with Z ≥ 15 on a deep space mission [3].
The hippocampus is essential for multiple neurocognitive process, including associative recognition memory [4] and the formation and retrieval of both new and old episodic memories [5,6]. Ground-based rodent studies have shown that exposure to <20 cGy SR impairs performance in several hippocampus-dependent cognitive tasks, including spatial memory in both rats [7,8,9,10,11,12] and mice [13,14,15], while, at the cohort level, SR exposure impairs spatial memory performance; at the individual levels, some irradiated rats maintain a spatial memory performance comparable to that seen in the sham-irradiated rats, while others have severely compromised performance [10,11,12]. These data suggest that some individuals are able to ameliorate the deleterious effects of SR while others are unable to do so. This bifurcating response of neurocognitive processes to SR exposure has important consequences for risk assessments but also provides a unique opportunity to establish the impact of SR on neurophysiology, and the subsequent adaptive responses associated with the impairment or preservation of neurocognitive performance. SR exposure induces multiple changes in the functionality of hippocampal neurons: changes in the complexity of the dendritic tree in the hippocampus [15,16,17,18], long-term suppression of glutamatergic transmission and NR2A receptor levels in the hippocampus [19], homeostatic synaptic plasticity [18,19,20,21,22,23], as well as autophagy and persistent oxidative stress [24]. Recent studies also suggest that SR-induced neurocognitive decrements may be regulated by non-neuronal components, with the activation status of the microglia in the first few days after exposure being important [25,26,27]. Collectively, these studies indicate that SR exposure alters numerous processes within the hippocampus, and that no single process will likely dominate the cognitive performance of an irradiated individual. Taking all these factors into consideration, it seems likely that a systems biology approach will be necessary to identify why some individuals retain a functional spatial memory, while others have impaired performance after SR exposure.
We have previously employed a robust label-free mass spectrometry (MS)-based unbiased proteomic profiling approach to characterize the composition of the hippocampal proteome in juvenile [28] and adult [29] male Wistar rats exposed to ≤20 cGy of 1 GeV/n 56Fe. These studies identified proteins whose expression are altered with respect to (1) radiation exposure and (2) impaired spatial memory performance. Nearly a quarter of the proteins found in the hippocampus of adult sham rats were lost or had reduced expression in the irradiated hippocampus [29]. These data are consistent with the SR-induced changes in the DNA methylation status within the hippocampus [30,31,32,33,34]. The biological significance of these methylation changes is quite pronounced since SR-induced loss of hippocampal-dependent memory updating and LTP within the hippocampus were reversed by the use of histone deacetylase 3 inhibitors [35].
While SR-induced changes in DNA methylation may be an important factor in influencing the composition of the hippocampal proteome, protein abundance is not only determined by transcriptional activity, but also by translation rates, sequestration of proteins into complexes, as well as protein turnover and degradation. SR exposure upregulates several components of the proteasome/protein degradation pathway in the hippocampus [28], and also alters protein homeostasis, with marked changes in ubiquitination [36]. While such SR-induced changes in ubiquitin-mediated cellular protein homeostasis will undoubtedly have profound effects on neuronal physiology and functionality, the ubiquitin-proteome system (UPS) plays a key role in regulating memory formation and consolidation [37,38,39]. Thus, differences in the UPS may serve to modulate the impact of SR-induced changes in neurotransmission on neurocognitive performance, and may explain the poor correlation between these parameters in our previous studies [18,21]. We thus established the composition of the hippocampal proteome in adult male rats exposed to 15 cGy 1 GeV/n 48Ti ions and identified protein signatures that are biomarkers of (1) Ti exposure and (2) spatial memory performance, and whether these biomarker proteins are involved with the UPS.

