1
|
Malek N, Gladysz R, Stelmach N, Drag M. Targeting Microglial Immunoproteasome: A Novel Approach in Neuroinflammatory-Related Disorders. ACS Chem Neurosci 2024; 15:2532-2544. [PMID: 38970802 PMCID: PMC11258690 DOI: 10.1021/acschemneuro.4c00099] [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: 02/15/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024] Open
Abstract
It is widely acknowledged that the aging process is linked to the accumulation of damaged and misfolded proteins. This phenomenon is accompanied by a decrease in proteasome (c20S) activity, concomitant with an increase in immunoproteasome (i20S) activity. These changes can be attributed, in part, to the chronic neuroinflammation that occurs in brain tissues. Neuroinflammation is a complex process characterized by the activation of immune cells in the central nervous system (CNS) in response to injury, infection, and other pathological stimuli. In certain cases, this immune response becomes chronic, contributing to the pathogenesis of various neurological disorders, including chronic pain, Alzheimer's disease, Parkinson's disease, brain traumatic injury, and others. Microglia, the resident immune cells in the brain, play a crucial role in the neuroinflammatory response. Recent research has highlighted the involvement of i20S in promoting neuroinflammation, increased activity of which may lead to the presentation of self-antigens, triggering an autoimmune response against the CNS, exacerbating inflammation, and contributing to neurodegeneration. Furthermore, since i20S plays a role in breaking down accumulated proteins during inflammation within the cell body, any disruption in its activity could lead to a prolonged state of inflammation and subsequent cell death. Given the pivotal role of i20S in neuroinflammation, targeting this proteasome subtype has emerged as a potential therapeutic approach for managing neuroinflammatory diseases. This review delves into the mechanisms of neuroinflammation and microglia activation, exploring the potential of i20S inhibitors as a promising therapeutic strategy for managing neuroinflammatory disorders.
Collapse
Affiliation(s)
- Natalia Malek
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Radoslaw Gladysz
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Natalia Stelmach
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marcin Drag
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| |
Collapse
|
2
|
Oganezovi N, Lagani V, Kikvidze M, Gamkrelidze G, Tsverava L, Lepsveridze E, Kelly KM, Solomonia R. Long-term effects of myo-inositol on traumatic brain injury: Epigenomic and transcriptomic studies. IBRO Neurosci Rep 2024; 16:291-299. [PMID: 38374956 PMCID: PMC10875114 DOI: 10.1016/j.ibneur.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/27/2024] [Indexed: 02/21/2024] Open
Abstract
Background and purpose Traumatic brain injury (TBI) and its consequences remain great challenges for neurology. Consequences of TBI are associated with various alterations in the brain but little is known about long-term changes of epigenetic DNA methylation patterns. Moreover, nothing is known about potential treatments that can alter these epigenetic changes in beneficial ways. Therefore, we have examined myo-inositol (MI), which has positive effects on several pathological conditions. Methods TBI was induced in mice by controlled cortical impact (CCI). One group of CCI animals received saline injections for two months (TBI+SAL), another CCI group received MI treatment (TBI+MI) for the same period and one group served as a sham-operated control. Mice were sacrificed 4 months after CCI and changes in DNA methylome and transcriptomes were examined. Results For the first time we: (i) provide comprehensive map of long-term DNA methylation changes after CCI in the hippocampus; (ii) identify differences by methylation sites between the groups; (iii) characterize transcriptome changes; (iv) provide association between DNA methylation sites and gene expression. MI treatment is linked with upregulation of genes covering 33 biological processes, involved in immune response and inflammation. In support of these findings, we have shown that expression of BATF2, a transcription factor involved in immune-regulatory networks, is upregulated in the hippocampus of the TBI+MI group where the BATF2 gene is demethylated. Conclusion TBI is followed by long-term epigenetic and transcriptomic changes in hippocampus. MI treatment has a significant effect on these processes by modulation of immune response and biological pathways of inflammation.
Collapse
Affiliation(s)
- Nino Oganezovi
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Vincenzo Lagani
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Marine Kikvidze
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Georgi Gamkrelidze
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Lia Tsverava
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
- Iv. Beritashvili Centre of Experimental Biomedicine, Tbilisi, Georgia
| | - Eka Lepsveridze
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
| | - Kevin M. Kelly
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Neurology, Philadelphia, PA, United States
- Department of Neurology, Allegheny General Hospital, Pittsburgh, PA, United States
- Center for Neuroscience Research, Allegheny Health Network Research Institute, Pittsburgh, PA, United States, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Revaz Solomonia
- School of Natural Sciences and Medicine, Institute of Chemical Biology, Ilia State University, Tbilisi, Georgia
- Iv. Beritashvili Centre of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
3
|
Mi Z, Ma J, Zeh DJ, Rose ME, Henchir JJ, Liu H, Ma X, Cao G, Dixon CE, Graham SH. Systemic treatment with ubiquitin carboxy terminal hydrolase L1 TAT protein ameliorates axonal injury and reduces functional deficits after traumatic brain injury in mice. Exp Neurol 2024; 373:114650. [PMID: 38092186 PMCID: PMC10939891 DOI: 10.1016/j.expneurol.2023.114650] [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: 06/01/2023] [Revised: 11/17/2023] [Accepted: 12/09/2023] [Indexed: 12/21/2023]
Abstract
Traumatic brain injury (TBI) is often associated with axonal injury that leads to significant motor and cognitive deficits. Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is highly expressed in neurons and loss of its activity plays an important role in the pathogenesis of TBI. Fusion protein was constructed containing wild type (WT) UCHL1 and the HIV trans-activator of transcription capsid protein transduction domain (TAT-UCHL1) that facilitates transport of the protein into neurons after systemic administration. Additional mutant proteins bearing cysteine to alanine UCHL1 mutations at cysteine 152 (C152A TAT-UCHL1) that prevents nitric oxide and reactive lipid binding of C152, and at cysteine 220 (C220A TAT-UCHL1) that inhibits farnesylation of the C220 site were also constructed. WT, C152A, and C220A TAT-UCHL1 proteins administered to mice systemically after controlled cortical impact (CCI) were detectable in brain at 1 h, 4 h and 24 h after CCI by immunoblot. Mice treated with C152A or WT TAT-UCHL1 decreased axonal injury detected by NF200 immunohistochemistry 24 h after CCI, but C220A TAT-UCHL1 treatment had no significant effect. Further study indicated that WT TAT-UCHL1 treatment administered 24 h after CCI alleviated axonal injury as detected by SMI32 immunoreactivity 7 d after CCI, improved motor and cognitive deficits, reduced accumulation of total and K48-linked poly-Ub proteins, and attenuated the increase of the autophagy marker Beclin-1. These results suggest that UCHL1 activity contributes to the pathogenesis of white matter injury, and that restoration of UCHL1 activity by systemic treatment with WT TAT-UCHL1 after CCI may improve motor and cognitive deficits. These results also suggest that farnesylation of the C220 site may be required for the protective effects of UCHL1.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dennis J Zeh
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeremy J Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina
| | - Xiecheng Ma
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Guodong Cao
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Reiners JC, Leopold L, Hallebach V, Sinske D, Meier P, Amoroso M, Langgartner D, Reber SO, Knöll B. Acute stress modulates the outcome of traumatic brain injury-associated gene expression and behavioral responses. FASEB J 2023; 37:e23218. [PMID: 37779443 DOI: 10.1096/fj.202301035r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023]
Abstract
Psychological stress and traumatic brain injury (TBI) result in long-lasting emotional and behavioral impairments in patients. So far, the interaction of psychological stress with TBI not only in the brain but also in peripheral organs is poorly understood. Herein, the impact of acute stress (AS) occurring immediately before TBI is investigated. For this, a mouse model of restraint stress and TBI was employed, and their influence on behavior and gene expression in brain regions, the hypothalamic-pituitary-adrenal (HPA) axis, and peripheral organs was analyzed. Results demonstrate that, compared to single AS or TBI exposure, mice treated with AS prior to TBI showed sex-specific alterations in body weight, memory function, and locomotion. The induction of immediate early genes (IEGs, e.g., c-Fos) by TBI was modulated by previous AS in several brain regions. Furthermore, IEG upregulation along the HPA axis (e.g., pituitary, adrenal glands) and other peripheral organs (e.g., heart) was modulated by AS-TBI interaction. Proteomics of plasma samples revealed proteins potentially mediating this interaction. Finally, the deletion of Atf3 diminished the TBI-induced induction of IEGs in peripheral organs but left them largely unaltered in the brain. In summary, AS immediately before brain injury affects the brain and, to a strong degree, also responses in peripheral organs.
