1
|
Liu J, Ji Z, He Q, Chen H, Xu X, Mei Q, Hu Y, Zhang H. Direct conversion of human umbilical cord mesenchymal stem cells into dopaminergic neurons for Parkinson's disease treatment. Neurobiol Dis 2024; 201:106683. [PMID: 39343249 DOI: 10.1016/j.nbd.2024.106683] [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: 06/26/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits due to the depletion of nigrostriatal dopamine. Stem cell differentiation therapy emerges as a promising treatment option for sustained symptom relief. In this study, we successfully developed a one-step differentiation system using the YFBP cocktail (Y27632, Forskolin, SB431542, and SP600125) to effectively convert human umbilical cord mesenchymal stem cells (hUCMSCs) into dopaminergic neurons without genetic modification. This approach addresses the challenge of rapidly and safely generating functional neurons on a large scale. After a 7-day induction period, over 80 % of the cells were double-positive for TUBB3 and NEUN. Transcriptome analysis revealed the dual roles of the cocktail in inducing fate erasure in mesenchymal stem cells and activating the neuronal program. Notably, these chemically induced cells (CiNs) did not express HLA class II genes, preserving their immune-privileged status. Further study indicated that YFBP significantly downregulated p53 signaling and accelerated the differentiation process when Pifithrin-α, a p53 signaling inhibitor, was applied. Additionally, Wnt/β-catenin signaling was transiently activated within one day, but the prolonged activation hindered the neuronal differentiation of hUCMSCs. Upon transplantation into the striatum of mice, CiNs survived well and tested positive for dopaminergic neuron markers. They exhibited typical action potentials and sodium and potassium ion channel activity, demonstrating neuronal electrophysiological activity. Furthermore, CiNs treatment significantly increased the number of tyrosine hydroxylase-positive cells and the concentration of dopamine in the striatum, effectively ameliorating movement disorders in PD mice. Overall, our study provides a secure and reliable framework for cell replacement therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Jinming Liu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Zhongqing Ji
- Department of Orthopedics, Suzhou Yongding Hospital, Suzhou 215200, China
| | - Qisheng He
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Huanhuan Chen
- The Suqian Clinical College of Xuzhou Medical University, Suqian 223800, China
| | - Xiaojing Xu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Qiuhao Mei
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Ya'nan Hu
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| | - Huanxiang Zhang
- Department of Cell Biology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
2
|
Benarroch E. What Is the Role of the Rho-ROCK Pathway in Neurologic Disorders? Neurology 2023; 101:536-543. [PMID: 37722862 PMCID: PMC10516277 DOI: 10.1212/wnl.0000000000207779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 09/20/2023] Open
|
3
|
Lu W, Wen J. H 2S-RhoA/ROCK Pathway and Glial Cells in Axonal Remyelination After Ischemic Stroke. Mol Neurobiol 2023; 60:5493-5504. [PMID: 37322287 DOI: 10.1007/s12035-023-03422-8] [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: 12/05/2022] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Henning L, Unichenko P, Bedner P, Steinhäuser C, Henneberger C. Overview Article Astrocytes as Initiators of Epilepsy. Neurochem Res 2023; 48:1091-1099. [PMID: 36244037 PMCID: PMC10030460 DOI: 10.1007/s11064-022-03773-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 08/22/2022] [Accepted: 09/27/2022] [Indexed: 10/17/2022]
Abstract
Astrocytes play a dual role in the brain. On the one hand, they are active signaling partners of neurons and can for instance control synaptic transmission and its plasticity. On the other hand, they fulfill various homeostatic functions such as clearance of glutamate and K+ released from neurons. The latter is for instance important for limiting neuronal excitability. Therefore, an impairment or failure of glutamate and K+ clearance will lead to increased neuronal excitability, which could trigger or aggravate brain diseases such as epilepsy, in which neuronal hyperexcitability plays a role. Experimental data indicate that astrocytes could have such a causal role in epilepsy, but the role of astrocytes as initiators of epilepsy and the relevant mechanisms are under debate. In this overview, we will discuss the potential mechanisms with focus on K+ clearance, glutamate uptake and homoeostasis and related mechanisms, and the evidence for their causative role in epilepsy.
Collapse
Affiliation(s)
- Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Petr Unichenko
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.
| |
Collapse
|
5
|
Hong N, Kim HJ, Kang K, Park JO, Mun S, Kim HG, Kang BH, Chung PS, Lee MY, Ahn JC. Photobiomodulation improves the synapses and cognitive function and ameliorates epileptic seizure by inhibiting downregulation of Nlgn3. Cell Biosci 2023; 13:8. [PMID: 36635704 PMCID: PMC9837965 DOI: 10.1186/s13578-022-00949-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Temporal lobe epilepsy (TLE) remains one of the most drug-resistant focal epilepsies. Glutamate excitotoxicity and neuroinflammation which leads to loss of synaptic proteins and neuronal death appear to represent a pathogen that characterizes the neurobiology of TLE. Photobiomodulation (PBM) is a rapidly growing therapy for the attenuation of neuronal degeneration harboring non-invasiveness benefits. However, the detailed effects of PBM on excitotoxicity or neuroinflammation remain unclear. We investigated whether tPBM exerts neuroprotective effects on hippocampal neurons in epilepsy mouse model by regulating synapse and synapse-related genes. METHODS In an in vitro study, we performed imaging analysis and western blot in primary hippocampal neurons from embryonic (E17) rat pups. In an in vivo study, RNA sequencing was performed to identify the gene regulatory by PBM. Histological stain and immunohistochemistry analyses were used to assess synaptic connections, neuroinflammation and neuronal survival. Behavioral tests were used to evaluate the effects of PBM on cognitive functions. RESULTS PBM was upregulated synaptic connections in an in vitro. In addition, it was confirmed that transcranial PBM reduced synaptic degeneration, neuronal apoptosis, and neuroinflammation in an in vivo. These effects of PBM were supported by RNA sequencing results showing the relation of PBM with gene regulatory networks of neuronal functions. Specifically, Nlgn3 showed increase after PBM and silencing the Nlgn3 reversed the positive effect of PBM in in vitro. Lastly, behavioral alterations including hypoactivity, anxiety and impaired memory were recovered along with the reduction of seizure score in PBM-treated mice. CONCLUSIONS Our findings demonstrate that PBM attenuates epileptic excitotoxicity, neurodegeneration and cognitive decline induced by TLE through inhibition of the Nlgn3 gene decrease induced by excitotoxicity.
Collapse
Affiliation(s)
- Namgue Hong
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Hee Jung Kim
- grid.411982.70000 0001 0705 4288Department of Physiology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Keunsoo Kang
- grid.411982.70000 0001 0705 4288Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea
| | - Ji On Park
- grid.411982.70000 0001 0705 4288Department of Medicine, Graduate School of Dankook University, Dankook University, Cheonan, Republic of Korea
| | - Seyoung Mun
- grid.411982.70000 0001 0705 4288Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan, Republic of Korea
| | - Hyung-Gun Kim
- grid.411982.70000 0001 0705 4288Department of Pharmacology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Bong Hui Kang
- grid.411982.70000 0001 0705 4288Department of Neurology, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Phil-Sang Chung
- grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Min Young Lee
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| | - Jin-Chul Ahn
- grid.411982.70000 0001 0705 4288Medical Laser Research Center, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Department of Biomedical Science, College of Medicine, Dankook University, Cheonan, Republic of Korea ,grid.411982.70000 0001 0705 4288Beckman Laser Institute Korea, Dankook University Hospital, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
6
|
Maurer-Morelli CV, de Vasconcellos JF, Bruxel EM, Rocha CS, do Canto AM, Tedeschi H, Yasuda CL, Cendes F, Lopes-Cendes I. Gene expression profile suggests different mechanisms underlying sporadic and familial mesial temporal lobe epilepsy. Exp Biol Med (Maywood) 2022; 247:2233-2250. [PMID: 36259630 PMCID: PMC9899983 DOI: 10.1177/15353702221126666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Most patients with pharmacoresistant mesial temporal lobe epilepsy (MTLE) have hippocampal sclerosis on the postoperative histopathological examination. Although most patients with MTLE do not refer to a family history of the disease, familial forms of MTLE have been reported. We studied surgical specimens from patients with MTLE who had epilepsy surgery for medically intractable seizures. We assessed and compared gene expression profiles of the tissue lesion found in patients with familial MTLE (n = 3) and sporadic MTLE (n = 5). In addition, we used data from control hippocampi obtained from a public database (n = 7). We obtained expression profiles using the Human Genome U133 Plus 2.0 (Affymetrix) microarray platform. Overall, the molecular profile identified in familial MTLE differed from that in sporadic MTLE. In the tissue of patients with familial MTLE, we found an over-representation of the biological pathways related to protein response, mRNA processing, and synaptic plasticity and function. In sporadic MTLE, the gene expression profile suggests that the inflammatory response is highly activated. In addition, we found enrichment of gene sets involved in inflammatory cytokines and mediators and chemokine receptor pathways in both groups. However, in sporadic MTLE, we also found enrichment of epidermal growth factor signaling, prostaglandin synthesis and regulation, and microglia pathogen phagocytosis pathways. Furthermore, based on the gene expression signatures, we identified different potential compounds to treat patients with familial and sporadic MTLE. To our knowledge, this is the first study assessing the mRNA profile in surgical tissue obtained from patients with familial MTLE and comparing it with sporadic MTLE. Our results clearly show that, despite phenotypic similarities, both forms of MTLE present distinct molecular signatures, thus suggesting different underlying molecular mechanisms that may require distinct therapeutic approaches.
