1
|
Shi Z, Mao L, Chen S, Du Z, Xiang J, Shi M, Wang Y, Wang Y, Chen X, Xu Z, Gao Y. Reversing Persistent PTEN Activation after Traumatic Brain Injury Fuels Long-Term Axonal Regeneration via Akt/mTORC1 Signaling Cascade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410136. [PMID: 39680734 PMCID: PMC11809353 DOI: 10.1002/advs.202410136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/24/2024] [Indexed: 12/18/2024]
Abstract
Traumatic brain injury (TBI) often leads to enduring axonal damage and persistent neurological deficits. While PTEN's role in neuronal growth is recognized, its long-term activation changes post-TBI and its effects on sensory-motor circuits are not well understood. Here, it is demonstrated that the neuronal knockout of PTEN (PTEN-nKO) significantly enhances both structural and functional recovery over the long term after TBI. Importantly, in vivo, DTI-MRI revealed that PTEN-nKO promotes white matter repair post-TBI. Additionally, calcium imaging and electromyographic recordings indicated that PTEN-nKO facilitates cortical remapping and restores sensory-motor pathways. Mechanistically, PTEN negatively regulates the Akt/mTOR pathway by inhibiting Akt, thereby suppressing mTOR. Raptor is a key component of mTORC1 and its suppression impedes axonal regeneration. The restoration of white matter integrity and the improvements in neural function observed in PTEN-nKO TBI-treated mice are reversed by a PTEN/Raptor double knockout (PTEN/Raptor D-nKO), suggesting that mTORC1 acts as a key mediator. These findings highlight persistent alterations in the PTEN/Akt/mTORC1 axis are critical for neural circuit remodeling and cortical remapping post-TBI, offering new insights into TBI pathophysiology and potential therapeutic targets.
Collapse
Affiliation(s)
- Ziyu Shi
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Leilei Mao
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Shuning Chen
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Zhuoying Du
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Jiakun Xiang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Minghong Shi
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yana Wang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yuqing Wang
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Xingdong Chen
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Zhi‐Xiang Xu
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yanqin Gao
- State Key Laboratory of Medical NeurobiologyMOE Frontiers Center for Brain Scienceand Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
2
|
Rada CC, Yuki K, Ding J, Kuo CJ. Regulation of the Blood-Brain Barrier in Health and Disease. Cold Spring Harb Perspect Med 2023; 13:a041191. [PMID: 36987582 PMCID: PMC10691497 DOI: 10.1101/cshperspect.a041191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The neurovascular unit is a dynamic microenvironment with tightly controlled signaling and transport coordinated by the blood-brain barrier (BBB). A properly functioning BBB allows sufficient movement of ions and macromolecules to meet the high metabolic demand of the central nervous system (CNS), while protecting the brain from pathogenic and noxious insults. This review describes the main cell types comprising the BBB and unique molecular signatures of these cells. Additionally, major signaling pathways for BBB development and maintenance are highlighted. Finally, we describe the pathophysiology of BBB diseases, their relationship to barrier dysfunction, and identify avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Cara C Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
3
|
Moreira NCDS, Tamarozzi ER, Lima JEBDF, Piassi LDO, Carvalho I, Passos GA, Sakamoto-Hojo ET. Novel Dual AChE and ROCK2 Inhibitor Induces Neurogenesis via PTEN/AKT Pathway in Alzheimer's Disease Model. Int J Mol Sci 2022; 23:ijms232314788. [PMID: 36499116 PMCID: PMC9737254 DOI: 10.3390/ijms232314788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and complex neurodegenerative disease. Acetylcholinesterase inhibitors (AChEIs) are a major class of drugs used in AD therapy. ROCK2, another promising target for AD, has been associated with the induction of neurogenesis via PTEN/AKT. This study aimed to characterize the therapeutic potential of a novel donepezil-tacrine hybrid compound (TA8Amino) to inhibit AChE and ROCK2 protein, leading to the induction of neurogenesis in SH-SY5Y cells. Experiments were carried out with undifferentiated and neuron-differentiated SH-SY5Y cells submitted to treatments with AChEIs (TA8Amino, donepezil, and tacrine) for 24 h or 7 days. TA8Amino was capable of inhibiting AChE at non-cytotoxic concentrations after 24 h. Following neuronal differentiation for 7 days, TA8Amino and donepezil increased the percentage of neurodifferentiated cells and the length of neurites, as confirmed by β-III-tubulin and MAP2 protein expression. TA8Amino was found to participate in the activation of PTEN/AKT signaling. In silico analysis showed that TA8Amino can stably bind to the active site of ROCK2, and in vitro experiments in SH-SY5Y cells demonstrate that TA8Amino significantly reduced the expression of ROCK2 protein, contrasting with donepezil and tacrine. Therefore, these results provide important information on the mechanism underlying the action of TA8Amino with regard to multi-target activities.