Collapse
Affiliation(s)
| | - Laura Leopold
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Vera Hallebach
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Daniela Sinske
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| | - Philip Meier
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Mattia Amoroso
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Bernd Knöll
- Institute of Neurobiochemistry, Ulm University, Ulm, Germany
| |
Collapse
|
5
|
Mi Z, Liu H, Rose ME, Ma J, Reay DP, Ma X, Henchir JJ, Dixon CE, Graham SH. Mutation of a Ubiquitin Carboxy Terminal Hydrolase L1 Lipid Binding Site Alleviates Cell Death, Axonal Injury, and Behavioral Deficits After Traumatic Brain Injury in Mice. Neuroscience 2021; 475:127-136. [PMID: 34508847 DOI: 10.1016/j.neuroscience.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022]
Abstract
Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is a protein highly expressed in neurons that may play important roles in the ubiquitin proteasome pathway (UPP) in neurons, axonal integrity, and motor function after traumatic brain injury (TBI). Binding of reactive lipid species to cysteine 152 of UCHL1 results in unfolding, aggregation, and inactivation of the enzyme. To test the role of this mechanism in TBI, mice bearing a cysteine to alanine mutation at site 152 (C152A mice) that renders UCHL1 resistant to inactivation by reactive lipids were subjected to the controlled cortical impact model (CCI) of TBI and compared to wild type (WT) controls. Alterations in protein ubiquitination and activation of autophagy pathway markers in traumatized brain were detected by immunoblotting. Cell death and axonal injury were determined by histological assessment and anti-amyloid precursor protein (APP) immunohistochemistry. Behavioral outcomes were determined using the beam balance and Morris water maze tests. C152A mice had reduced accumulation of ubiquitinated proteins, decreased activation of the autophagy markers Beclin-1 and LC3B, a decreased number of abnormal axons, decreased CA1 cell death, and improved motor and cognitive function compared to WT controls after CCI; no significant change in spared tissue volume was observed. These results suggest that binding of lipid substrates to cysteine 152 of UCHL1 is important in the pathogenesis of injury and recovery after TBI and may be a novel target for future therapeutic approaches.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Jeremy J Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
6
|
Oleuropein Activates Neonatal Neocortical Proteasomes, but Proteasome Gene Targeting by AAV9 Is Variable in a Clinically Relevant Piglet Model of Brain Hypoxia-Ischemia and Hypothermia. Cells 2021; 10:cells10082120. [PMID: 34440889 PMCID: PMC8391411 DOI: 10.3390/cells10082120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 10/26/2022] Open
Abstract
Cerebral hypoxia-ischemia (HI) compromises the proteasome in a clinically relevant neonatal piglet model. Protecting and activating proteasomes could be an adjunct therapy to hypothermia. We investigated whether chymotrypsin-like proteasome activity differs regionally and developmentally in the neonatal brain. We also tested whether neonatal brain proteasomes can be modulated by oleuropein, an experimental pleiotropic neuroprotective drug, or by targeting a proteasome subunit gene using recombinant adeno-associated virus-9 (AAV). During post-HI hypothermia, we treated piglets with oleuropein, used AAV-short hairpin RNA (shRNA) to knock down proteasome activator 28γ (PA28γ), or enforced PA28γ using AAV-PA28γ with green fluorescent protein (GFP). Neonatal neocortex and subcortical white matter had greater proteasome activity than did liver and kidney. Neonatal white matter had higher proteasome activity than did juvenile white matter. Lower arterial pH 1 h after HI correlated with greater subsequent cortical proteasome activity. With increasing brain homogenate protein input into the assay, the initial proteasome activity increased only among shams, whereas HI increased total kinetic proteasome activity. OLE increased the initial neocortical proteasome activity after hypothermia. AAV drove GFP expression, and white matter PA28γ levels correlated with proteasome activity and subunit levels. However, AAV proteasome modulation varied. Thus, neonatal neocortical proteasomes can be pharmacologically activated. HI slows the initial proteasome performance, but then augments ongoing catalytic activity. AAV-mediated genetic manipulation in the piglet brain holds promise, though proteasome gene targeting requires further development.
Collapse
|
7
|
Saikumar J, Bonini NM. Synergistic effects of brain injury and aging: common mechanisms of proteostatic dysfunction. Trends Neurosci 2021; 44:728-740. [PMID: 34301397 DOI: 10.1016/j.tins.2021.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023]
Abstract
The aftermath of TBI is associated with an acute stress response and the accumulation of insoluble protein aggregates. Even after the symptoms of TBI are resolved, insidious molecular processes continue to develop, which often ultimately result in the development of age-associated neurodegenerative disorders. The precise molecular cascades that drive unhealthy brain aging are still largely unknown. In this review, we discuss proteostatic dysfunction as a converging mechanism contributing to accelerated brain aging after TBI. We examine evidence from human tissue and in vivo animal models, spanning both the aging and injury contexts. We conclude that TBI has a sustained debilitating effect on the proteostatic machinery, which may contribute to the accelerated pathological and cognitive hallmarks of aging that are observed following injury.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Anderson EN, Morera AA, Kour S, Cherry JD, Ramesh N, Gleixner A, Schwartz JC, Ebmeier C, Old W, Donnelly CJ, Cheng JP, Kline AE, Kofler J, Stein TD, Pandey UB. Traumatic injury compromises nucleocytoplasmic transport and leads to TDP-43 pathology. eLife 2021; 10:e67587. [PMID: 34060470 PMCID: PMC8169113 DOI: 10.7554/elife.67587] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a predisposing factor for many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and chronic traumatic encephalopathy (CTE). Although defects in nucleocytoplasmic transport (NCT) is reported ALS and other neurodegenerative diseases, whether defects in NCT occur in TBI remains unknown. We performed proteomic analysis on Drosophila exposed to repeated TBI and identified resultant alterations in several novel molecular pathways. TBI upregulated nuclear pore complex (NPC) and nucleocytoplasmic transport (NCT) proteins as well as alter nucleoporin stability. Traumatic injury disrupted RanGAP1 and NPC protein distribution in flies and a rat model and led to coaggregation of NPC components and TDP-43. In addition, trauma-mediated NCT defects and lethality are rescued by nuclear export inhibitors. Importantly, genetic upregulation of nucleoporins in vivo and in vitro triggered TDP-43 cytoplasmic mislocalization, aggregation, and altered solubility and reduced motor function and lifespan of animals. We also found NUP62 pathology and elevated NUP62 concentrations in postmortem brain tissues of patients with mild or severe CTE as well as co-localization of NUP62 and TDP-43 in CTE. These findings indicate that TBI leads to NCT defects, which potentially mediate the TDP-43 pathology in CTE.