Collapse
Affiliation(s)
- Claudia V Maurer-Morelli
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Jaira F de Vasconcellos
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Department of Biology, James Madison
University, Harrisonburg, VA 22807, USA
| | - Estela M Bruxel
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Cristiane S Rocha
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Amanda M do Canto
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil
| | - Helder Tedeschi
- Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Clarissa L Yasuda
- Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Fernando Cendes
- Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Department of Neurology, School of
Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine,
School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888,
Brazil,Brazilian Institute of Neuroscience and
Neurotechnology (BRAINN), Campinas 13083-888, Brazil,Iscia Lopes-Cendes.
| |
Collapse
|
7
|
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
|
8
|
Murray TE, Richards CM, Robert-Gostlin VN, Bernath AK, Lindhout IA, Klegeris A. Potential neurotoxic activity of diverse molecules released by astrocytes. Brain Res Bull 2022; 189:80-101. [PMID: 35988785 DOI: 10.1016/j.brainresbull.2022.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Astrocytes are the main support cells of the central nervous system. They also participate in neuroimmune reactions. In response to pathological and immune stimuli, astrocytes transform to reactive states characterized by increased release of inflammatory mediators. Some of these molecules are neuroprotective and inflammation resolving while others, including reactive oxygen species (ROS), nitric oxide (NO), matrix metalloproteinase (MMP)- 9, L-glutamate, and tumor necrosis factor α (TNF), are well-established toxins known to cause damage to surrounding cells and tissues. We hypothesized that similar to microglia, the brain immune cells, reactive astrocytes can release a broader set of diverse molecules that are potentially neurotoxic. A literature search was conducted to identify such molecules using the following two criteria: 1) evidence of their expression and secretion by astrocytes and 2) direct neurotoxic action. This review describes 14 structurally diverse molecules as less-established astrocyte neurotoxins, including C-X-C motif chemokine ligand (CXCL)10, CXCL12/CXCL12(5-67), FS-7-associated surface antigen ligand (FasL), macrophage inflammatory protein (MIP)- 2α, TNF-related apoptosis inducing ligand (TRAIL), pro-nerve growth factor (proNGF), pro-brain-derived neurotrophic factor (proBDNF), chondroitin sulfate proteoglycans (CSPGs), cathepsin (Cat)B, group IIA secretory phospholipase A2 (sPLA2-IIA), amyloid beta peptides (Aβ), high mobility group box (HMGB)1, ceramides, and lipocalin (LCN)2. For some of these molecules, further studies are required to establish either their direct neurotoxic effects or the full spectrum of stimuli that induce their release by astrocytes. Only limited studies with human-derived astrocytes and neurons are available for most of these potential neurotoxins, which is a knowledge gap that should be addressed in the future. We also summarize available evidence of the role these molecules play in select neuropathologies where reactive astrocytes are a key feature. A comprehensive understanding of the full spectrum of neurotoxins released by reactive astrocytes is key to understanding neuroinflammatory diseases characterized by the adverse activation of these cells and may guide the development of novel treatment strategies.
Collapse
Affiliation(s)
- Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Victoria N Robert-Gostlin
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Anna K Bernath
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Ivan A Lindhout
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
9
|
Wang X, Wu J, Wang W, Zhang Y, He D, Xiao B, Zhang H, Song A, Xing Y, Li B. Reprogramming of Rat Fibroblasts into Induced Neurons by Small-Molecule Compounds In Vitro and In Vivo. ACS Chem Neurosci 2022; 13:2099-2109. [PMID: 35723446 DOI: 10.1021/acschemneuro.2c00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cell replacement is a promising approach for neurodegenerative disease treatment. Somatic cells such as fibroblasts can be induced to differentiate into neurons by specific transcription factors; however, the potential of viral vectors used for reprogramming to integrate into the genome raises concerns about the potential clinical applications of this approach. Here, we directly reprogrammed rat embryonic skin fibroblasts into induced neurons (iNs) via six small-molecule compounds (SMs) (VPA, CHIR99021, forskolin, Y-27632, Repsox, and P7C3-A20). iNs exhibit typical neuronal morphology, and immunofluorescence showed that more than 96% of the iNs expressed the early neuronal marker class III beta-tubulin (TUJ1) and that more than 91% of iNs expressed the mature neuronal marker neuron-specific enolase (NSE) after 10 days of reprogramming. Quantitative real-time polymerase chain reaction also showed that most iNs expressed the dopaminergic neuron marker tyrosine hydroxylase, the neural marker Nur correlation factor 1, the (γ-aminobutyric acid, GABA) GABAergic neuronal marker GABA, and the cholinergic neuron marker choline acetyltransferase. In addition, we found that cell proliferation decreased during reprogramming and that protein synthesis increased initially and then decreased. SMs were mixed with hydrogels, and the hydrogels were implanted subcutaneously into the backs of rats. After 7 days, the TUJ1 and NSE proteins were expressed in surrounding tissues, indicating that SMs caused reprogramming in vivo. In summary, rat skin fibroblasts can be efficiently reprogrammed into iNs by SMs in vitro and in vivo.
Collapse
Affiliation(s)
- Xueyun Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Jing Wu
- Department of Paediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001 Henan, P.R. China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Yuanwang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Dixin He
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Boying Xiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Haohao Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Anqi Song
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Bo Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| |
Collapse
|
10
|
Wang Z, Ren D, Zheng P. The role of Rho/ROCK in epileptic seizure-related neuronal damage. Metab Brain Dis 2022; 37:881-887. [PMID: 35119588 PMCID: PMC9042975 DOI: 10.1007/s11011-022-00909-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/06/2022] [Indexed: 01/09/2023]
Abstract
Epilepsy is one of the most severe neurological disorders characterized by spontaneous recurrent seizures. Although more than two-thirds of patients can be cured with anti-epileptic drugs (AEDs), the rest one-third of epilepsy patients are resistant to AEDs. A series of studies have demonstrated Rho/Rho-associated kinase (ROCK) pathway might be involved in the pathogenesis of epilepsy in the recent twenty years. Several related pathway inhibitors of Rho/ROCK have been used in the treatment of epilepsy. We searched PubMed from Jan 1, 2000 to Dec 31, 2020, using the terms "epilepsy AND Rho AND ROCK" and "seizure AND Rho AND ROCK". We selected articles that characterized Rho/ROCK in animal models of epilepsy and patients. We then chose the most relevant research studies including in-vitro, in-vivo and clinical trials. The expression of Rho/ROCK could be a potential non-invasive biomarker to apply in treatment for patients with epilepsy. RhoA and ROCK show significant upregulation in the acute and chronic stage of epilepsy. ROCK inhibitors can reduce the epilepsy, epileptic seizure-related neuronal death and comorbidities. These findings demonstrate the novel development for diagnosis and treatment for patients with epilepsy. Rho/ROCK signaling pathway inhibitors may show more promising effects in epilepsy and related neurological diseases.
Collapse
Affiliation(s)
- Zhihan Wang
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Dabin Ren
- Department of Neurusurgery & Key Laboratory, Shanghai Pudong New area People's Hospital, Shanghai, 201299, China
| | - Ping Zheng
- Department of Neurusurgery & Key Laboratory, Shanghai Pudong New area People's Hospital, Shanghai, 201299, China.
| |
Collapse
|
11
|
Zhu Z, Lu J, Wang S, Peng W, Yang Y, Chen C, Zhou X, Yang X, Xin W, Chen X, Pi J, Yin W, Yao L, Pi R. Acrolein, an endogenous aldehyde induces synaptic dysfunction in vitro and in vivo: Involvement of RhoA/ROCK2 pathway. Aging Cell 2022; 21:e13587. [PMID: 35315217 PMCID: PMC9009232 DOI: 10.1111/acel.13587] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/19/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Acrolein, an unsaturated aldehyde, is increased in the brain of Alzheimer's disease (AD) patients and identified as a potential inducer of sporadic AD. Synaptic dysfunction, as a typical pathological change occurring in the early stage of AD, is most closely associated with the severity of dementia. However, there remains a lack of clarity on the mechanisms of acrolein inducing AD-like pathology and synaptic impairment. In this study, acrolein-treated primary cultured neurons and mice were applied to investigate the effects of acrolein on cognitive impairment and synaptic dysfunction and their signaling mechanisms. In vitro, ROCK inhibitors, Fasudil, and Y27632, could attenuate the axon ruptures and synaptic impairment caused by acrolein. Meanwhile, RNA-seq distinct differentially expressed genes in acrolein models and initially linked activated RhoA/Rho-kinase2 (ROCK2) to acrolein-induced synaptic dysfunction, which could regulate neuronal cytoskeleton and neurite. The Morris water maze test and in vivo field excitatory postsynaptic potential (fEPSP) were performed to evaluate spatial memory and long-term potential (LTP), respectively. Acrolein induced cognitive impairment and attenuated LTP. Furthermore, the protein level of Synapsin 1 and postsynaptic density 95 (PSD95) and dendritic spines density were also decreased in acrolein-exposed mice. These changes were improved by ROCK2 inhibitor Fasudil or in ROCK2+/- mice. Together, our findings suggest that RhoA/ROCK2 signaling pathway plays a critical role in acrolein-induced synaptic damage and cognitive dysfunction, suggesting inhibition of ROCK2 should benefit to the early AD.