2. Results

The spatial memory performance of the retired breeder (about nine months at the time of irradiation) rats was established at 90 ± 14 days post irradiation using the Barnes Maze. Each rat was tested on the Barnes’ Maze over a three-day period, with two tests per day. A key element of the Barnes Maze is that rats progressively learn and recall the location of the escape box, as measured by the time to find its location i.e., escape latency (ESL).
For each rat, an individualized metric of their relative increase (or decrease) in performance between days 1 and 3 of testing i.e., the Relative Escape Latency (REL3) was calculated (where the Day 3 ESL for a specific rat is expressed as a decimal fraction of its Day 1 ESL). The REL3 for rats that improve in spatial memory between days 1 and 3 is less than unity. Alternatively, if spatial memory is impaired by Day 3, the REL3 remains near unity or becomes greater than unity.
The mean REL3 for the entire population exposed to 15 cGy 48Ti (0.81 +/− 0.12, n = 24) was significantly higher (p = 0.046, Mann–Whitney) than the mean REL3 of the sham-irradiated rats (0.53 +/− 0.05, n = 55). Rats were classified as having impaired spatial memory (15/Impaired) if their REL3 value was greater than 0.7. The distribution of the REL3 values within each cohort is shown in Figure 1. Representative rats from each cohort (Sham-irradiated: 8 (REL3 < 0.69), 15/Functional: 4 (REL3 < 0.55), 15/Impaired: 4 (REL3 > 1.1 < 2.5)) were selected for subsequent proteomic analysis, the REL3 values for those rats are separately depicted in Figure 1.
An analysis of the composition of the hippocampal proteome of the representative rats from each cohort (Sham-irradiated (n = 8), 15/Impaired ((REL3 > 1.1 < 2.5) (n = 4)), 15/Functional ((REL3 < 0.55) (n = 4)), revealed that there were 974–1099 proteins that satisfied our vigorous inclusion criteria (quantifiable in >66% of technical replicates, and present in >66% of the biological replicates) in the various cohorts of rats (Sham-functional: 1099; 15/Functional: 954; 15/Impaired: 974). A complete list of the identified proteins within each cohort is provided in the Supplemental Table S1 There were 732 proteins that were detected in all three cohorts, hereafter referred to as “common” proteins (Figure 2).
There were 159 (88 fully mapped) proteins that were detected in the proteome of the sham rats but were not detected in the irradiated rats (irrespective of their spatial memory performance status), hereafter referred to as “Sham only” signature proteins. The Uniprot accession numbers of the 88 fully mapped “Sham Only” protein are listed in Table 1. The gene/protein names of all proteins detected in the study are listed with the appropriate Uniprot numbers in the Supplemental Table S1.
While these proteins are classified as “sham only” proteins, they are also technically negative biomarkers of Ti exposure. There were also 41 positive biomarkers of Ti exposure, i.e., were detected in both the 15/Functional and 15/Impaired cohorts, but not in the sham proteomic profile, hereafter referred to as “Ti exposure marker (TEM)” signature proteins. The 20 fully annotated and reviewed TEM proteins are listed in Table 2.
In addition to the TEM proteins, the proteome of the Ti-exposed rats contained other unique proteins that reflected the spatial memory performance of the irradiated rats. There were 84 proteins that were found only in rats who had impaired spatial memory performance (15/Impaired), hereafter referred to as “spatial memory impairment (SMI)” signature proteins. The 37 fully annotated and reviewed SMI proteins are listed in Table 3. There were an additional 24 upregulated TEM proteins (italicized in Table 3).
The hippocampal proteome of the irradiated rats that exhibited sham-like spatial memory performance (15/functional) rats was characterized by the presence of 91 uniquely expressed proteins, hereafter referred to as “preservation of spatial memory impairment (PSM)” signature proteins. The 41 fully annotated and reviewed PSM proteins are listed in Table 4. There were an additional 14 upregulated PSM proteins (italicized in Table 4).
Our unique ability to identify proteomic signatures that are associated with a phenotypic endpoint (spatial memory performance) provides an opportunity to investigate how SR exposure can lead to spatial memory impairment in some rats (15/Impaired) and not others (15/Functional). As observed in our previous studies on Fe-induced changes in the hippocampal proteome [28,29], there were significant Ti-induced changes in the expression of proteins involved with neuronal homeostasis, axonogenesis, pre-synaptic membrane organization, G-protein coupled receptors oxidative damage response, calcium transport, and signaling. One notable change was the unique presence of Glia-maturation factor B within the PSM signature.
Gene Ontology (GO) enrichment analysis (using FunRich) of the six protein signatures (i.e., “Sham only”, “TEM”, “SMI”, “PSM”, “Impaired” Phenotype (TEM and SMI), and “Functional” (TEM and PSM) proteins) identified five processes that satisfied our inclusion criteria: GO:0044877-protein-containing complex binding; GO:0042802-identical protein binding; GO:0046872-metal ion binding; GO:0005524-ATP binding; and GO:0019901-protein kinase binding. Over 30% of the SMI proteins were identified as having an important role in the GO:0042802-identical protein binding and GO:0005524-ATP binding processes (Table 5).
The overlap with the ubiquitin-proteome system (UPS) UniProt database for each of the six protein signatures, revealed that roughly a quarter of the proteins in the TEM, SMI, and Impaired signatures have a direct role in the UPS (Table 6). The Sham signature was unique in that it was the only one to contain proteins that exhibited deubiquitinase (DUB) activity, with two deubiquitinases (Q4VSI4/Usp7; Q9R085/Usp15) being absent in all irradiated cohorts. The UPS is a key regulator of protein homeostasis within the cell; we utilized a simple ubiquitin homeostasis algorithm to determine if there were any indications of SR-induced changes in UPS functionality within the various signatures. The Sham signature yielded a pro-de-ubiquitination weighting (−4), whereas both the Functional (TEM and PSM) and Impaired (TEM and SMI) signatures yield a pro- ubiquitination weighting (7.5 and 8.5 respectively). The TEM signature in isolation had a +2 weighting factor.

3. Discussion

The present study is one of only a few [28,29,40,41,42,43] to utilize a bottom-up proteomic profiling strategy to identify changes in the hippocampal proteome that occur after radiation exposure. The proteomic profiles generated in this study are essentially a snapshot of the functional status quo within the hippocampus at the time of sampling (approximately three months after exposure). There are unique protein changes that are observed in all irradiated rats (loss of sham only and de novo expression of TEM proteins). These changes may have occurred as an immediate response to SR exposure, or may be the result of compensatory responses invoked over time in response to the SR exposure. Superimposed on these “exposure” related changes, there are further protein signatures that are found in rats that have functional (PSM proteins) or impaired spatial memory (SMI proteins) performance. A unique feature of our study has been the identification of protein signatures (biomarkers) that are associated with a specific phenotypic (spatial memory functionality) endpoint, in this case spatial memory performance, assessed just prior to recovery of the brain tissue. Thus, there is a reasonable likelihood of a casual association with the observed proteomic (PSM and SMI) changes to spatial memory performance. However, as previously reported [28,29], there are complex changes in the composition of the proteome that will most likely require advanced analysis (such as identification of reciprocal up- and downregulation of multiple proteins/pathways) to identify which of the observed changes have phenotypic significance.
One of the most striking features of the proteomic profiling was that ~14% of the proteins found in the hippocampus of sham rats were lost in the irradiated hippocampus (Figure 2, Table 2). While these proteins have been classified as “sham only” signature proteins (since they were only detected in unirradiated rats), these proteins are also technically negative biomarkers of Ti exposure. Reduced expression of hippocampal proteins may be a hallmark (biomarker) of SR exposure since we have previously observed a SR (Fe)—induced loss of protein expression [28,29]. SR (Si) exposure elevates DNA methylation levels in the hippocampus [30], which would certainly be consistent with a reduction in protein expression. However, SR-induced DNA methylation levels would appear to not be the sole determinant of the observed changes in the hippocampal proteome, given that there were many additional (and complex) changes that occur in the hippocampal proteome of SR-exposed rats, including the upregulation (or de novo) expression of many proteins. The functionality of the hippocampus in SR-exposed rats may be reflected by the changes in the expression of the TEM proteins. In our previous studies [28,29], these SR exposure markers included many involved in neuronal homeostasis, axonogenesis, pre-synaptic membrane organization, G-protein coupled receptors oxidative damage response, calcium transport, and signaling. There are further proteomic changes in the hippocampus of irradiated rats that are associated with the loss of spatial memory performance, while the proteome of rats that have been able to preserve spatial memory performance have another proteomic signature. Notable amongst the PSM signature proteins was the unique presence of Glia-maturation factor B (Table 4). Several studies have demonstrated that SR-induced behavioral and memory deficits are associated with altered microglial activation [25,26,27]. We have included the proteomic expression data from our various rat cohorts (as Supplementary Material) for others to mine using the new generation of bioinformatics approaches to determine the full spectrum of changes present in these rats. However, it should be noted that the proteomic signatures of the various cohorts identified in this study do need orthogonal validation. While Western blotting is frequently used for such purposes, the linearity of antibody-generated signals needs extensive validation to get truly quantitative data. A more feasible approach with the more widespread use of “Orbitrap” mass spectrometers would be to use multiple reaction monitoring (MRM)/parallel reaction monitoring (PRM) of specific peptides from the proteins of interest [44].
Collectively, the specific upregulation (or de novo) expression of multiple proteins suggest that SR-induced methylation cannot be the sole determinant of SR-induced changes in the hippocampal proteome. Protein expression is a complex balance between transcriptional/translational activity and protein turnover/degradation. The maintenance of protein homeostasis is an important role of the ubiquitin-proteasome system, which is responsible for roughly 80% of all intracellular protein degradation [45]. SR exposure has recently been shown to alter the ubiquitin-mediated protein homeostatic response in cultured neural cells, associated with an elevated number of ubiquitination-site linkages [36]. Nearly a quarter of the proteins in the TEM, SMI, and Impaired signatures identified in this study have a direct role in the UPS (Table 6). Furthermore, only the Sham signature contains proteins that exhibited deubiquitinase activity, with two deubiquitinases (Q4VSI4/Usp7; Q9R085/Usp15) being absent in all irradiated cohorts. While both of these hydrolases impact a wide range of ubiquitin-dependent processes, the Usp7 protein is of special interest in the context of SR-induced loss of neurocognition. The Usp7 protein involved in maintaining DNA methylation, and the loss of Usp7 in the irradiated rats may thus be related to the SR-induced epigenetic changes [30]. Usp7 also deubiquitinates the repressor element 1 silencing transcription factor (REST), thereby stabilizing REST and promoting the maintenance of neural progenitor cells [45]. Radiation-induced changes in the Usp7-mediated deubiquitination of REST could thus account for the neural precursor-cell dysfunction reported following low-LET radiation exposures [46]. REST also plays an important role in the shaping of the synaptic output of adult NMDAR dependent neurons [47], so the loss of neurocognitive function following SR exposure could in part be mediated via Usp7-mediated deubiquitination of REST.
Our data would suggest that the SR-induced changes in UPS in cultured neuronal cells [36] also occurs in vivo, at least within the hippocampus of rats exposed to 15 cGy Ti ions. However, while the UPS system does play a key role in “bulk” protein homeostasis, UPS-mediated activity-dependent protein degradation is required for memory consolidation [48], consolidation of spatial memory [49,50], contextual fear conditioning, and inhibitory avoidance [37,38,51,52]. The loss of deubiquitinase activity after Ti exposure could certainly contribute to the loss of spatial memory performance. The very simplistic ubiquitin homeostasis algorithm that we developed has provided some insight into how SR may lead to imbalances in the UPS system, and suggests that SR-exposure is associated with a pro-ubiquitination balance, whereas, in Sham rats, there is a pro-deubiquitination balance. However, this conclusion is based upon the very select protein signatures identified in this study, the importance of which to the “global” ubiquitination balance may be small. Specifically designed experiments utilizing ubiquitinated protein enrichment approaches would probably be necessary to definitively establish how SR exposure alters the functionality of the UPS system.

