1
|
Zhang ZZ, Nasir A, Li D, Khan S, Bai Q, Yuan F. Effect of dexmedetomidine on ncRNA and mRNA profiles of cerebral ischemia-reperfusion injury in transient middle cerebral artery occlusion rats model. Front Pharmacol 2024; 15:1437445. [PMID: 39170713 PMCID: PMC11335533 DOI: 10.3389/fphar.2024.1437445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Ischemic stroke poses a significant global health burden, with rapid revascularization treatments being crucial but often insufficient to mitigate ischemia-reperfusion (I/R) injury. Dexmedetomidine (DEX) has shown promise in reducing cerebral I/R injury, but its potential molecular mechanism, particularly its interaction with non-coding RNAs (ncRNAs), remains unclear. This study investigates DEX's therapeutic effect and potential molecular mechanisms in reducing cerebral I/R injury. A transient middle cerebral artery obstruction (tMACO) model was established to simulate cerebral I/R injury in adult rats. DEX was administered pre-ischemia and post-reperfusion. RNA sequencing and bioinformatic analyses were performed on the ischemic cerebral cortex to identify differentially expressed non-coding RNAs (ncRNAs) and mRNAs. The sequencing results showed 6,494 differentially expressed (DE) mRNA and 2698 DE circRNA between the sham and tMCAO (I/R) groups. Additionally, 1809 DE lncRNA, 763 DE mRNA, and 2795 DE circRNA were identified between the I/R group and tMCAO + DEX (I/R + DEX) groups. Gene ontology (GO) analysis indicated significant enrichment in multicellular biogenesis, plasma membrane components, and protein binding. KEGG analysis further highlighted the potential mechanism of DEX action in reducing cerebral I/R injury, with hub genes involved in inflammatory pathways. This study demonstrates DEX's efficacy in reducing cerebral I/R injury and offers insights into its brain-protective effects, especially in ischemic stroke. Further research is warranted to fully understand DEX's neuroprotective mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Zhen Zhen Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Abdul Nasir
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dong Li
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Suliman Khan
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Bai
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Medical Research Center, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Yuan
- Department of Anesthesiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Zhang D, Li D, Wang X, Sui Y, Ma F, Dai Y, Wang M, Qin W. Urine Proteomic Signatures of Mild Hypothermia Treatment in Cerebral Ischemia-Reperfusion Injury in Rats. Cell Mol Neurobiol 2024; 44:49. [PMID: 38836960 PMCID: PMC11153299 DOI: 10.1007/s10571-024-01483-4] [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: 01/25/2024] [Accepted: 05/27/2024] [Indexed: 06/06/2024]
Abstract
Mild hypothermia (MH) is an effective measure to alleviate cerebral ischemia-reperfusion (I/R) injury. However, the underlying biological mechanisms remain unclear. This study set out to investigate dynamic changes in urinary proteome due to MH in rats with cerebral I/R injury and explore the neuroprotective mechanisms of MH. A Pulsinelli's four-vessel occlusion (4-VO) rat model was used to mimic global cerebral I/R injury. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was employed to profile the urinary proteome of rats with/without MH (32 °C) treatment after I/R injury. Representative differentially expressed proteins (DEPs) associated with MH were validated by western blotting in hippocampus. A total of 597 urinary proteins were identified, among which 119 demonstrated significant changes associated with MH. Gene Ontology (GO) annotation of the DEPs revealed that MH significantly enriched in endopeptidase activity, inflammatory response, aging, response to oxidative stress and reactive oxygen species, blood coagulation, and cell adhesion. Notably, changes in 12 DEPs were significantly reversed by MH treatment. Among them, 8 differential urinary proteins were previously reported to be closely associated with brain disease, including NP, FZD1, B2M, EPCR, ATRN, MB, CA1and VPS4A. Two representative proteins (FZD1, B2M) were further validated by western blotting in the hippocampus and the results were shown to be consistent with urinary proteomic analysis. Overall, this study strengthens the idea that urinary proteome can sensitively reflect pathophysiological changes in the brain, and appears to be the first study to explore the neuroprotective effects of MH by urinary proteomic analysis. FZD1 and B2M may be involved in the most fundamental molecular biological mechanisms of MH neuroprotection.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Dapeng Li
- Department of Bone and Joint Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Xueting Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Yanyan Sui
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Fuguo Ma
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Yuting Dai
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Mingshan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China.
| | - Weiwei Qin
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China.
| |
Collapse
|
3
|
Zhang H, Feng Y, Si Y, Lu C, Wang J, Wang S, Li L, Xie W, Yue Z, Yong J, Dai S, Zhang L, Li X. Shank3 ameliorates neuronal injury after cerebral ischemia/reperfusion via inhibiting oxidative stress and inflammation. Redox Biol 2024; 69:102983. [PMID: 38064762 PMCID: PMC10755590 DOI: 10.1016/j.redox.2023.102983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/01/2024] Open
Abstract
Shank3, a key molecule related to the development and deterioration of autism, has recently been found to downregulate in the murine brain after ischemia/reperfusion (I/R). Despite this discovery, however, its effects on neuronal injury and the mechanism underlying the effects remain to be clarified. To address this, in this study, based on genetically modified mice models, we revealed that the expression of Shank3 showed a time-dependent change in murine hippocampal neurons after I/R, and that conditional knockout (cko) of Shank3 in neurons resulted in aggravated neuronal injuries. The protective effects of Shank3 against oxidative stress and inflammation after I/R were achieved through direct binding STIM1 and subsequent proteasome-mediated degradation of STIM1. The STIM1 downregulation induced the phosphorylation of downstream Nrf2 Ser40, which subsequently translocated to the nucleus, and further increased the expression of antioxidant genes such as NQO1 and HO-1 in HT22 cells. In vivo, the study has further confirmed that double knockout of Shank3 and Stim1 alleviated oxidative stress and inflammation after I/R in Shank3cko mice. In conclusion, the present study has demonstrated that Shank3 interacts with STIM1 and inhibits post-I/R neuronal oxidative stress and inflammatory response via the Nrf2 pathway. This interaction can potentially contribute to the development of a promising method for I/R treatment.
Collapse
Affiliation(s)
- Hongchen Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Feng
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanfang Si
- Department of Ophthalmology, The Eighth Medical Center, Affiliated to the Senior Department of Ophthalmology, The Third Medical Center, Chinese People's Liberation Army General Hospital, Beijing, 100091, China
| | - Chuanhao Lu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiquan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenyu Xie
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheming Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Yong
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Du X, Amin N, Xu L, Botchway BOA, Zhang B, Fang M. Pharmacological intervention of curcumin via the NLRP3 inflammasome in ischemic stroke. Front Pharmacol 2023; 14:1249644. [PMID: 37915409 PMCID: PMC10616488 DOI: 10.3389/fphar.2023.1249644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Ischemic-induced neuronal injury arises due to low oxygen/nutrient levels and an inflammatory response that exacerbates neuronal loss. NOD-like receptor family pyrin domain-containing 3 (NLRP3) is an important regulator of inflammation after ischemic stroke, with its inhibition being involved in nerve regeneration. Curcumin, a main active ingredient in Chinese herbs, plays a positive role in neuronal repair and neuroprotection by regulating the NLRP3 signaling pathway. Nevertheless, the signaling mechanisms relating to how curcumin regulates NLRP3 inflammasome in inflammation and neural restoration following ischemic stroke are unknown. In this report, we summarize the main biological functions of the NLRP3 inflammasome along with the neuroprotective effects and underlying mechanisms of curcumin via impairment of the NLRP3 pathway in ischemic brain injury. We also discuss the role of medicinal interventions that target the NLRP3 and potential pathways, as well as possible directions for curcumin therapy to penetrate the blood-brain barrier (BBB) and hinder inflammation in ischemic stroke. This report conclusively demonstrates that curcumin has neuroprotective properties that inhibit inflammation and prevent nerve cell loss, thereby delaying the progression of ischemic brain damage.
Collapse
Affiliation(s)
- Xiaoxue Du
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nashwa Amin
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Linhao Xu
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Benson O. A. Botchway
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Pharmacy Department, Bupa Cromwell Hospital, London, United Kingdom
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Marong Fang
- Department of Neurology, Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
| |
Collapse
|
5
|
Wang J, Sun X, Dai Y, Ma Y, Wang M, Li X, Qin W. Proteome profiling of hippocampus reveals the neuroprotective effect of mild hypothermia on global cerebral ischemia-reperfusion injury in rats. Sci Rep 2023; 13:14450. [PMID: 37660166 PMCID: PMC10475051 DOI: 10.1038/s41598-023-41766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/31/2023] [Indexed: 09/04/2023] Open
Abstract
Cerebral ischemia is one of the leading causes of disability and mortality worldwide. Blood reperfusion of ischemic cerebral tissue may cause cerebral ischemia-reperfusion (IR) injury. In this study, a rat model of global cerebral I/R injury was established via Pulsinelli's four-vessel occlusion (4-VO) method. The liquid chromatography-tandem mass spectrometry (LC-MS/MS) and bioinformatics analysis were employed to examine the ipsilateral hippocampus proteome profiles of rats with/without MH (32 °C) treatment after IR injury. Totally 2 122 proteins were identified, among which 153 proteins were significantly changed associated with MH (n = 7 per group, fold change-1.5, p < 0.05). GO annotation of the differentially expressed proteins (DEPs) revealed that cellular oxidant detoxification, response to zinc ions, aging, oxygen transport, negative regulation of catalytic activity, response to hypoxia, regulation of protein phosphorylation, and cellular response to vascular endothelial growth factor stimulus were significantly enriched with MH treatment. The KEGG analysis indicated that metabolic pathways, thermogenesis, pathways of neurodegeneration, chemical carcinogenesis-reactive oxygen species, fluid shear stress and atherosclerosis, and protein processing in endoplasmic reticulum were significantly enriched with MH treatment. Importantly, changes in 16 DEPs were reversed by MH treatment. Among them, VCAM-1, S100A8, CaMKK2 and MKK7 were verified as potential markers associated with MH neuroprotection by Western blot analysis. This study is one of the first to investigate the neuroprotective effects of MH on the hippocampal proteome of experimental models of cerebral IR injury. These DEPs may be involved in the most fundamental molecular mechanisms of MH neuroprotection, and provide a scientific foundation for further promotion of reparative strategies in cerebral IR injury.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Xiaopeng Sun
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, China
| | - Yuting Dai
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Yuan Ma
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Mingshan Wang
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
| | - Xiaoxia Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China
| | - Weiwei Qin
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China.
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
6
|
Proteomic investigations of acute ischemic stroke in animal models: a narrative review. JOURNAL OF BIO-X RESEARCH 2022. [DOI: 10.1097/jbr.0000000000000134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
7
|
Datta A, Chen C, Gao YG, Sze SK. Quantitative Proteomics of Medium-Sized Extracellular Vesicle-Enriched Plasma of Lacunar Infarction for the Discovery of Prognostic Biomarkers. Int J Mol Sci 2022; 23:ijms231911670. [PMID: 36232970 PMCID: PMC9569577 DOI: 10.3390/ijms231911670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Lacunar infarction (LACI), a subtype of acute ischemic stroke, has poor mid- to long-term prognosis due to recurrent vascular events or incident dementia which is difficult to predict using existing clinical data. Herein, we aim to discover blood-based biomarkers for LACI as a complementary prognostic tool. Convalescent plasma was collected from forty-five patients following a non-disabling LACI along with seventeen matched control subjects. The patients were followed up prospectively for up to five years to record an occurrence of adverse outcome and grouped accordingly (i.e., LACI-no adverse outcome, LACI-recurrent vascular event, and LACI-cognitive decline without any recurrence of vascular events). Medium-sized extracellular vesicles (MEVs), isolated from the pooled plasma of four groups, were analyzed by stable isotope labeling and 2D-LC-MS/MS. Out of 573 (FDR < 1%) quantified proteins, 146 showed significant changes in at least one LACI group when compared to matched healthy control. A systems analysis revealed that major elements (~85%) of the MEV proteome are different from the proteome of small-sized extracellular vesicles obtained from the same pooled plasma. The altered MEV proteins in LACI patients are mostly reduced in abundance. The majority of the shortlisted MEV proteins are not linked to commonly studied biological processes such as coagulation, fibrinolysis, or inflammation. Instead, they are linked to oxygen-glucose deprivation, endo-lysosomal trafficking, glucose transport, and iron homeostasis. The dataset is provided as a web-based data resource to facilitate meta-analysis, data integration, and targeted large-scale validation.