Collapse
Affiliation(s)
- Zeyu Zhu
- School of Medicine Sun Yat‐Sen University Guangzhou China
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Junfeng Lu
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
- Department of Internal Medicine The Affiliated Tumor Hospital of Zhengzhou University Zhengzhou China
| | - Shuyi Wang
- School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Weijia Peng
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Yang Yang
- School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Chen Chen
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Xin Zhou
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen Center for Disease Control and Prevention
| | - Wenjun Xin
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Xinyi Chen
- School of Pharmaceutical Sciences South China Research Center for Acupuncture and Moxibustion Guangzhou University of Chinese Medicine Guangzhou China
| | - Jiakai Pi
- Guangzhou Foreign Language School Guangzhou China
| | - Wei Yin
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Lin Yao
- Research Institute of Acupuncture and Moxibustion Shandong University of Traditional Chinese Medicine Jinan China
| | - Rongbiao Pi
- School of Medicine Sun Yat‐Sen University Guangzhou China
- International Joint Laboratory<SYSU‐PolyU HK> of Novel Anti‐Dementia Drugs of Guangzhou Guangzhou China
- Guangdong Province Key Laboratory of Brain Function and Disease Sun Yat‐sen University Guangzhou China
| |
Collapse
|
12
|
Ye Z, Izadi A, Gurkoff GG, Rickerl K, Sharp F, Ander B, Bauer SZ, Lui A, Lyeth BG, Liu D. Combined Inhibition of Fyn and c-Src Protects Hippocampal Neurons and Improves Spatial Memory via ROCK after Traumatic Brain Injury. J Neurotrauma 2022; 39:520-529. [PMID: 35109711 PMCID: PMC8978569 DOI: 10.1089/neu.2021.0311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Our previous studies demonstrated that TBI and ventricular administration of thrombin caused hippocampal neuron loss and cognitive dysfunction via activation of Src family kinases (SFKs). Based on SFK localization in brain, we hypothesized SFK subtypes Fyn and c-Src as well as SFK downstream molecule Rho-associated protein kinase (ROCK) contribute to cell death and cognitive dysfunction after TBI. We administered nanoparticle wrapped siRNA-Fyn and siRNA-c-Src, or ROCK inhibitor Y-27632 to adult rats subjected to moderate lateral fluid percussion (LFP) induced TBI. Spatial memory function was assessed from 12 to 16 days, and NeuN stained hippocampal neurons were assessed 16 days after TBI. The combination of siRNA-Fyn and siRNA-c-Src, but neither alone, prevented hippocampal neuron loss and spatial memory deficits after TBI. The ROCK inhibitor Y-27632 also prevented hippocampal neuronal loss and spatial memory deficits after TBI. The data suggest that the combined actions of three kinases (Fyn, c-Src, ROCK) mediate hippocampal neuronal cell death and spatial memory deficits produced by LFP-TBI, and that inhibiting this pathway prevents the TBI-induced cell death and memory deficits.
Collapse
Affiliation(s)
- Zhouheng Ye
- University of California at Davis Medical Center, Department of Neurology, Davis, California, United States;
| | - Ali Izadi
- University of California, Davis, Neurological Surgery, 1515 Newton Ct, Room 502, Davis, California, United States, 95618;
| | - Gene Gabriel Gurkoff
- University of California, Davis, Neurological Surgery, 1515 Newton Ct, Room 502, Davis, California, United States, 95618;
| | - Kaitlin Rickerl
- University of California at Davis Medical Center, Department of Neurology, Davis, California, United States;
| | - Frank Sharp
- University of California Davis, MIND Institute, Davis, United States;
| | - Bradley Ander
- University of California at Davis Medical Center, Department of Neurology and the M.I.N.D. Institute, Sacramento, California, United States;
| | - Sawyer Z Bauer
- University of California at Davis Medical Center, Department of Neurology, Davis, California, United States;
| | - Austin Lui
- University of California at Davis Medical Center, Department of Neurology, Davis, California, United States;
| | - Bruce G Lyeth
- U.C. Davis, Neurological Surgery, One Shields Ave, Davis, California, United States, 95616;
| | - DaZhi Liu
- University of California at Davis Medical Center, Department of Neurology, Davis, California, United States;
| |
Collapse
|
13
|
Song LJ, Zhang H, Qu XP, Jin JG, Wang C, Jiang X, Gao L, Li G, Wang DL, Shen LL, Liu B. Increased expression of Rho-associated protein kinase 2 confers astroglial Stat3 pathway activation during epileptogenesis. Neurosci Res 2021; 177:25-37. [PMID: 34740726 DOI: 10.1016/j.neures.2021.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 10/19/2022]
Abstract
Patients with TLE are prone to tolerance to antiepileptic drugs. Based on the perspective of molecular targets for drug resistance, it is necessary to explore effective drug resistant genes and signaling pathways for the treatment of TLE. We performed gene expression profiles in hippocampus of patients with drug-resistant TLE and identified ROCK2 as one of the 20 most significantly increased genes in hippocampus. In vitro and in vivo experiments were performed to identify the potential role of ROCK2 in epileptogenesis. In addition, the activity of Stat3 pathway was tested in rat hippocampal tissues and primary cultured astrocytes. The expression levels of ROCK2 in the hippocampus of TLE patients were significantly increased compared with the control group, which was due to the hypomethylation of ROCK2 promoter. Fasudil, a specific Rho-kinase inhibitor, alleviated epileptic seizures in the pilocarpine rat model of TLE. Furthermore, ROCK2 activated the Stat3 pathway in pilocarpine-treated epilepsy rats, and the spearman correlation method confirmed that ROCK2 is associated with Stat3 activation in TLE patients. In addition, ROCK2 was predominantly expressed in astrocytes during epileptogenesis, and induced epileptogenesis by activating astrocyte cell cycle progression via Stat3 pathway. The overexpressed ROCK2 plays an important role in the pathogenesis of drug-resistant epilepsy. ROCK2 accelerates astrocytes cell cycle progression via the activation of Stat3 pathway likely provides the key to explaining the process of epileptogenesis.
Collapse
Affiliation(s)
- Li-Jia Song
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Peng Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jun-Gong Jin
- Department of Neurosurgery, Xi'an International Medical Center, Xi'an, China
| | - Chao Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xue Jiang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Da-Li Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang-Liang Shen
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China.
| | - Bei Liu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
14
|
Kimura T, Horikoshi Y, Kuriyagawa C, Niiyama Y. Rho/ROCK Pathway and Noncoding RNAs: Implications in Ischemic Stroke and Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222111573. [PMID: 34769004 PMCID: PMC8584200 DOI: 10.3390/ijms222111573] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 01/18/2023] Open
Abstract
Ischemic strokes (IS) and spinal cord injuries (SCI) are major causes of disability. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of the RhoA/ROCK pathway contributes to neuronal apoptosis, neuroinflammation, blood-brain barrier dysfunction, astrogliosis, and axon growth inhibition in IS and SCI. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), were previously considered to be non-functional. However, they have attracted much attention because they play an essential role in regulating gene expression in physiological and pathological conditions. There is growing evidence that ROCK inhibitors, such as fasudil and VX-210, can reduce injury in IS and SCI in animal models and clinical trials. Recently, it has been reported that miRNAs are decreased in IS and SCI, while lncRNAs are increased. Inhibiting the Rho/ROCK pathway with miRNAs alleviates apoptosis, neuroinflammation, oxidative stress, and axon growth inhibition in IS and SCI. Further studies are required to explore the significance of ncRNAs in IS and SCI and to establish new strategies for preventing and treating these devastating diseases.
Collapse
Affiliation(s)
- Tetsu Kimura
- Correspondence: ; Tel.: +81-18-884-6175; Fax: +81-18-884-6448
| | | | | | | |
Collapse
|
15
|
Xiang Y, Niu Y, Xie Y, Chen S, Zhu F, Shen W, Zeng LH. Inhibition of RhoA/Rho kinase signaling pathway by fasudil protects against kainic acid-induced neurite injury. Brain Behav 2021; 11:e2266. [PMID: 34156163 PMCID: PMC8413774 DOI: 10.1002/brb3.2266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
AIM RhoA/Rho kinase pathway is essential for regulating cytoskeletal structure. Although its effect on normal neurite outgrowth has been demonstrated, the role of this pathway in seizure-induced neurite injury has not been revealed. The research examined the phosphorylation level of RhoA/Rho kinase signaling pathway and to clarify the effect of fasudil on RhoA/Rho kinase signaling pathway and neurite outgrowth in kainic acid (KA)-treated Neuro-2A cells and hippocampal neurons. METHOD Western blotting analysis was used to investigate the expression of key proteins of RhoA/Rho kinase signaling pathway and the depolymerization of actin. After incubated without serum to induce neurite outgrowth, Neuro-2A cells were fixed, and immunofluorescent assay of rhodamine-phalloidin was applied to detect the cellular morphology and neurite length. The influence of KA on neurons was detected in primary hippocampal neurons. Whole-cell patch clamp was conducted in cultured neurons or hippocampal slices to record action potentials. RESULT KA at the dose of 100-200 μmol/L induced the increase in phosphorylation of Rho-associated coiled-coil-containing protein kinase and decrease in phosphorylation of Lin11, Isl-1 and Mec-3 kinase and cofilin. The effect of 200 μmol/L KA was peaked at 1-2 hours, and then gradually returned to baseline after 8 hours. Pretreatment with Rho kinase inhibitor fasudil reversed KA-induced activation of RhoA/Rho kinase pathway and increase in phosphorylation of slingshot and 14-3-3, which consequently reduced the ratio of G/F-actin. KA treatment induced inhibition of neurite outgrowth and decrease in spines both in Neuro-2a cells and in cultured hippocampal neurons, and pretreatment with fasudil alleviated KA-induced neurite outgrowth inhibition and spine loss. CONCLUSION These data indicate that inhibiting RhoA/Rho kinase pathway might be a potential treatment for seizure-induced injury.
Collapse
Affiliation(s)
- Yingchun Xiang
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Yumiao Niu
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang, China.,Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Yacong Xie
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang, China.,Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Shishuo Chen
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang, China.,Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Feng Zhu
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Weida Shen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Ling-Hui Zeng
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, Zhejiang, China.,Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Li B, Xu Y, Quan Y, Cai Q, Le Y, Ma T, Liu Z, Wu G, Wang F, Bao C, Li H. Inhibition of RhoA/ROCK Pathway in the Early Stage of Hypoxia Ameliorates Depression in Mice via Protecting Myelin Sheath. ACS Chem Neurosci 2020; 11:2705-2716. [PMID: 32667781 DOI: 10.1021/acschemneuro.0c00352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neuroplasticity and connectivity in the central nervous system (CNS) are easily damaged after hypoxia. Long-term exposure to an anoxic environment can lead to neuropsychiatric symptoms and increases the likelihood of depression. Demyelination is an important lesion of CNS injury that may occur in depression. Previous studies have found that the RhoA/ROCK pathway is upregulated in neuropsychiatric disorders such as multiple sclerosis, stroke, and neurodegenerative diseases. Therefore, the chief aim of this study is to explore the regulatory role of the RhoA/ROCK pathway in the development of depression after hypoxia by behavioral tests, Western blotting, immunostaining as well as electron microscopy. Results showed that HIF-1α, S100β, RhoA/ROCK, and immobility time in FST were increased, sucrose water preference ratio in SPT was decreased, and the aberrant activity of neurocyte and demyelination occurred after hypoxia. After the administration of Y-27632 and fluoxetine in hypoxia, these alterations were improved. Lingo1, a negative regulatory factor, was also overexpressed after hypoxia and its expression was decreased when the pathway blocked. However, fluoxetine had no effect on the expression of Lingo1. Then, we demonstrated that demyelination was associated with failures of oligodendrocyte precursor cell proliferation and differentiation and increased apoptosis of oligodendrocytes. Collectively, our data indicate that the RhoA/ROCK pathway plays a vital role in the initial depression during hypoxia. Blocking this pathway in the early stage of hypoxia can enhance the effectiveness of antidepressants, rescue myelin damage, and reduce the expression of the negative regulatory protein of myelination. The findings provide new insight into the prophylaxis and treatment of depression.