4. Materials and Methods

4.1. Irradiation Procedure

The rats used in this study are a subset of the 185 male Wistar retired breeder rats (HSD:WI; Harlan Sprague–Dawley, Inc., Indianapolis, IN, USA) that were used in our previous study [12]. The rats were delivered directly from the supplier to BNL, where they were group housed, maintained on a 12:12 light/dark cycle and given ad libitum access to autoclaved Purina Rodent Chow 5001 and municipal water by bottle. After at least one week of acclimatization, the rats were irradiated with 15 cGy 1 GeV/n 48Ti exposure at the NASA Space Radiation Laboratory at a dose rate of 10 cGy/min. After irradiation, the rats were implanted with ID-100us RFID transponders (Electronic ID Devices Ltd., Santa Barbara, CA, USA to facilitate identification of individual animals. One week after irradiation, the rats were transported to EVMS, where they were group housed, and given ad libitum access to Teklad 2014 rat chow and municipal water by bottle. The rats were maintained on a reversed 12:12 light/dark cycle, i.e., lights were switched off during working hours, resulting in the rats being in their active phase when tested for spatial memory performance.

4.2. Spatial Memory Testing

Spatial memory performance was established at 90 ± 14 days post irradiation. The rats were tested on the Barnes Maze in accordance with our previously published protocol [12]. A key element of the Barnes Maze is that rats progressively learn and recall the location of the escape box, as measured by the time to find its location i.e., Escape Latency. For each animal, an individualized metric of their relative increase (or decrease) in performance between days 1 and 3 of testing was calculated i.e., the Relative Escape Latency 3 (REL3), where the Day 3 ESL for a specific rat is expressed as a decimal fraction of its Day 1 ESL. The REL3 is less than unity if rats improve their spatial memory between days 1 and 3 of testing, but, if spatial memory is impaired, the REL3 remains at or above unity.

4.3. Hippocampal Protein Extraction and Mass Spectrometric Profiling

The protocol followed for peptide and protein identification for the brain tissue lysate has been published in a previous paper [29]. Briefly, experiments were executed on the Q-Exactive (QE) mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) using ‘bottom-up’ proteomics. A label-free precursor ion detection method (Proteome Discoverer, version 1.4, Thermo Scientific) was used because of the accurate mass measurements on proteins/peptides with specific retention times on precursors/fragments within 5 ppm mass accuracy. These factors combine to afford protein/peptide identifications with high confidence and high sequence coverage. The Sequest algorithm, a search engine employed by Proteome Discoverer (version 1.4, Thermo Scientific), was used to identify peptides from the resulting MS/MS spectra by searching against the combined Rat protein database (a total of 25,320 sequences) extracted from Swissprot (version 57) using taxonomy “Rattus”. Searching parameters for parent and fragment ion tolerances was set as 15 ppm and 80 mmu for the QE, and trypsin was set as the protease with a maximum of 2 missed cleavages. Only those proteins that have >2 peptides identified (or >50% of protein covered by a single peptide) were included in the comparative quantitative analysis steps, and result in a correct protein identification probability of p < 0.05.
Quantitation of a protein within a given technical replicate was achieved by calculating the area under the curve (AUC) for the respective de-isotoped peptide and charge reduced multiple tryptic peptides. A protein is classified as being “present” if it is identified in two of the three technical replicate samples for an individual rat hippocampus sample. In the event that a protein is not detectable in a particular cohort, an AUC value of 1 is assigned for that protein. The mean AUC value for each individual rat is calculated. A mean cohort AUC value (and the SEM) was then calculated for any protein that was “present” in the majority of the individual rats within that cohort. In those instances where the SEM exceeded the mean AUC, those proteins were removed from further analysis.