Collapse
Affiliation(s)
- Arnab Datta
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore 575018, Karnataka, India
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Correspondence: or (A.D.); (S.K.S.)
| | - Christopher Chen
- Memory, Aging and Cognition Centre, National University Health System, Singapore 119228, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Yong-Gui Gao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
- Correspondence: or (A.D.); (S.K.S.)
| |
Collapse
|
8
|
Differential Regulation of the Immune System in Peripheral Blood Following Ischemic Stroke. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2747043. [PMID: 35722467 PMCID: PMC9200570 DOI: 10.1155/2022/2747043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/09/2022] [Indexed: 11/18/2022]
Abstract
Method 108 IS samples and 47 matched controls were obtained from the GEO database. Immune-related genes (IRGs) and their associated drugs were collected from the ImmPort and PharmGBK databases, respectively. Random forest (RF) regression and least absolute shrinkage and selection operator (LASSO) logistic regression were applied to identify immune-related genetic biomarkers (IRGBs) of IS, and accuracy was verified using neural network models. Finally, proportion changes of various immune cells in peripheral blood of IS patients were evaluated using CIBERSORT and xCell and correlation analyses were performed between IRGBs and differentially distributed immune cells. Results A total of 537 genes were differentially expressed between IS and control samples. Four immune-related differential expressed genes identified by regression analysis presented strong predictive power (AUC = 0.909) which we suggeseted them as immune-related genetic biomarkers (IRGBs). We also demonstrated six immune-related genes targeted by known drugs. In addition, post-IS immune system presented an increase in the proportion of innate immune cells and a decrease in adaptive immune cells in the peripheral circulation, and IRGBs showing significance were associated with this process. Conclusion The study identified CARD11, ICAM2, VIM, and CD19 as immune-related genetic biomarkers of IS. Six immune-related DEGs targeted by known drugs were found and provide new candidate drug targets for modulating the post-IS immune system. The innate immune cells and adaptive immune cells are diversified in the post-IS immune system, and IRGBs might play important role during this process.
Collapse
|
9
|
Jiang W, Zhang P, Yang P, Kang N, Liu J, Aihemaiti Y, Tu H. Phosphoproteome Analysis Identifies a Synaptotagmin-1-Associated Complex Involved in Ischemic Neuron Injury. Mol Cell Proteomics 2022; 21:100222. [PMID: 35257887 PMCID: PMC9043414 DOI: 10.1016/j.mcpro.2022.100222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cerebral stroke is one of the leading causes of death in adults worldwide. However, the molecular mechanisms of stroke-induced neuron injury are not fully understood. Here, we obtained phosphoproteomic and proteomic profiles of the acute ischemic hippocampus by LC–MS/MS analysis. Quantitative phosphoproteomic analyses revealed that the dysregulated phosphoproteins were involved in synaptic components and neurotransmission. We further demonstrated that phosphorylation of Synaptotagmin-1 (Syt1) at the Thr112 site in cultured hippocampal neurons aggravated oxygen-glucose deprivation–induced neuronal injury. Immature neurons with low expression of Syt1 exhibit slight neuronal injury in a cerebral ischemia model. Administration of the Tat-Syt1T112A peptide protects neurons against cerebral ischemia-induced injury in vitro and in vivo. Surprisingly, potassium voltage-gated channel subfamily KQT member 2 (Kcnq2) interacted with Syt1 and Annexin A6 (Anxa6) and alleviated Syt1-mediated neuronal injury upon oxygen-glucose deprivation treatment. These results reveal a mechanism underlying neuronal injury and may provide new targets for neuroprotection after acute cerebral ischemia onset. Established the phosphoproteome profiles of acute cerebral ischemic hippocampus. Phosphoproteomic profile reveals phosphorylation of Syt1 and Kcnq2, which are upregulated. Phosphorylation of Syt1 aggravates neuron injury, which is relieved by Tat-Syt1T112A. Kcnq2 interacts with Syt1 and Anxa6 and alleviates Syt1-mediated neuronal injury.
Collapse
Affiliation(s)
- Wei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Pei Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Peng Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Na Kang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Yilixiati Aihemaiti
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Babu M, Singh N, Datta A. In Vitro Oxygen Glucose Deprivation Model of Ischemic Stroke: A Proteomics-Driven Systems Biological Perspective. Mol Neurobiol 2022; 59:2363-2377. [PMID: 35080759 DOI: 10.1007/s12035-022-02745-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/11/2022] [Indexed: 01/17/2023]
Abstract
Oxygen glucose deprivation (OGD) of brain cells is the commonest in vitro model of ischemic stroke that is used extensively for basic and preclinical stroke research. Protein mass spectrometry is one of the most promising and rapidly evolving technologies in biomedical research. A systems-level understanding of cell-type-specific responses to oxygen and glucose deprivation without systemic influence is a prerequisite to delineate the response of the neurovascular unit following ischemic stroke. In this systematic review, we summarize the proteomics studies done on different OGD models. These studies have followed an expression or interaction proteomics approach. They have been primarily used to understand the cellular pathophysiology of ischemia-reperfusion injury or to assess the efficacy of interventions as potential treatment options. We compile the limitations of OGD model and downstream proteomics experiment. We further show that despite having limitations, several proteins shortlisted as altered in in vitro OGD-proteomics studies showed comparable regulation in ischemic stroke patients. This showcases the translational potential of this approach for therapeutic target and biomarker discovery. We next discuss the approaches that can be adopted for cell-type-specific validation of OGD-proteomics results in the future. Finally, we briefly present the research questions that can be addressed by OGD-proteomics studies using emerging techniques of protein mass spectrometry. We have also created a web resource compiling information from OGD-proteomics studies to facilitate data sharing for community usage. This review intends to encourage preclinical stroke community to adopt a hypothesis-free proteomics approach to understand cell-type-specific responses following ischemic stroke.
Collapse
Affiliation(s)
- Manju Babu
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Nikhil Singh
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Arnab Datta
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
11
|
Chen Y, Song F, Tu M, Wu S, He X, Liu H, Xu C, Zhang K, Zhu Y, Zhou R, Jin C, Wang P, Zhang H, Tian M. Quantitative proteomics revealed extensive microenvironmental changes after stem cell transplantation in ischemic stroke. Front Med 2021; 16:429-441. [PMID: 34241786 DOI: 10.1007/s11684-021-0842-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022]
Abstract
The local microenvironment is essential to stem cell-based therapy for ischemic stroke, and spatiotemporal changes of the microenvironment in the pathological process provide vital clues for understanding the therapeutic mechanisms. However, relevant studies on microenvironmental changes were mainly confined in the acute phase of stroke, and long-term changes remain unclear. This study aimed to investigate the microenvironmental changes in the subacute and chronic phases of ischemic stroke after stem cell transplantation. Herein, induced pluripotent stem cells (iPSCs) and neural stem cells (NSCs) were transplanted into the ischemic brain established by middle cerebral artery occlusion surgery. Positron emission tomography imaging and neurological tests were applied to evaluate the metabolic and neurofunctional alterations of rats transplanted with stem cells. Quantitative proteomics was employed to investigate the protein expression profiles in iPSCs-transplanted brain in the subacute and chronic phases of stroke. Compared with NSCs-transplanted rats, significantly increased glucose metabolism and neurofunctional scores were observed in iPSCs-transplanted rats. Subsequent proteomic data of iPSCs-transplanted rats identified a total of 39 differentially expressed proteins in the subacute and chronic phases, which are involved in various ischemic stroke-related biological processes, including neuronal survival, axonal remodeling, antioxidative stress, and mitochondrial function restoration. Taken together, our study indicated that iPSCs have a positive therapeutic effect in ischemic stroke and emphasized the wide-ranging microenvironmental changes in the subacute and chronic phases.
Collapse
Affiliation(s)
- Yao Chen
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China.,Department of Radiology, Zhejiang Hospital, Hangzhou, 310030, China
| | - Fahuan Song
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Mengjiao Tu
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China.,Department of PET Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Shuang Wu
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Xiao He
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Hao Liu
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Caiyun Xu
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Kai Zhang
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Yuankai Zhu
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Rui Zhou
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Chentao Jin
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China
| | - Ping Wang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310027, China.,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Hong Zhang
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China. .,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China. .,Shanxi Medical University, Taiyuan, 030001, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, 310027, China. .,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China.
| | - Mei Tian
- Department of Nuclear Medicine and Medical PET Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China. .,Institute of Nuclear Medicine and Molecular Imaging, Zhejiang University, Hangzhou, 310009, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, 310009, China.
| |
Collapse
|
12
|
Gu RF, Fang T, Nelson A, Gyoneva S, Gao B, Hedde J, Henry K, Peterson E, Burkly LC, Wei R. Proteomic Characterization of the Dynamics of Ischemic Stroke in Mice. J Proteome Res 2021; 20:3689-3700. [PMID: 34085531 PMCID: PMC8256414 DOI: 10.1021/acs.jproteome.1c00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Novel therapies and biomarkers are
needed for the treatment of
acute ischemic stroke (AIS). This study aimed to provide comprehensive
insights into the dynamic proteome changes and underlying molecular
mechanisms post-ischemic stroke. TMT-coupled proteomic analysis was
conducted on mouse brain cortex tissue from five time points up to
4 weeks poststroke in the distal hypoxic-middle cerebral artery occlusion
(DH-MCAO) model. We found that nearly half of the detected proteome
was altered following stroke, but only ∼8.6% of the changes
were at relatively large scales. Clustering on the changed proteome
defined four distinct expression patterns characterized by temporal
and quantitative changes in innate and adaptive immune response pathways
and cytoskeletal and neuronal remodeling. Further analysis on a subset
of 309 “top hits”, which temporally responded to stroke
with relatively large and sustained changes, revealed that they were
mostly secreted proteins, highly correlated to different cortical
cytokines, and thereby potential pharmacodynamic biomarker candidates
for inflammation-targeting therapies. Closer examination of the top
enriched neurophysiologic pathways identified 57 proteins potentially
associated with poststroke recovery. Altogether, our study generated
a rich dataset with candidate proteins worthy of further validation
as biomarkers and/or therapeutic targets for stroke. The proteomics
data are available in the PRIDE Archive with identifier PXD025077.
Collapse
Affiliation(s)
- Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Terry Fang
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ashley Nelson
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Stefka Gyoneva
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Benbo Gao
- Chemical Biology and Proteomics, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Joe Hedde
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Kate Henry
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Emily Peterson
- Medicinal Chemistry, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Linda C Burkly
- Genetic and Neurodevelopmental Disorders Research, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ru Wei
- Chemical Biology and Proteomics, Biogen, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
13
|
Novel Roles of SH2 and SH3 Domains in Lipid Binding. Cells 2021; 10:cells10051191. [PMID: 34068055 PMCID: PMC8152464 DOI: 10.3390/cells10051191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/07/2023] Open
Abstract
Signal transduction, the ability of cells to perceive information from the surroundings and alter behavior in response, is an essential property of life. Studies on tyrosine kinase action fundamentally changed our concept of cellular regulation. The induced assembly of subcellular hubs via the recognition of local protein or lipid modifications by modular protein interactions is now a central paradigm in signaling. Such molecular interactions are mediated by specific protein interaction domains. The first such domain identified was the SH2 domain, which was postulated to be a reader capable of finding and binding protein partners displaying phosphorylated tyrosine side chains. The SH3 domain was found to be involved in the formation of stable protein sub-complexes by constitutively attaching to proline-rich surfaces on its binding partners. The SH2 and SH3 domains have thus served as the prototypes for a diverse collection of interaction domains that recognize not only proteins but also lipids, nucleic acids, and small molecules. It has also been found that particular SH2 and SH3 domains themselves might also bind to and rely on lipids to modulate complex assembly. Some lipid-binding properties of SH2 and SH3 domains are reviewed here.