Collapse
Affiliation(s)
- Baichuan Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Yang Xu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Yong Quan
- Department of Teaching Experiment Center, Army Medical University, Chongqing 400038, China
| | - Qiyan Cai
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Yifan Le
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Teng Ma
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Zhi Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Guangyan Wu
- Department of Teaching Experiment Center, Army Medical University, Chongqing 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Chuncha Bao
- Department of Teaching Experiment Center, Army Medical University, Chongqing 400038, China
| | - Hongli Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
- Department of Teaching Experiment Center, Army Medical University, Chongqing 400038, China
| |
Collapse
|
17
|
Zhou X, Zhao B, Poonit K, Weng W, Yao C, Sun C, Yan H. An aligned nanofiber nerve conduit that inhibits painful traumatic neuroma formation through regulation of the RhoA/ROCK signaling pathway. J Neurosurg 2020; 132:837-846. [PMID: 30717043 DOI: 10.3171/2018.9.jns18360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/28/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Traumatic neuromas represent a prevalent source of neuropathic pain. As of yet, there has been no single treatment method that can guarantee permanent relief of symptoms. Although nerve-capping techniques have shown promise, their exact mechanisms remain elusive. The authors' aim was to examine the role of the RhoA/ROCK signaling pathway in the prevention of neuroma formation after neurectomy utilizing a nerve-capping technique. METHODS An aligned nanofiber tube was fabricated to cap the sciatic nerve in Sprague Dawley rats. The rats (n = 60) were randomly divided into the aligned SF/P (LLA-CL) capping group (capping group, n = 20), the capping and Y-27632 (ROCK pathway inhibitor) intervention group (intervention group, n = 20), and the no-capping group (control group, n = 20). The authors undertook a comprehensive assessment of the capping group, examining the animals' behavior, the extent of neuroma development, histology, gene and protein expression, and ultrastructural changes associated with the RhoA/ROCK signaling pathway. These findings were compared with those in the intervention and control groups. RESULTS The inciting injury resulted in the expression of the RhoA/ROCK signaling pathway, as well as its further upregulation in peripheral neurons. Axon outgrowth was significantly increased when RhoA/ROCK signaling pathway was suppressed. The average autotomy score in the capping group was observed to be much lower than that of the intervention and control groups. At 30 days postneurectomy, the capping group displayed no obvious neuroma formation, while a bulbous neuroma was found in the nerve stumps of both the control and intervention groups. Quantitative real-time polymerase chain reaction and the Western blot analysis demonstrated that the expression of myelin-associated glycoprotein was substantially upregulated in the capping group; in contrast, the expression of NF-200 was significantly downregulated. The expression of myosin light chain was notably lower in the intervention group, but there was no significant difference when compared with the control group (p > 0.05). CONCLUSIONS The RhoA/ROCK signaling pathway has emerged as a critical player in the process of traumatic neuroma formation after neurectomy. It is possible that the nerve-capping technique could generate a "regenerative brake" based on the regulation of the RhoA/ROCK signaling pathway in this event. These findings may provide concrete evidence that could help develop new strategies for the management of painful neuromas.
Collapse
|
18
|
Mulherkar S, Tolias KF. RhoA-ROCK Signaling as a Therapeutic Target in Traumatic Brain Injury. Cells 2020; 9:E245. [PMID: 31963704 PMCID: PMC7016605 DOI: 10.3390/cells9010245] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. TBIs, which range in severity from mild to severe, occur when a traumatic event, such as a fall, a traffic accident, or a blow, causes the brain to move rapidly within the skull, resulting in damage. Long-term consequences of TBI can include motor and cognitive deficits and emotional disturbances that result in a reduced quality of life and work productivity. Recovery from TBI can be challenging due to a lack of effective treatment options for repairing TBI-induced neural damage and alleviating functional impairments. Central nervous system (CNS) injury and disease are known to induce the activation of the small GTPase RhoA and its downstream effector Rho kinase (ROCK). Activation of this signaling pathway promotes cell death and the retraction and loss of neural processes and synapses, which mediate information flow and storage in the brain. Thus, inhibiting RhoA-ROCK signaling has emerged as a promising approach for treating CNS disorders. In this review, we discuss targeting the RhoA-ROCK pathway as a therapeutic strategy for treating TBI and summarize the recent advances in the development of RhoA-ROCK inhibitors.
Collapse
Affiliation(s)
- Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA;
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
19
|
Shu L, Chen B, Chen B, Xu H, Wang G, Huang Y, Zhao Y, Gong H, Jiang M, Chen L, Liu X, Wang Y. Brain ischemic insult induces cofilin rod formation leading to synaptic dysfunction in neurons. J Cereb Blood Flow Metab 2019; 39:2181-2195. [PMID: 29932353 PMCID: PMC6827117 DOI: 10.1177/0271678x18785567] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Ischemic stroke not only induces neuron death in the infarct area but also structural and functional damage of the surviving neurons in the surrounding peri-infarct area. In the present study, we first identified cofilin rod, a pathological rod-like aggregation, formed in neurons of in vivo ischemic stroke animal model and induced neuronal impairment. Cofilin rods formed only on the ipsilateral side of the middle cerebral artery occlusion and reperfusion (MCAO-R) rat brain and showed the highest density in peri-infarct area. Our real-time live cell imaging, immunostaining and patch clamp studies showed that cofilin rod formation in neurons led to dendritic mitochondrial transportation failure, as well as impairment of synaptic structure and functions. Overexpression of LIM kinase or activation of its upstream regulator Rho, suppressed ischemia-induced cofilin rod formation and showed protective effect on synaptic function and structure impairment in both cultured neurons and MCAO-R rat model. In summary, our results demonstrate a novel mechanism of ischemic stroke-induced neuron injury in peri-infarct area and provide a potential target for the protection of neuronal structure and function against brain ischemia insult.
Collapse
Affiliation(s)
- Liang Shu
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ben Chen
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Chen
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Hai Xu
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoxiang Wang
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yian Huang
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingya Zhao
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Gong
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Jiang
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lidian Chen
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Collaborative Innovation Center for Rehabilitation Technology, TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Xu Liu
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- Department of Neurology, State Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Song X, He R, Han W, Li T, Xie L, Cheng L, Chen H, Xie M, Jiang L. Protective effects of the ROCK inhibitor fasudil against cognitive dysfunction following status epilepticus in male rats. J Neurosci Res 2018; 97:506-519. [PMID: 30421453 DOI: 10.1002/jnr.24355] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/30/2018] [Accepted: 10/17/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Xiaojie Song
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
- Department of Neurology Children’s Hospital of Chongqing Medical University Chongqing China
| | - Rong He
- Pediatric department University‐Town Hospital of Chongqing Medical University Chongqing China
| | - Wei Han
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
| | - Tianyi Li
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
- Department of Neurology Children’s Hospital of Chongqing Medical University Chongqing China
| | - Lingling Xie
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
- Department of Neurology Children’s Hospital of Chongqing Medical University Chongqing China
| | - Li Cheng
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
| | - Hengsheng Chen
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
| | - Mingdan Xie
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
| | - Li Jiang
- Pediatric Research Institute Children’s Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics Chongqing China
- Department of Neurology Children’s Hospital of Chongqing Medical University Chongqing China
| |
Collapse
|
21
|
ROCK inhibition in models of neurodegeneration and its potential for clinical translation. Pharmacol Ther 2018; 189:1-21. [DOI: 10.1016/j.pharmthera.2018.03.008] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Suppressive effect of Rho-kinase inhibitors Y-27632 and fasudil on spike-and-wave discharges in genetic absence epilepsy rats from Strasbourg (GAERS). Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1275-1283. [PMID: 30073384 DOI: 10.1007/s00210-018-1546-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022]
Abstract
Rho/Rho-kinase (ROCK) signaling contributes to neuroinflammation, epileptogenesis, and seizures in convulsive-type epilepsies. However, this pathway has not been investigated in absence epilepsy. We investigated RhoA activity in genetic absence epilepsy rats from Strasburg (GAERS) and the effects of ROCK inhibitors Y-27632 and fasudil on spike-and-wave discharges (SWDs) of GAERS. ROCK level and activity were measured by Western blot analysis in the brain areas involved in absence seizures (i.e., cortex and thalamus) and hippocampus. Male GAERS were stereotaxically implanted with bilateral cortical electrodes for electroencephalogram (EEG) recordings and/or guide cannula into the right ventricle. ROCK inhibitors were administered by intraperitoneal injection (1-10 mg/kg for Y-27632 or fasudil) or intracerebroventricular injection (7-20 nmol/5 μl for Y-27632 or 10-100 nmol/5 μl for fasudil). EEG was recorded under freely moving conditions. Compared with Wistar rats, GAERS exhibited increased RhoA activity in the somatosensory cortex but not in the thalamus or hippocampus. The single systemic administration of Y-27632 and fasudil partially suppressed the duration and frequency of absence seizure, respectively. However, local brain administration caused a widespread suppressive effect on the total seizure duration, number of seizures, and the average individual seizure length. In summary, Rho/ROCK signaling may be involved in the pathophysiology of absence epilepsy. Furthermore, ROCK inhibitors can control the expression of absence seizure in GAERS, thus indicating that Y-27632 and fasudil have the potential to be used as novel anti-absence drugs.