Collapse
Affiliation(s)
- Eric N Anderson
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical CenterPittsburghUnited States
| | - Andrés A Morera
- Department of Chemistry and Biochemistry, University of ArizonaTucsonUnited States
| | - Sukhleen Kour
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical CenterPittsburghUnited States
| | - Jonathan D Cherry
- Department of Pathology and Laboratory Medicine, Boston University School of MedicineBostonUnited States
- Boston VA Healthcare SystemBostonUnited States
| | - Nandini Ramesh
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical CenterPittsburghUnited States
| | - Amanda Gleixner
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
- LiveLike Lou Center for ALS Research, Brain Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Jacob C Schwartz
- Department of Chemistry and Biochemistry, University of ArizonaTucsonUnited States
| | - Christopher Ebmeier
- Molecular, Cellular & Developmental Biology, University of ColoradoBoulderUnited States
| | - William Old
- Molecular, Cellular & Developmental Biology, University of ColoradoBoulderUnited States
| | - Christopher J Donnelly
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
- LiveLike Lou Center for ALS Research, Brain Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation; Safar Center for Resuscitation Research, University of PittsburghPittsburghUnited States
| | - Anthony E Kline
- Physical Medicine & Rehabilitation; Safar Center for Resuscitation Research, University of PittsburghPittsburghUnited States
- Center for Neuroscience; Center for the Neural Basis of Cognition; Critical Care Medicine, University of PittsburghPittsburghUnited States
| | - Julia Kofler
- Department of Pathology, University of PittsburghPittsburghUnited States
| | - Thor D Stein
- Department of Pathology and Laboratory Medicine, Boston University School of MedicineBostonUnited States
- Boston VA Healthcare SystemBostonUnited States
| | - Udai Bhan Pandey
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical CenterPittsburghUnited States
- Department of Human Genetics, University of Pittsburgh School of Public HealthPittsburghUnited States
| |
Collapse
|
9
|
Mladenovic Djordjevic AN, Kapetanou M, Loncarevic-Vasiljkovic N, Todorovic S, Athanasopoulou S, Jovic M, Prvulovic M, Taoufik E, Matsas R, Kanazir S, Gonos ES. Pharmacological intervention in a transgenic mouse model improves Alzheimer's-associated pathological phenotype: Involvement of proteasome activation. Free Radic Biol Med 2021; 162:88-103. [PMID: 33279620 PMCID: PMC7889698 DOI: 10.1016/j.freeradbiomed.2020.11.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/23/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide, characterized by a progressive decline in a variety of cognitive and non-cognitive functions. The amyloid beta protein cascade hypothesis places the formation of amyloid beta protein aggregates on the first position in the complex pathological cascade leading to neurodegeneration, and therefore AD might be considered to be a protein-misfolding disease. The Ubiquitin Proteasome System (UPS), being the primary protein degradation mechanism with a fundamental role in the maintenance of proteostasis, has been identified as a putative therapeutic target to delay and/or to decelerate the progression of neurodegenerative disorders that are characterized by accumulated/aggregated proteins. The purpose of this study was to test if the activation of proteasome in vivo can alleviate AD pathology. Specifically by using two compounds with complementary modes of proteasome activation and documented antioxidant and redox regulating properties in the 5xFAD transgenic mice model of AD, we ameliorated a number of AD related deficits. Shortly after proteasome activation we detected significantly reduced amyloid-beta load correlated with improved motor functions, reduced anxiety and frailty level. Essentially, to our knowledge this is the first report to demonstrate a dual activation of the proteasome and its downstream effects. In conclusion, these findings open up new directions for future therapeutic potential of proteasome-mediated proteolysis enhancement.
Collapse
Affiliation(s)
- Aleksandra N Mladenovic Djordjevic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia.
| | - Marianna Kapetanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635, Athens, Greece
| | - Natasa Loncarevic-Vasiljkovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia; Molecular Nutrition and Health Lab, CEDOC - Centro de Estudos de Doenças Crónicas, NOVA Medical School / Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Edifício CEDOC II, Rua Câmara Pestana 6, 1150-082, Lisboa, Portugal
| | - Smilja Todorovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia
| | - Sofia Athanasopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635, Athens, Greece; Department of Biology, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Milena Jovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia
| | - Milica Prvulovic
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia
| | - Era Taoufik
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521, Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521, Athens, Greece
| | - Selma Kanazir
- Department for Neurobiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Boulevard Despota Stefana, 142, 11000, Belgrade, Serbia
| | - Efstathios S Gonos
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., 11635, Athens, Greece.
| |
Collapse
|
10
|
Mi Z, Liu H, Rose ME, Ma X, Reay DP, Ma J, Henchir J, Dixon CE, Graham SH. Abolishing UCHL1's hydrolase activity exacerbates TBI-induced axonal injury and neuronal death in mice. Exp Neurol 2020; 336:113524. [PMID: 33159930 DOI: 10.1016/j.expneurol.2020.113524] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 01/13/2023]
Abstract
Ubiquitin (Ub) C-terminal hydrolase L1 (UCHL1) is a multifunctional protein that is expressed in neurons throughout brain at high levels. UCHL1 deletion is associated with axonal degeneration, progressive sensory motor ataxia, and premature death in mice. UCHL1 has been hypothesized to play a role in the pathogenesis of neurodegenerative diseases and recovery after neuronal injury. UCHL1 hydrolyzes Ub from polyubiquitinated (poly-Ub) proteins, but also may ligate Ub to select neuronal proteins, and interact with cytoskeletal proteins. These and other mechanisms have been hypothesized to underlie UCHL1's role in neurodegeneration and response to brain injury. A UCHL1 knockin mouse containing a C90A mutation (C90A) devoid of hydrolase activity was constructed. The C90A mouse did not develop the sensory and motor deficits, degeneration of the gracile nucleus and tract, or premature death as seen in UCHL1 deficient mice. C90A and wild type (WT) mice were subjected to the controlled cortical impact (CCI) model of traumatic brain injury (TBI), and cell death, axonal injury and behavioral outcome were assessed. C90A mice exhibited decreased spared tissue volume, greater loss of CA1 hippocampal neurons and greater axonal injury as detected using anti-amyloid precursor protein (APP) antibody and anti- non-phosphorylated neurofilament H (SMI-32) antibody immunohistochemistry after CCI compared to WT controls. Poly-Ub proteins and Beclin-1 were elevated after CCI in C90A mice compared to WT controls. Vestibular motor deficits assessed using the beam balance test resolved by day 5 after CCI in WT mice but not in C90A mice. These results suggest that the hydrolase activity of UCHL1 does not account for the progressive neurodegeneration and premature death seen in mice that do not express full length UCHL1. The hydrolase activity of UCHL1 contributes to the function of the ubiquitin proteasome pathway (UPP), ameliorates activation of autophagy, and improves motor recovery after CCI. Thus, UCHL1 hydrolase activity plays an important role in acute injury response after TBI.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Jeremy Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA.