Collapse
|
14
|
Zhang X, Yin L, Jia X, Zhang Y, Liu T, Zhang L. iTRAQ-based Quantitative Proteomic Analysis of Dural Tissues Reveals Upregulated Haptoglobin to be a Potential Biomarker of Moyamoya Disease. CURR PROTEOMICS 2021. [DOI: 10.2174/1570164617666191210103652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Moyamoya Disease (MMD) is a rare cerebrovascular disease with a high rate
of disability and mortality. Immune reactions have been implicated in the pathogenesis of MMD, however,
the underlying mechanism is still unclear.
Objective:
To identify proteins related to MMD specially involved in the immunogenesis, we performed
a proteomic study.
Methods:
In this work, dural tissues or plasma from 98 patients with MMD, 17 disease controls without
MMD, and 12 healthy donors were included. Proteomic profiles of dural tissues from 4 MMD and
4 disease controls were analyzed by an isobaric tag for relative and absolute quantitation (iTRAQ)-
based proteomics. The immune-related proteins were explored by bioinformatics and the key MMDrelated
proteins were verified by western blot, multiple reaction monitoring methods, enzyme-linked
immunosorbent assay, and tissue microarray.
Results:
1,120 proteins were identified, and 82 MMD-related proteins were found with more than 1.5
fold difference compared with those in the control samples. Gene Ontology analysis showed that 29
proteins were immune-related. In particular, Haptoglobin (HP) was up-regulated in dural tissue and
plasma of MMD samples compared to the controls, and its up-regulation was found to be sex- and
MMD Suzuki grade dependent. Through Receiver Operating Characteristic (ROC) analysis, HP can
well discriminate MMD and healthy donors with the Area Under the Curve (AUC) of 0.953.
Conclusion:
We identified the biggest protein database of the dura mater. 29 out of 82 differentially
expressed proteins in MMD are involved in the immune process. Of which, HP was up-regulated in
dural tissue and plasma of MMD, with sex- and MMD Suzuki grade-dependence. HP might be a potential
biomarker of MMD.
Collapse
Affiliation(s)
- Xiaojun Zhang
- The 85th Hospital of the Chinese People's Liberation Army, Shanghai 200052, China
| | - Lin Yin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaofang Jia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yujiao Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Tiefu Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lijun Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
15
|
Solution NMR Structure of the SH3 Domain of Human Caskin1 Validates the Lack of a Typical Peptide Binding Groove and Supports a Role in Lipid Mediator Binding. Cells 2021; 10:cells10010173. [PMID: 33467043 PMCID: PMC7830187 DOI: 10.3390/cells10010173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
SH3 domains constitute an important class of protein modules involved in a variety of cellular functions. They participate in protein-protein interactions via their canonical ligand binding interfaces composed of several evolutionarily conserved aromatic residues forming binding grooves for typical (PxxP) and atypical (PxxxPR, RxxK, RKxxY) binding motifs. The calcium/calmodulin-dependent serine protein kinase (CASK)-interacting protein 1, or Caskin1, a multidomain scaffold protein regulating the cortical actin filaments, is enriched in neural synapses in mammals. Based on its known interaction partners and knock-out animal studies, Caskin1 may play various roles in neural function and it is thought to participate in several pathological processes of the brain. Caskin1 has a single, atypical SH3 domain in which key aromatic residues are missing from the canonical binding groove. No protein interacting partner for this SH3 domain has been identified yet. Nevertheless, we have recently demonstrated the specific binding of this SH3 domain to the signaling lipid mediator lysophospatidic acid (LPA) in vitro. Here we report the solution NMR structure of the human Caskin1 SH3 domain and analyze its structural features in comparison with other SH3 domains exemplifying different strategies in target selectivity. The key differences revealed by our structural study show that the canonical binding groove found in typical SH3 domains accommodating proline-rich motifs is missing in Caskin1 SH3, most likely excluding a bona fide protein target for the domain. The LPA binding site is distinct from the altered protein binding groove. We conclude that the SH3 domain of Caskin1 might mediate the association of Caskin1 with membrane surfaces with locally elevated LPA content.
Collapse
|
16
|
Heese K. Gastrodia elata Blume (Tianma): Hope for Brain Aging and Dementia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:8870148. [PMID: 33424999 PMCID: PMC7781687 DOI: 10.1155/2020/8870148] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Since aging-related diseases, including dementia, represent major public health threats to our society, physician-scientists must develop innovative, interdisciplinary strategies to open new avenues for development of alternative therapies. One such novel approach may lie in traditional Chinese medicine (TCM). Gastrodia elata Blume (G. elata, tianma) is a TCM frequently used for treatment of cerebrocardiovascular diseases (CCVDs). Recent studies of G. elata-based treatment modalities, which have investigated its pharmacologically relevant activity, potential efficacy, and safety, have employed G. elata in well-characterized, aging-related disease models, with a focus on models of aging-related dementia, such as Alzheimer's disease (AD). Here, I examine results from previous studies of G. elata, as well as related herbal preparations and pure natural products, as prophylaxis and remedies for aging-related CCVDs and dementia. Concluding, data suggest that tianma treatment may be used as a promising complementary therapy for AD.
Collapse
Affiliation(s)
- Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, Republic of Korea
| |
Collapse
|
17
|
Phosphoglycerate mutase 1 reduces neuronal damage in the hippocampus following ischemia/reperfusion through the facilitation of energy utilization. Neurochem Int 2019; 133:104631. [PMID: 31836547 DOI: 10.1016/j.neuint.2019.104631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/15/2019] [Accepted: 12/08/2019] [Indexed: 02/08/2023]
Abstract
In a previous study, we observed the effect of phosphoglycerate mutase 1 (PGAM1) on proliferating cells and neuroblasts in the subgranular zone of mouse dentate gyrus. In the present study, we examined the roles of PGAM1 in the HT22 hippocampal cell line and in gerbil hippocampus after H2O2-induced oxidative stress and after ischemia/reperfusion, respectively. Control-PGAM1 and Tat-PGAM1 proteins were synthesized using Tat-1 expression vector since Tat-1 fusion proteins can easily cross the blood-brain barrier and cell membranes. We found that transduction of Tat-PGAM1 protein into HT22 cells was dose- and time-dependent. Delivery of the protein to the cytoplasm was confirmed by western blotting and immunocytochemistry. Treatment of HT22 cells with Tat-PGAM1 protein showed a concentration-dependent reduction in cell damage and decreased formation of reactive oxygen species after H2O2 exposure. Tat-PGAM1 administration significantly ameliorated the ischemia-induced hyperactivity in gerbils at 1 day after ischemia/reperfusion. Additionally, a pronounced decrease in neuronal damage and reactive gliosis were observed in the hippocampal CA1 region of the Tat-PGAM1-treated group at 4 days after ischemia/reperfusion compared to that in the vehicle (Tat peptide) or control-PGAM1-treated groups. Administration of Tat-PGAM1 mitigated the changes in ATP content, succinate dehydrogenase activity, pH, and 4-hydroxynonenal levels in the hippocampus at 4 and 7 days after ischemia/reperfusion compared to that in the vehicle-treated group. In addition, administration of Tat-PGAM1 significantly ameliorated the ischemia-induced increases of lactate levels in the hippocampus at 15 min and 6 h after ischemia/reperfusion than in the vehicle or control-PGAM1-treated groups. These results suggest that Tat-PGAM1 can be used as a therapeutic agent to prevent neuronal damage from oxidative stress or ischemia.
Collapse
|
18
|
Liu JC, Xue DF, Wang XQ, Ai DB, Qin PJ. MiR-101 relates to chronic peripheral neuropathic pain through targeting KPNB1 and regulating NF-κB signaling. Kaohsiung J Med Sci 2019; 35:139-145. [PMID: 30887716 DOI: 10.1002/kjm2.12025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022] Open
Abstract
Accumulating evidences indicates that chronic neuropathic pain is a kind of neuro-immune disorder with enhanced activation of the immune system. Although the prevalence is very high, neuropathic pain remains extremely difficult to cure. miRNAs are a group of short nonprotein coding RNAs, regulating target genes expression via targeting 3'-untranslated region. More and more research indicates that altered miRNAs expression profile relates to the pathogenesis of neuropathic pain. In this study, we firstly detected the expression of six candidate miRNAs in the plasma samples from 23 patients with neuropathic pain and 10 healthy controls. Subsequently, the level of miR-132 and miR-101 was detected in the sural nerve biopsies. We found miR-101 level was significantly repressed in both the plasma samples and sural nerve biopsies from neuropathic pain patients. Predicted by bioinformatics tools and confirmed by dual luciferase assay and immunoblotting, we identified that KPNB1 is a direct target of miR-101. The negative correlation between miR-101 and KPNB1 was also confirmed in the sural nerve biopsies, and miR-101 reduction relates to the activation of NF-κB signaling in vivo and in vitro which contributes to the pathogenesis of neuropathic pain.
Collapse
Affiliation(s)
- Jun-Chao Liu
- Department of Anesthesia and Surgery, Qingdao Municipal Hospital, Qingdao Clinical Anesthesia Research Center, Qingdao clinical pain research center, Qingdao, Puerto Rico, China
| | - Dong-Fang Xue
- Department of Anesthesia and Surgery, Qingdao Municipal Hospital, Qingdao Clinical Anesthesia Research Center, Qingdao clinical pain research center, Qingdao, Puerto Rico, China
| | - Xiao-Qian Wang
- Department of Surgery, Qingdao University Affiliated Hospital, Qingdao, Puerto Rico, China
| | - Deng-Bin Ai
- Department of Anesthesia and Surgery, Qingdao Municipal Hospital, Qingdao Clinical Anesthesia Research Center, Qingdao clinical pain research center, Qingdao, Puerto Rico, China
| | - Pei-Juan Qin
- Department of Anesthesia and Surgery, Qingdao Municipal Hospital, Qingdao Clinical Anesthesia Research Center, Qingdao clinical pain research center, Qingdao, Puerto Rico, China
| |
Collapse
|
19
|
Wen M, Jin Y, Zhang H, Sun X, Kuai Y, Tan W. Proteomic Analysis of Rat Cerebral Cortex in the Subacute to Long-Term Phases of Focal Cerebral Ischemia-Reperfusion Injury. J Proteome Res 2019; 18:3099-3118. [PMID: 31265301 DOI: 10.1021/acs.jproteome.9b00220] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Stroke is a leading cause of mortality and disability, and ischemic stroke accounts for more than 80% of the disease occurrence. Timely reperfusion is essential in the treatment of ischemic stroke, but it is known to cause ischemia-reperfusion (I/R) injury and the relevant studies have mostly focused on the acute phase. Here we reported on a global proteomic analysis to investigate the development of cerebral I/R injury in the subacute and long-term phases. A rat model was used, with 2 h-middle cerebral artery occlusion (MCAO) followed with 1, 7, and 14 days of reperfusion. The proteins of cerebral cortex were analyzed by SDS-PAGE, whole-gel slicing, and quantitative LC-MS/MS. Totally 5621 proteins were identified, among which 568, 755, and 492 proteins were detected to have significant dys-regulation in the model groups with 1, 7, and 14 days of reperfusion, respectively, when compared with the corresponding sham groups (n = 4, fold change ≥1.5 or ≤0.67 and p ≤ 0.05). Bioinformatic analysis on the functions and reperfusion time-dependent dys-regulation profiles of the proteins exhibited changes of structures and biological processes in cytoskeleton, synaptic plasticity, energy metabolism, inflammation, and lysosome from subacute to long-term phases of cerebral I/R injury. Disruption of cytoskeleton and synaptic structures, impairment of energy metabolism processes, and acute inflammation responses were the most significant features in the subacute phase. With the elongation of reperfusion time to the long-term phase, a tendency of recovery was detected on cytoskeleton, while inflammation pathways different from the subacute phase were activated. Also, lysosomal structures and functions might be restored. This is the first work reporting the proteome changes that occurred at different time points from the subacute to long-term phases of cerebral I/R injury and we expect it would provide useful information to improve the understanding of the mechanisms involved in the development of cerebral I/R injury and suggest candidates for treatment.