Collapse
|
23
|
Liu A, Wu Y, Li L, Wang Y. The roles of interleukin‐1 and RhoA signaling pathway in rat epilepsy model treated with low‐frequency electrical stimulation. J Cell Biochem 2017; 119:2535-2544. [DOI: 10.1002/jcb.26415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/21/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Ai‐Hua Liu
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Ya‐Ting Wu
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Li‐Ping Li
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| | - Yu‐Ping Wang
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingP.R. China
- Beijing Intensive Epilepsy CenterBeijing Key Laboratory of NeuromodulationBeijingP.R. China
| |
Collapse
|
24
|
Liu DZ, Waldau B, Ander BP, Zhan X, Stamova B, Jickling GC, Lyeth BG, Sharp FR. Inhibition of Src family kinases improves cognitive function after intraventricular hemorrhage or intraventricular thrombin. J Cereb Blood Flow Metab 2017; 37:2359-2367. [PMID: 27624844 PMCID: PMC5531336 DOI: 10.1177/0271678x16666291] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Intraventricular hemorrhage causes spatial memory loss, but the mechanism remains unknown. Our recent studies demonstrated that traumatic brain injury activates Src family kinases, which cause spatial memory loss. To test whether the spatial memory loss was due to blood in the ventricles, which activated Src family kinases, we infused autologous whole blood or thrombin into the lateral ventricles of adult rats to model non-traumatic intraventricular hemorrhage. Hippocampal neuron loss was examined 1 day to 5 weeks later. Spatial memory function was assessed 29 to 33 days later using the Morris water maze. Five weeks after the ventricular injections of blood or thrombin, there was death of most hippocampal neurons and significant memory deficits compared with sham operated controls. These data show that intraventricular thrombin is sufficient to kill hippocampal neurons and produce spatial memory loss. In addition, systemic administration of the non-specific Src family kinase inhibitor PP2 or intraventricular injection of siRNA-Fyn, a Src family kinase family member, prevented hippocampal neuronal loss and spatial memory deficits following intraventricular hemorrhage. The data support the conclusions that thrombin mediates the hippocampal neuronal cell death and spatial memory deficits produced by intraventricular blood and that these can be blocked by non-specific inhibition of Src family kinases or by inhibiting Fyn.
Collapse
Affiliation(s)
- Da Zhi Liu
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| | - Ben Waldau
- 2 Department of Neurological Surgery, University of California at Davis, Davis, USA
| | - Bradley P Ander
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| | - Xinhua Zhan
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| | - Boryana Stamova
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| | - Glen C Jickling
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| | - Bruce G Lyeth
- 2 Department of Neurological Surgery, University of California at Davis, Davis, USA
| | - Frank R Sharp
- 1 Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, USA
| |
Collapse
|
25
|
Niego B, Lee N, Larsson P, De Silva TM, Au AEL, McCutcheon F, Medcalf RL. Selective inhibition of brain endothelial Rho-kinase-2 provides optimal protection of an in vitro blood-brain barrier from tissue-type plasminogen activator and plasmin. PLoS One 2017; 12:e0177332. [PMID: 28510599 PMCID: PMC5433693 DOI: 10.1371/journal.pone.0177332] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022] Open
Abstract
Rho-kinase (ROCK) inhibition, broadly utilised in cardiovascular disease, may protect the blood-brain barrier (BBB) during thrombolysis from rt-PA-induced damage. While the use of nonselective ROCK inhibitors like fasudil together with rt-PA may be hindered by possible hypotensive side-effects and inadequate capacity to block detrimental rt-PA activity in brain endothelial cells (BECs), selective ROCK-2 inhibition may overcome these limitations. Here, we examined ROCK-2 expression in major brain cells and compared the ability of fasudil and KD025, a selective ROCK-2 inhibitor, to attenuate rt-PA-induced BBB impairment in an in vitro human model. ROCK-2 was highly expressed relative to ROCK-1 in all human and mouse brain cell types and particularly enriched in rodent brain endothelial cells and astrocytes compared to neurons. KD025 was more potent than fasudil in attenuation of rt-PA- and plasminogen-induced BBB permeation under normoxia, but especially under stroke-like conditions. Importantly, only KD025, but not fasudil, was able to block rt-PA-dependent permeability increases, morphology changes and tight junction degradation in isolated BECs. Selective ROCK-2 inhibition further diminished rt-PA-triggered myosin phosphorylation, shape alterations and matrix metalloprotease activation in astrocytes. These findings highlight ROCK-2 as the key isoform driving BBB impairment and brain endothelial damage by rt-PA and the potential of KD025 to optimally protect the BBB during thrombolysis.
Collapse
Affiliation(s)
- Be’eri Niego
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| | - Natasha Lee
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Pia Larsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - T. Michael De Silva
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Amanda E-Ling Au
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Fiona McCutcheon
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Chang J, Yao X, Zou H, Wang L, Lu Y, Zhang Q, Zhao H. BDNF/PI3K/Akt and Nogo-A/RhoA/ROCK signaling pathways contribute to neurorestorative effect of Houshiheisan against cerebral ischemia injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:1032-1042. [PMID: 27833029 DOI: 10.1016/j.jep.2016.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/06/2016] [Accepted: 11/04/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Houshiheisan (HSHS), a classic traditional medicine prescription, has notable effects on patients with stroke AIM OF THE STUDY: To investigate the neurorestorative effects of HSHS on ischemic stroke and explore its mode of action. MATERIALS AND METHODS Focal cerebral ischemia models were induced by permanent middle cerebral artery occlusion (pMCAO). Male Sprague-Dawley (SD) rats were randomly divided into 5 experimental groups: sham vehicle, ischemia vehicle, pMCAO+HSHS at 5.1, 10.2g/kg, and pMCAO+Ginaton 0.028g/kg. HSHS or Ginaton was administrated 6h after pMCAO onset. Neurological function was assessed and then rats were sacrificed 7 days after MCAO. Cerebral ischemic injury was evaluated by hematoxylin and eosin (HE) staining and Neuronal nuclear antigen (NeuN) immunofluorescence analysis. The levels of BDNF were detected by enzyme linked immunosorbent assay (ELISA), and the expression levels of PI3K/Akt and Nogo-A/RhoA/ROCK2 signaling pathway were detected by western blot and quantitative real-time PCR (qRT-PCR). RESULTS Compared with those results of pMCAO group, HSHS 5.1 and HSHS 10.2 groups markedly improved neurological function, alleviated pathological damage, promoted the neuronal survival, increased the expression of BDNF, PI3K, Akt, in protein and mRNA, decreased the expression of Nogo-A, NgR, RhoA and ROCK2 in protein and mRNA 7 days after pMCAO. CONCLUSIONS The findings demonstrate that HSHS had significant therapeutic effects on ischemic stroke and it perhaps worked through the activation of BDNF/PI3K/Akt and down-regulation of Nogo-A/RhoA/ROCK signaling pathways.
Collapse
Affiliation(s)
- Jiahui Chang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China
| | - Xiaoquan Yao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China
| | - Haiyan Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China
| | - Yue Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China
| | - Qiuxia Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China.
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing 100069, China.
| |
Collapse
|
27
|
Combined application of Rho-ROCKII and GSK-3β inhibitors exerts an improved protective effect on axonal regeneration in rats with spinal cord injury. Mol Med Rep 2016; 14:5180-5188. [PMID: 27840930 PMCID: PMC5355718 DOI: 10.3892/mmr.2016.5918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 10/17/2016] [Indexed: 12/03/2022] Open
Abstract
Previous studies have reported that the Rho-associated coiled-coil containing protein kinase 2 (ROCKII) and glycogen synthase kinase-3β (GSK)-3β signaling pathways are involved in axonal regeneration. The present study investigated the effects of the combined application of Y27632 (a ROCKII inhibitor) and 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8; a GSK-3β inhibitor) on neurite outgrowth and functional recovery in rats with spinal cord injury (SCI). A total of 90 female Sprague-Dawley rats were randomly allocated into six groups, and the SCI rats received daily administration of 1.6 mg/kg Y27632 for 2 weeks and/or 1 mg/kg TDZD-8 for 3 weeks via a catheter. Cellular apoptosis in the injured spinal cords was measured at each time point using a terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. The expression levels of growth-associated protein-43 (GAP-43) were determined by immunohistochemical staining. In addition, an anterograde tracer was used to analyze axonal regeneration, the Basso Beattie Bresnahan locomotor rating scale (BBB) was analyzed, and the somatosensory evoked potential (SEP) test was conducted. The results demonstrated that SCI upregulated the number of apoptotic cells, increased GAP-43 expression and enhanced the latent periods of SEP, as compared with in mice that underwent a sham operation. Furthermore, SCI decreased the BBB scores and the SEP amplitudes. These injuries in the spinal cord were reduced following treatment with Y27632, TDZD-8, or their combined application, as detected by decreased apoptosis, the induction of axonal regeneration, and the promotion of functional recovery of the lower limbs. Although the BBB scores, and SEP amplitudes and latent periods were not significantly different among the three drug treatment groups, the combined application of Y27632 and TDZD-8 resulted in stronger axonal regenerative potency and a greater protective effect on secondary SCI. These results indicated that the combined application of Y27632 and TDZD-8 may more effectively protect against secondary SCI by inhibiting cellular apoptosis, enhancing GAP-43 expression and promoting neurite outgrowth in SCI rats, compared with Y27632 or TDZD-8 alone.