| |
Collapse
|
11
|
Saikumar J, Byrns CN, Hemphill M, Meaney DF, Bonini NM. Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17269-17277. [PMID: 32611818 PMCID: PMC7382229 DOI: 10.1073/pnas.2003909117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - China N Byrns
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104;
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
12
|
Feldmann LK, Le Prieult F, Felzen V, Thal SC, Engelhard K, Behl C, Mittmann T. Proteasome and Autophagy-Mediated Impairment of Late Long-Term Potentiation (l-LTP) after Traumatic Brain Injury in the Somatosensory Cortex of Mice. Int J Mol Sci 2019; 20:ijms20123048. [PMID: 31234472 PMCID: PMC6627835 DOI: 10.3390/ijms20123048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) can lead to impaired cognition and memory consolidation. The acute phase (24–48 h) after TBI is often characterized by neural dysfunction in the vicinity of the lesion, but also in remote areas like the contralateral hemisphere. Protein homeostasis is crucial for synaptic long-term plasticity including the protein degradation systems, proteasome and autophagy. Still, little is known about the acute effects of TBI on synaptic long-term plasticity and protein degradation. Thus, we investigated TBI in a controlled cortical impact (CCI) model in the motor and somatosensory cortex of mice ex vivo-in vitro. Late long-term potentiation (l-LTP) was induced by theta-burst stimulation in acute brain slices after survival times of 1–2 days. Protein levels for the plasticity related protein calcium/calmodulin-dependent protein kinase II (CaMKII) was quantified by Western blots, and the protein degradation activity by enzymatical assays. We observed missing maintenance of l-LTP in the ipsilateral hemisphere, however not in the contralateral hemisphere after TBI. Protein levels of CaMKII were not changed but, interestingly, the protein degradation revealed bidirectional changes with a reduced proteasome activity and an increased autophagic flux in the ipsilateral hemisphere. Finally, LTP recordings in the presence of pharmacologically modified protein degradation systems also led to an impaired synaptic plasticity: bath-applied MG132, a proteasome inhibitor, or rapamycin, an activator of autophagy, both administered during theta burst stimulation, blocked the induction of LTP. These data indicate that alterations in protein degradation pathways likely contribute to cognitive deficits in the acute phase after TBI, which could be interesting for future approaches towards neuroprotective treatments early after traumatic brain injury.
Collapse
Affiliation(s)
- Lucia K Feldmann
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Florie Le Prieult
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Vanessa Felzen
- Institute for Pathobiochemistry, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Serge C Thal
- Clinics for Anaesthesiology, UMC of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Kristin Engelhard
- Clinics for Anaesthesiology, UMC of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Christian Behl
- Institute for Pathobiochemistry, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| | - Thomas Mittmann
- Institute for Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|
13
|
Adelöf J, Andersson M, Porritt M, Petersen A, Zetterberg M, Wiseman J, Hernebring M. PA28αβ overexpression enhances learning and memory of female mice without inducing 20S proteasome activity. BMC Neurosci 2018; 19:70. [PMID: 30400847 PMCID: PMC6218978 DOI: 10.1186/s12868-018-0468-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/27/2018] [Indexed: 11/16/2022] Open
Abstract
Background The proteasome system plays an important role in synaptic plasticity. Induction and maintenance of long term potentiation is directly dependent on selective targeting of proteins for proteasomal degradation. The 20S proteasome activator PA28αβ activates hydrolysis of small nonubiquitinated peptides and possesses protective functions upon oxidative stress and proteinopathy. The effect of PA28αβ activity on behavior and memory function is, however, not known. We generated a mouse model that overexpresses PA28α (PA28αOE) to understand PA28αβ function during healthy adult homeostasis via assessment of physiological and behavioral profiles, focusing on female mice. Results PA28α and PA28β protein levels were markedly increased in all PA28αOE tissues analyzed. PA28αOE displayed reduced depressive-like behavior in the forced swim test and improved memory/learning function assessed by intersession habituation in activity box and shuttle box passive avoidance test, with no significant differences in anxiety or general locomotor activity. Nor were there any differences found when compared to WT for body composition or immuno-profile. The cognitive effects of PA28αOE were female specific, but could not be explained by alterations in estrogen serum levels or hippocampal regulation of estrogen receptor β. Further, there were no differences in hippocampal protein expression of neuronal or synaptic markers between PA28αOE and WT. Biochemical analysis of hippocampal extracts demonstrated that PA28α overexpression did not increase PA28–20S peptidase activity or decrease K48-polyubiquitin levels. Instead, PA28αOE exhibited elevated efficiency in preventing aggregation in the hippocampus. Conclusions This study reveals, for the first time, a connection between PA28αβ and neuronal function. We found that PA28α overexpressing female mice displayed reduced depressive-like behavior and enhanced learning and memory. Since the positive effects of PA28α overexpression arose without an activation of 20S proteasome capacity, they are likely independent of PA28αβ’s role as a 20S proteasome activator and instead depend on a recognized chaperone-like function. These findings suggest that proteostasis in synaptic plasticity is more diverse than previously reported, and demonstrates a novel function of PA28αβ in the brain. Electronic supplementary material The online version of this article (10.1186/s12868-018-0468-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Adelöf
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,IMED Biotech Unit, Discovery Biology, Discovery Sciences, AstraZeneca, Gothenburg, Sweden
| | - My Andersson
- Department of Clinical Sciences, Epilepsy Centre, Lund University, Lund, Sweden
| | - Michelle Porritt
- IMED Biotech Unit, Discovery Biology, Discovery Sciences, AstraZeneca, Gothenburg, Sweden
| | - Anne Petersen
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Madeleine Zetterberg
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - John Wiseman
- IMED Biotech Unit, Discovery Biology, Discovery Sciences, AstraZeneca, Gothenburg, Sweden
| | - Malin Hernebring
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden. .,IMED Biotech Unit, Discovery Biology, Discovery Sciences, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
14
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
15
|
Moritz KE, McCormack NM, Abera MB, Viollet C, Yauger YJ, Sukumar G, Dalgard CL, Burnett BG. The role of the immunoproteasome in interferon-γ-mediated microglial activation. Sci Rep 2017; 7:9365. [PMID: 28839214 PMCID: PMC5571106 DOI: 10.1038/s41598-017-09715-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/14/2017] [Indexed: 11/12/2022] Open
Abstract
Microglia regulate the brain microenvironment by sensing damage and neutralizing potentially harmful insults. Disruption of central nervous system (CNS) homeostasis results in transition of microglia to a reactive state characterized by morphological changes and production of cytokines to prevent further damage to CNS tissue. Immunoproteasome levels are elevated in activated microglia in models of stroke, infection and traumatic brain injury, though the exact role of the immunoproteasome in neuropathology remains poorly defined. Using gene expression analysis and native gel electrophoresis we characterize the expression and assembly of the immunoproteasome in microglia following interferon-gamma exposure. Transcriptome analysis suggests that the immunoproteasome regulates multiple features of microglial activation including nitric oxide production and phagocytosis. We show that inhibiting the immunoproteasome attenuates expression of pro-inflammatory cytokines and suppresses interferon-gamma-dependent priming of microglia. These results imply that targeting immunoproteasome function following CNS injury may attenuate select microglial activity to improve the pathophysiology of neurodegenerative conditions or the progress of inflammation-mediated secondary injury following neurotrauma.