Collapse
Affiliation(s)
| | - Elvira Regina Tamarozzi
- Department of Biotechnology, School of Arts, Sciences and Humanities—USP, São Paulo 03828-000, Brazil
| | | | - Larissa de Oliveira Piassi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-900, Brazil
| | - Geraldo Aleixo Passos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-901, Brazil
- Correspondence: ; Tel.: +55-16-3315-3827
| |
Collapse
|
4
|
Erickson MA, Banks WA. Transcellular routes of blood-brain barrier disruption. Exp Biol Med (Maywood) 2022; 247:788-796. [PMID: 35243912 DOI: 10.1177/15353702221080745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Disruption of the blood-brain barrier (BBB) can occur through different mechanisms and pathways. As these pathways result in increased permeability to different classes of substances, it is likely that the neurological insults that occur will also differ for these pathways. The major categories of BBB disruption are paracellular (between cells) and transcellular (across cells) with a subcategory of transcellular leakage involving vesicles (transcytotic). Older literature, as well as more recent studies, highlights the importance of the transcellular pathways in BBB disruption. Of the various transcytotic mechanisms that are thought to be active at the BBB, some are linked to receptor-mediated transcytosis, whereas others are likely involved in BBB disruption. For most capillary beds, transcytotic mechanisms are less clearly linked to permeability than are membrane spanning canaliculi and fenestrations. Disruption pathways share cellular mechanisms to some degree as exemplified by transcytotic caveolar and transcellular canaliculi formations. The discovery of some of the cellular components involved in transcellular mechanisms of BBB disruption and the ability to measure them are adding greatly to our classic knowledge, which is largely based on ultrastructural studies. Future work will likely address the conditions and diseases under which the various pathways of disruption are active, the different impacts that they have, and the cellular biology that underlies the different pathways to disruption.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| |
Collapse
|
5
|
Li Y, Fan C, Wang L, Lan T, Gao R, Wang W, Yu SY. MicroRNA-26a-3p rescues depression-like behaviors in male rats via preventing hippocampal neuronal anomalies. J Clin Invest 2021; 131:e148853. [PMID: 34228643 DOI: 10.1172/jci148853] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Depression is a neuropsychiatric disease associated with neuronal anomalies within specific brain regions. In the present study, we screened microRNA (miRNA) expression profiles in the dentate gyrus (DG) of the hippocampus and found that miR-26a-3p was markedly downregulated in a rat model of depression, whereas upregulation of miR-26a-3p within DG regions rescued the neuronal deterioration and depression-like phenotypes resulting from stress exposure, effects that appear to be mediated by the PTEN pathway. The knockdown of miR-26a-3p in DG regions of normal control rats induced depression-like behaviors, effects that were accompanied by activation of the PTEN/PI3K/Akt signaling pathway and neuronal deterioration via suppression of autophagy, impairments in synaptic plasticity, and promotion of neuronal apoptosis. In conclusion, these results suggest that miR-26a-3p deficits within the hippocampal DG mediated the neuronal anomalies contributing to the display of depression-like behaviors. This miRNA may serve as a potential therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Ye Li
- Department of Physiology and
| | | | - Liyan Wang
- Morphological Experimental Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | | | - Rui Gao
- Department of Microorganism, Jinan Nursing Vocational College, Lvyoulu Road, Jinan, Shandong Province, China
| | | | - Shu Yan Yu
- Department of Physiology and.,Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
6
|
Sargolzaei S, Kaushik A, Soltani S, Amini MH, Khalghani MR, Khoshavi N, Sargolzaei A. Preclinical Western Blot in the Era of Digital Transformation and Reproducible Research, an Eastern Perspective. Interdiscip Sci 2021; 13:490-499. [PMID: 34080131 DOI: 10.1007/s12539-021-00442-7] [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: 07/07/2020] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/25/2022]
Abstract
The current research is an interdisciplinary endeavor to develop a necessary tool in preclinical protein studies of diseases or disorders through western blotting. In the era of digital transformation and open access principles, an interactive cloud-based database called East-West Blot ( https://rancs-lab.shinyapps.io/WesternBlots ) is designed and developed. The online interactive subject-specific database built on the R shiny platform facilitates a systematic literature search on the specific subject matter, here set to western blot studies of protein regulation in the preclinical model of TBI. The tool summarizes the existing publicly available knowledge through a data visualization technique and easy access to the critical data elements and links to the study itself. The application compiled a relational database of PubMed-indexed western blot studies labeled under HHS public access, reporting downstream protein regulations presented by fluid percussion injury model of traumatic brain injury. The promises of the developed tool include progressing toward implementing the principles of 3Rs (replacement, reduction, and refinement) for humane experiments, cultivating the prerequisites of reproducible research in terms of reporting characteristics, paving the ways for a more collaborative experimental design in basic science, and rendering an up-to-date and summarized perspective of current publicly available knowledge.
Collapse
Affiliation(s)
- Saman Sargolzaei
- Department of Engineering, University of Tennessee at Martin, Martin, TN, USA.