4.4. Post Analysis of the Proteins Identified by Proteome Discoverer

The online visual analysis tool Venny 2.1 [53] was used to establish protein signatures that were common to all cohorts, and those that were uniquely present in specific cohorts. Proteins that were up- or downregulated in the irradiated cohorts (i.e., all Ti exposed rats (TEM proteins), Ti-exposed rats that retained sham-like spatial memory performance (PSM proteins), and Ti-exposed rats that had impaired spatial memory performance (SMI proteins)) were then identified. The AUC values of the proteins in the irradiated cohorts were compared to the respective AUC values in the sham cohort and those proteins whose expression index (relative to the sham AUC value) was ≥1.5 (upregulated) or ≤0.66 (down-regulated) identified. The Wilcoxon–Mann–Whitney test was used to identify proteins whose expression differed from that seen in the sham-irradiated rats at the 5% significance level.
The functional significance of the protein signatures that characterized each of the phenotypic groups, (i.e., Shams, all Ti exposed rats, Ti-exposed rats that retained sham-like spatial memory performance and Ti-exposed rats that had impaired spatial memory performance) was established using the Compare gene enrichment function in FunRich version 3.1.3 [54], a freely available software tool. Prior to analyses, the Rodent (Taxon ID: 9989) database was downloaded from UniProt and configured as the background database within FunRich. The cohort protein signatures were characterized against known UniProt molecular functions and biological processes, with an inclusion criterion of ≥10% of proteins within the signature must be linked to that function, and that the function was identified in at least two of the four proteins (i.e., Sham, PSM, TEM, or SMI) signatures.
The online visual analysis tool Venny 2.1 [40] was used to identify the proteins within each signature that were present in the UniProt reference base for the UPS. A FunRich-generated report detailing the cellular component, molecular function, and biological process data of each protein was then generated.
To provide some insight into the overall ubiquitination activity implicated by the various protein signatures, a simplistic ubiquitin homeostasis algorithm was developed. Signature proteins identified as being in the UniProt UPS database were assigned impact values based on how many and the types of ubiquitin-regulated functions they exhibited. Ubiquitination-promoting activity garnered positive scores, and ubiquitination-depressing (or deubiquitination) activity earned negative scores. For each instance wherein ubiquitination or deubiquitination was a primary function of that protein (that is, the protein is immediately responsible for ubiquitination or deubiquitination), they were given a +1 or −1 score, respectively. Non-primary functions were assigned +0.5 or −0.5; this includes any ubiquitination-adjacent function whose dysfunction does not immediately impact ubiquitination activity, but would likely lead to disruptions in the UPS (e.g., ubiquitin ligase activity, component of the 26S proteasome, positive or negative regulation of the catabolic process, etc.). Ambivalent proteins (i.e., proteins that were identified as overlapping with the UPS, but whose function or impact is not yet confirmed) were assigned a neutral 0. Once each value was assigned, scores were totaled by cohort in order to obtain a net impact value; positive scores indicate an abundance of ubiquitination activity, negative scores indicating less pro-ubiquitination activity.

4.5. Statistical Analysis

All statistical analysis was conducted using GraphPad Prism version 9.1.2 for Windows, GraphPad Software, San Diego, CA, USA, www.graphpad.com, assessed on 11 July 2021.

5. Conclusions

In conclusion, this study has provided further insight into the complex changes that occur in the hippocampus of SR-exposed rats, and have identified specific protein signatures that characterize the hippocampus of rats that developed spatial-memory impairment as well as those that have managed to maintain sham-like performance levels. In addition, our study has demonstrated that there may be marked changes in the functionality of the UPS system that warrants further investigation, and specifically its role in regulating SR-induced neurocognitive impairment.

Supplementary Materials

Supplementary Table S1 is available online at https://www.mdpi.com/article/10.3390/ijms22147713/s1.

Author Contributions

Conceptualization, R.A.B. and S.M.D.; methodology, S.M.D., A.S.F., V.D.D. and W.K.R.; formal analysis, R.A.B., S.M.D., A.T. and W.K.R.; resources, R.A.B., W.K.R. and S.M.D.; data curation R.A.B., A.T. and S.M.D.; writing—original draft preparation, A.T. and R.A.B.; writing: review and editing, R.A.B., A.T., S.M.D., A.S.F., V.D.D. and W.K.R.; visualization, A.T. and R.A.B.; supervision R.A.B. and W.K.R.; project administration, R.A.B.; funding acquisition, R.A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by NASA grant support NNX11AC56G and NNX14AE73G.