Collapse
|
28
|
Wang Y, Liu Q, Xu Y, Zhang Y, Lv Y, Tan Y, Jiang N, Cao G, Ma X, Wang J, Cao Z, Yu B, Kou J. Ginsenoside Rg1 Protects against Oxidative Stress-induced Neuronal Apoptosis through Myosin IIA-actin Related Cytoskeletal Reorganization. Int J Biol Sci 2016; 12:1341-1356. [PMID: 27877086 PMCID: PMC5118780 DOI: 10.7150/ijbs.15992] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/08/2016] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress-induced cytoskeletal dysfunction of neurons has been implicated as a crucial cause of cell apoptosis or death in the central nervous system (CNS) diseases, such as neurodegenerative and psychiatric diseases. The application of neuroprotectants rescuing the neurons from cytoskeletal damage and apoptosis can be a potential treatment for these CNS diseases. Ginsenoside Rg1 (Rg1), one of the major active components of ginseng, has been reported possessing notable neuroprotective activities. However, there is rare report about its effect on cytoskeleton and its undergoing mechanism. The current study is to reveal the regulatory effects of Rg1 on cytoskeletal and morphological lesion in oxidative stress-induced neuronal apoptosis. The results demonstrated that pre-treatment with Rg1 (0.1-10 μM) attenuated hydrogen peroxide (H2O2)-induced neuronal apoptosis and oxidative stress through reducing the intracellular reactive oxygen species (ROS) production and methane dicarboxylic aldehyde (MDA) level. The Rg1 treatment also abolished H2O2-induced morphological changes, including cell rounding, membrane blebbing, neurite retraction and nuclei condensation, which were generated by myosin IIA-actin interaction. These effects were mediated via the down-regulation of caspase-3, ROCK1 (Rho-associated kinase1) activation and myosin light chain (MLC, Ser-19) phosphorylation. Furthermore, inhibiting myosin II activity with blebbistatin partly blocked the neuroprotective effects of Rg1. The computer-aided homology modelling revealed that Rg1 preferentially positioned in the actin binding cleft of myosin IIA and might block the binding of myosin IIA to actin filaments. Accordingly, the neuroprotective mechanism of Rg1 is related to the activity that inhibits myosin IIA-actin interaction and the caspase-3/ROCK1/MLC signaling pathway. These findings put some insights into the unique neuroprotective properties of Rg1 associated with the regulation of myosin IIA-actin cytoskeletal structure under oxidative stress and provide experimental evidence for Rg1 in CNS diseases.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Qian Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002, China.; School of Dentistry, Cardiff Institute of Tissue Engineering and Repair, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Yingqiong Xu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Yanni Lv
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang, 330006, China
| | - Yisha Tan
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Nan Jiang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Guosheng Cao
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Xiaonan Ma
- Cellular and Molecular Biology Center, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Jingrong Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Zhengyu Cao
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Boyang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| | - Junping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, China
| |
Collapse
|
29
|
Hollanders K, Hove IV, Sergeys J, Bergen TV, Lefevere E, Kindt N, Castermans K, Vandewalle E, van Pelt J, Moons L, Stalmans I. AMA0428, A Potent Rock Inhibitor, Attenuates Early and Late Experimental Diabetic Retinopathy. Curr Eye Res 2016; 42:260-272. [PMID: 27399806 DOI: 10.1080/02713683.2016.1183030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE Diabetic retinopathy (DR) is characterized by an early stage of inflammation and vessel leakage, and an advanced vasoproliferative stage. Also, neurodegeneration might play an important role in disease pathogenesis. The aim of this study was to investigate the effect of the Rho kinase (ROCK) inhibitor, AMA0428, on these processes. METHODS The response to ROCK inhibition by AMA0428 (1 µg) was studied in vivo using the murine model for streptozotocin (STZ)-induced diabetes, focusing on early non-proliferative DR features and the oxygen-induced retinopathy (OIR) model to investigate proliferative DR. Intravitreal (IVT) administration of AMA0428 was compared with murine anti-VEGF-R2 antibody (DC101, 6.2 µg) and placebo (H2O/PEG; 1C8). Outcome was assessed by analyzing leukostasis using fluorescein isothiocyanate coupled concanavalin A (FITC-ConA) and vessel leakage (bovine serum albumin conjugated with fluorescein isothiocyanate; FITC-BSA)/neovascularization and neurodegeneration by immunohistological approaches (hematoxylin and eosin (H&E), terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL), Brn3a). ELISA and Western blotting were employed to unravel the consequences of ROCK inhibition (1 µM AMA0428) on myosin phosphatase target protein (MYPT)-1 phosphorylation, endothelial nitric oxide synthase (eNOS) phosphorylation, and vascular endothelial growth factor (VEGF) levels in retinas of diabetic mice, on NF-κβ activity and ICAM-1 expression in endothelial cells (ECs). RESULTS In vivo, AMA0428 significantly reduced vessel leakage and neovascularization, respectively, in the STZ and OIR model, comparable to DC101 therapy. Additionally, the ROCK inhibitor decreased neurodegeneration in both models and inhibited leukostasis by 30% (p < 0.05) in the STZ model (p < 0.05), while DC101 had no positive effect on the outcome of these latter processes. ROCK activity was upregulated in the diabetic retina and AMA0428 administration resulted in decreased phospho-MYPT-1, enhanced phospho-eNOS, and reduced VEGF levels. In vitro, AMA0428 interfered with NF-κβ activity, thereby inhibiting ICAM-1 expression in ECs. CONCLUSIONS Targeting ROCK with AMA0428 effectively attenuated outcome in an early DR model (STZ) and a late vasoproliferative retinopathy model (OIR). These findings make AMA0428 a promising candidate with an additional anti-inflammatory and neuroprotective benefit for DR patients, as compared with anti-VEGF treatment.
Collapse
Affiliation(s)
- Karolien Hollanders
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,b Department of Ophthalmology , University Hospitals Ghent , Ghent , Belgium
| | - Inge Van Hove
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Jurgen Sergeys
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Tine Van Bergen
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium
| | - Evy Lefevere
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | | | | | - Evelien Vandewalle
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,e Department of Ophthalmology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| | - Jos van Pelt
- f Department of Hepatology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| | - Lieve Moons
- c Department of Biology, KU Leuven-University of Leuven , Leuven , Belgium
| | - Ingeborg Stalmans
- a Department of Ophthalmology , KU Leuven-University of Leuven , Leuven , Belgium.,e Department of Ophthalmology , University Hospitals Leuven, KU Leuven-University of Leuven , Leuven , Belgium
| |
Collapse
|
30
|
Henneberger C. Does rapid and physiological astrocyte-neuron signalling amplify epileptic activity? J Physiol 2016; 595:1917-1927. [PMID: 27106234 DOI: 10.1113/jp271958] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022] Open
Abstract
The hippocampus is a key brain region in the pathophysiology of mesial temporal lobe epilepsy. Long-term changes of its architecture and function on the network and cellular level are well documented in epilepsy. Astrocytes can control many aspects of neuronal function and their long-term alterations over weeks, months and years play an important role in epilepsy. However, a pathophysiological transformation of astrocytes does not seem to be required for astrocytes to contribute to epileptic activity. Some of the properties of physiological astrocyte-neuron communication could allow these cells to exacerbate or synchronize neuronal firing on shorter time scales of milliseconds to minutes. Therefore, these astrocyte-neuron interactions are increasingly recognized as potential contributors to epileptic activity. Fast and reciprocal communication between astrocytes and neurons is enabled by a diverse set of mechanisms that could both amplify and counteract epileptic activity. They may thus promote or cause development of epileptic activity or inhibit it. Mechanisms of astrocyte-neuron interactions that can quickly increase network excitability involve, for example, astrocyte Ca2+ and Na+ signalling, K+ buffering, gap junction coupling and metabolism. However, rapid changes of astrocyte neurotransmitter uptake and morphology may also underlie or support development of network hyperexcitability. The temporal characteristics of these interactions, their ability to synchronize neuronal activity and their net effect on network activity will determine their contribution to the emergence or maintenance of epileptic activity.
Collapse
Affiliation(s)
- Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,UCL Institute of Neurology, London, UK
| |
Collapse
|
31
|
Bye N, Christie KJ, Turbic A, Basrai HS, Turnley AM. Rho kinase inhibition following traumatic brain injury in mice promotes functional improvement and acute neuron survival but has little effect on neurogenesis, glial responses or neuroinflammation. Exp Neurol 2016; 279:86-95. [DOI: 10.1016/j.expneurol.2016.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/27/2022]
|
32
|
Leclerc JL, Lampert AS, Diller MA, Doré S. PGE2-EP3 signaling exacerbates intracerebral hemorrhage outcomes in 24-mo-old mice. Am J Physiol Heart Circ Physiol 2016; 310:H1725-34. [PMID: 27084388 DOI: 10.1152/ajpheart.00638.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 04/07/2016] [Indexed: 01/15/2023]
Abstract
With the population aging at an accelerated rate, the prevalence of stroke and financial burden of stroke-related health care costs are expected to continue to increase. Intracerebral hemorrhage (ICH) is a devastating stroke subtype more commonly affecting the elderly population, who display increased mortality and worse functional outcomes compared with younger patients. This study aimed to investigate the contribution of the prostaglandin E2 (PGE2) E prostanoid (EP) receptor subtype 3 in modulating anatomical outcomes and functional recovery following ICH in 24-mo-old mice. EP3 is the most abundant EP receptor in the brain and we have previously shown that signaling through the PGE2-EP3 axis exacerbates ICH outcomes in young mice. Here, we show that EP3 receptor deletion results in 17.9 ± 6.1% less ICH-induced brain injury (P < 0.05) and improves neurological functional recovery (P < 0.01), as identified by lower neurological deficit scores, decreased resting time, and more gross and fine motor movements. Immunohistological staining was performed to investigate possible mechanisms of EP3-mediated neurotoxicity. Identified mechanisms include reduced blood accumulation and modulation of angiogenic and astroglial responses. Using this aged cohort of mice, we have confirmed and extended our previous results in young mice demonstrating the deleterious role of the PGE2-EP3 signaling axis in modulating brain injury and functional recovery after ICH, further supporting the notion of the EP3 receptor as a putative therapeutic avenue for the treatment of ICH.