| | - Ajeet Kaushik
- Department of Natural Sciences, Florida Polytechnic University, Lakeland, FL, USA
| | - Seyed Soltani
- Mechanical Engineering Department, Florida Polytechnic University, Lakeland, FL, USA
| | - M Hadi Amini
- School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Mohammad Reza Khalghani
- Electrical and Computer Engineering Department, Florida Polytechnic University, Lakeland, FL, USA
| | - Navid Khoshavi
- Computer Science Department, Florida Polytechnic University, Lakeland, FL, USA
| | - Arman Sargolzaei
- Department of Mechanical Engineering, Tennessee Technological University, Cookeville, TN, USA
| |
Collapse
|
7
|
Moreira NCDS, Lima JEBDF, Chierrito TPC, Carvalho I, Sakamoto-Hojo ET. Novel Hybrid Acetylcholinesterase Inhibitors Induce Differentiation and Neuritogenesis in Neuronal Cells in vitro Through Activation of the AKT Pathway. J Alzheimers Dis 2020; 78:353-370. [PMID: 32986667 DOI: 10.3233/jad-200425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a progressive loss of episodic memory associated with amyloid-β peptide aggregation and the abnormal phosphorylation of the tau protein, leading to the loss of cholinergic function. Acetylcholinesterase (AChE) inhibitors are the main class of drugs used in AD therapy. OBJECTIVE The aim of the current study was to evaluate the potential of two tacrine-donepezil hybrid molecules (TA8Amino and TAHB3), which are AChE inhibitors, to induce neurodifferentiation and neuritogenesis in SH-SY5Y cells. METHODS The experiments were carried out to characterize neurodifferentiation, cellular changes related to responses to oxidative stress and pathways of cell survival in response to drug treatments. RESULTS The results indicated that the compounds did not present cytotoxic effects in SH-SY5Y or HepG2 cells. TA8Amino and TAHB3 induced neurodifferentiation and neuritogenesis in SH-SY5Y cells. These cells showed increased levels of intracellular and mitochondrial reactive oxygen species; the induction of oxidative stress was also demonstrated by an increase in SOD1 expression in TA8Amino and TAHB3-treated cells. Cells treated with the compounds showed an increase in PTEN(Ser380/Thr382/383) and AKT(Ser473) expression, suggesting the involvement of the AKT pathway. CONCLUSION Our results demonstrated that TA8Amino and TAHB3 present advantages as potential drugs for AD therapy and that they are capable of inducing neurodifferentiation and neuritogenesis.
Collapse
Affiliation(s)
| | | | | | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
8
|
Chen H, Jing Y, Xu Z, Yang D, Ju S, Guo Y, Tian H, Xue L. Upregulation of C Terminus of Hsc70-Interacting Protein Attenuates Apoptosis and Procoagulant Activity and Facilitates Brain Repair After Traumatic Brain Injury. Front Neurosci 2020; 14:925. [PMID: 33013306 PMCID: PMC7506102 DOI: 10.3389/fnins.2020.00925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) could highly induce coagulopathy through breaking the dynamic balance between coagulation and fibrinolysis systems, which may be a major contributor to the progressive secondary injury cascade that occurs after TBI. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibition is reported to exert neuroprotection in TBI, making it a potential regulatory target involved in TBI-induced coagulation disorder. PTEN level is controlled in a major way by E3 ligase-mediated degradation through the ubiquitin-proteasome system. The C terminus of Hsc70-interacting protein (CHIP) has been shown to regulate proteasomal degradation and ubiquitination level of PTEN. In the present study, CHIP was overexpressed and knocked down in mouse brain microvascular endothelial cells (bEnd.3) and tissues during the early phase of TBI. In vitro cell proliferation, cell apoptosis, migration capacity, and invasion capacity were determined. The changes of procoagulant and apoptosis molecules after TBI were also detected as well as the micrangium density and blood-brain barrier permeability after in vivo TBI. In vitro results demonstrated that CHIP overexpression facilitated bEnd.3 cell proliferation, migration, and invasion and downregulated cell apoptosis and the expressions of procoagulant molecules through promoting PTEN ubiquitination in a simulated TBI model with stretch-induced injury treatment. In vivo experiments also demonstrated that CHIP overexpression suppressed post-TBI apoptosis and procoagulant protein expressions, as well as increased microvessel density, reduced hemorrhagic injury, and blood-brain barrier permeability. These findings suggested that the upregulation of CHIP may attenuate apoptosis and procoagulant activity, facilitate brain repair, and thus exerts neuroprotective effects in TBI.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yao Jing
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Zhiming Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Dianxu Yang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shiming Ju
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yan Guo
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hengli Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Lixia Xue
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
9
|
Li Z, Xu R, Zhu X, Li Y, Wang Y, Xu W. MicroRNA-23a-3p improves traumatic brain injury through modulating the neurological apoptosis and inflammation response in mice. Cell Cycle 2019; 19:24-38. [PMID: 31818176 DOI: 10.1080/15384101.2019.1691763] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Secondary brain damage plays an important role in Traumatic brain injury (TBI) and inhibition of this damage has benefit for TBI treatment. However, the pathogenesis of secondary brain damage remains largely unknown. Here, we tried to explore the influence of microRNAs (miRNAs) on neuron apoptosis and inflammatory response after TBI. Firstly, the miRNA expression profiles were analyzed in the cerebral cortex tissues from the TBI mice model (controlled cortical impact) using miRNA microarray. miR-23a-3p (miR-23a) attracted our attention as its suppressive effects on apoptosis and inflammation. The further results showed that miR-23a upregulation improved long-term neurological function, the neuron apoptosis, and inhibited neuroinflammation, whereas knockdown of miR-23a had an opposite result. Using etoposide-induced primary cortical neurons injury model, we found that miR-23a was decreased in this cell model and miR-23a overexpression-suppressed etoposide induced the activity of caspase 3 and the releases of inflammatory mediators in primary cortical neurons. Phosphatase and tensin homolog (PTEN), a well‑known regulator of the AKT/mTOR pathway, was found to be a direct target of miR‑23a in the primary cortical neurons. Most importantly, it was found that miR-23a overexpression reactivated the AKT/mTOR pathway in TBI mice model, as demonstrated by the upregulation of phosphorylated (p‑)AKT and p‑mTOR. Taken together, these data indicate that miR-23a may serve as a therapeutic target for the treatment of TBI.