Collapse
Affiliation(s)
- Meiling Wen
- School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , P. R. China
| | - Ya Jin
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Yihe Kuai
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences , Guangdong University of Technology , Guangzhou 510006 , P. R. China
| |
Collapse
|
20
|
Liu T, Zhou J, Cui H, Li P, Luo J, Li T, He F, Wang Y, Tang T. iTRAQ-based quantitative proteomics reveals the neuroprotection of rhubarb in experimental intracerebral hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2019; 232:244-254. [PMID: 30502478 DOI: 10.1016/j.jep.2018.11.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/26/2018] [Accepted: 11/20/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhubarb is a traditional Chinese medicine(TCM), that possesses neuroprotective, anti-inflammatory, antibacterial, antioxidative, purgative and anticancer properties, and has been used to treat intracerebral hemorrhage (ICH) and many other diseases. AIMS OF THE STUDY This study aimed to investigate the changes of brain protein in ICH rats treated with rhubarb and to explore the multi-target mechanism of rhubarb in the treatment of ICH via bioinformatics analysis of differentially expressed proteins (DEPs). MATERIALS AND METHODS Rats were subjected to collagenase-induced ICH and then treated orally with 3 or 12 g/kg rhubarb daily for 2 days following ICH. After sacrifice, total protein of brain tissue was extracted, and isobaric tag for relative and absolute quantification (iTRAQ)-based liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was employed to quantitatively identify of the DEPs in two treatment groups compared with the vehicle group. The DEPs were analyzed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and STRING databases. Bioinformatics Analysis Tool for Molecular mechanism of TCM (BATMAN-TCM) was used to predict the target of rhubarb and western blotting was used for verification. RESULTS In total, 1356 proteins were identified with a 1% false discovery rate (FDR). Among them, 55 DEPs were significantly altered in the sham, vehicle, low dose rhubarb group (LDR, 3 g/kg), and high dose rhubarb group (HDR, 12 g/kg). Enrichment analysis of GO annotations indicated that rhubarb mainly regulated expression of some neuron projection proteins involved in the response to drug and nervous system development. The dopaminergic synapse pathway was found to be the most significant DEP in the combined analysis of the KEGG and BATMAN-TCM databases. Based on the results of the STRING analysis, oxidative stress (OS), calcium binding protein regulation, vascularization, and energy metabolism were important in the rhubarb therapeutic process. CONCLUSION Rhubarb achieves its effects mainly through the dopaminergic synapse pathway in ICH treatment. The ICH-treating mechanisms of rhubarb may also involve anti-OS, calcium binding protein regulation, angiogenic regulation, and energy metabolism improvement. This study adds new evidence to clinical applications of rhubarb for ICH.
Collapse
Affiliation(s)
- Tao Liu
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China; Department of Gerontology, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, 830000 Urumqi, China
| | - Jing Zhou
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Hanjin Cui
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Pengfei Li
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Jiekun Luo
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Teng Li
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
| | - Feng He
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yang Wang
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China; National Research Center of geriatrics, Xiangya Hospital, Central South University, China.
| | - Tao Tang
- Institute of Integrative Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China; National Research Center of geriatrics, Xiangya Hospital, Central South University, China.
| |
Collapse
|
21
|
Kim AY, Baik EJ. Glutamate Dehydrogenase as a Neuroprotective Target Against Neurodegeneration. Neurochem Res 2018; 44:147-153. [PMID: 29357018 DOI: 10.1007/s11064-018-2467-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 10/18/2022]
Abstract
Regulation of glutamate metabolism via glutamate dehydrogenase (GDH) might be the promising therapeutic approach for treating neurodegenerative disorders. In the central nervous system, glutamate functions both as a major excitatory neurotransmitter and as a key intermediate metabolite for neurons. GDH converts glutamate to α-ketoglutarate, which serves as a TCA cycle intermediate. Dysregulated GDH activity in the central nervous system is highly correlated with neurological disorders. Indeed, studies conducted with mutant mice and allosteric drugs have shown that deficient or overexpressed GDH activity in the brain can regulate whole body energy metabolism and affect early onset of Parkinson's disease, Alzheimer's disease, temporal lobe epilepsy, and spinocerebellar atrophy. Moreover, in strokes with excitotoxicity as the main pathophysiology, mice that overexpressed GDH exhibited smaller ischemic lesion than mice with normal GDH expression. In additions, GDH activators improve lesions in vivo by increasing α-ketoglutarate levels. In neurons exposed to an insult in vitro, enhanced GDH activity increases ATP levels. Thus, in an energy crisis, neuronal mitochondrial activity is improved and excitotoxic risk is reduced. Consequently, modulating GDH activity in energy-depleted conditions could be a sound strategy for maintaining the mitochondrial factory in neurons, and thus, protect against metabolic failure.
Collapse
Affiliation(s)
- A Young Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, South Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, 16499, South Korea.
| |
Collapse
|
22
|
Huang L, Shang E, Fan W, Li X, Li B, He S, Fu Y, Zhang Y, Li Y, Fang W. S-oxiracetam protect against ischemic stroke via alleviating blood brain barrier dysfunction in rats. Eur J Pharm Sci 2017; 109:40-47. [DOI: 10.1016/j.ejps.2017.07.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/20/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
|
23
|
Datta A, Chai YL, Tan JM, Lee JH, Francis PT, Chen CP, Sze SK, Lai MKP. An iTRAQ-based proteomic analysis reveals dysregulation of neocortical synaptopodin in Lewy body dementias. Mol Brain 2017; 10:36. [PMID: 28800743 PMCID: PMC5553757 DOI: 10.1186/s13041-017-0316-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/21/2017] [Indexed: 02/08/2023] Open
Abstract
Lewy body dementias are the second most common cause of neurodegenerative dementia in the elderly after Alzheimer's disease (AD). The two clinical subgroups of Lewy body dementias, namely, dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), are differentiated by the chronology of cognitive symptoms relative to parkinsonism. At present, there remains a debate on whether DLB and PDD are separate disease entities, or fall within the same spectrum of Lewy body dementias. In this study, we compared the detergent-soluble proteome via an 8-plex isobaric tag for relative and absolute quantitation (iTRAQ) analysis of pooled lysates from the prefrontal cortex (BA9) of DLB (n = 19) and PDD (n = 21) patients matched a priori for amyloid (total Aβ42) burden, semi-quantitative scores for Lewy bodies and neurofibrillary tangles together with age-matched control (n = 21) subjects. A total of 1914 proteins were confidently identified by iTRAQ (false discovery rate = 0%). None of the proteins showed a significant yet opposite regulation in between DLB and PDD when compared to aged controls in the proteomic data set as well as following immunoblot analysis of the pooled and individual lysates involving all 61 subjects. The postsynaptic protein, synaptopodin (SYNPO) was significantly down-regulated in both DLB and PDD subgroups, suggesting a defective synaptic transmission in the demented patients. In conclusion, the largely similar proteome of DLB and PDD matched for amyloid burden suggests that variations in concomitant AD-related pathology, abnormal post-translational modifications or protein-protein interactions, defective intracellular trafficking or misfolding of proteins could play a part in driving the clinically observed differences between these two subgroups of Lewy body dementias. This further indicates that amyloid-targeting therapeutic strategies may show different efficacies in DLB versus PDD.
Collapse
Affiliation(s)
- Arnab Datta
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore. .,Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, Building 10 Room 6N318, Bethesda, MD 20814, USA.
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Jing Min Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Jasinda H Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Paul T Francis
- Wolfson Centre for Age-related Diseases, King's College London, Guy's Campus, St Thomas Street, London SE1 1UL, UK
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, Singapore 117599, Singapore. .,Wolfson Centre for Age-related Diseases, King's College London, Guy's Campus, St Thomas Street, London SE1 1UL, UK.
| |
Collapse
|
24
|
Abstract
Despite the success of cART, greater than 50% of HIV infected people develop cognitive and motor deficits termed HIV-associated neurocognitive disorders (HAND). Macrophages are the major cell type infected in the CNS. Unlike for T cells, the virus does not kill macrophages and these long-lived cells may become HIV reservoirs in the brain. They produce cytokines/chemokines and viral proteins that promote inflammation and neuronal damage, playing a key role in HIV neuropathogenesis. HIV Tat is the transactivator of transcription that is essential for replication and transcriptional regulation of the virus and is the first protein to be produced after HIV infection. Even with successful cART, Tat is produced by infected cells. In this study we examined the role of the HIV Tat protein in the regulation of gene expression in human macrophages. Using THP-1 cells, a human monocyte/macrophage cell line, and their infection with lentivirus, we generated stable cell lines that express Tat-Flag. We performed ChIP-seq analysis of these cells and found 66 association sites of Tat in promoter or coding regions. Among these are C5, CRLF2/TSLPR, BDNF, and APBA1/Mint1, genes associated with inflammation/damage. We confirmed the association of Tat with these sequences by ChIP assay and expression of these genes in our THP-1 cell lines by qRT-PCR. We found that HIV Tat increased expression of C5, APBA1, and BDNF, and decreased CRLF2. The K50A Tat-mutation dysregulated expression of these genes without affecting the binding of the Tat complex to their gene sequences. Our data suggest that HIV Tat, produced by macrophage HIV reservoirs in the brain despite successful cART, contributes to neuropathogenesis in HIV-infected people.
Collapse
|
25
|
Evolution of blood-brain-barrier permeability after acute ischemic stroke. PLoS One 2017; 12:e0171558. [PMID: 28207745 PMCID: PMC5313141 DOI: 10.1371/journal.pone.0171558] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 01/22/2017] [Indexed: 11/29/2022] Open
Abstract
The dynamics of BBB permeability after AIS in humans are not well understood. In the present study we measured the evolution of BBB permeability after AIS in humans using MRI. Patients presenting to our institution with a diagnosis of AIS underwent a single dynamic contrast-enhanced MRI (DCE-MRI) sequence to measure BBB permeability during their initial workup. Forty-two patients were included in the final analysis. The patient sample underwent DCE-MRI at a mean time of 23.8hrs after the onset of AIS symptoms (range: 1.3–90.7hrs). At all time-points the BBB permeability within the infarct region of the brain as defined on DWI/ADC was higher compared to the homologous region of the contralateral hemisphere (p<0.005). BBB permeability, expressed as a ratio of infarct permeability to contralateral permeability, was greatest at 6-48hrs after the onset of AIS. Although the data was not acquired longitudinally, these findings suggest that the permeability of the BBB is continually elevated following AIS, which contradicts previous assertions that BBB permeability after AIS follows a biphasic course. Knowledge of BBB dynamics following AIS may provide insight into future treatments for AIS, especially BBB stabilizing agents.
Collapse
|
26
|
Wang H, Song Y, Hao D, Du L. Molecular mechanisms for N G-nitro-L-arginine methyl ester action against cerebral ischemia–reperfusion injury-induced blood–brain barrier dysfunction. ASIAN BIOMED 2017. [DOI: 10.5372/1905-7415.0802.277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Background: Ischemic stroke, an acute neurological injury lacking an effective therapy, is a leading cause of death worldwide. The unmet need in stroke research is to identify viable therapeutic targets and to understand their interplay during cerebral ischemia-reperfusion (I/R) injury.
Objective: To explore the protective effects and molecular mechanism of NG-nitro-L-arginine methyl ester (L-NAME) in cerebral ischemia-reperfusion injury-induced blood-brain barrier (BBB) dysfunction.
Methods: Two hundred fifty-six rats were randomly assigned to a sham operation group, I/R group, and I/R with L-NAME treatment group. Brain water content was determined by calculating dry/wet weight. The permeability of the BBB was observed using an electron microscope and by determining the Evans Blue leakage from brain tissue on the ischemic side. The expression of brain MMP-9 and GFAP was determined using an immunohistochemical method. The expression of ZO-1 protein was determined by western blotting.