Institutional Review Board Statement

This study was conducted in accordance with the National Research Council’s “Guide for the Care and Use of Laboratory Animals (8th Edition)”, at facilities of Eastern Virginia Medical School (EVMS) and Brookhaven National Laboratory (BNL), both of which are accredited by the Association for Assessment and Accreditation of Laboratory Animal Care, International. All procedures where approved by the Institutional Animal Care and Use Committee of both EVMS and BNL.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cucinotta, F.A.; Alp, M.; Sulzman, F.M.; Wang, M. Space radiation risks to the central nervous system. Life Sci. Sp. Res. 2014, 2, 54–69. [Google Scholar] [CrossRef] [Green Version]
  2. Slaba, T.C.; Blattnig, S.R.; Norbury, J.W.; Rusek, A.; La Tessa, C. Reference field specification and preliminary beam selection strategy for accelerator-based GCR simulation. Life Sci. Sp. Res. 2016, 8, 52–67. [Google Scholar] [CrossRef]
  3. Curtis, S.B.; Vazquez, M.E.; Wilson, J.W.; Atwell, W.; Kim, M.; Capala, J. Cosmic ray hit frequencies in critical sites in the central nervous system. Adv. Sp. Res. 1998. [Google Scholar] [CrossRef]
  4. Barker, G.R.I.; Warburton, E.C. Object-in-place associative recognition memory depends on glutamate receptor neurotransmission within two defined hippocampal-cortical circuits: A critical role for AMPA and NMDA receptors in the hippocampus, perirhinal, and prefrontal cortices. Cereb. Cortex 2015, 25, 472–481. [Google Scholar] [CrossRef] [Green Version]
  5. Moscovitch, M.; Nadel, L.; Winocur, G.; Gilboa, A.; Rosenbaum, R.S. The cognitive neuroscience of remote episodic, semantic and spatial memory. Curr. Opin. Neurobiol. 2006, 16, 179–190. [Google Scholar] [CrossRef]
  6. Rekkas, P.V.; Todd Constable, R. Evidence that autobiographic memory retrieval does not become independent of the hippocampus: An fMRI study contrasting very recent with remote events. J. Cogn. Neurosci. 2005, 17, 1950–1961. [Google Scholar] [CrossRef] [PubMed]
  7. Shukitt-Hale, B.; Casadesus, G.; McEwen, J.J.; Rabin, B.M.; Joseph, J.A. Spatial learning and memory deficits induced by exposure to iron-56- particle radiation. Radiat. Res. 2000. [Google Scholar] [CrossRef]
  8. Denisova, N.A.; Shukitt-Hale, B.; Rabin, B.M.; Joseph, J.A. Brain signaling and behavioral responses induced by exposure to 56Fe-particle radiation. Radiat. Res. 2002. [Google Scholar] [CrossRef]
  9. Rabin, B.M.; Miller, M.G.; Larsen, A.; Spadafora, C.; Zolnerowich, N.N.; Dell’Acqua, L.A.; Shukitt-Hale, B. Effects of exposure to 12C and 4He particles on cognitive performance of intact and ovariectomized female rats. Life Sci. Sp. Res. 2019, 22, 47–54. [Google Scholar] [CrossRef]
  10. Britten, R.A.; Davis, L.K.; Johnson, A.M.; Keeney, S.; Siegel, A.; Sanford, L.D.; Singletary, S.J.; Lonart, G. Low (20 cGy) doses of 1 GeV/u 56Fe-particle radiation lead to a persistent reduction in the spatial learning ability of rats. Radiat. Res. 2012, 177, 146–151. [Google Scholar] [CrossRef] [PubMed]
  11. Britten, R.A.; Jewell, J.S.; Miller, V.D.; Davis, L.K.; Hadley, M.M.; Wyrobek, A.J. Impaired Spatial Memory Performance in Adult Wistar Rats Exposed to Low (5-20 cGy) Doses of 1 GeV/n 56Fe Particles. Radiat. Res. 2016, 185, 332–337. [Google Scholar] [CrossRef] [PubMed]
  12. Britten, R.A.; Jewell, J.S.; Duncan, V.D.; Davis, L.K.; Hadley, M.M.; Wyrobek, A.J. Spatial Memory Performance of Socially Mature Wistar Rats is Impaired after Exposure to Low (5 cGy) Doses of 1 GeV/n 48Ti Particles. Radiat. Res. 2017, 187, 60–65. [Google Scholar] [CrossRef] [PubMed]
  13. Villasana, L.E.; Rosenthal, R.A.; Doctrow, S.R.; Pfankuch, T.; Zuloaga, D.G.; Garfinkel, A.M.C.; Raber, J. Effects of alpha-lipoic acid on associative and spatial memory of sham-irradiated and 56Fe-irradiated C57BL/6J male mice. Pharmacol. Biochem. Behav. 2013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Haley, G.E.; Yeiser, L.; Olsen, R.H.J.; Davis, M.J.; Johnson, L.A.; Raber, J. Early effects of whole-body 56Fe irradiation on hippocampal function in C57BL/6J mice. Radiat. Res. 2013, 179, 590–596. [Google Scholar] [CrossRef] [Green Version]
  15. Parihar, V.K.; Maroso, M.; Syage, A.; Allen, B.D.; Angulo, M.C.; Soltesz, I.; Limoli, C.L. Persistent nature of alterations in cognition and neuronal circuit excitability after exposure to simulated cosmic radiation in mice. Exp. Neurol. 2018, 305, 44–55. [Google Scholar] [CrossRef]
  16. Parihar, V.K.; Allen, B.; Tran, K.K.; Macaraeg, T.G.; Chu, E.M.; Kwok, S.F.; Chmielewski, N.N.; Craver, B.M.; Baulch, J.E.; Acharya, M.M.; et al. What happens to your brain on the way to Mars. Sci. Adv. 2015, 1. [Google Scholar] [CrossRef] [Green Version]
  17. Ingiosi, A.M.; Hayworth, C.R.; Harvey, D.O.; Singletary, K.G.; Rempe, M.J.; Wisor, J.P.; Frank, M.G. A Role for Astroglial Calcium in Mammalian Sleep and Sleep Regulation. Curr. Biol. 2020, 30, 4373–4383.e7. [Google Scholar] [CrossRef]
  18. Krishnan, B.; Natarajan, C.; Bourne, K.Z.; Alikhani, L.; Wang, J.; Sowa, A.; Groen, K.; Perry, B.; Dickstein, D.L.; Baulch, J.E.; et al. Chronic low dose neutron exposure results in altered neurotransmission properties of the hippocampus-prefrontal cortex axis in both mice and rats. Int. J. Mol. Sci. 2021, 22, 3668. [Google Scholar] [CrossRef]
  19. MacHida, M.; Lonart, G.; Britten, R.A. Low (60 cGy) doses of 56Fe HZE-particle radiation lead to a persistent reduction in the glutamatergic readily releasable pool in rat hippocampal synaptosomes. Radiat. Res. 2010, 174, 618–623. [Google Scholar] [CrossRef]