Collapse
Affiliation(s)
- Zhikun Li
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ruijun Xu
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaodong Zhu
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yifan Li
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yi Wang
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Orthopedic Surgery, TongRen Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Walker CL, Wu X, Liu NK, Xu XM. Bisperoxovanadium Mediates Neuronal Protection through Inhibition of PTEN and Activation of PI3K/AKT-mTOR Signaling after Traumatic Spinal Injuries. J Neurotrauma 2019; 36:2676-2687. [PMID: 30672370 DOI: 10.1089/neu.2018.6294] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although mechanisms involved in progression of cell death in spinal cord injury (SCI) have been studied extensively, few are clear targets for translation to clinical application. One of the best-understood mechanisms of cell survival in SCI is phosphatidylinositol-3-kinase (PI3K)/Akt and associated downstream signaling. Clear therapeutic efficacy of a phosphatase and tensin homologue (PTEN) inhibitor called bisperoxovanadium (bpV) has been shown in SCI, traumatic brain injury, stroke, and other neurological disease models in both neuroprotection and functional recovery. The present study aimed to elucidate mechanistic influences of bpV activity in neuronal survival in in vitro and in vivo models of SCI. Treatment with 100 nM bpV(pic) reduced cell death in a primary spinal neuron injury model (p < 0.05) in vitro, and upregulated both Akt and ribosomal protein S6 (pS6) activity (p < 0.05) compared with non-treated injured neurons. Pre-treatment of spinal neurons with a PI3K inhibitor, LY294002 or mammalian target of rapamycin (mTOR) inhibitor, rapamycin blocked bpV activation of Akt and ribosomal protein S6 activity, respectively. Treatment with bpV increased extracellular signal-related kinase (Erk) activity after scratch injury in vitro, and rapamycin reduced influence by bpV on Erk phosphorylation. After a cervical hemicontusive SCI, Akt phosphorylation decreased in total tissue via Western blot analysis (p < 0.01) as well as in penumbral ventral horn motor neurons throughout the first week post-injury (p < 0.05). Conversely, PTEN activity appeared to increase over this period. As observed in vitro, bpV also increased Erk activity post-SCI (p < 0.05). Our results suggest that PI3K/Akt signaling is the likely primary mechanism of bpV action in mediating neuroprotection in injured spinal neurons.
Collapse
Affiliation(s)
- Chandler L Walker
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana.,Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
11
|
Cell death after traumatic brain injury: Detrimental role of anoikis in healing. Clin Chim Acta 2018; 482:149-154. [DOI: 10.1016/j.cca.2018.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/19/2022]
|
12
|
Meconi A, Wortman RC, Wright DK, Neale KJ, Clarkson M, Shultz SR, Christie BR. Repeated mild traumatic brain injury can cause acute neurologic impairment without overt structural damage in juvenile rats. PLoS One 2018; 13:e0197187. [PMID: 29738554 PMCID: PMC5940222 DOI: 10.1371/journal.pone.0197187] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/27/2018] [Indexed: 11/19/2022] Open
Abstract
Repeated concussion is becoming increasingly recognized as a serious public health concern around the world. Moreover, there is a greater awareness amongst health professionals of the potential for repeated pediatric concussions to detrimentally alter the structure and function of the developing brain. To better study this issue, we developed an awake closed head injury (ACHI) model that enabled repeated concussions to be performed reliably and reproducibly in juvenile rats. A neurological assessment protocol (NAP) score was generated immediately after each ACHI to help quantify the cumulative effects of repeated injury on level of consciousness, and basic motor and reflexive capacity. Here we show that we can produce a repeated ACHI (4 impacts in two days) in both male and female juvenile rats without significant mortality or pain. We show that both single and repeated injuries produce acute neurological deficits resembling clinical concussion symptoms that can be quantified using the NAP score. Behavioural analyses indicate repeated ACHI acutely impaired spatial memory in the Barnes maze, and an interesting sex effect was revealed as memory impairment correlated moderately with poorer NAP score performance in a subset of females. These cognitive impairments occurred in the absence of motor impairments on the Rotarod, or emotional changes in the open field and elevated plus mazes. Cresyl violet histology and structural magnetic resonance imaging (MRI) indicated that repeated ACHI did not produce significant structural damage. MRI also confirmed there was no volumetric loss in the cortex, hippocampus, or corpus callosum of animals at 1 or 7 days post-ACHI. Together these data indicate that the ACHI model can provide a reliable, high throughput means to study the effects of concussions in juvenile rats.