Results: We found that L-NAME remarkably attenuated the permeability of the BBB after I/R as assessed by Evans Blue leakage and brain water content (p < 0.05). This was further confirmed by examination of the ultrastructural morphology of the BBB using a transmission electron microscope. Furthermore, we found that expression of the zonae occludens-1 (ZO-1) was decreased in endothelial cells, and expression of MMP-9 and GFAP was increased in the basement membrane and astrocyte end-feet in vehicle control groups, respectively, but these changes could be prevented by L-NAME pretreatment.
Conclusion: These results suggested that the neuroprotective effects of L-NAME against BBB damage induced by I/R might be related to the upregulation of tight junction proteins and inhibition of MMP-9 and GFAP expression. L-NAME can be used as a potential MMP-9-based multiple targeting therapeutic strategy in cerebral I/R injury.
Collapse
Affiliation(s)
- Hanghui Wang
- Department of Ultrasound, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China China
- Hong Hui Hospital, Xi’an Jiaotong University College of Medicine, Shaanxi 710054, China
| | - Yixin Song
- Department of Ultrasound, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China China
- Hong Hui Hospital, Xi’an Jiaotong University College of Medicine, Shaanxi 710054, China
| | - Dingjun Hao
- Department of Ultrasound, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China China
- Correspondence to: Hong Hui Hospital, Xi’an Jiaotong University College of Medicine, Shaanxi, 710054, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai First People’s Hospital Afiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200080, China China
- Hong Hui Hospital, Xi’an Jiaotong University College of Medicine, Shaanxi 710054, China
| |
Collapse
|
27
|
Law HCH, Szeto SSW, Quan Q, Zhao Y, Zhang Z, Krakovska O, Lui LT, Zheng C, Lee SMY, Siu KWM, Wang Y, Chu IK. Characterization of the Molecular Mechanisms Underlying the Chronic Phase of Stroke in a Cynomolgus Monkey Model of Induced Cerebral Ischemia. J Proteome Res 2017; 16:1150-1166. [PMID: 28102082 DOI: 10.1021/acs.jproteome.6b00651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stroke is one of the main causes of mortality and long-term disability worldwide. The pathophysiological mechanisms underlying this disease are not well understood, particularly in the chronic phase after the initial ischemic episode. In this study, a Macaca fascicularis stroke model consisting of two sample groups, as determined by MRI-quantified infarct volumes as a measure of the stroke severity 28 days after the ischemic episode, was evaluated using qualitative and quantitative proteomics analyses. By using multiple online multidimensional liquid chromatography platforms, 8790 nonredundant proteins were identified that condensed to 5223 protein groups at 1% global false discovery rate (FDR). After the application of a conservative criterion (5% local FDR), 4906 protein groups were identified from the analysis of cerebral cortex. Of the 2068 quantified proteins, differential proteomic analyses revealed that 31 and 23 were dysregulated in the elevated- and low-infarct-volume groups, respectively. Neurogenesis, synaptogenesis, and inflammation featured prominently as the cellular processes associated with these dysregulated proteins. Protein interaction network analysis revealed that the dysregulated proteins for inflammation and neurogenesis were highly connected, suggesting potential cross-talk between these processes in modulating the cytoskeletal structure and dynamics in the chronic phase poststroke. Elucidating the long-term consequences of brain tissue injuries from a cellular prospective, as well as the molecular mechanisms that are involved, would provide a basis for the development of new potentially neurorestorative therapies.
Collapse
Affiliation(s)
- Henry C H Law
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| | - Samuel S W Szeto
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| | - Quan Quan
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| | - Yun Zhao
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, College of Pharmacy, Jinan University , Guangzhou 510632, China
| | - Olga Krakovska
- Department of Chemistry and Centre for Research in Mass Spectrometry, York University , Toronto, Ontario M3J 1P3, Canada
| | - Leong Ting Lui
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| | - Chengyou Zheng
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, College of Pharmacy, Jinan University , Guangzhou 510632, China
| | - Simon M-Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau , Avenue Padre Tomás Pereira S.J., Taipa, Macau 999078, China
| | - K W Michael Siu
- Department of Chemistry and Centre for Research in Mass Spectrometry, York University , Toronto, Ontario M3J 1P3, Canada.,Department of Chemistry and Biochemistry, University of Windsor , Windsor, Ontario N9B 3P4, Canada
| | - Yuqiang Wang
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, College of Pharmacy, Jinan University , Guangzhou 510632, China
| | - Ivan K Chu
- Department of Chemistry, The University of Hong Kong , Hong Kong, China
| |
Collapse
|
28
|
Kim AY, Jeong KH, Lee JH, Kang Y, Lee SH, Baik EJ. Glutamate dehydrogenase as a neuroprotective target against brain ischemia and reperfusion. Neuroscience 2016; 340:487-500. [PMID: 27845178 DOI: 10.1016/j.neuroscience.2016.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/02/2016] [Accepted: 11/06/2016] [Indexed: 01/13/2023]
Abstract
Deregulation of glutamate homeostasis is associated with degenerative neurological disorders. Glutamate dehydrogenase (GDH) is important for glutamate metabolism and plays a central role in expanding the pool of tricarboxylic acid (TCA) cycle intermediate alpha-ketoglutarate (α-KG), which improves overall bioenergetics. Under high energy demand, maintenance of ATP production results in functionally active mitochondria. Here, we tested whether the modulation of GDH activity can rescue ischemia/reperfusion-induced neuronal death in an in vivo mouse model of middle artery occlusion and an in vitro oxygen/glucose depletion model. Iodoacetate, an inhibitor of glycolysis, was also used in a model of energy failure, remarkably depleting ATP and α-KG. To stimulate GDH activity, the GDH activator 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid and potential activator beta-lapachone were used. The GDH activators restored α-KG and ATP levels in the injury models and provided potent neuroprotection. We also found that beta-lapachone increased glutamate utilization, accompanied by a reduction in extracellular glutamate. Thus, our hypothesis that mitochondrial GDH activators increase α-KG production as an alternative energy source for use in the TCA cycle under energy-depleted conditions was confirmed. Our results suggest that increasing GDH-mediated glutamate oxidation represents a new therapeutic intervention for neurodegenerative disorders, including stoke.
Collapse
Affiliation(s)
- A Young Kim
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Kyeong-Hoon Jeong
- Gachon University of Medicine and Science, Incheon 406-840, Republic of Korea
| | - Jae Ho Lee
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Yup Kang
- Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Soo Hwan Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Republic of Korea.
| |
Collapse
|
29
|
Adav SS, Sze SK. Insight of brain degenerative protein modifications in the pathology of neurodegeneration and dementia by proteomic profiling. Mol Brain 2016; 9:92. [PMID: 27809929 PMCID: PMC5094070 DOI: 10.1186/s13041-016-0272-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/16/2016] [Indexed: 02/06/2023] Open
Abstract
Dementia is a syndrome associated with a wide range of clinical features including progressive cognitive decline and patient inability to self-care. Due to rapidly increasing prevalence in aging society, dementia now confers a major economic, social, and healthcare burden throughout the world, and has therefore been identified as a public health priority by the World Health Organization. Previous studies have established dementia as a 'proteinopathy' caused by detrimental changes in brain protein structure and function that promote misfolding, aggregation, and deposition as insoluble amyloid plaques. Despite clear evidence that pathological cognitive decline is associated with degenerative protein modifications (DPMs) arising from spontaneous chemical modifications to amino acid side chains, the molecular mechanisms that promote brain DPMs formation remain poorly understood. However, the technical challenges associated with DPM analysis have recently become tractable due to powerful new proteomic techniques that facilitate detailed analysis of brain tissue damage over time. Recent studies have identified that neurodegenerative diseases are associated with the dysregulation of critical repair enzymes, as well as the misfolding, aggregation and accumulation of modified brain proteins. Future studies will further elucidate the mechanisms underlying dementia pathogenesis via the quantitative profiling of the human brain proteome and associated DPMs in distinct phases and subtypes of disease. This review summarizes recent developments in quantitative proteomic technologies, describes how these techniques have been applied to the study of dementia-linked changes in brain protein structure and function, and briefly outlines how these findings might be translated into novel clinical applications for dementia patients. In this review, only spontaneous protein modifications such as deamidation, oxidation, nitration glycation and carbamylation are reviewed and discussed.
Collapse
Affiliation(s)
- Sunil S. Adav
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551 Singapore
| | - Siu Kwan Sze
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551 Singapore
| |
Collapse
|
30
|
Shao WH, Chen JJ, Fan SH, Lei Y, Xu HB, Zhou J, Cheng PF, Yang YT, Rao CL, Wu B, Liu HP, Xie P. Combined Metabolomics and Proteomics Analysis of Major Depression in an Animal Model: Perturbed Energy Metabolism in the Chronic Mild Stressed Rat Cerebellum. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:383-92. [PMID: 26134254 DOI: 10.1089/omi.2014.0164] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a highly prevalent, debilitating mental illness of importance for global health. However, its molecular pathophysiology remains poorly understood. Combined proteomics and metabolomics approaches should provide a comprehensive understanding of MDD's etiology. The present study reports novel "-omics" insights from a rodent model of MDD. Cerebellar samples from chronic mild stressed (CMS)-treated depressed rats and controls were compared with a focus on the differentially expressed proteins and metabolites using isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics and gas chromotography/mass spectrometry (GC-MS) metabolomics techniques, respectively. The combined analyses found significant alterations associated with cerebellar energy metabolism, as indicated by (1) abnormal amino acid metabolism accompanied by corresponding metabolic enzymatic alterations and disturbed protein turnover, (2) increased glycolytic and tricarboxylic acid (TCA) cycle enzyme levels paralleled by changes in the concentrations of associated metabolites, and (3) perturbation of ATP biosynthesis through adenosine accompanied by perturbation of the mitochondrial respiratory chain. To the best of our knowledge, this study is the first to integrate proteomics and metabolomics analyses to examine the pathophysiological mechanism(s) underlying MDD in a CMS rodent model of depression. These results can offer important insights into the pathogenesis of MDD.
Collapse
Affiliation(s)
- Wei-hua Shao
- 1 Department of Respiratory Medicine, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian-jun Chen
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Song-hua Fan
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yang Lei
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hong-bo Xu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Jian Zhou
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng-fei Cheng
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Yong-tao Yang
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Cheng-long Rao
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Bo Wu
- 3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Hai-peng Liu
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| | - Peng Xie
- 2 Department of Neurology, First Affiliated Hospital of Chongqing Medical University , Chongqing, China .,3 Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University , Chongqing, China .,4 Chongqing Key Laboratory of Neurobiology , Chongqing, China
| |
Collapse
|
31
|
White TE, Surles-Zeigler MC, Ford GD, Gates AS, Davids B, Distel T, LaPlaca MC, Ford BD. Bilateral gene interaction hierarchy analysis of the cell death gene response emphasizes the significance of cell cycle genes following unilateral traumatic brain injury. BMC Genomics 2016; 17:130. [PMID: 26912237 PMCID: PMC4765060 DOI: 10.1186/s12864-016-2412-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 01/26/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Delayed or secondary cell death that is caused by a cascade of cellular and molecular processes initiated by traumatic brain injury (TBI) may be reduced or prevented if an effective neuroprotective strategy is employed. Microarray and subsequent bioinformatic analyses were used to determine which genes, pathways and networks were significantly altered 24 h after unilateral TBI in the rat. Ipsilateral hemi-brain, the corresponding contralateral hemi-brain, and naïve (control) brain tissue were used for microarray analysis. RESULTS Ingenuity Pathway Analysis showed cell death and survival (CD) to be a top molecular and cellular function associated with TBI on both sides of the brain. One major finding was that the overall gene expression pattern suggested an increase in CD genes in ipsilateral brain tissue and suppression of CD genes contralateral to the injury which may indicate an endogenous protective mechanism. We created networks of genes of interest (GOI) and ranked the genes by the number of direct connections each had in the GOI networks, creating gene interaction hierarchies (GIHs). Cell cycle was determined from the resultant GIHs to be a significant molecular and cellular function in post-TBI CD gene response. CONCLUSIONS Cell cycle and apoptosis signalling genes that were highly ranked in the GIHs and exhibited either the inverse ipsilateral/contralateral expression pattern or contralateral suppression were identified and included STAT3, CCND1, CCND2, and BAX. Additional exploration into the remote suppression of CD genes may provide insight into neuroprotective mechanisms that could be used to develop therapies to prevent cell death following TBI.