  20. Vlkolinský, R.; Krucker, T.; Nelson, G.A.; Obenaus, A. 56Fe-particle radiation reduces neuronal output and attenuates lipopolysaccharide-induced inhibition of long-term potentiation in the mouse hippocampus. Radiat. Res. 2008, 169, 523–530. [Google Scholar] [CrossRef]
  21. Britten, R.A.; Duncan, V.D.; Fesshaye, A.; Rudobeck, E.; Nelson, G.A.; Vlkolinsky, R. Altered Cognitive Flexibility and Synaptic Plasticity in the Rat Prefrontal Cortex after Exposure to Low (≤15 cGy) Doses of 28Si Radiation. Radiat. Res. 2020, 193, 223–235. [Google Scholar] [CrossRef]
  22. Acharya, M.M.; Baulch, J.E.; Klein, P.M.; Baddour, A.A.D.; Apodaca, L.A.; Kramár, E.A.; Alikhani, L.; Garcia, C.; Angulo, M.C.; Batra, R.S.; et al. New concerns for neurocognitive function during deep space exposures to chronic, low dose-rate, neutron radiation. eNeuro 2019, 6. [Google Scholar] [CrossRef]
  23. Klein, P.M.; Parihar, V.K.; Szabo, G.G.; Zöldi, M.; Angulo, M.C.; Allen, B.D.; Amin, A.N.; Nguyen, Q.A.; Katona, I.; Baulch, J.E.; et al. Detrimental impacts of mixed-ion radiation on nervous system function. Neurobiol. Dis. 2021, 151. [Google Scholar] [CrossRef] [PubMed]
  24. Poulose, S.M.; Bielinski, D.F.; Carrihill-Knoll, K.; Rabin, B.M.; Shukitt-Hale, B. Exposure to 16O-particle radiation causes aging-like decrements in rats through increased oxidative stress, inflammation and loss of autophagy. Radiat. Res. 2011, 176, 761–769. [Google Scholar] [CrossRef] [PubMed]
  25. Allen, B.D.; Syage, A.R.; Maroso, M.; Baddour, A.A.D.; Luong, V.; Minasyan, H.; Giedzinski, E.; West, B.L.; Soltesz, I.; Limoli, C.L.; et al. Mitigation of helium irradiation-induced brain injury by microglia depletion. J. Neuroinflamm. 2020, 17, 159. [Google Scholar] [CrossRef]
  26. Krukowski, K.; Feng, X.; Paladini, M.S.; Chou, A.; Sacramento, K.; Grue, K.; Riparip, L.K.; Jones, T.; Campbell-Beachler, M.; Nelson, G.; et al. Temporary microglia-depletion after cosmic radiation modifies phagocytic activity and prevents cognitive deficits. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef]
  27. Parihar, V.K.; Angulo, M.C.; Allen, B.D.; Syage, A.; Usmani, M.T.; Passerat de la Chapelle, E.; Amin, A.N.; Flores, L.; Lin, X.; Giedzinski, E.; et al. Sex-Specific Cognitive Deficits Following Space Radiation Exposure. Front. Behav. Neurosci. 2020. [Google Scholar] [CrossRef] [PubMed]
  28. Britten, R.A.; Jewell, J.S.; Davis, L.K.; Miller, V.D.; Hadley, M.M.; Semmes, O.J.; Lonart, G.; Dutta, S.M. Changes in the hippocampal proteome associated with spatial memory impairment after exposure to low (20 cGy) doses of 1 GeV/n 56Fe radiation. Radiat. Res. 2017, 187, 287–297. [Google Scholar] [CrossRef] [PubMed]
  29. Dutta, S.M.; Hadley, M.M.; Peterman, S.; Jewell, J.S.; Duncan, V.D.; Britten, R.A. Quantitative Proteomic Analysis of the Hippocampus of Rats with GCR-Induced Spatial Memory Impairment. Radiat. Res. 2018, 189, 136–145. [Google Scholar] [CrossRef]
  30. Acharya, M.M.; Baddour, A.A.D.; Kawashita, T.; Allen, B.D.; Syage, A.R.; Nguyen, T.H.; Yoon, N.; Giedzinski, E.; Yu, L.; Parihar, V.K.; et al. Epigenetic determinants of space radiation-induced cognitive dysfunction. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef]
  31. Impey, S.; Pelz, C.; Tafessu, A.; Marzulla, T.; Turker, M.S.; Raber, J. Proton irradiation induces persistent and tissue-specific DNA methylation changes in the left ventricle and hippocampus. BMC Genom. 2016, 17. [Google Scholar] [CrossRef] [Green Version]
  32. Impey, S.; Jopson, T.; Pelz, C.; Tafessu, A.; Fareh, F.; Zuloaga, D.; Marzulla, T.; Riparip, L.K.; Stewart, B.; Rosi, S.; et al. Short- and long-term effects of 56Fe irradiation on cognition and hippocampal DNA methylation and gene expression. BMC Genom. 2016, 17. [Google Scholar] [CrossRef] [Green Version]
  33. Impey, S.; Jopson, T.; Pelz, C.; Tafessu, A.; Fareh, F.; Zuloaga, D.; Marzulla, T.; Riparip, L.K.; Stewart, B.; Rosi, S.; et al. Bi-directional and shared epigenomic signatures following proton and 56Fe irradiation. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Torres, E.R.S.; Hall, R.; Bobe, G.; Choi, J.; Impey, S.; Pelz, C.; Lindner, J.R.; Stevens, J.F.; Raber, J. Integrated Metabolomics-DNA Methylation Analysis Reveals Significant Long-Term Tissue-Dependent Directional Alterations in Aminoacyl-tRNA Biosynthesis in the Left Ventricle of the Heart and Hippocampus Following Proton Irradiation. Front. Mol. Biosci. 2019, 6, 77. [Google Scholar] [CrossRef] [PubMed]
  35. Keiser, A.A.; Kramár, E.A.; Dong, T.; Shanur, S.; Pirodan, M.; Ru, N.; Acharya, M.M.; Baulch, J.E.; Limoli, C.L.; Wood, M.A. Systemic HDAC3 inhibition ameliorates impairments in synaptic plasticity caused by simulated galactic cosmic radiation exposure in male mice. Neurobiol. Learn. Mem. 2021, 178, 107367. [Google Scholar] [CrossRef] [PubMed]
  36. Shaler, T.; Lin, H.; Bakke, J.; Chen, S.; Grover, A.; Chang, P. Particle radiation-induced dysregulation of protein homeostasis in primary human and mouse neuronal cells. Life Sci. Sp. Res. 2020, 25, 9–17. [Google Scholar] [CrossRef]
  37. Reis, D.S.; Jarome, T.J.; Helmstetter, F.J. Memory formation for trace fear conditioning requires ubiquitin-proteasome mediated protein degradation in the prefrontal cortex. Front. Behav. Neurosci. 2013. [Google Scholar] [CrossRef] [Green Version]
  38. Lee, S.H.; Choi, J.H.; Lee, N.; Lee, H.R.; Kim, J.I.; Yu, N.K.; Choi, S.L.; Lee, S.H.; Kim, H.; Kaang, B.K. Synaptic protein degradation underlies destabilization of retrieved fear memory. Science 2008, 319, 1253–1256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Jarome, T.J.; Helmstetter, F.J. The ubiquitin–proteasome system as a critical regulator of synaptic plasticity and long-term memory formation. Neurobiol. Learn. Mem. 2013, 105, 107–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kempf, S.J.; Casciati, A.; Buratovic, S.; Janik, D.; von Toerne, C.; Ueffing, M.; Neff, F.; Moertl, S.; Stenerlöw, B.; Saran, A.; et al. The cognitive defects of neonatally irradiated mice are accompanied by changed synaptic plasticity, adult neurogenesis and neuroinflammation. Mol. Neurodegener. 2014. [Google Scholar] [CrossRef] [Green Version]