Collapse
Affiliation(s)
- Alicia Meconi
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Ryan C. Wortman
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - David K. Wright
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Katie J. Neale
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Melissa Clarkson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Sandy R. Shultz
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Centre for Brain Health and Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
13
|
Zhao D, Chen J, Zhang Y, Liao HB, Zhang ZF, Zhuang Y, Pan MX, Tang JC, Liu R, Lei Y, Wang S, Qin XP, Feng YG, Chen Y, Wan Q. Glycine confers neuroprotection through PTEN/AKT signal pathway in experimental intracerebral hemorrhage. Biochem Biophys Res Commun 2018; 501:85-91. [PMID: 29698679 DOI: 10.1016/j.bbrc.2018.04.171] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/21/2018] [Indexed: 01/12/2023]
Abstract
Glycine has been shown to protect against ischemic stroke through various mechanisms. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) which antagonize Akt-dependent cell survival has been linked to neuronal damage. However, whether glycine has a neuroprotective property in intracerebral hemorrhage (ICH) was unknown. This study aimed to determine the protective effect of glycine in rats ICH. Adult male Sprague-Dawley (SD) rats were subjected to left striatum infusion of autologous blood. ICH animals received glycine (0.2-3 mg/kg, icv) at 1 h after ICH with or without pre-injection of Akt Inhibitor IV (100 μM, 2 μl, icv) 0.5 h prior to glycine treatment. Our results showed that in the perihematomal area PTEN was up-regulated in the early stage after ICH. However, glycine treatment decreased PTEN protein level and increased the phosphorylation level of AKT (p-AKT) in the perihematomal area. With the administration of glycine, neuronal death was significantly reduced and Evans blue leakage was alleviated as well as the brain edema after ICH. Moreover, hematoma volume was decreased and neurobehavioral outcome was improved. Nevertheless, Akt Inhibitor IV abolished the neuroprotective effects of glycine after ICH. Together, our findings demonstrate, for the first time, the protective role of glycine on ICH rats, and suggest that the neuroprotective effect of glycine was mediated through PTEN/Akt signal pathway.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China; Department of Biomedical Engineering, School of Basic Medical Sciences, WuhanUniversity, Wuhan 430071, China; Department of Physiology, School of Basic Medical Sciences, Hubei University of Medicine, 30 South Renmin Road, Shiyan, Hubei, 442000 China
| | - Juan Chen
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China; Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science & Technology, 26 Shengli Street, Wuhan, 430013, China
| | - Ya Zhang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Hua-Bao Liao
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Zhi-Feng Zhang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China; Department of Physiology, School of Basic Medical Sciences, Hubei University of Medicine, 30 South Renmin Road, Shiyan, Hubei, 442000 China
| | - Yang Zhuang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Meng-Xian Pan
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Jun-Chun Tang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Rui Liu
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Yang Lei
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Shu Wang
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Xing-Ping Qin
- Department of Physiology, Collaborative Innovation Center for Brain Science, School of Basic Medical Sciences, Wuhan University School of Medicine, 185 Donghu Street, Wuhan 430071, China
| | - Yu-Gong Feng
- Institute of Neuroregeneration& Neurorehabilitation, Department of Neurosurgery of the Affiliated Hospital, Qingdao University, 308 Ningxia Street, Qingdao, 266071, China
| | - Yun Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, WuhanUniversity, Wuhan 430071, China.
| | - Qi Wan
- Institute of Neuroregeneration& Neurorehabilitation, Department of Neurosurgery of the Affiliated Hospital, Qingdao University, 308 Ningxia Street, Qingdao, 266071, China.
| |
Collapse
|
14
|
Pulido R. PTEN Inhibition in Human Disease Therapy. Molecules 2018; 23:molecules23020285. [PMID: 29385737 PMCID: PMC6017825 DOI: 10.3390/molecules23020285] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor suppressor PTEN is a major homeostatic regulator, by virtue of its lipid phosphatase activity against phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], which downregulates the PI3K/AKT/mTOR prosurvival signaling, as well as by its protein phosphatase activity towards specific protein targets. PTEN catalytic activity is crucial to control cell growth under physiologic and pathologic situations, and it impacts not only in preventing tumor cell survival and proliferation, but also in restraining several cellular regeneration processes, such as those associated with nerve injury recovery, cardiac ischemia, or wound healing. In these conditions, inhibition of PTEN catalysis is being explored as a potentially beneficial therapeutic intervention. Here, an overview of human diseases and conditions in which PTEN inhibition could be beneficial is presented, together with an update on the current status of specific small molecule inhibitors of PTEN enzymatic activity, their use in experimental models, and their limitations as research or therapeutic drugs.
Collapse
Affiliation(s)
- Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
15
|
Najem D, Rennie K, Ribecco-Lutkiewicz M, Ly D, Haukenfrers J, Liu Q, Nzau M, Fraser DD, Bani-Yaghoub M. Traumatic brain injury: classification, models, and markers. Biochem Cell Biol 2018; 96:391-406. [PMID: 29370536 DOI: 10.1139/bcb-2016-0160] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide. Due to its high incidence rate and often long-term sequelae, TBI contributes significantly to increasing costs of health care expenditures annually. Unfortunately, advances in the field have been stifled by patient and injury heterogeneity that pose a major challenge in TBI prevention, diagnosis, and treatment. In this review, we briefly discuss the causes of TBI, followed by its prevalence, classification, and pathophysiology. The current imaging detection methods and animal models used to study brain injury are examined. We discuss the potential use of molecular markers in detecting and monitoring the progression of TBI, with particular emphasis on microRNAs as a novel class of molecular modulators of injury and its repair in the neural tissue.