Collapse
Affiliation(s)
- Todd E White
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Monique C Surles-Zeigler
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Gregory D Ford
- Division of Natural Sciences and Physical Education, Georgia Highlands College, 5441 Highway 20, NE, Cartersville, GA, 30121, USA.
| | - Alicia S Gates
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Benem Davids
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| | - Timothy Distel
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
- University of California-Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.
| | - Michelle C LaPlaca
- Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Byron D Ford
- Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
- University of California-Riverside School of Medicine, 900 University Ave., Riverside, CA, 92521, USA.
| |
Collapse
|
32
|
Hoque A, Hossain MI, Ameen SS, Ang CS, Williamson N, Ng DCH, Chueh AC, Roulston C, Cheng HC. A beacon of hope in stroke therapy-Blockade of pathologically activated cellular events in excitotoxic neuronal death as potential neuroprotective strategies. Pharmacol Ther 2016; 160:159-79. [PMID: 26899498 DOI: 10.1016/j.pharmthera.2016.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Excitotoxicity, a pathological process caused by over-stimulation of ionotropic glutamate receptors, is a major cause of neuronal loss in acute and chronic neurological conditions such as ischaemic stroke, Alzheimer's and Huntington's diseases. Effective neuroprotective drugs to reduce excitotoxic neuronal loss in patients suffering from these neurological conditions are urgently needed. One avenue to achieve this goal is to clearly define the intracellular events mediating the neurotoxic signals originating from the over-stimulated glutamate receptors in neurons. In this review, we first focus on the key cellular events directing neuronal death but not involved in normal physiological processes in the neurotoxic signalling pathways. These events, referred to as pathologically activated events, are potential targets for the development of neuroprotectant therapeutics. Inhibitors blocking some of the known pathologically activated cellular events have been proven to be effective in reducing stroke-induced brain damage in animal models. Notable examples are inhibitors suppressing the ion channel activity of neurotoxic glutamate receptors and those disrupting interactions of specific cellular proteins occurring only in neurons undergoing excitotoxic cell death. Among them, Tat-NR2B9c and memantine are clinically effective in reducing brain damage caused by some acute and chronic neurological conditions. Our second focus is evaluation of the suitability of the other inhibitors for use as neuroprotective therapeutics. We also discuss the experimental approaches suitable for bridging our knowledge gap in our current understanding of the excitotoxic signalling mechanism in neurons and discovery of new pathologically activated cellular events as potential targets for neuroprotection.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ching-Seng Ang
- Bio21 Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Dominic C H Ng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia; School of Biomedical Science, University of Queensland, St. Lucia, QLD, Australia
| | - Anderly C Chueh
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Carli Roulston
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
33
|
Chen HJ, Shen YC, Shiao YJ, Liou KT, Hsu WH, Hsieh PH, Lee CY, Chen YR, Lin YL. Multiplex Brain Proteomic Analysis Revealed the Molecular Therapeutic Effects of Buyang Huanwu Decoction on Cerebral Ischemic Stroke Mice. PLoS One 2015; 10:e0140823. [PMID: 26492191 PMCID: PMC4619651 DOI: 10.1371/journal.pone.0140823] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022] Open
Abstract
Stroke is the second-leading cause of death worldwide, and tissue plasminogen activator (TPA) is the only drug used for a limited group of stroke patients in the acute phase. Buyang Huanwu Decoction (BHD), a traditional Chinese medicine prescription, has long been used for improving neurological functional recovery in stroke. In this study, we characterized the therapeutic effect of TPA and BHD in a cerebral ischemia/reperfusion (CIR) injury mouse model using multiplex proteomics approach. After the iTRAQ-based proteomics analysis, 1310 proteins were identified from the mouse brain with <1% false discovery rate. Among them, 877 quantitative proteins, 10.26% (90/877), 1.71% (15/877), and 2.62% (23/877) of the proteins was significantly changed in the CIR, BHD treatment, and TPA treatment, respectively. Functional categorization analysis showed that BHD treatment preserved the integrity of the blood–brain barrier (BBB) (Alb, Fga, and Trf), suppressed excitotoxicity (Grm5, Gnai, and Gdi), and enhanced energy metabolism (Bdh), thereby revealing its multiple effects on ischemic stroke mice. Moreover, the neurogenesis marker doublecortin was upregulated, and the activity of glycogen synthase kinase 3 (GSK-3) and Tau was inhibited, which represented the neuroprotective effects. However, TPA treatment deteriorated BBB breakdown. This study highlights the potential of BHD in clinical applications for ischemic stroke.
Collapse
Affiliation(s)
- Hong-Jhang Chen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Kuo-Tong Liou
- Department of Chinese Martial Arts and Graduate Institute of Sport Coaching Science, Chinese Culture University, Taipei, Taiwan
| | - Wei-Hsiang Hsu
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Pei-Hsuan Hsieh
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Ying Lee
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Yet-Ran Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
- * E-mail: (YLL); (YRC)
| | - Yun-Lian Lin
- National Research Institute of Chinese Medicine, Taipei, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
- * E-mail: (YLL); (YRC)
| |
Collapse
|
34
|
Datta A, Sze SK. Data for iTRAQ profiling of micro-vesicular plasma specimens: In search of potential prognostic circulatory biomarkers for Lacunar infarction. Data Brief 2015; 4:510-7. [PMID: 26966714 PMCID: PMC4783520 DOI: 10.1016/j.dib.2015.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/07/2015] [Accepted: 07/07/2015] [Indexed: 01/11/2023] Open
Abstract
To discover potential prognostic biomarkers of Lacunar infarction (LACI), here we present quantitative proteomics data of plasma microvesicle-enriched fraction derived by comparative isobaric profiling of three groups of prospectively followed-up LACI patients (LACI – no adverse outcome, LACI –recurrent vascular event and LACI – cognitive decline) and a demographically matched control group. We confidently (unused prot score >3, FDR=1.1%) identified 183 proteins, 43 out of which were significantly regulated (p-value<0.05) in at least one of the three LACI groups in comparison to control group. Bioinformatics analysis and data mining revealed upregulation of brain-specific proteins including myelin basic protein, proteins of coagulation cascade (e.g., fibrinogen alpha chain, fibrinogen beta chain) and focal adhesion (e.g., integrin alpha-IIb, talin-1, and filamin-A) while albumin was downregulated in both groups of patients with adverse outcome. The data of this study are also in line with our previously published article entitled “Discovery of prognostic biomarker candidates of Lacunar infarction by quantitative proteomics of microvesicles enriched plasma” by Datta et al. (2014). The raw data had been deposited to the ProteomeXchange consortium with identifier PXD000748.
Collapse
Affiliation(s)
- Arnab Datta
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
35
|
Brea D, Agulla J, Staes A, Gevaert K, Campos F, Sobrino T, Blanco M, Dávalos A, Castillo J, Ramos-Cabrer P. Study of Protein Expression in Peri-Infarct Tissue after Cerebral Ischemia. Sci Rep 2015; 5:12030. [PMID: 26153530 PMCID: PMC4495553 DOI: 10.1038/srep12030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/11/2015] [Indexed: 11/26/2022] Open
Abstract
In this work, we report our study of protein expression in rat peri-infarct tissue, 48 h after the induction of permanent focal cerebral ischemia. Two proteomic approaches, gel electrophoresis with mass spectrometry and combined fractional diagonal chromatography (COFRADIC), were performed using tissue samples from the periphery of the induced cerebral ischemic lesions, using tissue from the contra-lateral hemisphere as a control. Several protein spots (3408) were identified by gel electrophoresis, and 11 showed significant differences in expression between peri-infarct and contra-lateral tissues (at least 3-fold, p < 0.05). Using COFRADIC, 5412 proteins were identified, with 72 showing a difference in expression. Apart from blood-related proteins (such as serum albumin), both techniques showed that the 70 kDa family of heat shock proteins were highly expressed in the peri-infarct tissue. Further studies by 1D and 2D western blotting and immunohistochemistry revealed that only one member of this family (the inducible form, HSP72 or HSP70i) is specifically expressed by the peri-infarct tissue, while the majority of this family (the constitutive form, HSC70 or HSP70c) is expressed in the whole brain. Our data support that HSP72 is a suitable biomarker of peri-infarct tissue in the ischemic brain.
Collapse
Affiliation(s)
- David Brea
- 1] Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain [2] Cellular and Molecular Neurobiology Research Group and Grup de Recerça en Neurociencies del IGTP, Department of Neurosciences, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias I Pujol-Universitat Autónoma de Barcelona, Badalona, Spain
| | - Jesús Agulla
- 1] Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain [2] Research Unit, University Hospital of Salamanca and Institute of Health Sciences of Castilla and Leon, Salamanca, Spain
| | - An Staes
- 1] Department of Medical Protein Research, VIB, Ghent, Belgium [2] Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Kris Gevaert
- 1] Department of Medical Protein Research, VIB, Ghent, Belgium [2] Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Francisco Campos
- Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Tomás Sobrino
- Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Miguel Blanco
- Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Antoni Dávalos
- Cellular and Molecular Neurobiology Research Group and Grup de Recerça en Neurociencies del IGTP, Department of Neurosciences, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias I Pujol-Universitat Autónoma de Barcelona, Badalona, Spain
| | - José Castillo
- Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| | - Pedro Ramos-Cabrer
- Neurology Department, Neurovascular Area, Clinical Neurosciences Research Laboratory, University Clinical Hospital, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Spain
| |
Collapse
|
36
|
Uncovering Neurodegenerative Protein Modifications via Proteomic Profiling. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 121:87-116. [DOI: 10.1016/bs.irn.2015.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
37
|
Elliott AS, Huber JD, O'Callaghan JP, Rosen CL, Miller DB. A review of sleep deprivation studies evaluating the brain transcriptome. SPRINGERPLUS 2014; 3:728. [PMID: 25932362 PMCID: PMC4409616 DOI: 10.1186/2193-1801-3-728] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/25/2014] [Indexed: 12/14/2022]
Abstract
Epidemiological studies show a positive association between adequate sleep and good health. Further, disrupted sleep may increase the risk for CNS diseases, such as stroke and Alzheimer’s disease. However, there has been limited progress in determining how sleep is linked to brain health or how sleep disruption may increase susceptibility to brain insult and disease. Animal studies can aid in understanding these links. In reviewing the animal literature related to the effects of sleep disruption on the brain, we found most of the work was directed toward investigating and characterizing the role of various brain areas or structures in initiating and regulating sleep. In contrast, limited effort has been directed towards understanding how sleep disruption alters the brain’s health or susceptibility to insult. We also note many current studies have determined the changes in the brain following compromised sleep by examining, for example, the brain transcriptome or to a more limited extent the proteome. However, these studies have utilized almost exclusively total sleep deprivation (e.g., 24 out of 24 hours) paradigms or single short periods of limited acute sleep deprivation (e.g., 3 out of 24 hours). While such strategies are beneficial in understanding how sleep is controlled, they may not have much translational value for determining links between sleep and brain health or for determining how sleep disruption may increase brain susceptibility to insult. Surprisingly, few studies have determined how the duration and recurrence of sleep deprivation influence the effects seen after sleep deprivation. Our aim in this review was to identify relevant rodent studies from 1980 through 2012 and analyze those that use varying durations of sleep deprivation or restriction in their effort to evaluate the effects of sleep deprivation on the brain transcriptome and to a more limited extent the proteome. We examined how differences in the duration of sleep deprivation affect gene and protein expression to better understand the full consequences of repeated sleep disruption on the brain. Future research needs to consider and emphasize how the type and extent of the sleep deprivation exposure impacts the conclusions reached concerning the influence of sleep disruption on the brain. We identified relevant studies between 1980 and 2012 by searching the electronic databases of PubMed, Medline (Ovid), Embase (Ovid), and Web of Science using the terms “sleep” AND “disrupt”, “deprivation”, “restrict”, “fragment”, “loss”, “disturb”, “disorder”, “dysfunction”, “brain”, “cortex”, striatum”, hypothalamus”, “hippocampus”, “gene”, “protein”, “genomics”, “proteomics”, “polymerase chain reaction”, “pcr”, “microarray”, “molecular”, “rodent” “rat”, “rats”, “mouse”, “mice”. All searches were limited to rodent studies in English and the reference lists of retrieved articles were searched for additional pertinent studies.