Collapse
Affiliation(s)
- Kasey E Moritz
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Nikki M McCormack
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Mahlet B Abera
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Coralie Viollet
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Young J Yauger
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Clifton L Dalgard
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Barrington G Burnett
- Neuroscience Program, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, MD, USA. .,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
16
|
Engel T, Martinez-Villarreal J, Henke C, Jimenez-Mateos EM, Sanz-Rodriguez A, Alves M, Hernandez-Santana Y, Brennan GP, Kenny A, Campbell A, Lucas JJ, Henshall DC. Spatiotemporal progression of ubiquitin-proteasome system inhibition after status epilepticus suggests protective adaptation against hippocampal injury. Mol Neurodegener 2017; 12:21. [PMID: 28235423 PMCID: PMC5324261 DOI: 10.1186/s13024-017-0163-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 02/17/2017] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND The ubiquitin-proteasome-system (UPS) is the major intracellular pathway leading to the degradation of unwanted and/or misfolded soluble proteins. This includes proteins regulating cellular survival, synaptic plasticity and neurotransmitter signaling; processes controlling excitability thresholds that are altered by epileptogenic insults. Dysfunction of the UPS has been reported to occur in a brain region- and cell-specific manner and contribute to disease progression in acute and chronic brain diseases. Prolonged seizures, status epilepticus, may alter UPS function but there has been no systematic attempt to map when and where this occurs in vivo or to determine the consequences of proteasome inhibition on seizure-induced brain injury. METHOD To determine whether seizures lead to an impairment of the UPS, we used a mouse model of status epilepticus whereby seizures are triggered by an intra-amygdala injection of kainic acid. Status epilepticus in this model causes cell death in selected brain areas, in particular the ipsilateral CA3 subfield of the hippocampus, and the development of epilepsy after a short latent period. To monitor seizure-induced dysfunction of the UPS we used a UPS inhibition reporter mouse expressing the ubiquitin fusion degradation substrate ubiquitinG76V-green fluorescent protein. Treatment with the specific proteasome inhibitor epoxomicin was used to establish the impact of proteasome inhibition on seizure-induced pathology. RESULTS AND CONCLUSIONS Our studies show that status epilepticus induced by intra-amygdala kainic acid causes select spatio-temporal UPS inhibition which is most evident in damage-resistant regions of the hippocampus, including CA1 pyramidal and dentate granule neurons then appears later in astrocytes. In support of this exerting a beneficial effect, injection of mice with the proteasome inhibitor epoxomicin protected the normally vulnerable hippocampal CA3 subfield from seizure-induced neuronal death in the model. These studies reveal brain region- and cell-specific UPS impairment occurs after seizures and suggest UPS inhibition can protect against seizure-induced brain damage. Identifying networks or pathways regulated through the proteasome after seizures may yield novel target genes for the treatment of seizure-induced cell death and possibly epilepsy.
Collapse
Affiliation(s)
- Tobias Engel
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland.
| | - Jaime Martinez-Villarreal
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Christine Henke
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland.,Medical Clinic III, University Clinic Dresden, TU Dresden, Dresden, Germany
| | - Eva M Jimenez-Mateos
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Amaya Sanz-Rodriguez
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Mariana Alves
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Yasmina Hernandez-Santana
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Gary P Brennan
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Aidan Kenny
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Aoife Campbell
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| | - Jose J Lucas
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 111 St. Stephen's Green, Dublin, 02 YN77, Ireland
| |
Collapse
|
17
|
Zhang Q, Zhang M, Huang X, Liu X, Li W. Inhibition of cytoskeletal protein carbonylation may protect against oxidative damage in traumatic brain injury. Exp Ther Med 2017; 12:4107-4112. [PMID: 28101189 DOI: 10.3892/etm.2016.3889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 09/27/2016] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress is the principal factor in traumatic brain injury (TBI) that initiates protracted neuronal dysfunction and remodeling. Cytoskeletal proteins are known to be carbonylated under oxidative stress; however, the complex molecular and cellular mechanisms of cytoskeletal protein carbonylation remain poorly understood. In the present study, the expression levels of glutathione (GSH) and thiobarbituric acid reactive substances (TBARS) were investigated in PC12 cells treated with H2O2. Western blot analysis was used to monitor the carbonylation levels of β-actin and β-tubulin. The results indicated that oxidative stress was increased in PC12 cells that were treated with H2O2 for 24 or 48 h. In addition, increased carbonylation levels of β-actin and β-tubulin were detected in H2O2-treated cells. However, these carbonylation levels were reduced by pretreatment with aminoguanidine, a type of reactive carbonyl species chelating agent, and a similar trend was observed following overexpression of proteasome β5 via transgenic technology. In conclusion, the present study results suggested that the development of TBI may cause carbonylation of cytoskeletal proteins, which would then undermine the stability of cytoskeletal proteins. Thus, the development of TBI may be improved via the inhibition of cytoskeletal protein carbonylation.
Collapse
Affiliation(s)
- Qiusheng Zhang
- The Clinical College of Shenzhen Second Hospital, Anhui Medical University, Shenzhen, Guangdong 518035, P.R. China; Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 508035, P.R. China
| | - Meng Zhang
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 508035, P.R. China
| | - Xianjian Huang
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 508035, P.R. China
| | - Xiaojia Liu
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 508035, P.R. China
| | - Weiping Li
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, Guangdong 508035, P.R. China
| |
Collapse
|
18
|
Peptide Pharmacological Approaches to Treating Traumatic Brain Injury: a Case for Arginine-Rich Peptides. Mol Neurobiol 2016; 54:7838-7857. [PMID: 27844291 DOI: 10.1007/s12035-016-0287-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/02/2016] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury (TBI) has a devastating effect on victims and their families, and has profound negative societal and economic impacts, a situation that is further compounded by the lack of effective treatments to minimise injury after TBI. The current strategy for managing TBI is partly through preventative measures and partly through surgical and rehabilitative interventions. Secondary brain damage remains the principal focus for the development of a neuroprotective therapeutic. However, the complexity of TBI pathophysiology has meant that single-action pharmacological agents have been largely unsuccessful in combatting the associated brain injury cascades, while combination therapies to date have proved equally ineffective. Peptides have recently emerged as promising lead agents for the treatment of TBI, especially those rich in the cationic amino acid, arginine. Having been shown to lessen the impact of ischaemic stroke in animal models, there are reasonable grounds to believe that arginine-rich peptides may have neuroprotective therapeutic potential in TBI. Here, we review a range of peptides previously examined as therapeutic agents for TBI. In particular, we focus on cationic arginine-rich peptides -- a new class of agents that growing evidence suggests acts through multiple neuroprotective mechanisms.