Collapse
Affiliation(s)
- Dema Najem
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Kerry Rennie
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Maria Ribecco-Lutkiewicz
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Dao Ly
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Julie Haukenfrers
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
| | - Qing Liu
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada.,b Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Munyao Nzau
- c Paediatric Neurosurgery, Children's Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Douglas D Fraser
- d Children's Health Research Institute, London, ON N6C 2V5, Canada.,e Departments of Pediatrics and Clinical Neurological Sciences, Western University, London, ON N6A 3K7, Canada
| | - Mahmud Bani-Yaghoub
- a Department of Translational Bioscience, National Research Council Canada, Ottawa, ON K1A 0R6, Canada.,f Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
16
|
Liu X, Huang S, Liu C, Liu X, Shen Y, Cui Z. PPP1CC is associated with astrocyte and microglia proliferation after traumatic spinal cord injury in rats. Pathol Res Pract 2017; 213:1355-1364. [DOI: 10.1016/j.prp.2017.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 10/18/2022]
|
17
|
Inhibition of PTEN Attenuates Endoplasmic Reticulum Stress and Apoptosis via Activation of PI3K/AKT Pathway in Alzheimer’s Disease. Neurochem Res 2017; 42:3052-3060. [PMID: 28819903 DOI: 10.1007/s11064-017-2338-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 12/14/2022]
|
18
|
Liu XY, Zhang LJ, Chen Z, Liu LB. The PTEN inhibitor bpV(pic) promotes neuroprotection against amyloid β-peptide (25-35)-induced oxidative stress and neurotoxicity. Neurol Res 2017; 39:758-765. [PMID: 28436304 DOI: 10.1080/01616412.2017.1317916] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Xiao-Ying Liu
- Department of Endocrinology, Fujian Institute of Endocrinology, Union Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Jing Zhang
- Zhejiang Department of Pharmacology, Pharmaceutical college, Ningbo, China
| | - Zhou Chen
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Li-Bin Liu
- Department of Endocrinology, Fujian Institute of Endocrinology, Union Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
19
|
Pulido R. PTEN: a yin-yang master regulator protein in health and disease. Methods 2016; 77-78:3-10. [PMID: 25843297 DOI: 10.1016/j.ymeth.2015.02.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 01/16/2023] Open
Abstract
The PTEN gene is a tumor suppressor gene frequently mutated in human tumors, which encodes a ubiquitous protein whose major activity is to act as a lipid phosphatase that counteracts the action of the oncogenic PI3K. In addition, PTEN displays protein phosphatase- and catalytically-independent activities. The physiologic control of PTEN function, and its inactivation in cancer and other human diseases, including some neurodevelopmental disorders, is upon the action of multiple regulatory mechanisms. This provides a wide spectrum of potential therapeutic approaches to reconstitute PTEN activity. By contrast, inhibition of PTEN function may be beneficial in a different group of human diseases, such as type 2 diabetes or neuroregeneration-related pathologies. This makes PTEN a functionally dual yin-yang protein with high potential in the clinics. Here, a brief overview on PTEN and its relation with human disease is presented.
Collapse
Affiliation(s)
- Rafael Pulido
- BioCruces Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
20
|
A Single Bolus of Docosahexaenoic Acid Promotes Neuroplastic Changes in the Innervation of Spinal Cord Interneurons and Motor Neurons and Improves Functional Recovery after Spinal Cord Injury. J Neurosci 2016; 35:12733-52. [PMID: 26377463 DOI: 10.1523/jneurosci.0605-15.2015] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Docosahexaenoic acid (DHA) is an ω-3 polyunsaturated fatty acid that is essential in brain development and has structural and signaling roles. Acute DHA administration is neuroprotective and promotes functional recovery in animal models of adult spinal cord injury (SCI). However, the mechanisms underlying this recovery have not been fully characterized. Here we investigated the effects of an acute intravenous bolus of DHA delivered after SCI and characterized DHA-induced neuroplasticity within the adult injured spinal cord. We found robust sprouting of uninjured corticospinal and serotonergic fibers in a rat cervical hemisection SCI model. A mouse pyramidotomy model was used to confirm that this robust sprouting was not species or injury model specific. Furthermore, we demonstrated that corticospinal fibers sprouting to the denervated side of the cord following pyramidotomy contact V2a interneurons. We also demonstrated increased serotonin fibers and synaptophysin in direct contact with motor neurons. DHA also increased synaptophysin in rat cortical cell cultures. A reduction in phosphatase and tensin homolog (PTEN) has been shown to be involved in axonal regeneration and synaptic plasticity. We showed that DHA significantly upregulates miR-21 and downregulates PTEN in corticospinal neurons. Downregulation of PTEN and upregulation of phosphorylated AKT by DHA were also seen in primary cortical neuron cultures and were accompanied by increased neurite outgrowth. In summary, acute DHA induces anatomical and synaptic plasticity in adult injured spinal cord. This study shows that DHA has therapeutic potential in cervical SCI and provides evidence that DHA could exert its beneficial effects in SCI via enhancement of neuroplasticity. SIGNIFICANCE STATEMENT In this study, we show that an acute intravenous injection of docosahexaenoic acid (DHA) 30 min after spinal cord injury induces neuroplasticity. We found robust sprouting of uninjured corticospinal and serotonergic fibers in a rat hemisection spinal cord injury model. A mouse pyramidotomy model was used to confirm that the robust sprouting involved V2a interneurons. We show that DHA significantly upregulates miR-21 and phosphorylated AKT, and downregulates phosphatase and tensin homolog (PTEN), which is involved in suppressing anatomical plasticity, in corticospinal neurons and in primary cortical neuron cultures. We conclude that acute DHA can induce anatomical and synaptic plasticity. This provides direct evidence that DHA could exert its beneficial effects in spinal cord injury via neuroplasticity enhancement.