Collapse
Affiliation(s)
- Alisa S Elliott
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Jason D Huber
- School of Pharmacy, West Virginia University, Morgantown, WV USA
| | - James P O'Callaghan
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| | - Charles L Rosen
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Diane B Miller
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| |
Collapse
|
38
|
Lu H, Hu H, He Z, Han X, Chen J, Tu R. Therapeutic imaging window of cerebral infarction revealed by multisequence magnetic resonance imaging: An animal and clinical study. Neural Regen Res 2014; 7:2446-55. [PMID: 25337095 PMCID: PMC4200719 DOI: 10.3969/j.issn.1673-5374.2012.31.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/09/2012] [Indexed: 12/17/2022] Open
Abstract
In this study, we established a Wistar rat model of right middle cerebral artery occlusion and observed pathological imaging changes (T2-weighted imaging [T2WI], T2FLAIR, and diffusion-weighted imaging [DWI]) following cerebral infarction. The pathological changes were divided into three phases: early cerebral infarction, middle cerebral infarction, and late cerebral infarction. In the early cerebral infarction phase (less than 2 hours post-infarction), there was evidence of intracellular edema, which improved after reperfusion. This improvement was defined as the ischemic penumbra. In this phase, a high DWI signal and a low apparent diffusion coefficient were observed in the right basal ganglia region. By contrast, there were no abnormal T2WI and T2FLAIR signals. For the middle cerebral infarction phase (2–4 hours post-infarction), a mixed edema was observed. After reperfusion, there was a mild improvement in cell edema, while the angioedema became more serious. A high DWI signal and a low apparent diffusion coefficient signal were observed, and some rats showed high T2WI and T2FLAIR signals. For the late cerebral infarction phase (4–6 hours post-infarction), significant angioedema was visible in the infarction site. After reperfusion, there was a significant increase in angioedema, while there was evidence of hemorrhage and necrosis. A mixed signal was observed on DWI, while a high apparent diffusion coefficient signal, a high T2WI signal, and a high T2FLAIR signal were also observed. All 86 cerebral infarction patients were subjected to T2WI, T2FLAIR, and DWI. MRI results of clinic data similar to the early infarction phase of animal experiments were found in 51 patients, for which 10 patients (10/51) had an onset time greater than 6 hours. A total of 35 patients had MRI results similar to the middle and late infarction phase of animal experiments, of which eight patients (8/35) had an onset time less than 6 hours. These data suggest that defining the “therapeutic time window” as the time 6 hours after infarction may not be suitable for all patients. Integrated application of MRI sequences including T2WI, T2FLAIR, DW-MRI, and apparent diffusion coefficient mapping should be used to examine the ischemic penumbra, which may provide valuable information for identifying the “therapeutic time window”.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Hui Hu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Zhanping He
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Xiangjun Han
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Jing Chen
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Haikou Municipal People's Hospital), Haikou 570208, Hainan Province, China
| | - Rong Tu
- Department of Radiology, Affiliated Hospital of Hainan Medical University, Haikou 570102, Hainan Province, China
| |
Collapse
|
39
|
Merali Z, Gao MM, Bowes T, Chen J, Evans K, Kassner A. Neuroproteome changes after ischemia/reperfusion injury and tissue plasminogen activator administration in rats: a quantitative iTRAQ proteomics study. PLoS One 2014; 9:e98706. [PMID: 24879061 PMCID: PMC4039533 DOI: 10.1371/journal.pone.0098706] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 05/02/2014] [Indexed: 01/02/2023] Open
Abstract
The thrombolytic, recombinant tissue plasminogen activator (rt-PA) is the only approved therapy for acute ischemic stroke (AIS). When administered after AIS, rt-PA has many adverse pleiotropic actions, which are currently poorly understood. The identification of proteins showing differential expression after rt-PA administration may provide insight into these pleiotropic actions. In this study we used a 2D-LC MS/MS iTRAQ proteomic analysis, western blotting, and pathway analysis to analyze changes in protein expression 24-hours after rt-PA administration in the cortical brain tissue of 36 rats that underwent a sham or transient middle cerebral artery occlusion surgery. After rt-PA administration we reported alterations in the expressions of 18 proteins, many of which were involved in excitatory neurotransmitter function or cytoskeletal structure. The expression changes of GAD2 and EAAT1 were validated with western blot. The interactions between the identified proteins were analyzed with the IPA pathway analysis tool and three proteins: DPYSL2, RTN4, and the NF-kB complex, were found to have characteristics of being key proteins in the network. The differential protein expressions we observed may reflect pleiotropic actions of rt-PA after experimental stroke, and shine light on the mechanisms of rt-PA's adverse effects. This may have important implications in clinical settings where thrombolytic therapy is used to treat AIS.
Collapse
Affiliation(s)
- Zamir Merali
- Physiology and Experimental Medicine, The Hospital For Sick Children, Toronto, Ontario, Canada
| | - Meah MingYang Gao
- Department of Medical Imaging, University Of Toronto, Toronto, Ontario, Canada
| | - Tim Bowes
- Department of Medical Imaging, University Of Toronto, Toronto, Ontario, Canada
| | - Jian Chen
- Ontario Cancer Biomarker Network, Toronto, Ontario Canada
| | - Kenneth Evans
- Ontario Cancer Biomarker Network, Toronto, Ontario Canada
| | - Andrea Kassner
- Department of Medical Imaging, University Of Toronto, Toronto, Ontario, Canada
- Physiology and Experimental Medicine, The Hospital For Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Datta A, Chen CP, Sze SK. Discovery of prognostic biomarker candidates of lacunar infarction by quantitative proteomics of microvesicles enriched plasma. PLoS One 2014; 9:e94663. [PMID: 24752076 PMCID: PMC3994162 DOI: 10.1371/journal.pone.0094663] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 03/18/2014] [Indexed: 11/25/2022] Open
Abstract
Background Lacunar infarction (LACI) is a subtype of acute ischemic stroke affecting around 25% of all ischemic stroke cases. Despite having an excellent recovery during acute phase, certain LACI patients have poor mid- to long-term prognosis due to the recurrence of vascular events or a decline in cognitive functions. Hence, blood-based biomarkers could be complementary prognostic and research tools. Methods and Finding Plasma was collected from forty five patients following a non-disabling LACI along with seventeen matched control subjects. The LACI patients were monitored prospectively for up to five years for the occurrence of adverse outcomes and grouped accordingly (i.e., LACI-no adverse outcome, LACI-recurrent vascular event, and LACI-cognitive decline without any recurrence of vascular events). Microvesicles-enriched fractions isolated from the pooled plasma of four groups were profiled by an iTRAQ-guided discovery approach to quantify the differential proteome. The data have been deposited to the ProteomeXchange with identifier PXD000748. Bioinformatics analysis and data mining revealed up-regulation of brain-specific proteins including myelin basic protein, proteins of coagulation cascade (e.g., fibrinogen alpha chain, fibrinogen beta chain) and focal adhesion (e.g., integrin alpha-IIb, talin-1, and filamin-A) while albumin was down-regulated in both groups of patients with adverse outcome. Conclusion This data set may offer important insight into the mechanisms of poor prognosis and provide candidate prognostic biomarkers for validation on larger cohort of individual LACI patients.
Collapse
Affiliation(s)
- Arnab Datta
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher P. Chen
- Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- * E-mail:
| |
Collapse
|
41
|
Datta A, Qian J, Chong R, Kalaria RN, Francis P, Lai MKP, Chen CP, Sze SK. Novel pathophysiological markers are revealed by iTRAQ-based quantitative clinical proteomics approach in vascular dementia. J Proteomics 2014; 99:54-67. [PMID: 24448401 PMCID: PMC4024194 DOI: 10.1016/j.jprot.2014.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 12/10/2013] [Accepted: 01/08/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED Vascular dementia (VaD) is a leading cause of dementia in the elderly together with Alzheimer's disease with limited treatment options. Poor understanding of the pathophysiology underlying VaD is hindering the development of new therapies. Hence, to unravel its underlying molecular pathology, an iTRAQ-2D-LC-MS/MS strategy was used for quantitative analysis of pooled lysates from Brodmann area 21 of pathologically confirmed cases of VaD and matched non-neurological controls. A total of 144 differentially expressed proteins out of 2281 confidently identified proteins (false discovery rate=0.3%) were shortlisted for bioinformatics analysis. Western blot analysis of selected proteins using samples from individual patients (n=10 per group) showed statistically significant increases in the abundance of SOD1 and NCAM and reduced ATP5A in VaD. This suggested a state of hypometabolism and vascular insufficiency along with an inflammatory condition during VaD. Elevation of SOD1 and increasing trend for iron-storage proteins (FTL, FTH1) may be indicative of an oxidative imbalance that is accompanied by an aberrant iron metabolism. The synaptic proteins did not exhibit a generalized decrease in abundance (e.g. syntaxin) in the VaD subjects. This reported proteome offers a reference data set for future basic or translational studies on VaD. BIOLOGICAL SIGNIFICANCE Our study is the first quantitative clinical proteomic study where iTRAQ-2D-LC-MS/MS strategy has been used to identify the differential proteome in the VaD cortex by comparing VaD and matched control subjects. We generate testable hypothesis about the involvement of various proteins in the vascular and parenchymal events during the evolution of VaD that finally leads to malfunction and demise of brain cells. This study also establishes quantitative proteomics as a complementary approach and viable alternative to existing neurochemical, electron microscopic and neuroimaging techniques that are traditionally being used to understand the molecular pathology of VaD. Our study could inspire fellow researchers to initiate similar retrospective studies targeting various ethnicities, age-groups or sub-types of VaD using brain samples available from brain banks across the world. Meta-analysis of these studies in the future may be able to shortlist candidate proteins or pathways for rationale exploration of therapeutic targets or biomarkers for VaD.
Collapse
Affiliation(s)
- Arnab Datta
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jingru Qian
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ruifen Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Raj N Kalaria
- Institute for Ageing Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Paul Francis
- Wolfson Centre for Age-related Diseases, King's College London, London, UK
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore.
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
42
|
Datta A, Akatsu H, Heese K, Sze SK. Quantitative clinical proteomic study of autopsied human infarcted brain specimens to elucidate the deregulated pathways in ischemic stroke pathology. J Proteomics 2013; 91:556-68. [PMID: 24007662 DOI: 10.1016/j.jprot.2013.08.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/15/2013] [Accepted: 08/20/2013] [Indexed: 11/26/2022]
Abstract
UNLABELLED Ischemic stroke, still lacking an effective neuroprotective therapy is the third leading cause of global mortality and morbidity. Here, we have applied an 8-plex iTRAQ-based 2D-LC-MS/MS strategy to study the commonly regulated infarct proteome from three different brain regions (putamen, thalamus and the parietal lobe) of female Japanese patients. Infarcts were compared with age-, post-mortem interval- and location-matched control specimens. The iTRAQ experiment confidently identified 1520 proteins with 0.1% false discovery rate. Bioinformatics data mining and immunochemical validation of pivotal perturbed proteins revealed a global failure of the cellular energy metabolism in the infarcted tissues as seen by the parallel down-regulation of proteins related to glycolysis, pyruvate dehydrogenase complex, TCA cycle and oxidative phosphorylation. The concomitant down-regulation of all participating proteins (SLC25A11, SLC25A12, GOT2 and MDH2) of malate-aspartate shuttle might be responsible for the metabolic in-coordination between the cytosol and mitochondria resulting in the failure of energy metabolism. The levels of proteins related to reactive gliosis (VIM, GFAP) and anti-inflammatory response (ANXA1, ANXA2) showed an increasing trend. The elevation of ferritin (FTL, FTH1) may indicate an iron-mediated oxidative imbalance aggravating the mitochondrial failure and neurotoxicity. The deregulated proteins could be useful as potential therapeutic targets or biomarkers for ischemic stroke. BIOLOGICAL SIGNIFICANCE Clinical proteomics of stroke has been lagging behind other areas of clinical proteomics like Alzheimer's disease or schizophrenia. Our study is the first quantitative clinical proteomics study where iTRAQ-2D-LC-MS/MS has been utilized in the area of ischemic stroke to obtain a comparative profile of human ischemic infarcts and age-, sex-, location- and post-mortem interval-matched control brain specimens. Different pathological attributes of ischemic stroke well-known through basic and pre-clinical research such as failure of cellular energy metabolism, reactive gliosis, activation of anti-inflammatory response and aberrant iron metabolism have been observed at the bedside. Our dataset could act as a reference for similar studies done in the future using ischemic brain samples from various brain banks across the world. A meta-analysis of these studies could help to map the pathological proteome specific to ischemic stroke that will guide the scientific community to better evaluate the pros and cons of the pre-clinical models for efficacy and mechanistic studies. Infarct being the core of injury should have the most intense regulation for several key proteins involved in the pathophysiology of ischemic stroke. Hence, a part of the up-regulated proteome could leak into the general circulation that may offer candidates of interest as potential biomarkers. In support of our proposed hypothesis, we report ferritin in the current study as one of the most elevated proteins in the infarct, which has been documented as a biomarker in the context of ischemic stroke by an independent study. Overall, our approach has the potential to identify probable therapeutic targets and biomarkers in the area of ischemic stroke.