Collapse
Affiliation(s)
- Jenna L Leclerc
- Department of Anesthesiology, University of Florida, Gainesville, Florida; Department of Neuroscience, University of Florida, Gainesville, Forida; and
| | - Andrew S Lampert
- Department of Anesthesiology, University of Florida, Gainesville, Florida
| | - Matthew A Diller
- Department of Anesthesiology, University of Florida, Gainesville, Florida
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, Florida; Department of Neuroscience, University of Florida, Gainesville, Forida; and Departments of Neurology, Psychiatry, Psychology and Pharmaceutics, University of Florida, Gainesville, Florida
| |
Collapse
|
33
|
Wang X, Wang B, Li Z, Zhu G, Heng L, Zhu X, Yang Q, Ma J, Gao G. Neuroprotection effect of Y-27632 against H2O2-induced cell apoptosis of primary cultured cortical neurons. RSC Adv 2016. [DOI: 10.1039/c6ra03284b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Y-27632 protects the cortical neurons from H2O2-induced apoptosis by inhibiting oxidative stress and activation of JNK and p38 MAPKs pathways.
Collapse
Affiliation(s)
- Xingqin Wang
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Bao Wang
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Zhenzhen Li
- Department of Burns and Cutaneous Surgery
- Xijing Hospital
- Fourth Military Medical University
- Xi'an 710032
- China
| | - Gang Zhu
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Lijun Heng
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Xianke Zhu
- Department of Orthopedics
- No. 150 Central Hospital of PLA
- Luoyang
- China
| | - Qian Yang
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Jie Ma
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| | - Guodong Gao
- Department of Neurosurgery
- Tangdu Hospital
- Fourth Military Medical University
- Xi'an 710038
- China
| |
Collapse
|
34
|
Newell-Litwa KA, Horwitz R, Lamers ML. Non-muscle myosin II in disease: mechanisms and therapeutic opportunities. Dis Model Mech 2015; 8:1495-515. [PMID: 26542704 PMCID: PMC4728321 DOI: 10.1242/dmm.022103] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The actin motor protein non-muscle myosin II (NMII) acts as a master regulator of cell morphology, with a role in several essential cellular processes, including cell migration and post-synaptic dendritic spine plasticity in neurons. NMII also generates forces that alter biochemical signaling, by driving changes in interactions between actin-associated proteins that can ultimately regulate gene transcription. In addition to its roles in normal cellular physiology, NMII has recently emerged as a critical regulator of diverse, genetically complex diseases, including neuronal disorders, cancers and vascular disease. In the context of these disorders, NMII regulatory pathways can be directly mutated or indirectly altered by disease-causing mutations. NMII regulatory pathway genes are also increasingly found in disease-associated copy-number variants, particularly in neuronal disorders such as autism and schizophrenia. Furthermore, manipulation of NMII-mediated contractility regulates stem cell pluripotency and differentiation, thus highlighting the key role of NMII-based pharmaceuticals in the clinical success of stem cell therapies. In this Review, we discuss the emerging role of NMII activity and its regulation by kinases and microRNAs in the pathogenesis and prognosis of a diverse range of diseases, including neuronal disorders, cancer and vascular disease. We also address promising clinical applications and limitations of NMII-based inhibitors in the treatment of these diseases and the development of stem-cell-based therapies.
Collapse
Affiliation(s)
- Karen A Newell-Litwa
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rick Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Marcelo L Lamers
- Department of Morphological Sciences, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-010, Brazil
| |
Collapse
|
35
|
Kourdougli N, Varpula S, Chazal G, Rivera C. Detrimental effect of post Status Epilepticus treatment with ROCK inhibitor Y-27632 in a pilocarpine model of temporal lobe epilepsy. Front Cell Neurosci 2015; 9:413. [PMID: 26557054 PMCID: PMC4615811 DOI: 10.3389/fncel.2015.00413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/28/2015] [Indexed: 01/18/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults where 20-30% of the patients are refractory to currently available anti-epileptic drugs. The RhoA/Rho-kinase signaling pathway activation has been involved in inflammatory responses, neurite outgrowth and neuronal death under pathological conditions such as epileptic insults. Acute preventive administration of ROCK inhibitor has been reported to have beneficial outcomes in Status Epilepticus (SE) epilepsy. In the present study, we evaluate the effect of chronic post SE treatment with the ROCK inhibitor Y-27632 in a rat pilocarpine model of TLE. We used chronic i.p. injections of Y-27632 for 5 days in 6 week old control rats or rats subjected to pilocarpine treatment as a model of TLE. Surprisingly, our findings demonstrate that a systemic administration of Y-27632 in pilocarpine-treated rats increases neuronal death in the CA3 region and ectopic recurrent mossy fiber sprouting (rMFS) in the dentate gyrus of the hippocampal formation. Interestingly, we found that chronic treatment with Y-27632 exacerbates the down-regulation and pathological distribution of the K(+)-Cl(-) cotransporter KCC2, thus providing a putative mechanism for post SE induced neuronal death. The involvement of astrogliosis in this mechanism appears to be intricate as ROCK inhibition reduces reactive astrogliosis in pilocarpine rats. Conversely, in control rats, chronic Y-27632 treatment increases astrogliosis. Together, our findings suggest that Y-27632 has a detrimental effect when chronically used post SE in a rat pilocarpine model of TLE.
Collapse
Affiliation(s)
- Nazim Kourdougli
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
| | - Saara Varpula
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| | - Genevieve Chazal
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
| | - Claudio Rivera
- INSERM Unité 901, INMEDMarseille, France
- Aix-Marseille Université, UMR S901Marseille, France
- Neuroscience Center, University of HelsinkiHelsinki, Finland
| |
Collapse
|
36
|
Leclerc JL, Lampert AS, Diller MA, Doré S. Genetic deletion of the prostaglandin E2 E prostanoid receptor subtype 3 improves anatomical and functional outcomes after intracerebral hemorrhage. Eur J Neurosci 2015; 41:1381-91. [PMID: 25847406 PMCID: PMC4696550 DOI: 10.1111/ejn.12909] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/05/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhage (ICH) is a stroke subtype associated with high mortality and morbidity. Following ICH, excitotoxicity and inflammation significantly contribute to secondary brain injury and poor outcomes. Prostaglandin E2 (PGE2 ) levels rise locally with insult to the nervous system, and PGE2 is known to modulate these processes mainly through its E prostanoid (EP) receptors, EP1-4. EP receptor subtype 3 (EP3) is the most abundant EP receptor in the brain and we have previously shown that signaling through the PGE2 -EP3 axis exacerbates excitotoxicity and ischemic stroke outcomes. This study aimed to investigate the contribution of this pathway in modulating anatomical outcomes and functional recovery following ICH. Genetic deletion of EP3 resulted in 48.2 ± 7.3% less ICH-induced brain injury (P < 0.005) and improved functional recovery (P < 0.05), as identified by neurological deficit scoring. To start investigating the mechanisms involved in neuroprotection with impaired PGE2 -EP3 signaling, histological staining was performed to evaluate blood and ferric iron accumulation, neuroinflammation, blood-brain barrier dysfunction, and peripheral neutrophil infiltration. After ICH, EP3 knockout mice demonstrated 49.5 ± 8.8% and 42.8 ± 13.1% less blood (P < 0.01) and ferric iron (P < 0.05), respectively. Furthermore, EP3 knockout mice had significantly reduced astrogliosis, microglial activation, blood-brain barrier breakdown, and neutrophil infiltration. Collectively, these results suggest an injurious role for the PGE2 -EP3 signaling axis in modulating brain injury, inflammation, and neurological functional recovery after ICH. Modulation of the PGE2 -EP3 signaling axis may represent a putative therapeutic avenue for the treatment of ICH.
Collapse
Affiliation(s)
- Jenna L Leclerc
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Andrew S Lampert
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | - Matthew A Diller
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
- Departments of Neurology, Psychiatry, and Pharmaceutics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
37
|
Contribution of protease-activated receptor 1 in status epilepticus-induced epileptogenesis. Neurobiol Dis 2015; 78:68-76. [PMID: 25843668 DOI: 10.1016/j.nbd.2015.03.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/28/2015] [Accepted: 03/26/2015] [Indexed: 12/28/2022] Open
Abstract
Clinical observations and studies on different animal models of acquired epilepsy consistently demonstrate that blood-brain barrier (BBB) leakage can be an important risk factor for developing recurrent seizures. However, the involved signaling pathways remain largely unclear. Given the important role of thrombin and its major receptor in the brain, protease-activated receptor 1 (PAR1), in the pathophysiology of neurological injury, we hypothesized that PAR1 may contribute to status epilepticus (SE)-induced epileptogenesis and that its inhibition shortly after SE will have neuroprotective and antiepileptogenic effects. Adult rats subjected to lithium-pilocarpine SE were administrated with SCH79797 (a PAR1 selective antagonist) after SE termination. Thrombin and PAR1 levels and neuronal cell survival were evaluated 48h following SE. The effect of PAR1 inhibition on animal survival, interictal spikes (IIS) and electrographic seizures during the first two weeks after SE and behavioral seizures during the chronic period was evaluated. SE resulted in a high mortality rate and incidence of IIS and seizures in the surviving animals. There was a marked increase in thrombin, decrease in PAR1 immunoreactivity and hippocampal cell loss in the SE-treated rats. Inhibition of PAR1 following SE resulted in a decrease in mortality and morbidity, increase in neuronal cell survival in the hippocampus and suppression of IIS, electrographic and behavioral seizures following SE. These data suggest that the PAR1 signaling pathway contributes to epileptogenesis following SE. Because breakdown of the BBB occurs frequently in brain injuries, PAR1 inhibition may have beneficial effects in a variety of acquired injuries leading to epilepsy.