Collapse
|
21
|
Selective inhibition of PTEN preserves ischaemic post-conditioning cardioprotection in STZ-induced Type 1 diabetic rats: role of the PI3K/Akt and JAK2/STAT3 pathways. Clin Sci (Lond) 2015; 130:377-92. [PMID: 26666444 DOI: 10.1042/cs20150496] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/14/2015] [Indexed: 12/23/2022]
Abstract
Patients with diabetes are vulnerable to MI/R (myocardial ischaemia/reperfusion) injury, but are not responsive to IPostC (ischaemic post-conditioning) which activates PI3K (phosphoinositide 3-kinase)/Akt (also known as PKB or protein kinase B) and JAK2 (Janus kinase 2)/STAT3 (signal transducer and activator of transcription 3) pathways to confer cardioprotection. We hypothesized that increased cardiac PTEN (phosphatase and tensin homologue deleted on chromosome 10), a major negative regulator of PI3K/Akt, is responsible for the loss of diabetic heart sensitivity to IPostC cardioprotecton. In STZ (streptozotocin)-induced Type 1 diabetic rats subjected to MI/R (30 min coronary occlusion and 120 min reperfusion), the post-ischaemic myocardial infarct size, CK-MB (creatine kinase-MB) and 15-F2t-isoprostane release, as well as cardiac PTEN expression were significantly higher than those in non-diabetic controls, concomitant with more severe cardiac dysfunction and lower cardiac Akt, STAT3 and GSK-3β (glycogen synthase kinase 3β) phosphorylation. IPostC significantly attenuated post-ischaemic infarct size, decreased PTEN expression and further increased Akt, STAT3 and GSK-3β phosphorylation in non-diabetic, but not in diabetic rats. Application of the PTEN inhibitor BpV (bisperoxovanadium) (1.0 mg/kg) restored IPostC cardioprotection in diabetic rats. HPostC (hypoxic post-conditioning) in combination with PTEN gene knockdown, but not HPostC alone, significantly reduced H/R (hypoxia/reoxygenation) injury in cardiac H9c2 cells exposed to high glucose as was evident from reduced apoptotic cell death and JC-1 monomer in cells, accompanied by increased phosphorylation of Akt, STAT3 and GSK-3β. PTEN inhibition/gene knockdown mediated restoration of IPostC/HPostC cardioprotection was completely reversed by the PI3K inhibitor wortmannin, and partially reversed by the JAK2 inhibitor AG490. Increased cardiac PTEN, by impairing PI3K/Akt and JAK2/STAT3 pathways, is a major mechanism that rendered diabetic hearts not responsive to post-conditioning cardioprotection.
Collapse
|
22
|
Alluri H, Wiggins-Dohlvik K, Davis ML, Huang JH, Tharakan B. Blood-brain barrier dysfunction following traumatic brain injury. Metab Brain Dis 2015; 30:1093-104. [PMID: 25624154 DOI: 10.1007/s11011-015-9651-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/13/2015] [Indexed: 01/24/2023]
Abstract
Traumatic brain injury is a serious cause of morbidity and mortality worldwide. After traumatic brain injury, the blood-brain barrier, the protective barrier between the brain and the intravascular compartment, becomes dysfunctional, leading to leakage of proteins, fluid, and transmigration of immune cells. As this leakage has profound clinical implications, including edema formation, elevated intracranial pressure and decreased perfusion pressure, much interest has been paid to better understanding the mechanisms responsible for these events. Various molecular pathways and numerous mediators have been found to be involved in the intricate process of regulating blood-brain barrier permeability following traumatic brain injury. This review provides an update to the existing knowledge about the various pathophysiological pathways and advancements in the field of blood-brain barrier dysfunction and hyperpermeability following traumatic brain injury, including the role of various tight junction proteins involved in blood-brain barrier integrity and regulation. We also address pitfalls of existing systems and propose strategies to improve the various debilitating functional deficits caused by this progressive epidemic.