Collapse
|
19
|
Zhao Y, Liu X, He Z, Niu X, Shi W, Ding JM, Zhang L, Yuan T, Li A, Yang W, Lu L. Essential role of proteasomes in maintaining self-renewal in neural progenitor cells. Sci Rep 2016; 6:19752. [PMID: 26804982 PMCID: PMC4726439 DOI: 10.1038/srep19752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/17/2015] [Indexed: 11/29/2022] Open
Abstract
Protein turnover and homeostasis are regulated by the proteasomal system, which is critical for cell function and viability. Pluripotency of stem cells also relies on normal proteasomal activity that mitigates senescent phenotypes induced by intensive cell replications, as previously demonstrated in human bone marrow stromal cells. In this study, we investigated the role of proteasomes in self-renewal of neural progenitor cells (NPCs). Through both in vivo and in vitro analyses, we found that the expression of proteasomes was progressively decreased during aging. Likewise, proliferation and self-renewal of NPCs were also impaired in aged mice, suggesting that the down-regulation of proteasomes might be responsible for this senescent phenotype. Lowering proteasomal activity by loss-of-function manipulations mimicked the senescence of NPCs both in vitro and in vivo; conversely, enhancing proteasomal activity restored and improved self-renewal in aged NPCs. These results collectively indicate that proteasomes work as a key regulator in promoting self-renewal of NPCs. This potentially provides a promising therapeutic target for age-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Yunhe Zhao
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| | - Xueqin Liu
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| | - Zebin He
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaojie Niu
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| | - Weijun Shi
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| | - Jian M. Ding
- Department of Physiology, East Carolina University Medical School, Greenville, 27834, USA
| | - Li Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, 510632, China
| | - Tifei Yuan
- School of Psychology, Nanjing Normal University, Nanjing, 210097, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, 510632, China
| | - Wulin Yang
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Li Lu
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
20
|
Sarkar C, Zhao Z, Aungst S, Sabirzhanov B, Faden AI, Lipinski MM. Impaired autophagy flux is associated with neuronal cell death after traumatic brain injury. Autophagy 2015; 10:2208-22. [PMID: 25484084 PMCID: PMC4502690 DOI: 10.4161/15548627.2014.981787] [Citation(s) in RCA: 250] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of autophagy contributes to neuronal cell death in several neurodegenerative and lysosomal storage diseases. Markers of autophagy are also increased after traumatic brain injury (TBI), but its mechanisms and function are not known. Following controlled cortical impact (CCI) brain injury in GFP-Lc3 (green fluorescent protein-LC3) transgenic mice, we observed accumulation of autophagosomes in ipsilateral cortex and hippocampus between 1 and 7 d. This accumulation was not due to increased initiation of autophagy but rather to a decrease in clearance of autophagosomes, as reflected by accumulation of the autophagic substrate SQSTM1/p62 (sequestosome 1). This was confirmed by ex vivo studies, which demonstrated impaired autophagic flux in brain slices from injured as compared to control animals. Increased SQSTM1 peaked at d 1-3 but resolved by d 7, suggesting that the defect in autophagy flux is temporary. The early impairment of autophagy is at least in part caused by lysosomal dysfunction, as evidenced by lower protein levels and enzymatic activity of CTSD (cathepsin D). Furthermore, immediately after injury both autophagosomes and SQSTM1 accumulated predominantly in neurons. This was accompanied by appearance of SQSTM1 and ubiquitin-positive puncta in the affected cells, suggesting that, similar to the situation observed in neurodegenerative diseases, impaired autophagy may contribute to neuronal injury. Consistently, GFP-LC3 and SQSTM1 colocalized with markers of both caspase-dependent and caspase-independent cell death in neuronal cells proximal to the injury site. Taken together, our data indicated for the first time that autophagic clearance is impaired early after TBI due to lysosomal dysfunction, and correlates with neuronal cell death.
Collapse
Key Words
- ACTB, actin
- AIFM1, apoptosis-inducing factor, mitochondrion-associated, 1
- APC, adenomatous polyposis coli
- ATG12, autophagy-related 12
- ATG5, autophagy-related 5
- ATG7, autophagy-related 7
- CAPS12, caspase 12
- CASP3, caspase 3
- CCI, controlled cortical impact
- CD68, CD68 molecule
- CSPG4, chondroitin sulfate proteoglycan 4
- CTSD, cathepsin D
- GFP, green fluorescent protein
- LAMP1, lysosomal-associated membrane protein 1
- LAMP2, lysosomal-associated membrane protein 2
- LC3, microtubule associated protein 1 light chain 3
- RBFOX3, RNA binding protein, fox-1 homolog (C. elegans) 3
- SPTAN1, spectrin, α, non-erythrocytic 1
- SQSTM1, sequestosome 1
- TBI, traumatic brain injury
- ULK1, unc-51 like autophagy activating kinase 1
- autophagy
- autophagy flux
- lysosome
- neuronal cell death
- traumatic brain injury
- β; AIF1/IBA1, allograft inflammatory factor 1
Collapse
Affiliation(s)
- Chinmoy Sarkar
- a Shock, Trauma and Anesthesiology Research (STAR) Center; Department of Anesthesiology ; University of Maryland School of Medicine ; Baltimore , MD USA
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Since the original descriptions of postconcussive pathophysiology, there has been a significant increase in interest and ongoing research to study the biological underpinnings of concussion. The initial ionic flux and glutamate release result in significant energy demands and a period of metabolic crisis for the injured brain. These physiological perturbations can now be linked to clinical characteristics of concussion, including migrainous symptoms, vulnerability to repeat injury, and cognitive impairment. Furthermore, advanced neuroimaging now allows a research window to monitor postconcussion pathophysiology in humans noninvasively. There is also increasing concern about the risk for chronic or even progressive neurobehavioral impairment after concussion/mild traumatic brain injury. Critical studies are underway to better link the acute pathobiology of concussion with potential mechanisms of chronic cell death, dysfunction, and neurodegeneration. This "new and improved" article summarizes in a translational fashion and updates what is known about the acute neurometabolic changes after concussive brain injury. Furthermore, new connections are proposed between this neurobiology and early clinical symptoms as well as to cellular processes that may underlie long-term impairment.
Collapse
Affiliation(s)
- Christopher C Giza
- *Division of Pediatric Neurology, Department of Pediatrics, Mattel Children's Hospital-UCLA, Los Angeles, California; ‡Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California; §Interdepartmental Programs for Neuroscience and Biomedical Engineering, UCLA, Los Angeles, California; ¶Department of Medical and Molecular Pharmacology, UCLA, Los Angeles, California
| | | |
Collapse
|
22
|
Inhibition of immunoproteasome reduces infarction volume and attenuates inflammatory reaction in a rat model of ischemic stroke. Cell Death Dis 2015; 6:e1626. [PMID: 25633295 PMCID: PMC4669779 DOI: 10.1038/cddis.2014.586] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/19/2014] [Accepted: 12/15/2014] [Indexed: 11/26/2022]
Abstract
The detailed knowledge about the contribution of immunoproteasome to the neuroinflammation in ischemic stroke is still not available. The immunoreactivity of low molecular mass peptide 2 (LMP2) and low molecular mass peptide 7 (LMP7) was evident in the ipsilateral ischemic cerebral cortex and striatum following transient middle cerebral artery occlusion (MCAO). Both LMP2 and LMP7 increased as early as 4 h after the MCAO, further increased at 24 h, peaked at 72 h and decreased 7 days later. LMP2 and LMP7 were mainly present in astrocytes and microglia/macrophage cells, respectively. LMP2 knockdown by shRNA (short hairpin RNA) markedly reduced the levels of LMP2 and LMP7 protein and caused 75.5 and 78.6% decrease in the caspase-like (C-L) and chymotrypsin-like (CT-L) activities, respectively. Compared with cont-shRNA group (39.7%, infarction volumes/total ipsilateral hemisphere), the infarction volumes were reduced to 22.5% in LMP2-shRNA group. Additionally, LMP2 knockdown significantly reduced activated astrocytes and microglia, the expression nuclear factor kappa B (NF-κB) p65, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) and caused less accumulation of ischemia-induced protein ubiquitination compared with MG132. These findings demonstrate that inhibition of LMP2 significantly attenuates inflammatory reaction and offers neuroprotection against focal cerebral ischemia in rats, suggesting that selective immunoproteasome inhibitors may be a promising strategy for stroke treatment.