Collapse
|
38
|
Stankiewicz TR, Ramaswami SA, Bouchard RJ, Aktories K, Linseman DA. Neuronal apoptosis induced by selective inhibition of Rac GTPase versus global suppression of Rho family GTPases is mediated by alterations in distinct mitogen-activated protein kinase signaling cascades. J Biol Chem 2015; 290:9363-76. [PMID: 25666619 DOI: 10.1074/jbc.m114.575217] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Indexed: 12/11/2022] Open
Abstract
Rho family GTPases play integral roles in neuronal differentiation and survival. We have shown previously that Clostridium difficile toxin B (ToxB), an inhibitor of RhoA, Rac1, and Cdc42, induces apoptosis of cerebellar granule neurons (CGNs). In this study, we compared the effects of ToxB to a selective inhibitor of the Rac-specific guanine nucleotide exchange factors Tiam1 and Trio (NSC23766). In a manner similar to ToxB, selective inhibition of Rac induces CGN apoptosis associated with enhanced caspase-3 activation and reduced phosphorylation of the Rac effector p21-activated kinase. In contrast to ToxB, caspase inhibitors do not protect CGNs from targeted inhibition of Rac. Also dissimilar to ToxB, selective inhibition of Rac does not inhibit MEK1/2/ERK1/2 or activate JNK/c-Jun. Instead, targeted inhibition of Rac suppresses distinct MEK5/ERK5, p90Rsk, and Akt-dependent signaling cascades known to regulate the localization and expression of the Bcl-2 homology 3 domain-only protein Bad. Adenoviral expression of a constitutively active mutant of MEK5 is sufficient to attenuate neuronal cell death induced by selective inhibition of Rac with NSC23766 but not apoptosis induced by global inhibition of Rho GTPases with ToxB. Collectively, these data demonstrate that global suppression of Rho family GTPases with ToxB causes a loss of MEK1/2/ERK1/2 signaling and activation of JNK/c-Jun, resulting in diminished degradation and enhanced transcription of the Bcl-2 homology 3 domain-only protein Bim. In contrast, selective inhibition of Rac induces CGN apoptosis by repressing unique MEK5/ERK5, p90Rsk, and Akt-dependent prosurvival pathways, ultimately leading to enhanced expression, dephosphorylation, and mitochondrial localization of proapoptotic Bad.
Collapse
Affiliation(s)
- Trisha R Stankiewicz
- From the Research Service, Veterans Affairs Medical Center, Denver, Colorado 80220, the Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, Colorado 80208
| | - Sai Anandi Ramaswami
- the Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, Colorado 80208
| | - Ron J Bouchard
- From the Research Service, Veterans Affairs Medical Center, Denver, Colorado 80220
| | - Klaus Aktories
- the Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, 79104 Freiburg, Germany, and
| | - Daniel A Linseman
- From the Research Service, Veterans Affairs Medical Center, Denver, Colorado 80220, the Department of Biological Sciences and Eleanor Roosevelt Institute, University of Denver, Denver, Colorado 80208, the Division of Clinical Pharmacology and Toxicology, Department of Medicine and Neuroscience Program, University of Colorado Denver, Aurora, Colorado 80045
| |
Collapse
|
39
|
Günther R, Saal KA, Suhr M, Scheer D, Koch JC, Bähr M, Lingor P, Tönges L. The rho kinase inhibitor Y-27632 improves motor performance in male SOD1(G93A) mice. Front Neurosci 2014; 8:304. [PMID: 25339858 PMCID: PMC4187656 DOI: 10.3389/fnins.2014.00304] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/09/2014] [Indexed: 12/11/2022] Open
Abstract
Disease progression in amyotrophic lateral sclerosis (ALS) is characterized by degeneration of motoneurons and their axons which results in a progressive muscle weakness and ultimately death from respiratory failure. The only approved drug, riluzole, lacks clinical efficacy so that more potent treatment options are needed. We have identified rho kinase (ROCK) as a target, which can be manipulated to beneficially influence disease progression in models of ALS. Here, we examined the therapeutic potential of the ROCK inhibitor Y-27632 in both an in vitro and in an in vivo paradigm of motoneuron disease. Application of Y-27632 to primary motoneurons in vitro increased survival and promoted neurite outgrowth. In vivo, SOD1G93A mice were orally treated with 2 or 30 mg/kg body weight of Y-27632. The 2 mg/kg group did not benefit from Y-27632 treatment, whereas treatment with 30 mg/kg resulted in improved motor function in male mice. Female mice showed only limited improvement and overall survival was not modified in both 2 and 30 mg/kg Y-27632 groups. In conclusion, we provide evidence that inhibition of ROCK by Y-27632 is neuroprotective in vitro but has limited beneficial effects in vivo being restricted to male mice. Therefore, the evaluation of ROCK inhibitors in preclinical models of ALS should always take gender differences into account.
Collapse
Affiliation(s)
- René Günther
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Kim-Ann Saal
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Martin Suhr
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - David Scheer
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medicine Göttingen Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| | - Lars Tönges
- Department of Neurology, University Medicine Göttingen Göttingen, Germany ; Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171-DFG Research Center 103 (CNMPB) Göttingen, Germany
| |
Collapse
|
40
|
Koch JC, Tönges L, Barski E, Michel U, Bähr M, Lingor P. ROCK2 is a major regulator of axonal degeneration, neuronal death and axonal regeneration in the CNS. Cell Death Dis 2014; 5:e1225. [PMID: 24832597 PMCID: PMC4047920 DOI: 10.1038/cddis.2014.191] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 12/19/2022]
Abstract
The Rho/ROCK/LIMK pathway is central for the mediation of repulsive environmental signals in the central nervous system. Several studies using pharmacological Rho-associated protein kinase (ROCK) inhibitors have shown positive effects on neurite regeneration and suggest additional pro-survival effects in neurons. However, as none of these drugs is completely target specific, it remains unclear how these effects are mediated and whether ROCK is really the most relevant target of the pathway. To answer these questions, we generated adeno-associated viral vectors to specifically downregulate ROCK2 and LIM domain kinase (LIMK)-1 in rat retinal ganglion cells (RGCs) in vitro and in vivo. We show here that specific knockdown of ROCK2 and LIMK1 equally enhanced neurite outgrowth of RGCs on inhibitory substrates and both induced substantial neuronal regeneration over distances of more than 5 mm after rat optic nerve crush (ONC) in vivo. However, only knockdown of ROCK2 but not LIMK1 increased survival of RGCs after optic nerve axotomy. Moreover, knockdown of ROCK2 attenuated axonal degeneration of the proximal axon after ONC assessed by in vivo live imaging. Mechanistically, we demonstrate here that knockdown of ROCK2 resulted in decreased intraneuronal activity of calpain and caspase 3, whereas levels of pAkt and collapsin response mediator protein 2 and autophagic flux were increased. Taken together, our data characterize ROCK2 as a specific therapeutic target in neurodegenerative diseases and demonstrate new downstream effects of ROCK2 including axonal degeneration, apoptosis and autophagy.
Collapse
Affiliation(s)
- J C Koch
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - L Tönges
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - E Barski
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - U Michel
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - M Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - P Lingor
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
41
|
Liu DZ, Sharp FR, Van KC, Ander BP, Ghiasvand R, Zhan X, Stamova B, Jickling GC, Lyeth BG. Inhibition of SRC family kinases protects hippocampal neurons and improves cognitive function after traumatic brain injury. J Neurotrauma 2014; 31:1268-76. [PMID: 24428562 DOI: 10.1089/neu.2013.3250] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) is often associated with intracerebral and intraventricular hemorrhage. Thrombin is a neurotoxin generated at bleeding sites fater TBI and can lead to cell death and subsequent cognitive dysfunction via activation of Src family kinases (SFKs). We hypothesize that inhibiting SFKs can protect hippocampal neurons and improve cognitive memory function after TBI. To test these hypotheses, we show that moderate lateral fluid percussion (LFP) TBI in adult rats produces bleeding into the cerebrospinal fluid (CSF) in both lateral ventricles, which elevates oxyhemoglobin and thrombin levels in the CSF, activates the SFK family member Fyn, and increases Rho-kinase 1(ROCK1) expression. Systemic administration of the SFK inhibitor, PP2, immediately after moderate TBI blocks ROCK1 expression, protects hippocampal CA2/3 neurons, and improves spatial memory function. These data suggest the possibility that inhibiting SFKs after TBI might improve clinical outcomes.
Collapse
Affiliation(s)
- Da Zhi Liu
- 1 Department of Neurology and the M.I.N.D. Institute, University of California , Davis, Medical Center, Sacramento, California
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yang X, Liu Y, Liu C, Xie W, Huang E, Huang W, Wang J, Chen L, Wang H, Qiu P, Xu J, Zhang F, Wang H. Inhibition of ROCK2 expression protects against methamphetamine-induced neurotoxicity in PC12 cells. Brain Res 2013; 1533:16-25. [DOI: 10.1016/j.brainres.2013.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 07/17/2013] [Accepted: 08/03/2013] [Indexed: 10/26/2022]
|
43
|
Sudarov A, Gooden F, Tseng D, Gan WB, Ross ME. Lis1 controls dynamics of neuronal filopodia and spines to impact synaptogenesis and social behaviour. EMBO Mol Med 2013; 5:591-607. [PMID: 23483716 PMCID: PMC3628102 DOI: 10.1002/emmm.201202106] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 01/29/2013] [Accepted: 01/31/2013] [Indexed: 11/08/2022] Open
Abstract
LIS1 (PAFAH1B1) mutation can impair neuronal migration, causing lissencephaly in humans. LIS1 loss is associated with dynein protein motor dysfunction, and disrupts the actin cytoskeleton through disregulated RhoGTPases. Recently, LIS1 was implicated as an important protein-network interaction node with high-risk autism spectrum disorder genes expressed in the synapse. How LIS1 might participate in this disorder has not been investigated. We examined the role of LIS1 in synaptogenesis of post-migrational neurons and social behaviour in mice. Two-photon imaging of actin-rich dendritic filopodia and spines in vivo showed significant reductions in elimination and turnover rates of dendritic protrusions of layer V pyramidal neurons in adolescent Lis1+/− mice. Lis1+/− filopodia on immature hippocampal neurons in vitro exhibited reduced density, length and RhoA dependent impaired dynamics compared to Lis1+/+. Moreover, Lis1+/− adolescent mice exhibited deficits in social interaction. Lis1 inactivation restricted to the postnatal hippocampus resulted in similar deficits in dendritic protrusion density and social interactions. Thus, LIS1 plays prominently in dendritic filopodia dynamics and spine turnover implicating reduced dendritic spine plasticity as contributing to developmental autistic-like behaviour.
Collapse
Affiliation(s)
- Anamaria Sudarov
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, USA
| | | | | | | | | |
Collapse
|