Collapse
Affiliation(s)
- Himakarnika Alluri
- Department of Surgery, Baylor Scott & White Health & Texas A&M University Health Science Center, College of Medicine, 702 S.W. H.K. Dodgen Loop, Temple, TX, 76504, USA
| | | | | | | | | |
Collapse
|
23
|
Administration of a PTEN inhibitor BPV(pic) attenuates early brain injury via modulating AMPA receptor subunits after subarachnoid hemorrhage in rats. Neurosci Lett 2015; 588:131-6. [PMID: 25575796 DOI: 10.1016/j.neulet.2015.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 12/31/2014] [Accepted: 01/03/2015] [Indexed: 01/08/2023]
Abstract
The aim of this study was to investigate whether the phosphatase and tensin homolog deleted on chromosome ten (PTEN) inhibitor dipotassium bisperoxo(pyridine-2-carboxyl) oxovanadate (BPV(pic)) attenuates early brain injury by modulating α-amino-3-hydroxy-5-methyl-4-isoxa-zolep-propionate (AMPA) receptor subunits after subarachnoid hemorrhage (SAH). A standard intravascular perforation model was used to produce the experimental SAH in Sprague-Dawley rats. BPV(pic) treatment (0.2mg/kg) was evaluated for effects on neurological score, brain water content, Evans blue extravasation, hippocampal neuronal death and AMPA receptor subunits alterations after SAH. We found that BPV(pic) is effective in attenuating BBB disruption, lowering edema, reducing hippocampal neural death and improving neurological outcomes. In addition, the AMPA receptor subunit GluR1 protein expression at cytomembrane was downregulated, whereas the expression of GluR2 and GluR3 was upregulated after BPV(pic) treatment. Our results suggest that PTEN inhibited by BPV(pic) plays a neuroprotective role in SAH pathophysiology, possibly by alterations in glutamate AMPA receptor subunits.
Collapse
|
24
|
Sandhir R, Gregory E, Berman NEJ. Differential response of miRNA-21 and its targets after traumatic brain injury in aging mice. Neurochem Int 2014; 78:117-21. [PMID: 25277076 DOI: 10.1016/j.neuint.2014.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 09/21/2014] [Accepted: 09/25/2014] [Indexed: 12/17/2022]
Abstract
The present study investigated the possible role of miR-21, a miRNA that has known prosurvival function, in poor outcomes in the elderly following traumatic brain injury compared to adults. Controlled cortical impact injury was induced in adult (5-6 months) and aged (22-24 months) C57/BL6 mice. miR-21 and four of its targets (PDCD4, TIMP3, RECK, PTEN) were analyzed at 1, 3, 7 days post injury in samples of injured cortex using real-time PCR analysis. Basal miR-21 expression was higher in the aged brain than in the adult brain. In the adult brain, miR-21 expression increased in response to injury, with the maximum increase 24 hours after injury followed by a gradual decrease, returning to baseline 7 days post-injury. In contrast, in aged mice, miR21 showed no injury response, and expression of miR-21 target genes (PTEN, PDCD4, RECK, TIMP3) was up-regulated at all post injury time points, with a maximal increase at 24 hours post injury. Based on these results, we conclude that the diminished miR21 injury response in the aged brain leads to up-regulation of its targets, with the potential to contribute to the poor prognosis following TBI in aging brain. Therefore, strategies aimed at up-regulation of miR-21 and/or down regulation of its targets might be useful in improving outcomes in the elderly following TBI.
Collapse
Affiliation(s)
- Rajat Sandhir
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Eugene Gregory
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Nancy E J Berman
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA.
| |
Collapse
|
25
|
van Vliet E, Aronica E, Gorter J. Role of blood–brain barrier in temporal lobe epilepsy and pharmacoresistance. Neuroscience 2014; 277:455-73. [DOI: 10.1016/j.neuroscience.2014.07.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 12/14/2022]
|
26
|
Kreis P, Leondaritis G, Lieberam I, Eickholt BJ. Subcellular targeting and dynamic regulation of PTEN: implications for neuronal cells and neurological disorders. Front Mol Neurosci 2014; 7:23. [PMID: 24744697 PMCID: PMC3978343 DOI: 10.3389/fnmol.2014.00023] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/12/2014] [Indexed: 01/13/2023] Open
Abstract
PTEN is a lipid and protein phosphatase that regulates a diverse range of cellular mechanisms. PTEN is mainly present in the cytosol and transiently associates with the plasma membrane to dephosphorylate PI(3,4,5)P3, thereby antagonizing the PI3-Kinase signaling pathway. Recently, PTEN has been shown to associate also with organelles such as the endoplasmic reticulum (ER), the mitochondria, or the nucleus, and to be secreted outside of the cell. In addition, PTEN dynamically localizes to specialized sub-cellular compartments such as the neuronal growth cone or dendritic spines. The diverse localizations of PTEN imply a tight temporal and spatial regulation, orchestrated by mechanisms such as posttranslational modifications, formation of distinct protein–protein interactions, or the activation/recruitment of PTEN downstream of external cues. The regulation of PTEN function is thus not only important at the enzymatic activity level, but is also associated to its spatial distribution. In this review we will summarize (i) recent findings that highlight mechanisms controlling PTEN movement and sub-cellular localization, and (ii) current understanding of how PTEN localization is achieved by mechanisms controlling posttranslational modification, by association with binding partners and by PTEN structural or activity requirements. Finally, we will discuss the possible roles of compartmentalized PTEN in developing and mature neurons in health and disease.
Collapse
Affiliation(s)
- Patricia Kreis
- MRC Centre for Developmental Neurobiology, King's College London London, UK
| | - George Leondaritis
- MRC Centre for Developmental Neurobiology, King's College London London, UK ; Institute of Biochemistry, Charité - Universitätsmedizin Berlin Berlin, Germany
| | - Ivo Lieberam
- MRC Centre for Developmental Neurobiology, King's College London London, UK
| | - Britta J Eickholt
- MRC Centre for Developmental Neurobiology, King's College London London, UK ; Institute of Biochemistry, Charité - Universitätsmedizin Berlin Berlin, Germany
| |
Collapse
|