  41. Kempf, S.J.; Moertl, S.; Sepe, S.; Von Toerne, C.; Hauck, S.M.; Atkinson, M.J.; Mastroberardino, P.G.; Tapio, S. Low-dose ionizing radiation rapidly affects mitochondrial and synaptic signaling pathways in murine hippocampus and cortex. J. Proteome Res. 2015. [Google Scholar] [CrossRef] [PubMed]
  42. Litvinchuk, A.V.; Zorina, E.S.; Kopylov, A.T.; Popova, V.O.; Legina, O.K.; Ronzhina, N.L.; Verlov, N.A.; Karlin, J.L.; Lysenko, V.V.; Ezhov, V.F.; et al. Research of the Effect of Proton Radiation on the Brain Proteome of Mouse. Biol. Bull. 2020. [Google Scholar] [CrossRef]
  43. Hladik, D.; Buratovic, S.; Von Toerne, C.; Azimzadeh, O.; Subedi, P.; Philipp, J.; Winkler, S.; Feuchtinger, A.; Samson, E.; Hauck, S.M.; et al. Combined treatment with low-dose ionizing radiation and ketamine induces adverse changes in CA1 neuronal structure in male murine hippocampi. Int. J. Mol. Sci. 2019, 20, 6103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Bhowmick, P.; Roome, S.; Borchers, C.H.; Goodlett, D.R.; Mohammed, Y. An Update on MRMAssayDB: A Comprehensive Resource for Targeted Proteomics Assays in the Community. J. Proteome Res. 2021, 20, 2105–2115. [Google Scholar] [CrossRef]
  45. Zheng, Q.; Huang, T.; Zhang, L.; Zhou, Y.; Luo, H.; Xu, H.; Wang, X. Dysregulation of Ubiquitin-Proteasome System in Neurodegenerative Diseases. Front. Aging Neurosci. 2016, 8, 303. [Google Scholar] [CrossRef]
  46. Monje, M.L.; Mizumatsu, S.; Fike, J.R.; Palmer, T.D. Irradiation induces neural precursor-cell dysfunction. Nat. Med. 2002, 8, 955–962. [Google Scholar] [CrossRef] [PubMed]
  47. Rodenas-Ruano, A.; Chávez, A.E.; Cossio, M.J.; Castillo, P.E.; Suzanne Zukin, R. REST-dependent epigenetic remodeling promotes the developmental switch in synaptic NMDA receptors. Nat. Neurosci. 2012. [Google Scholar] [CrossRef]
  48. Jarome, T.J.; Ferrara, N.C.; Kwapis, J.L.; Helmstetter, F.J. CaMKII regulates proteasome phosphorylation and activity and promotes memory destabilization following retrieval. Neurobiol. Learn. Mem. 2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Artinian, J.; McGauran, A.M.T.; De Jaeger, X.; Mouledous, L.; Frances, B.; Roullet, P. Protein degradation, as with protein synthesis, is required during not only long-term spatial memory consolidation but also reconsolidation. Eur. J. Neurosci. 2008. [Google Scholar] [CrossRef] [PubMed]
  50. Figueiredo, L.S.; Dornelles, A.S.; Petry, F.S.; Falavigna, L.; Dargél, V.A.; Köbe, L.M.; Aguzzoli, C.; Roesler, R.; Schröder, N. Two waves of proteasome-dependent protein degradation in the hippocampus are required for recognition memory consolidation. Neurobiol. Learn. Mem. 2015. [Google Scholar] [CrossRef]
  51. Lopez-Salon, M.; Alonso, M.; Vianna, M.R.M.; Viola, H.; Mello e Souza, T.; Izquierdo, I.; Pasquini, J.M.; Medina, J.H. The ubiquitin-proteasome cascade is required for mammalian long-term memory formation. Eur. J. Neurosci. 2001. [Google Scholar] [CrossRef] [PubMed]
  52. Cullen, P.K.; Ferrara, N.C.; Pullins, S.E.; Helmstetter, F.J. Context memory formation requires activitydependent protein degradation in the hippocampus. Learn. Mem. 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Oliveros, J.C. VENNY. An Interactive Tool for Comparing Lists with Venn Diagrams. 2007. Available online: http://bioinfogp.cnb.csic.es/tools/venny/index.html.Bioinfogp.Cnb.Csic.Es/Tools/Venny/Index.Html (accessed on 10 July 2021).
  54. Fonseka, P.; Pathan, M.; Chitti, S.V.; Kang, T.; Mathivanan, S. FunRich enables enrichment analysis of OMICs datasets. J. Mol. Biol. 2021, 433, 166747. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of 1 GeV/n 48Ti-exposure on the spatial memory performance of individual rats. Individual REL3 values for sham-irradiated rats (squares) or rats exposed to 15 cGy 1 GeV/n 48Ti (diamonds and triangles); horizontal bar denotes Median value of the REL3 values within a cohort. Cohort abbreviations: Sham: Sham-irradiated rats; 0/P: Sham-irradiated rats used for proteomic analysis, 15: all rats exposed to 15 cGy 1 GeV/n 48Ti; 15/F: all rats classified as having a functional spatial memory; 15/FP: 15/F rats used for proteomic analysis; 15/I: all rats classified as having impaired spatial memory; 15/IP: 15/I rats used for proteomic analysis.
Figure 1. Effect of 1 GeV/n 48Ti-exposure on the spatial memory performance of individual rats. Individual REL3 values for sham-irradiated rats (squares) or rats exposed to 15 cGy 1 GeV/n 48Ti (diamonds and triangles); horizontal bar denotes Median value of the REL3 values within a cohort. Cohort abbreviations: Sham: Sham-irradiated rats; 0/P: Sham-irradiated rats used for proteomic analysis, 15: all rats exposed to 15 cGy 1 GeV/n 48Ti; 15/F: all rats classified as having a functional spatial memory; 15/FP: 15/F rats used for proteomic analysis; 15/I: all rats classified as having impaired spatial memory; 15/IP: 15/I rats used for proteomic analysis.
Ijms 22 07713 g001
Figure 2. Venn diagram of the number and distribution of proteins identified within the hippocampus of the Sham, 15/Functional, and 15/Impaired rat cohorts. Numbers represent the number of proteins detected in each cohort.
Figure 2. Venn diagram of the number and distribution of proteins identified within the hippocampus of the Sham, 15/Functional, and 15/Impaired rat cohorts. Numbers represent the number of proteins detected in each cohort.