Collapse
|
23
|
Tylicka M, Matuszczak E, Dębek W, Hermanowicz A, Ostrowska H. Circulating proteasome activity following mild head injury in children. Childs Nerv Syst 2014; 30:1191-6. [PMID: 24700339 PMCID: PMC4072065 DOI: 10.1007/s00381-014-2409-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/24/2014] [Indexed: 01/04/2023]
Abstract
PURPOSE The aim of the study is to characterize changes in circulating proteasome (c-proteasome) activity following mild traumatic brain injury in children. METHODS Fifty children managed at the Department of Pediatric Surgery because of concussion-mild head injury was randomly included into the study. The children were aged 11 months to 17 years (median = 10.07 + -1.91 years). Plasma proteasome activity was assessed using Suc-Leu-Leu-Val-Tyr-AMC peptide substrate, 2-6 h, 12-16 h, and 2 days after injury. Twenty healthy children admitted for planned inguinal hernia repair served as controls. RESULTS Statistically significant elevation of plasma c-proteasome activity was noted in children with mild head injury 2-6 h, 12-16 h, and 2 days after the injury. CONCLUSIONS Authors observed a statistically significant upward trend in the c-proteasome activity between 2-6 and 12-16 h after the mild head injury, consistent with the onset of the symptoms of cerebral concussion and a downward trend in the c-proteasome activity in the plasma of children with mild head injury between 12-16 h and on the second day after the injury, consistent with the resolving of the symptoms of cerebral concussion. Further studies are needed to demonstrate that the proteasome activity could be a prognostic factor, which can help in further diagnostic and therapeutic decisions in patients with head injury.
Collapse
Affiliation(s)
- Marzena Tylicka
- Department of Biophysics, Medical University of Białystok, Mickiewicza 2A, 15-089 Białystok, Poland
| | - Ewa Matuszczak
- Department of Pediatric Surgery, Medical University of Białystok, 15-274 Białystok, Poland
| | - Wojciech Dębek
- Department of Pediatric Surgery, Medical University of Białystok, 15-274 Białystok, Poland
| | - Adam Hermanowicz
- Department of Pediatric Surgery, Medical University of Białystok, 15-274 Białystok, Poland
| | - Halina Ostrowska
- Department of Pediatric Surgery, Medical University of Białystok, 15-274 Białystok, Poland
| |
Collapse
|
24
|
Pla A, Pascual M, Renau-Piqueras J, Guerri C. TLR4 mediates the impairment of ubiquitin-proteasome and autophagy-lysosome pathways induced by ethanol treatment in brain. Cell Death Dis 2014; 5:e1066. [PMID: 24556681 PMCID: PMC3944260 DOI: 10.1038/cddis.2014.46] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 12/19/2013] [Accepted: 01/20/2014] [Indexed: 12/28/2022]
Abstract
New evidence indicates the involvement of protein degradation dysfunctions in neurodegeneration, innate immunity response and alcohol hepatotoxicity. We recently demonstrated that ethanol increases brain proinflammatory mediators and causes brain damage by activating Toll-like receptor 4 (TLR4) signaling in glia. However, it is uncertain if the ubiquitin-proteasome and autophagy-lysosome pathways are involved in ethanol-induced brain damage and whether the TLR4 response is implicated in proteolytic processes. Using the cerebral cortex of WT and TLR4-knockout mice with and without chronic ethanol treatment, we demonstrate that ethanol induces poly-ubiquitinated proteins accumulation and promotes immunoproteasome activation by inducing the expression of β2i, β5i and PA28α, although it decreases the 20S constitutive proteasome subunits (α2, β5). Ethanol also upregulates mTOR phosphorylation, leading to a downregulation of the autophagy-lysosome pathway (ATG12, ATG5, cathepsin B, p62, LC3) and alters the volume of autophagic vacuoles. Notably, mice lacking TLR4 receptors are protected against ethanol-induced alterations in protein degradation pathways. In summary, the present results provide the first evidence demonstrating that chronic ethanol treatment causes proteolysis dysfunctions in the mouse cerebral cortex and that these events are TLR4 dependent. These findings could provide insight into the mechanisms underlying ethanol-induced brain damage.
Collapse
Affiliation(s)
- A Pla
- Department of Cellular Pathology, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - M Pascual
- Department of Cellular Pathology, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - J Renau-Piqueras
- Section of Biology and Cellular Pathology, Centro Investigación Hospital La Fe, Avda. de Campanar 114, Valencia 46015, Spain
| | - C Guerri
- Department of Cellular Pathology, Centro de Investigación Príncipe Felipe, C/Eduardo Primo Yúfera 3, Valencia 46012, Spain
| |
Collapse
|
25
|
Ferrington DA, Gregerson DS. Immunoproteasomes: structure, function, and antigen presentation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 109:75-112. [PMID: 22727420 DOI: 10.1016/b978-0-12-397863-9.00003-1] [Citation(s) in RCA: 291] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Immunoproteasomes contain replacements for the three catalytic subunits of standard proteasomes. In most cells, oxidative stress and proinflammatory cytokines are stimuli that lead to elevated production of immunoproteasomes. Immune system cells, especially antigen-presenting cells, express a higher basal level of immunoproteasomes. A well-described function of immunoproteasomes is to generate peptides with a hydrophobic C terminus that can be processed to fit in the groove of MHC class I molecules. This display of peptides on the cell surface allows surveillance by CD8 T cells of the adaptive immune system for pathogen-infected cells. Functions of immunoproteasomes, other than generating peptides for antigen presentation, are emerging from studies in immunoproteasome-deficient mice, and are complemented by recently described diseases linked to mutations or single-nucleotide polymorphisms in immunoproteasome subunits. Thus, this growing body of literature suggests a more pleiotropic role in cell function for the immunoproteasome.
Collapse
Affiliation(s)
- Deborah A Ferrington
- Department of Ophthalmology, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
26
|
Ferrington DA, Roehrich H, Chang AA, Huang CW, Maldonado M, Bratten W, Rageh AA, Heuss ND, Gregerson DS, Nelson EF, Yuan C. Corneal wound healing is compromised by immunoproteasome deficiency. PLoS One 2013; 8:e54347. [PMID: 23365662 PMCID: PMC3554767 DOI: 10.1371/journal.pone.0054347] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 12/12/2012] [Indexed: 01/05/2023] Open
Abstract
Recent studies have revealed roles for immunoproteasome in regulating cell processes essential for maintaining homeostasis and in responding to stress and injury. The current study investigates how the absence of immunoproteasome affects the corneal epithelium under normal and stressed conditions by comparing corneas from wildtype (WT) mice and those deficient in two immunoproteasome catalytic subunits (lmp7(-/-)/mecl-1(-/-), L7M1). Immunoproteasome expression was confirmed in WT epithelial cells and in cells of the immune system that were present in the cornea. More apoptotic cells were found in both corneal explant cultures and uninjured corneas of L7M1 compared to WT mice. Following mechanical debridement, L7M1 corneas displayed delayed wound healing, including delayed re-epithelialization and re-establishment of the epithelial barrier, as well as altered inflammatory cytokine production compared to WT mice. These results suggest that immunoproteasome plays an important role in corneal homeostasis and wound healing.
Collapse
Affiliation(s)
- Deborah A. Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Angela A. Chang
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Craig W. Huang
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Marcela Maldonado
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wendy Bratten
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Abrar A. Rageh
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Neal D. Heuss
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Dale S. Gregerson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Elizabeth F. Nelson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ching Yuan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
27
|
Goldman SM, Kamel F, Ross GW, Jewell SA, Bhudhikanok GS, Umbach D, Marras C, Hauser RA, Jankovic J, Factor SA, Bressman S, Lyons KE, Meng C, Korell M, Roucoux DF, Hoppin JA, Sandler DP, Langston JW, Tanner CM. Head injury, α-synuclein Rep1, and Parkinson's disease. Ann Neurol 2012; 71:40-8. [PMID: 22275250 DOI: 10.1002/ana.22499] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To test the hypothesis that variability in SNCA Rep1, a polymorphic dinucleotide microsatellite in the promoter region of the gene encoding α-synuclein, modifies the association between head injury and Parkinson's disease (PD) risk. METHODS Participants in the Farming and Movement Evaluation (FAME) and the Study of Environmental Association and Risk of Parkinsonism using Case-Control Historical Interviews (SEARCH), 2 independent case-control studies, were genotyped for Rep1 and interviewed regarding head injuries with loss of consciousness or concussion prior to Parkinson's disease (PD) diagnosis. Logistic regression modeling adjusted for potential confounding variables and tested interaction between Rep1 genotype and head injury. RESULTS Consistent with prior reports, relative to medium-length Rep1, short Rep1 genotype was associated with reduced PD risk (pooled odds ratio [OR], 0.7; 95% confidence interval [CI], 0.5-0.9), and long Rep1 with increased risk (pooled OR, 1.4; 95% CI, 0.95-2.2). Overall, head injury was not significantly associated with PD (pooled OR, 1.3; 95% CI, 0.9-1.8). However, head injury was strongly associated with PD in those with long Rep1 (FAME OR, 5.4; 95% CI, 1.5-19; SEARCH OR, 2.3; 95% CI, 0.6-9.2; pooled OR, 3.5; 95% CI 1.4-9.2, p-interaction = 0.02). Individuals with both head injury and long Rep1 were diagnosed 4.9 years earlier than those with neither risk factor (p = 0.03). INTERPRETATION While head injury alone was not associated with PD risk, our data suggest head injury may initiate and/or accelerate neurodegeneration when levels of synuclein are high, as in those with Rep1 expansion. Given the high population frequency of head injury, independent verification of these results is essential.
Collapse
|
28
|
Cook C, Gass J, Dunmore J, Tong J, Taylor J, Eriksen J, McGowan E, Lewis J, Johnston J, Petrucelli L. Aging is not associated with proteasome impairment in UPS reporter mice. PLoS One 2009; 4:e5888. [PMID: 19517018 PMCID: PMC2690827 DOI: 10.1371/journal.pone.0005888] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 05/18/2009] [Indexed: 01/20/2023] Open
Abstract
Background Covalent linkage of ubiquitin regulates the function and, ultimately, the degradation of many proteins by the ubiquitin-proteasome system (UPS). Given its essential role in protein regulation, even slight perturbations in UPS activity can substantially impair cellular function. Methodology/Principal Findings We have generated and characterized a novel transgenic mouse model which expresses a previously described reporter for UPS function. This UPS reporter contains a degron sequence attached to the C-terminus of green fluorescent protein, and is predominantly expressed in neurons throughout the brain of our transgenic model. We then demonstrated that this reporter system is sensitive to UPS inhibition in vivo. Conclusions/Significance Given the obstacles associated with evaluating proteasomal function in the brain, our mouse model uniquely provides the capability to monitor UPS function in real time in individual neurons of a complex organism. Our novel mouse model now provides a useful resource with which to evaluate the impact of aging, as well as various genetic and/or pharmacological modifiers of neurodegenerative disease(s).
Collapse
|
29
|
Szabo Z, Ying Z, Radak Z, Gomez-Pinilla F. Voluntary exercise may engage proteasome function to benefit the brain after trauma. Brain Res 2009; 1341:25-31. [PMID: 19368831 DOI: 10.1016/j.brainres.2009.01.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 01/17/2009] [Accepted: 01/21/2009] [Indexed: 12/29/2022]
Abstract
Brain trauma is associated with long-term decrements in synaptic plasticity and cognitive function, which likely reside on the acute effects of the injury on protein structure and function. Based on the action of proteasome on protein synthesis and degradation we have examined the effects of brain injury on proteasome level/activity and the potential of exercise to interact with the effects of the injury. Exercise has a healing ability but its action on proteasome function is not understood. Male Sprague-Dawley adult rats (n=19) were performed mild brain fluid percussion injury (FPI) prior to exercise. Animals were assigned to four groups: sedentary (Sed) or exercise (Exc) with sham surgery (Sham) or FPI: Sham/Sed, Sham/Exc, FPI/Sed, FPI/Exc. Animals were sacrificed after 14 days of treatment. FPI elevated levels of carbonyl (160.1+/-9.6% SEM, p<0.01) and reduced synapsin I levels (58.3+/-4.3% SEM, p<0.01) in the ipsilateral side of caudal cerebral cortex (FPI/Sed compared to Sham/Sed controls), and it appears that increased levels of carbonyls were associated with increased chymotripsin like activity. These results seem to indicate that proteasome function may be associated with levels of oxidative stress, and that these events may contribute to the action of exercise on synaptic plasticity. Interestingly, exercise attenuated changes in carbonyls, proteasome activity, and synapsin I following FPI, which may indicate an action of exercise on the molecular substrates that control protein turnover following brain trauma. Levels of the regulatory transcription factor of proteasome, Zif 268 were reduced by exercise in Sham and FPI animals and changed in proportion with proteasome activity/content. The overall results indicate that the action of exercise interfaces with that of brain injury on molecular systems involved with protein fate and function, which may be significant for synaptic plasticity.
Collapse
Affiliation(s)
- Zsofia Szabo
- Department of Physiological Science, Brain Injury Research Center, UCLA, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
30
|
Ferrington DA, Hussong SA, Roehrich H, Kapphahn RJ, Kavanaugh SM, Heuss ND, Gregerson DS. Immunoproteasome responds to injury in the retina and brain. J Neurochem 2008; 106:158-69. [PMID: 18346202 PMCID: PMC4401486 DOI: 10.1111/j.1471-4159.2008.05345.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is well known that immunoproteasome generates peptides for MHC Class I occupancy and recognition by cytotoxic T lymphocytes (CTL). The present study focused on evidence for alternative roles for immunoproteasome. Retina and brain were analyzed for expression of immunoproteasome subunits using immunohistochemistry and western blotting under normal conditions and after injury/stress induced by CTL attack on glia (brain) or neurons (retina). Normal retina expressed substantial levels of immunoproteasome in glia, neurons, and retinal pigment epithelium. The basal level of immunoproteasome in retina was two-fold higher than in brain; CTL-induced retinal injury further up-regulated immunoproteasome expression. Immunoproteasome up-regulation was also observed in injured brain and corresponded with expression in Purkinje cells, microglia, astrocytes, and oligodendrocytes. These results suggest that the normal environment of the retina is sufficiently challenging to require on-going expression of immunoproteasome. Further, immunoproteasome up-regulation with retinal and brain injury implies a role in neuronal protection and/or repair of damage.
Collapse
Affiliation(s)
- Deborah A Ferrington
- Department of Ophthalmology, University of Minnesota, Minneapolis, Minnesota, USA.
| | | | | | | | | | | | | |
Collapse
|