Collapse
Affiliation(s)
- Arnab Datta
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | | | | | | |
Collapse
|
43
|
Yang HY, Kwon J, Kook MS, Kang SS, Kim SE, Sohn S, Jung S, Kwon SO, Kim HS, Lee JH, Lee TH. Proteomic analysis of gingival tissue and alveolar bone during alveolar bone healing. Mol Cell Proteomics 2013; 12:2674-88. [PMID: 23824910 DOI: 10.1074/mcp.m112.026740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bone tissue regeneration is orchestrated by the surrounding supporting tissues and involves the build-up of osteogenic cells, which orchestrate remodeling/healing through the expression of numerous mediators and signaling molecules. Periodontal regeneration models have proven useful for studying the interaction and communication between alveolar bone and supporting soft tissue. We applied a quantitative proteomic approach to analyze and compare proteins with altered expression in gingival soft tissue and alveolar bone following tooth extraction. For target identification and validation, hard and soft tissue were extracted from mini-pigs at the indicated times after tooth extraction. From triplicate experiments, 56 proteins in soft tissue and 27 proteins in alveolar bone were found to be differentially expressed before and after tooth extraction. The expression of 21 of those proteins was altered in both soft tissue and bone. Comparison of the activated networks in soft tissue and alveolar bone highlighted their distinct responsibilities in bone and tissue healing. Moreover, we found that there is crosstalk between identified proteins in soft tissue and alveolar bone with respect to cellular assembly, organization, and communication. Among these proteins, we examined in detail the expression patterns and associated networks of ATP5B and fibronectin 1. ATP5B is involved in nucleic acid metabolism, small molecule biochemistry, and neurological disease, and fibronectin 1 is involved in cellular assembly, organization, and maintenance. Collectively, our findings indicate that bone regeneration is accompanied by a profound interaction among networks regulating cellular resources, and they provide novel insight into the molecular mechanisms involved in the healing of periodontal tissue after tooth extraction.
Collapse
Affiliation(s)
- Hee-Young Yang
- Department of Oral Biochemistry, Dental Science Research Institute and the BK21 Project, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Craft GE, Chen A, Nairn AC. Recent advances in quantitative neuroproteomics. Methods 2013; 61:186-218. [PMID: 23623823 PMCID: PMC3891841 DOI: 10.1016/j.ymeth.2013.04.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 03/29/2013] [Accepted: 04/13/2013] [Indexed: 01/07/2023] Open
Abstract
The field of proteomics is undergoing rapid development in a number of different areas including improvements in mass spectrometric platforms, peptide identification algorithms and bioinformatics. In particular, new and/or improved approaches have established robust methods that not only allow for in-depth and accurate peptide and protein identification and modification, but also allow for sensitive measurement of relative or absolute quantitation. These methods are beginning to be applied to the area of neuroproteomics, but the central nervous system poses many specific challenges in terms of quantitative proteomics, given the large number of different neuronal cell types that are intermixed and that exhibit distinct patterns of gene and protein expression. This review highlights the recent advances that have been made in quantitative neuroproteomics, with a focus on work published over the last five years that applies emerging methods to normal brain function as well as to various neuropsychiatric disorders including schizophrenia and drug addiction as well as of neurodegenerative diseases including Parkinson's disease and Alzheimer's disease. While older methods such as two-dimensional polyacrylamide electrophoresis continued to be used, a variety of more in-depth MS-based approaches including both label (ICAT, iTRAQ, TMT, SILAC, SILAM), label-free (label-free, MRM, SWATH) and absolute quantification methods, are rapidly being applied to neurobiological investigations of normal and diseased brain tissue as well as of cerebrospinal fluid (CSF). While the biological implications of many of these studies remain to be clearly established, that there is a clear need for standardization of experimental design and data analysis, and that the analysis of protein changes in specific neuronal cell types in the central nervous system remains a serious challenge, it appears that the quality and depth of the more recent quantitative proteomics studies is beginning to shed light on a number of aspects of neuroscience that relates to normal brain function as well as of the changes in protein expression and regulation that occurs in neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- George E Craft
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Anshu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06508
- Yale/NIDA Neuroproteomics Center, Yale University School of Medicine, New Haven, CT, 06508
| |
Collapse
|
45
|
Crepaldi CR, Vitale PAM, Tesch AC, Laure HJ, Rosa JC, de Cerqueira César M. Application of 2D BN/SDS-PAGE coupled with mass spectrometry for identification of VDAC-associated protein complexes related to mitochondrial binding sites for type I brain hexokinase. Mitochondrion 2013; 13:823-30. [PMID: 23719229 DOI: 10.1016/j.mito.2013.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 02/27/2013] [Accepted: 05/14/2013] [Indexed: 11/25/2022]
Abstract
Two types of binding sites for hexokinase, designated as Type A or Type B sites, have been shown to coexist on brain mitochondria. The ratio of these sites varies between species. HK1 attaches by reversibly binding to the voltage dependent anion channel (VDAC). Regarding the nature of hexokinase binding sites, we investigated if it was linked to distinct VDAC interactomes. We approached this question by 2D BN/SDS-PAGE of mitochondria, followed by mass spectrometry. Our results are consistent with the possibility that the ratio of Type A/Type B sites is due to differential VDAC interactions in bovine and rat neuronal cells.
Collapse
Affiliation(s)
- Carla Rossini Crepaldi
- Department of Basic Sciences, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | | | | | | | | | | |
Collapse
|
46
|
Feng L, Manavalan A, Mishra M, Sze SK, Hu JM, Heese K. Tianma modulates blood vessel tonicity. Open Biochem J 2012; 6:56-65. [PMID: 22787517 PMCID: PMC3391654 DOI: 10.2174/1874091x01206010056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 04/10/2012] [Accepted: 04/24/2012] [Indexed: 01/23/2023] Open
Abstract
Tianma is a traditional Chinese medicine (TCM) often used for the treatment of hypertension and heart diseases. To elucidate the function of tianma at the molecular level, we investigated the effect of tianma on vascular functions and aortic protein metabolism. We found that long-term treatment with tianma (~2.5g/kg/day for three months) in one-year-old rats could enhance acetylcholine (ACh)-induced vasorelaxation in endothelium-intact thoracic aortic rings against both KCl (80 mM)- and phenylephrine (PE)-induced contraction. By using the iTRAQ (isobaric tag for relative and absolute quantification) technique, we confirmed from the functional data at the proteome level that tianma treatment down-regulated the expressions of contractile proteins (e.g. Acta2) and other related structural proteins (e.g. desmin), and up-regulated the expressions of extracellular matrix (ECM) glycoproteins (e.g. Fbln5) and anti-thrombotic proteins (e.g. Anxa2) in aortic tissue. By inductive reasoning, tianma could perform its vasodilatory effect not only by inhibiting vascular smooth muscle contraction, but also by enhancing blood vessel elasticity and stabilizing the arterial structure. Thus, tianma might become a novel therapeutic herbal medicine for cardiovascular diseases by regulating the aortic proteome metabolism.
Collapse
Affiliation(s)
- Lin Feng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | | | | | | | | | | |
Collapse
|
47
|
Ramachandran U, Manavalan A, Sundaramurthi H, Sze SK, Feng ZW, Hu JM, Heese K. Tianma modulates proteins with various neuro-regenerative modalities in differentiated human neuronal SH-SY5Y cells. Neurochem Int 2012; 60:827-36. [PMID: 22710396 DOI: 10.1016/j.neuint.2012.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/29/2012] [Accepted: 03/19/2012] [Indexed: 12/15/2022]
Abstract
Tianma (Rhizoma gastrodiae) is the dried rhizome of the plant Gastrodia elata Blume (Orchidaceae family). As a medicinal herb in traditional Chinese medicine (TCM) its functions are to control convulsions, pain, headache, dizziness, vertigo, seizure, epilepsy and others. In addition, tianma is frequently used for the treatment of neurodegenerative disorders though the mechanism of action is widely unknown. Accordingly, this study was designed to examine the effects of tianma on the proteome metabolism in differentiated human neuronal SH-SY5Y cells to explore its specific effects on neuronal signaling pathways. Using an iTRAQ (isobaric tags for relative and absolute quantitation)-based proteomics research approach, we identified 2390 modulated proteins, out of which 406 were found to be altered by tianma in differentiated human neuronal SH-SY5Y cells. Based on the observed data, we hypothesize that tianma promotes neuro-regenerative signaling cascades by controlling chaperone/proteasomal degradation pathways (e.g. CALR, FKBP3/4, HSP70/90) and mobilizing neuro-protective genes (such as AIP5) as well as modulating other proteins (RTN1/4, NCAM, PACSIN2, and PDLIM1/5) with various regenerative modalities and capacities related to neuro-synaptic plasticity.
Collapse
|
48
|
Manavalan A, Feng L, Sze SK, Hu JM, Heese K. New insights into the brain protein metabolism of Gastrodia elata-treated rats by quantitative proteomics. J Proteomics 2012; 75:2468-79. [PMID: 22402058 DOI: 10.1016/j.jprot.2012.02.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 02/13/2012] [Accepted: 02/20/2012] [Indexed: 01/05/2023]
Abstract
Gastrodia elata (tianma) is a traditional Chinese herbal medicine (TCM) often used for the treatment of cerebrovascular diseases. In this study, we investigated the effects of tianma on the brain protein metabolism by quantitative proteomics to gain evidence for a direct relationship between tianma treatment and brain functions. One-year-old rats were treated with tianma (~2.5 g/kg/day) for 3months and the brain tissue proteome was analyzed by using the iTRAQ (isobaric tag for relative and absolute quantification) technology. According to our results, the long-term treatment with tianma could modulate the brain protein metabolism at the proteome level by down-regulating the expressions of various proteins, such as Gnao1 and Dctn2, which are related to neuronal growth cone control and synaptic activities. In addition, tianma treatment also induced the up-regulation of molecular chaperons and proteins related to the misfolded protein response, like Anxa5, and also other proteins involved in Huntington's disease (HD) (e.g. Pacsin1 and Arf3). Concluding, tianma could eventually contribute to activities related to synaptic plasticity and neuro-restorative processes and thus might be a novel candidate agent for the treatment of neurodegenerative diseases by regulating the brain proteome.
Collapse
Affiliation(s)
- Arulmani Manavalan
- School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | | | | | | | | |
Collapse
|
49
|
Protein Biomarkers for Traumatic and Ischemic Brain Injury: From Bench to Bedside. Transl Stroke Res 2011; 2:455-62. [DOI: 10.1007/s12975-011-0137-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/07/2011] [Accepted: 11/08/2011] [Indexed: 12/31/2022]
|