Ijms 22 07713 g002
Table 1. Sham Only signature proteins.
Table 1. Sham Only signature proteins.
UniProt Accession Numbers §
A1A5R8P11951P38438Q4G017Q6AXV4Q8K4T4
B0BNE5P12001 *P53678Q4KLZ4Q6AY09 *Q99MZ4
B2DD29P13471 *P55053Q4VSI4 *Q6AY30Q99PF5
B2GV54P15429P56603Q5FVQ8Q6AY41Q9ESB5
B2RZ37P15431P60892Q5MJ12 *Q6AYH5Q9JHU0
B2RZ78P15800P62193 *Q5U2Z5Q6AYK6 *Q9QYL8
D3ZEY4P20069P62824Q62656Q6MG06Q9R085 *
D3ZGS3P22062P62959Q62824Q6MG49Q9R0I8
D4ABY2P23576P68255 *Q63009 *Q6MG55Q9R1N3
F1LSG8P23965P84586 *Q63100Q6MG60Q9WU70
O35274P26817P86182 *Q63633Q6P7S1Q9Z1C7
O54701P29411Q01066Q63881Q78P75Q9Z269 *
P00564P31422Q02356Q64232Q7M6Z5Q9Z2 × 5
P01026P31424Q1WIM2Q64640Q80W83 *
P04256 *P31977Q4FZT0Q66H20Q811X6
§ Fully mapped and annotated in UniProt. * Denotes matched to UPS data base.
Table 2. Titanium Exposure Marker (TEM) signature proteins.
Table 2. Titanium Exposure Marker (TEM) signature proteins.
UniProt Accession Numbers §
F1LQ48 *P13803Q07303Q4KLM4 *
O35763P14604Q02874Q5M9I5
O70511P18484P62243 *Q63803
P09330P21396P53676Q68FS2 *
P13638P26772P48679 *Q91XU8
§ Fully mapped and annotated in UniProt. * Denotes matched to UPS data base.
Table 3. Spatial Memory Impairment (SMI) signature proteins.
Table 3. Spatial Memory Impairment (SMI) signature proteins.
UniProt Accession Numbers §
B0BN85 *P15178P60901Q4KM49Q6P7Q4
B0BNF1P15865P61203Q4V8I7Q6QIX3
D4AE41 *P18163P61227Q505J8Q7TQ94
F1LNJ2 *P20171 *P62024Q569C2Q80W89
F1M386 *P21272 *P62250Q5HZA6Q8VHW5
O35179P29101P62914Q5U216Q9JHL4
O35314P31647P63045Q5XIE8Q9JJ31 *
O35346 *P37285P69682Q5XIF3Q9JKE3
P02650P40307P70619Q5XIG8Q9JMJ4
P04631P47863P84092Q5XIT1Q9WV63
P04785P48768Q08602Q62829 *Q9Z0W7
P07153 *P51635Q32ZG1Q63635Q9Z142
P09895P55051Q3B8Q0Q64591Q9Z1N1
P0C5X8P56536Q499P2Q66X93
P11232P59215Q4FZT2Q68FP1
§ Fully mapped and annotated in UniProt. Bold font denotes only identified in this signature. Italic font denotes proteins that were unregulated (>1.5 fold) in this signature compared to shams. * Denotes matched to UPS data base.
Table 4. Preservation of Spatial Memory (PSM) signature proteins.
Table 4. Preservation of Spatial Memory (PSM) signature proteins.
UniProt Accession Numbers §
B5DF41P25093P62890 *Q3ZB98Q8K4M9
D3ZBM7 *P25809P63029Q497B0Q91Z79
D4A6D8P40329P63088Q4V882Q99M64
O08557P41123 *P69735Q5M7U6Q99PE7
O35821 *P46101P83868Q5PQN0Q9ER24
O54922P46844P84087Q5U2T3Q9JLA3
O55043P47987P85970Q5XHY5Q9QVC8
P10686 *P61107P97837Q62717Q9WTT2
P13264P62268 *Q05695Q63228Q9WVK7
P19468P62853Q07205Q6RUV5Q9Z0V6
P20650P62859Q07310Q812D1Q9Z270
§ Fully mapped and annotated in UniProt. Bold font denotes only identified in this signature. Italic font denotes proteins that were unregulated (>1.5 fold) in this signature compared to shams. * Denotes matched to UPS data base.
Table 5. Relative abundance of signature proteins in the five operational GO processes (identified by FunRich analysis).
Table 5. Relative abundance of signature proteins in the five operational GO processes (identified by FunRich analysis).
Go ProcessDescription % Protein within Functional Signature Involved in the Process
ShamTEMSMIPSM
0019901Protein kinase binding.3.7516.679.0913.89
0042802Identical protein binding21.2511.1130.38.3
0044877Protein-containing complex binding10.011.1115.1511.11
0046872Metal ion binding10.011.116.0611.11
0005524ATP binding15.011.1130.313.89
TEM: Titanium Exposure Marker; SMI: Spatial Memory Impairment; PSM: Preservation of Spatial Memory. Numbers represent the relative abundance of the unique signature proteins as a percentage of all proteins involved in the GO process.
Table 6. Abundance of Ubiquitin-related proteins in the protein signatures associated with various levels of spatial memory functionality.
Table 6. Abundance of Ubiquitin-related proteins in the protein signatures associated with various levels of spatial memory functionality.
Protein Signature Classification
ShamTEMSMIPSMImpaired (TEM&SMI)Functional (TEM&PSM)
15/88 (17%) §5/20 (25%) § 10/37 (27%) § 7/41 (17%) §15/61 (24.5%) §12/57 (21.0%) §
P04256
P12001
P13471
P62193
P68255
P84586
P86182
Q4VSI4
Q5MJ12
Q63009
Q6AY09
Q6AYK6
Q80W83
Q9R085
Q9Z269
F1LQ48
P48679
P62243
Q4KLM4
Q68FS2
B0BN85
D4AE41
F1LNJ2
F1M386
O35346
P07153
P20171
P21272
Q62829
Q9JJ31
D3ZBM7
O35821
P10686
P41123
P62268
P62890
Q6RUV5
B0BN85
D4AE41
F1LNJ2
F1LQ48
F1M386
O35346
P07153
P20171
P21272
P48679
P62243
Q4KLM4
Q62829
Q68FS2
Q9JJ31
D3ZBM7
F1LQ48
O35821
P10686
P41123
P48679
P62243
P62268
P62890
Q4KLM4
Q68FS2
Q6RUV5
TEM: Titanium Exposure Marker; SMI: Spatial Memory Impairment; PSM: Preservation of Spatial Memory. § Numbers represent the number of URP proteins as a function of the total number of proteins found within the functional signature, and the calculated relative abundance of the Ubiquitin-related proteins (URP) proteins within each signature.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tidmore, A.; Dutta, S.M.; Fesshaye, A.S.; Russell, W.K.; Duncan, V.D.; Britten, R.A. Space Radiation-Induced Alterations in the Hippocampal Ubiquitin-Proteome System. Int. J. Mol. Sci. 2021, 22, 7713. https://doi.org/10.3390/ijms22147713

AMA Style

Tidmore A, Dutta SM, Fesshaye AS, Russell WK, Duncan VD, Britten RA. Space Radiation-Induced Alterations in the Hippocampal Ubiquitin-Proteome System. International Journal of Molecular Sciences. 2021; 22(14):7713. https://doi.org/10.3390/ijms22147713

Chicago/Turabian Style

Tidmore, Alyssa, Sucharita M. Dutta, Arriyam S. Fesshaye, William K. Russell, Vania D. Duncan, and Richard A. Britten. 2021. "Space Radiation-Induced Alterations in the Hippocampal Ubiquitin-Proteome System" International Journal of Molecular Sciences 22, no. 14: 7713. https://doi.org/10.3390/ijms22147713

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop