1
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMID: 39766257 PMCID: PMC11673996 DOI: 10.3390/biom14121550] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
2
|
Karimova D, Rostami E, Chubarev VN, Tarasov VV, Schiöth HB, Rask-Andersen M. Advances in development of biomarkers for brain damage and ischemia. Mol Biol Rep 2024; 51:803. [PMID: 39001884 PMCID: PMC11246271 DOI: 10.1007/s11033-024-09708-x] [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: 02/23/2024] [Accepted: 06/06/2024] [Indexed: 07/15/2024]
Abstract
Acquired brain injury is an urgent situation that requires rapid diagnosis and treatment. Magnetic resonance imaging (MRI) and computed tomography (CT) are required for accurate diagnosis. However, these methods are costly and require substantial infrastructure and specialized staff. Circulatory biomarkers of acute brain injury may help in the management of patients with acute cerebrovascular events and prevent poor outcome and mortality. The purpose of this review is to provide an overview of the development of potential biomarkers of brain damage to increase diagnostic possibilities. For this purpose, we searched the PubMed database of studies on the diagnostic potential of brain injury biomarkers. We also accessed information from Clinicaltrials.gov to identify any clinical trials of biomarker measurements for the diagnosis of brain damage. In total, we present 41 proteins, enzymes and hormones that have been considered as biomarkers for brain injury, of which 20 have been studied in clinical trials. Several microRNAs have also emerged as potential clinical biomarkers for early diagnosis. Combining multiple biomarkers in a panel, along with other parameters, is yielding promising outcomes.
Collapse
Affiliation(s)
- Diana Karimova
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala, University, Uppsala, Sweden
| | - Elham Rostami
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Vladimir N Chubarev
- Advanced Molecular Technology, Limited Liable Company (LLC), Moscow, 354340, Russia
| | - Vadim V Tarasov
- Advanced Molecular Technology, Limited Liable Company (LLC), Moscow, 354340, Russia
| | - Helgi B Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala, University, Uppsala, Sweden
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
3
|
Zhang W, Zhang J, Wang Y, Wang S, Wu Y, Zhang W, Wu M, Wang L, Xu G, Deng F, Liu W, Liu Z, Chen L, Xiao K, Zhang L. In Vitro Detection of S100B and Severity Evaluation of Traumatic Brain Injury Based on Biomimetic Peptide-Modified Nanochannels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306809. [PMID: 38009781 DOI: 10.1002/smll.202306809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/26/2023] [Indexed: 11/29/2023]
Abstract
The diagnosis and evaluation of traumatic brain injury (TBI) are crucial steps toward the treatment and prognosis of patients. A common question remains as to whether it is possible to introduce an ideal device for signal detection and evaluation that can directly connect digital signals with TBI, thereby enabling prompt response of the evaluation signal and sensitive and specific functioning of the detection process. Herein, a method is presented utilizing polymetric porous membranes with TRTK-12 peptide-modified nanochannels for the detection of S100B (a TBI biomarker) and assessment of TBI severity. The method leverages the specific bonding force between TRTK-12 peptide and S100B protein, along with the nanoconfinement effect of nanochannels, to achieve high sensitivity (LOD: 0.002 ng mL-1) and specificity (∆I/I0: 44.7%), utilizing ionic current change as an indicator. The proposed method, which is both sensitive and specific, offers a simple yet responsive approach for real-time evaluation of TBI severity. This innovative technique provides valuable scientific insights into the advancement of future diagnostic and therapeutic integration devices.
Collapse
Affiliation(s)
- Wenyuan Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, P. R. China
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, P. R. China
| | - Jianrui Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yijun Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Senyao Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yitian Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Wenchang Zhang
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, P. R. China
| | - Minghui Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Li Wang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Guoheng Xu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Fuan Deng
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Wenchao Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Zhengwei Liu
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, P. R. China
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, P. R. China
| | - Lu Chen
- School of Materials and Environmental Engineering, Shenzhen Polytechnic, Shenzhen, 518055, P. R. China
| | - Kai Xiao
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| |
Collapse
|
4
|
Samartsev IN, Zhivolupov SA, Gorbatenkova OV, Ponomarev VV, Butakova JS. [Biomarkers of neuroinflammation in patients with chronic cerebral ischemia during the therapy with vinpocetine (study INFLAMARK)]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:50-58. [PMID: 38147382 DOI: 10.17116/jnevro202312312150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
OBJECTIVE To evaluate the effect of vinpocetine therapy on clinical manifestations of chronic cerebral ischemia (CCI) and the blood concentrations of neuroinflammation markers (S100B, IL-1β). MATERIAL AND METHODS The study included 30 patients (mean age 61.6 [56.9; 67.9] years) with CCI that received vinpocetine (30 mg/day) for 3 months. Brain changes according to magnetic resonance imaging data were assessed using the STRIVE protocol. We analyzed the dynamics of changes in the clinical questionnaires: Montreal Cognitive Assessment Scale (MoCA), Hospital Anxiety and Depression Scale (HADS), Asthenic State Scale (ASS), Epworth Sleepiness Scale (ESS), general impressions of treatment (Global Rating of Change Scale, GRC). RESULTS In 3 months after vinpocetine therapy there was a significant improvement in cognitive status (MoCA: 25.1±2.1 vs 26.6±1.4 p<0.05), emotional state (HADS: 8.4±1.4 vs 7.1±1.8 (p<0.05)), daytime sleep parameters (ESS 8.4±2.1 vs 6.2±2.3 p<0.05) and reduction in asthenia (ASS: 72.2±18.1 vs 52.3±9.3, p<0.05). A significantly larger proportion of patients assessed the improvement from therapy as «moderate» and «pronounced» (GRC, n=22, 73.3%). Concentrations of S100B and IL-1β decreased significantly by the time therapy was completed. The overall severity of cerebrovascular changes according to MRI was significantly associated with blood levels of S100β, but not IL-1β: β=0.504, p=0.026, 95% CI 0.149-0.901, mainly due to periventricular changes in white matter (β=0.562, p=0.035, 95% CI (-0.024-0.820). Blood levels of S100β correlated with MoCA test results (r=0.6795), and IL-1β correlated with ESS scores (r=0. 6657). CONCLUSIONS The use of vinpocetine can significantly reduce the severity of cognitive and affective disorders, asthenia, normalize the circadian rhythm of sleep, suppress the expression S100β and IL-1β in patients with CCI. One of the vinpocetine's mechanisms of action may be the inhibition of neuroinflammation.
Collapse
Affiliation(s)
- I N Samartsev
- Kirov Military medical academy, St. Petersburg, Russia
| | | | | | | | - J S Butakova
- Novodvinsk Central City Hospital, Novodvinsk, Russia
| |
Collapse
|
5
|
Seol S, Kwon J, Kang HJ. Cell type characterization of spatiotemporal gene co-expression modules in Down syndrome brain. iScience 2022; 26:105884. [PMID: 36647384 PMCID: PMC9840153 DOI: 10.1016/j.isci.2022.105884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/02/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability and increases the risk of other brain-related dysfunctions, like seizures, early-onset Alzheimer's disease, and autism. To reveal the molecular profiles of DS-associated brain phenotypes, we performed a meta-data analysis of the developmental DS brain transcriptome at cell type and co-expression module levels. In the DS brain, astrocyte-, microglia-, and endothelial cell-associated genes show upregulated patterns, whereas neuron- and oligodendrocyte-associated genes show downregulated patterns. Weighted gene co-expression network analysis identified cell type-enriched co-expressed gene modules. We present eight representative cell-type modules for neurons, astrocytes, oligodendrocytes, and microglia. We classified the neuron modules into glutamatergic and GABAergic neurons and associated them with detailed subtypes. Cell type modules were interpreted by analyzing spatiotemporal expression patterns, functional annotations, and co-expression networks of the modules. This study provides insight into the mechanisms underlying brain abnormalities in DS and related disorders.
Collapse
Affiliation(s)
- Sihwan Seol
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Joonhong Kwon
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Hyo Jung Kang
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea,Corresponding author
| |
Collapse
|
6
|
Chu C, Zhong R, Cai M, Li N, Lin W. Elevated Blood S100B Levels in Patients With Migraine: A Systematic Review and Meta-Analysis. Front Neurol 2022; 13:914051. [PMID: 35911929 PMCID: PMC9329586 DOI: 10.3389/fneur.2022.914051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In recent years, a growing number of researches indicate that S100B may act in migraine, but the relationship between S100B and migraine remains controversial. Therefore, the current study aimed to perform a meta-analysis to quantitatively summarize S100B levels in migraine patients. Methods We used Stata 12.0 software to summarize eligible studies from PubMed, EMBASE, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI), and Wanfang databases. We applied standardized mean differences (SMDs) with 95% confidence intervals (95%CIs) to appraise the association between S100B and migraine. Results The combined results of nine case-control studies indicated that compared with healthy controls, overall migraine patients had significantly increased S100B levels in peripheral blood (SMD = 0.688, 95%CI: 0.341–1.036, P < 0.001). The S100B levels in migraineurs during ictal periods (SMD =1.123, 95%CI: 0.409–1.836, P = 0.002) and interictal periods (SMD = 0.487, 95%CI: 0313–0.661, P < 0.001), aura (SMD = 0.999, 95%CI: 0.598–1.400, P < 0.001) and without aura (SMD = 0.534, 95%CI: 0.286–0.783, P < 0.001) were significantly higher than those in the controls. The subgroup analyses by age, country, migraine assessment, and assay method of S100B also illustrated a statistically obvious association between S100B levels and migraine, indicating that age may be the most important source of heterogeneity. Sensitivity analysis showed that no individual study has a significant influence on the overall association between S100B and migraine. Conclusion This meta-analysis demonstrates that the level of S100B in peripheral blood of patients with migraine was significantly increased. Migraine may be associated with pathological reactions involving S100B, which is instrumental for the clinical diagnosis of migraine and therapy that considers S100B as a potential target.
Collapse
|
7
|
Michetti F, Di Sante G, Clementi ME, Sampaolese B, Casalbore P, Volonté C, Romano Spica V, Parnigotto PP, Di Liddo R, Amadio S, Ria F. Growing role of S100B protein as a putative therapeutic target for neurological- and nonneurological-disorders. Neurosci Biobehav Rev 2021; 127:446-458. [PMID: 33971224 DOI: 10.1016/j.neubiorev.2021.04.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023]
Abstract
S100B is a calcium-binding protein mainly expressed by astrocytes, but also localized in other definite neural and extra-neural cell types. While its presence in biological fluids is widely recognized as a reliable biomarker of active injury, growing evidence now indicates that high levels of S100B are suggestive of pathogenic processes in different neural, but also extra-neural, disorders. Indeed, modulation of S100B levels correlates with the occurrence of clinical and/or toxic parameters in experimental models of diseases such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, muscular dystrophy, multiple sclerosis, acute neural injury, inflammatory bowel disease, uveal and retinal disorders, obesity, diabetes and cancer, thus directly linking the levels of S100B to pathogenic mechanisms. In general, deletion/inactivation of the protein causes the improvement of the disease, whereas its over-expression/administration induces a worse clinical presentation. This scenario reasonably proposes S100B as a common therapeutic target for several different disorders, also offering new clues to individuate possible unexpected connections among these diseases.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| | - Gabriele Di Sante
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168 Rome, Italy.
| | - Maria Elisabetta Clementi
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Beatrice Sampaolese
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta" SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Patrizia Casalbore
- Institute for Systems Analysis and Computer Science, IASI-CNR, Largo Francesco Vito 1, 00168 Rome, Italy.
| | - Cinzia Volonté
- Institute for Systems Analysis and Computer Science, IASI-CNR, Largo Francesco Vito 1, 00168 Rome, Italy; Cellular Neurobiology Unit, Preclinical Neuroscience, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Vincenzo Romano Spica
- Department of Movement, Human and Health Sciences, Laboratory of Epidemiology and Biotechnologies, University of Rome "Foro Italico", Piazza Lauro De Bosis 6, 00135, Rome, Italy.
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, Padua, Italy.
| | - Rosa Di Liddo
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, Padua, Italy; Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy.
| | - Susanna Amadio
- Cellular Neurobiology Unit, Preclinical Neuroscience, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168 Rome, Italy.
| |
Collapse
|
8
|
Wang Z, Guo W, Yi F, Zhou T, Li X, Feng Y, Guo Q, Xu H, Song X, Cao L. The Regulatory Effect of SIRT1 on Extracellular Microenvironment Remodeling. Int J Biol Sci 2021; 17:89-96. [PMID: 33390835 PMCID: PMC7757024 DOI: 10.7150/ijbs.52619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
The sirtuins family is well known by its unique nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase function. The most-investigated member of the family, Sirtuin 1 (SIRT1), accounts for deacetylating a broad range of transcription factors and coregulators, such as p53, the Forkhead box O (FOXO), and so on. It serves as a pivotal regulator in various intracellular biological processes, including energy metabolism, DNA damage response, genome stability maintenance and tumorigenesis. Although the most attention has been focused on its intracellular functions, the regulatory effect on extracellular microenvironment remodeling of SIRT1 has been recognized by researchers recently. SIRT1 can regulate cell secretion process and participate in glucose metabolism, neuroendocrine function, inflammation and tumorigenesis. Here, we review the advances in the understanding of SIRT1 on remodeling the extracellular microenvironment, which may provide new ideas for pathogenesis investigation and guidance for clinical treatment.
Collapse
Affiliation(s)
- Zhuo Wang
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Wendong Guo
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Fei Yi
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Tingting Zhou
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Xiaoman Li
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Yanling Feng
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Qiqiang Guo
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Hongde Xu
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Xiaoyu Song
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| | - Liu Cao
- College of Basic Medical Science, Institute of Translational Medicine, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning Province, P.R. China, 110122
| |
Collapse
|
9
|
Yilmaz S. Serum NO, S100B, NSE concentrations in migraine and their relationship. J Clin Neurosci 2020; 82:32-35. [PMID: 33317735 DOI: 10.1016/j.jocn.2020.10.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/30/2020] [Accepted: 10/18/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Migraine pathogenesis still remains uncertain. Studies have found contradictory results regarding NO, S100B and NSE parameters in migraine patients. Therefore, in our study, we aimed to measure NO, S100B and NSE concentrations in migraine patients, compare them with the control group and find the relationship between these parameters. MATERIALS AND METHODS Fifty-two patients (35 women and 17 men) diagnosed with migraine according to the International Headache Classification II criteria were included in the study. 30 healthy participants without any history of disease were included in the control group. Serum NO, S100B and NSE levels were determined in all participants. RESULTS It was found that NO, S100B parameters increased compared to the control group, and NSE parameter decreased compared to the control group in the migraine patients participating in this study (p = 0,004, p = 0,002, p = 0,000) It was found that there was a moderate positive linear correlation between serum S100B and NSE in the migraine patients in our study (r = 442, p = 0.011). CONCLUSION In our study, the fact that there was a statistically significant difference in the NO, S100b and NSE parameters of migraine and control group patients indicates that these molecules can be effective in the pathogenesisof migraine. The moderate positive linear correlation found between serum S100B and NSE in migraine patients in our study demonstrates that these molecules together can be effective in the pathogenesis.
Collapse
Affiliation(s)
- Sedat Yilmaz
- Department of Medical Biochemistry, Medicine Faculty of Adiyaman University, Adiyaman, Turkey.
| |
Collapse
|
10
|
Wang J, Liu J, Li R, Wang C. Research and progress on biomarkers of neuromyelitis optica spectrum disorders. J Recept Signal Transduct Res 2020; 41:417-424. [PMID: 33019871 DOI: 10.1080/10799893.2020.1830109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are a demyelinating disorder of the central nervous system based on the involvement of the optic nerve and/or spinal cord. The disease is characterized by high recurrence and disability. NMOSD is mainly diagnosed by AQP4-IgG and MOG-IgG. However, there are still some patients with negative or undetermined double-antibody, and AQP4-IgG and MOG-IgG cannot indicate the clinical disease activity. Therefore, it is urgent to explore interesting biomarkers in serum and cerebrospinal fluid to promote early clinical diagnosis and/or as a target for diagnosis and treatment. This article summarized the research progress in serum and cerebrospinal fluid biomarkers of astrocytes, neurons, myelin sheath, and other damage after the onset of NMOSD. Besides the value of microglial activation-related proteins in the diagnosis and treatment of NMOSD was prospected, so as to promote the research progress of NMOSD.
Collapse
Affiliation(s)
- Jinyang Wang
- School of Laboratory Medicine, Weifang Medical College, Weifang, P. R. China.,Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Jiayu Liu
- Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Ruibing Li
- Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Chengbin Wang
- School of Laboratory Medicine, Weifang Medical College, Weifang, P. R. China.,Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| |
Collapse
|
11
|
Steliga A, Kowiański P, Czuba E, Waśkow M, Moryś J, Lietzau G. Neurovascular Unit as a Source of Ischemic Stroke Biomarkers-Limitations of Experimental Studies and Perspectives for Clinical Application. Transl Stroke Res 2020; 11:553-579. [PMID: 31701356 PMCID: PMC7340668 DOI: 10.1007/s12975-019-00744-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/13/2023]
Abstract
Cerebral stroke, which is one of the most frequent causes of mortality and leading cause of disability in developed countries, often leads to devastating and irreversible brain damage. Neurological and neuroradiological diagnosis of stroke, especially in its acute phase, is frequently uncertain or inconclusive. This results in difficulties in identification of patients with poor prognosis or being at high risk for complications. It also makes difficult identification of these stroke patients who could benefit from more aggressive therapies. In contrary to the cardiovascular disease, no single biomarker is available for the ischemic stroke, addressing the abovementioned issues. This justifies the need for identifying of effective diagnostic measures characterized by high specificity and sensitivity. One of the promising avenues in this area is studies on the panels of biomarkers characteristic for processes which occur in different types and phases of ischemic stroke and represent all morphological constituents of the brains' neurovascular unit (NVU). In this review, we present the current state of knowledge concerning already-used or potentially applicable biomarkers of the ischemic stroke. We also discuss the perspectives for identification of biomarkers representative for different types and phases of the ischemic stroke, as well as for different constituents of NVU, which concentration levels correlate with extent of brain damage and patients' neurological status. Finally, a critical analysis of perspectives on further improvement of the ischemic stroke diagnosis is presented.
Collapse
Affiliation(s)
- Aleksandra Steliga
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Przemysław Kowiański
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland.
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland.
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Monika Waśkow
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Cordeiro JL, Neves JD, Vizuete AF, Aristimunha D, Pedroso TA, Sanches EF, Gonçalves CA, Netto CA. Arundic Acid (ONO-2506), an Inhibitor of S100B Protein Synthesis, Prevents Neurological Deficits and Brain Tissue Damage Following Intracerebral Hemorrhage in Male Wistar Rats. Neuroscience 2020; 440:97-112. [PMID: 32474054 DOI: 10.1016/j.neuroscience.2020.05.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
Stroke is one of the leading causes of mortality and neurological morbidity. Intracerebral hemorrhage (ICH) has the poorest prognosis among all stroke subtypes and no treatment has been effective in improving outcomes. Following ICH, the observed high levels of S100B protein have been associated with worsening of injury and neurological deficits. Arundic acid (AA) exerts neuroprotective effects through inhibition of astrocytic synthesis of S100B in some models of experimental brain injury; however, it has not been studied in ICH. The aim of this study was to evaluate the effects of intracerebroventricular (ICV) administration of AA in male Wistar rats submitted to ICH model assessing the following variables: reactive astrogliosis, S100B levels, antioxidant defenses, cell death, lesion extension and neurological function. Firstly, AA was injected at different doses (0.02, 0.2, 2 and 20 μg/μl) in the left lateral ventricle in order to observe which dose would decrease GFAP and S100B striatal levels in non-injured rats. Following determination of the effective dose, ICH damage was induced by IV-S collagenase intrastrial injection and 2 μg/μl AA was injected through ICV route immediately before injury. AA treatment prevented ICH-induced neurological deficits and tissue damage, inhibited excessive astrocytic activation and cellular apoptosis, reduced peripheral and central S100B levels (in striatum, serum and cerebrospinal fluid), improved neuronal survival and enhanced the antioxidant defences after injury. Altogether, these results suggest that S100B is a viable target for treating ICH and highlight AA as an interesting strategy for improving neurological outcome after experimental brain hemorrhage.
Collapse
Affiliation(s)
- J L Cordeiro
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Post-graduation Program of Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-190, Brazil.
| | - J D Neves
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - A F Vizuete
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - D Aristimunha
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - T A Pedroso
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - E F Sanches
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Post-graduation Program of Phisiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-190, Brazil
| | - C A Gonçalves
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - C A Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| |
Collapse
|
13
|
Huang L, Zhang L, Liu Z, Zhao S, Xu D, Li L, Peng Q, Ai Y. Pentamidine protects mice from cecal ligation and puncture-induced brain damage via inhibiting S100B/RAGE/NF-κB. Biochem Biophys Res Commun 2019; 517:221-226. [DOI: 10.1016/j.bbrc.2019.07.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 07/17/2019] [Indexed: 02/08/2023]
|
14
|
Illouz T, Madar R, Biragyn A, Okun E. Restoring microglial and astroglial homeostasis using DNA immunization in a Down Syndrome mouse model. Brain Behav Immun 2019; 75:163-180. [PMID: 30389461 PMCID: PMC6358279 DOI: 10.1016/j.bbi.2018.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/22/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Down Syndrome (DS), the most common cause of genetic intellectual disability, is characterized by over-expression of the APP and DYRK1A genes, located on the triplicated chromosome 21. This chromosomal abnormality leads to a cognitive decline mediated by Amyloid-β (Aβ) overproduction and tau hyper-phosphorylation as early as the age of 40. In this study, we used the Ts65Dn mouse model of DS to evaluate the beneficial effect of a DNA vaccination against the Aβ1-11 fragment, in ameliorating Aβ-related neuropathology and rescue of cognitive and behavioral abilities. Anti-Aβ1-11 vaccination induced antibody production and facilitated clearance of soluble oligomers and small extracellular inclusions of Aβ from the hippocampus and cortex of Ts65Dn mice. This was correlated with reduced neurodegeneration and restoration of the homeostatic phenotype of microglial and astroglial cells. Vaccinated Ts65Dn mice performed better in spatial-learning tasks, exhibited reduced motor hyperactivity typical for this strain, and restored short-term memory abilities. Our findings support the hypothesis that DS individuals may benefit from active immunotherapy against Aβ from a young age by slowing the progression of dementia.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
15
|
The Role of Oxidative Stress and Bioenergetic Dysfunction in Sulfite Oxidase Deficiency: Insights from Animal Models. Neurotox Res 2018; 35:484-494. [DOI: 10.1007/s12640-018-9986-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/13/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023]
|
16
|
Michetti F, D'Ambrosi N, Toesca A, Puglisi MA, Serrano A, Marchese E, Corvino V, Geloso MC. The S100B story: from biomarker to active factor in neural injury. J Neurochem 2018; 148:168-187. [DOI: 10.1111/jnc.14574] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/19/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Fabrizio Michetti
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
- IRCCS San Raffaele Scientific Institute; Università Vita-Salute San Raffaele; Milan Italy
| | - Nadia D'Ambrosi
- Department of Biology; Università degli Studi di Roma Tor Vergata; Rome Italy
| | - Amelia Toesca
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | | | - Alessia Serrano
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| |
Collapse
|
17
|
S100B promotes microglia M1 polarization and migration to aggravate cerebral ischemia. Inflamm Res 2018; 67:937-949. [DOI: 10.1007/s00011-018-1187-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/05/2018] [Accepted: 09/14/2018] [Indexed: 01/20/2023] Open
|
18
|
Dyck S, Kataria H, Alizadeh A, Santhosh KT, Lang B, Silver J, Karimi-Abdolrezaee S. Perturbing chondroitin sulfate proteoglycan signaling through LAR and PTPσ receptors promotes a beneficial inflammatory response following spinal cord injury. J Neuroinflammation 2018; 15:90. [PMID: 29558941 PMCID: PMC5861616 DOI: 10.1186/s12974-018-1128-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) results in upregulation of chondroitin sulfate proteoglycans (CSPGs) by reactive glia that impedes repair and regeneration in the spinal cord. Degradation of CSPGs is known to be beneficial in promoting endogenous repair mechanisms including axonal sprouting/regeneration, oligodendrocyte replacement, and remyelination, and is associated with improvements in functional outcomes after SCI. Recent evidence suggests that CSPGs may regulate secondary injury mechanisms by modulating neuroinflammation after SCI. To date, the role of CSPGs in SCI neuroinflammation remains largely unexplored. The recent discovery of CSPG-specific receptors, leukocyte common antigen-related (LAR) and protein tyrosine phosphatase-sigma (PTPσ), allows unraveling the cellular and molecular mechanisms of CSPGs in SCI. In the present study, we have employed parallel in vivo and in vitro approaches to dissect the role of CSPGs and their receptors LAR and PTPσ in modulating the inflammatory processes in the acute and subacute phases of SCI. METHODS In a clinically relevant model of compressive SCI in female Sprague Dawley rats, we targeted LAR and PTPσ by two intracellular functionally blocking peptides, termed ILP and ISP, respectively. We delivered ILP and ISP treatment intrathecally to the injured spinal cord in a sustainable manner by osmotic mini-pumps for various time-points post-SCI. We employed flow cytometry, Western blotting, and immunohistochemistry in rat SCI, as well as complementary in vitro studies in primary microglia cultures to address our questions. RESULTS We provide novel evidence that signifies a key immunomodulatory role for LAR and PTPσ receptors in SCI. We show that blocking LAR and PTPσ reduces the population of classically activated M1 microglia/macrophages, while promoting alternatively activated M2 microglia/macrophages and T regulatory cells. This shift was associated with a remarkable elevation in pro-regenerative immune mediators, interleukin-10 (IL-10), and Arginase-1. Our parallel in vitro studies in microglia identified that while CSPGs do not induce an M1 phenotype per se, they promote a pro-inflammatory phenotype. Interestingly, inhibiting LAR and PTPσ in M1 and M2 microglia positively modulates their inflammatory response in the presence of CSPGs, and harnesses their ability for phagocytosis and mobilization. Interestingly, our findings indicate that CSPGs regulate microglia, at least in part, through the activation of the Rho/ROCK pathway downstream of LAR and PTPσ. CONCLUSIONS We have unveiled a novel role for LAR and PTPσ in regulating neuroinflammation in traumatic SCI. Our findings provide new insights into the mechanisms by which manipulation of CSPG signaling can promote recovery from SCI. More importantly, this work introduces the potential of ILP/ISP as a viable strategy for modulating the immune response following SCI and other neuroinflammatory conditions of the central nervous system.
Collapse
Affiliation(s)
- Scott Dyck
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Kallivalappil T Santhosh
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Bradley Lang
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jerry Silver
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
19
|
Crompton LA, Cordero‐Llana O, Caldwell MA. Astrocytes in a dish: Using pluripotent stem cells to model neurodegenerative and neurodevelopmental disorders. Brain Pathol 2017; 27:530-544. [PMID: 28585380 PMCID: PMC8028895 DOI: 10.1111/bpa.12522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023] Open
Abstract
Neuroscience and Neurobiology have historically been neuron biased, yet up to 40% of the cells in the brain are astrocytes. These cells are heterogeneous and regionally diverse but universally essential for brain homeostasis. Astrocytes regulate synaptic transmission as part of the tripartite synapse, provide metabolic and neurotrophic support, recycle neurotransmitters, modulate blood flow and brain blood barrier permeability and are implicated in the mechanisms of neurodegeneration. Using pluripotent stem cells (PSC), it is now possible to study regionalised human astrocytes in a dish and to model their contribution to neurodevelopmental and neurodegenerative disorders. The evidence challenging the traditional neuron-centric view of degeneration within the CNS is reviewed here, with focus on recent findings and disease phenotypes from human PSC-derived astrocytes. In addition we compare current protocols for the generation of regionalised astrocytes and how these can be further refined by our growing knowledge of neurodevelopment. We conclude by proposing a functional and phenotypical characterisation of PSC-derived astrocytic cultures that is critical for reproducible and robust disease modelling.
Collapse
Affiliation(s)
- Lucy A. Crompton
- School of Biochemistry, Medical Sciences BldUniversity of BristolBristolBS8 1TDUK
| | - Oscar Cordero‐Llana
- Bristol Medical School, Medical Sciences BldUniversity of BristolBristolBS8 1TDUK
| | - Maeve A. Caldwell
- Trinity College Institute for NeuroscienceTrinity College Dublin 2Ireland
| |
Collapse
|
20
|
Cognitive dysfunction correlates with elevated serum S100B concentration in drug-free acutely relapsed patients with schizophrenia. Psychiatry Res 2017; 247:6-11. [PMID: 27863321 DOI: 10.1016/j.psychres.2016.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 09/04/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023]
Abstract
S100B, a biomarker of glial dysfunction and blood-brain barrier (BBB) disruption, has been proposed to be involved in the pathophysiology of schizophrenia. In the present study, we aimed at exploring the association of serum S100B levels with cognitive deficits using MATRICS Consensus Cognitive Battery (MCCB) in schizophrenia, by excluding the impact of antipsychotics. Sixty-two unmedicated patients with schizophrenia during their acute phases were divided into a drug-naïve group (n=34) and a drug-free group (n=28). S100B serum concentrations were measured and MCCB was administered to all of the patients. Forty healthy controls donated their blood samples for S100B assessment. The results indicated that serum S100B was significantly elevated in the drug-naive/free acute-stage schizophrenic patients when compared to the healthy controls. In the drug-free group, the serum S100B level was an independent contributor to the global cognitive dysfunctions, particularly for the speed of processing, attention/vigilance, visual learning and reasoning/problem solving subscores. Nevertheless, no significant associations between S100B and MCCB composite score or any cognitive domain subscore were observed in the drug-naïve group. These findings support the hypothesis that glial dysfunction and associated marker protein S100B may contribute to the pathophysiologic development of neurocognitive deficits in the relapsed individuals with schizophrenia.
Collapse
|
21
|
S100B raises the alert in subarachnoid hemorrhage. Rev Neurosci 2016; 27:745-759. [DOI: 10.1515/revneuro-2016-0021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/26/2016] [Indexed: 12/19/2022]
Abstract
AbstractSubarachnoid hemorrhage (SAH) is a devastating disease with high mortality and mobility, the novel therapeutic strategies of which are essentially required. The calcium binding protein S100B has emerged as a brain injury biomarker that is implicated in pathogenic process of SAH. S100B is mainly expressed in astrocytes of the central nervous system and functions through initiating intracellular signaling or via interacting with cell surface receptor, such as the receptor of advanced glycation end products. The biological roles of S100B in neurons have been closely associated with its concentrations, resulting in either neuroprotection or neurotoxicity. The levels of S100B in the blood have been suggested as a biomarker to predict the progress or the prognosis of SAH. The role of S100B in the development of cerebral vasospasm and brain damage may result from the induction of oxidative stress and neuroinflammation after SAH. To get further insight into mechanisms underlying the role of S100B in SAH based on this review might help us to find novel therapeutic targets for SAH.
Collapse
|
22
|
Cuajungco MP, Lees GJ, Kydd RR, Tanzi RE, Bush AI. Zinc and Alzheimer's Disease: An Update. Nutr Neurosci 2016; 2:191-208. [DOI: 10.1080/1028415x.1999.11747277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
The Dual Function of Reactive Oxygen/Nitrogen Species in Bioenergetics and Cell Death: The Role of ATP Synthase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3869610. [PMID: 27034734 PMCID: PMC4806282 DOI: 10.1155/2016/3869610] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/15/2016] [Indexed: 01/11/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) targeting mitochondria are major causative factors in disease pathogenesis. The mitochondrial permeability transition pore (PTP) is a mega-channel modulated by calcium and ROS/RNS modifications and it has been described to play a crucial role in many pathophysiological events since prolonged channel opening causes cell death. The recent identification that dimers of ATP synthase form the PTP and the fact that posttranslational modifications caused by ROS/RNS also affect cellular bioenergetics through the modulation of ATP synthase catalysis reveal a dual function of these modifications in the cells. Here, we describe mitochondria as a major site of production and as a target of ROS/RNS and discuss the pathophysiological conditions in which oxidative and nitrosative modifications modulate the catalytic and pore-forming activities of ATP synthase.
Collapse
|
24
|
Cunha MP, Lieberknecht V, Ramos-Hryb AB, Olescowicz G, Ludka FK, Tasca CI, Gabilan NH, Rodrigues ALS. Creatine affords protection against glutamate-induced nitrosative and oxidative stress. Neurochem Int 2016; 95:4-14. [PMID: 26804444 DOI: 10.1016/j.neuint.2016.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 01/07/2016] [Accepted: 01/18/2016] [Indexed: 12/11/2022]
Abstract
Creatine has been reported to exert beneficial effects in several neurodegenerative diseases in which glutamatergic excitotoxicity and oxidative stress play an etiological role. The purpose of this study was to investigate the protective effects of creatine, as compared to the N-Methyl-d-Aspartate (NMDA) receptor antagonist dizocilpine (MK-801), against glutamate or hydrogen peroxide (H2O2)-induced injury in human neuroblastoma SH-SY5Y cells. Exposure of cells to glutamate (60-80 mM) or H2O2 (200-300 μM) for 24 h decreased cellular viability and increased dichlorofluorescein (DCF) fluorescence (indicative of increased reactive oxygen species, ROS) and nitric oxide (NO) production (assessed by mono-nitrogen oxides, NOx, levels). Creatine (1-10 mM) or MK-801 (0.1-10 μM) reduced glutamate- and H2O2-induced toxicity. The protective effect of creatine against glutamate-induced toxicity involves its antioxidant effect, since creatine, similar to MK-801, prevented the increase on DCF fluorescence induced by glutamate or H2O2. Furthermore, creatine or MK-801 blocked glutamate- and H2O2-induced increases in NOx levels. In another set of experiments, the repeated, but not acute, administration of creatine (300 mg/kg, po) in mice prevented the decreases on cellular viability and mitochondrial membrane potential (assessed by tetramethylrhodamine ethyl ester, TMRE, probe) of hippocampal slices incubated with glutamate (10 mM). Creatine concentration-dependent decreased the amount of nitrite formed in the reaction of oxygen with NO produced from sodium nitroprusside solution, suggesting that its protective effect against glutamate or H2O2-induced toxicity might be due to its scavenger activity. Overall, the results suggest that creatine may be useful as adjuvant therapy for neurodegenerative disease treatments.
Collapse
Affiliation(s)
- Mauricio P Cunha
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil.
| | - Vicente Lieberknecht
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Ana Belén Ramos-Hryb
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Gislaine Olescowicz
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Fabiana K Ludka
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Carla I Tasca
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Nelson H Gabilan
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, 88040-900, Florianópolis, SC, Brazil
| |
Collapse
|
25
|
de Souza DF, Wartchow KM, Lunardi PS, Brolese G, Tortorelli LS, Batassini C, Biasibetti R, Gonçalves CA. Changes in Astroglial Markers in a Maternal Immune Activation Model of Schizophrenia in Wistar Rats are Dependent on Sex. Front Cell Neurosci 2015; 9:489. [PMID: 26733814 PMCID: PMC4689875 DOI: 10.3389/fncel.2015.00489] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/03/2015] [Indexed: 01/26/2023] Open
Abstract
Data from epidemiological studies suggest that prenatal exposure to bacterial and viral infection is an important environmental risk factor for schizophrenia. The maternal immune activation (MIA) animal model is used to study how an insult directed at the maternal host can have adverse effects on the fetus, leading to behavioral and neurochemical changes later in life. We evaluated whether the administration of LPS to rat dams during late pregnancy affects astroglial markers (S100B and GFAP) of the offspring in later life. The frontal cortex and hippocampus were compared in male and female offspring on postnatal days (PND) 30 and 60. The S100B protein exhibited an age-dependent pattern of expression, being increased in the frontal cortex and hippocampus of the MIA group at PND 60, while at PND 30, male rats presented increased S100B levels only in the frontal cortex. Considering that S100B secretion is reduced by elevation of glutamate levels, we may hypothesize that this early increment in frontal cortex tissue of males is associated with elevated extracellular levels of glutamate and glutamatergic hypofunction, an alteration commonly associated with SCZ pathology. Moreover, we also found augmented GFAP in the frontal cortex of the LPS group at PND 30, but not in the hippocampus. Taken together data indicate that astroglial changes induced by MIA are dependent on sex and brain region and that these changes could reflect astroglial dysfunction. Such alterations may contribute to our understanding of the abnormal neuronal connectivity and developmental aspects of SCZ and other psychiatric disorders.
Collapse
Affiliation(s)
- Daniela F de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Krista M Wartchow
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Paula S Lunardi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Giovana Brolese
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Lucas S Tortorelli
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Cristiane Batassini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Regina Biasibetti
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| |
Collapse
|
26
|
Zhou Y, Duan S, Zhou Y, Yu S, Wu J, Wu X, Zhao J, Zhao Y. Sulfiredoxin-1 attenuates oxidative stress via Nrf2/ARE pathway and 2-Cys Prdxs after oxygen-glucose deprivation in astrocytes. J Mol Neurosci 2015; 55:941-50. [PMID: 25407820 DOI: 10.1007/s12031-014-0449-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/15/2014] [Indexed: 12/30/2022]
Abstract
Sulfiredoxin-1 (Srxn1), an endogenous antioxidant protein, is involved in keeping the balance of the cell's oxidation/reduction and can resist oxidative stress. However, the exact antioxidant effects of Srxn1 remain fully unclear. The study aims to examine the effects of Srxn1 on oxidative stress and explore the potential mechanisms in astrocytes with 6 h/oxygen-glucose deprivation (OGD), 24 h/respiration. In the study, silencing Srxn1 was performed before exposure to 6 h/OGD, 24 h/respiration in primary astrocytes. Decreased cell viability and increased cellular damage measured by CellTiter 96H AQueous Non-Radioactive Cell Proliferation Assay (MTS) and lactate dehydrogenase (LDH) were observed in Srxn1 silencing astrocytes. In addition, Srxn1 silencing resulted in a decrease in both intracellular superoxide dismutase (SOD) and glutathione (GSH). NF-E2-related factor 2 (Nrf2), a transcription factor known to influence susceptibility to oxidative stress, upregulated Srxn1 expression during oxidative stress caused by OGD in the astrocytes. Electromobility shift assay (EMSA) demonstrated a decreased binding of Nrf2 to oligomers containing Srxn1 ter-specific antioxidant response element (ARE)-binding site in Nrf2 silencing astrocytes. We also found that a reduction of peroxiredoxin (Prdx)-SO3 was closely dependent on Srxn1. In addition, 2-Cys Prdxs protein levels were increased in the astrocytes exposed to OGD, as evaluated by immunoblot analysis. All taken together, the study suggested that silencing Srxn1 would result into increasing sensitivity to OGD-induced oxidative stress injury in astrocytes. Furthermore, Nrf2/ARE pathway was involved into Srxn1, playing its antioxidant protection against oxidative stress, all of which would provide a novel therapeutic theory for treating acute ischemic brain injury.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Pathology, Chongqing Medical University, No. 1 Yixueyuan Street, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Casola C, Schiwek JE, Reinehr S, Kuehn S, Grus FH, Kramer M, Dick HB, Joachim SC. S100 Alone Has the Same Destructive Effect on Retinal Ganglion Cells as in Combination with HSP 27 in an Autoimmune Glaucoma Model. J Mol Neurosci 2015; 56:228-36. [DOI: 10.1007/s12031-014-0485-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/21/2014] [Indexed: 01/27/2023]
|
28
|
Wang P, Guan P, Wang T, Yu X, Guo J, Wang Z. Aggravation of Alzheimer's disease due to the COX-2-mediated reciprocal regulation of IL-1β and Aβ between glial and neuron cells. Aging Cell 2014; 13:605-15. [PMID: 24621265 PMCID: PMC4326948 DOI: 10.1111/acel.12209] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2014] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and displays the characteristics of chronic neurodegenerative disorders; amyloid plaques (AP) that contain amyloid β-protein (Aβ) accumulate in AD, which is also characterized by tau phosphorylation. Epidemiological evidence has demonstrated that long-term treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) markedly reduces the risk of AD by inhibiting the expression of cyclooxygenase 2 (COX-2). Although the levels of COX-2 and its metabolic product prostaglandin (PG)E2 are elevated in the brain of AD patients, the mechanisms for the development of AD remain unknown. Using human- or mouse-derived glioblastoma and neuroblastoma cell lines as model systems, we delineated the signaling pathways by which COX-2 mediates the reciprocal regulation of interleukin-1β (IL-1β) and Aβ between glial and neuron cells. In glioblastoma cells, COX-2 regulates the synthesis of IL-1β in a PGE2 -dependent manner. Moreover, COX-2-derived PGE2 signals the activation of the PI3-K/AKT and PKA/CREB pathways via cyclic AMP; these pathways transactivate the NF-κB p65 subunit via phosphorylation at Ser 536 and Ser 276, leading to IL-1β synthesis. The secretion of IL-1β from glioblastoma cells in turn stimulates the expression of COX-2 in human or mouse neuroblastoma cells. Similar regulatory mechanisms were found for the COX-2 regulation of BACE-1 expression in neuroblastoma cells. More importantly, Aβ deposition mediated the inflammatory response of glial cells via inducing the expression of COX-2 in glioblastoma cells. These findings not only provide new insights into the mechanisms of COX-2-induced AD but also initially define the therapeutic targets of AD.
Collapse
Affiliation(s)
- Pu Wang
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| | - Pei‐Pei Guan
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| | - Tao Wang
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| | - Xin Yu
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| | - Jian‐Jun Guo
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| | - Zhan‐You Wang
- College of Life and Health Sciences Northeastern University Shenyang 110819China
| |
Collapse
|
29
|
Chen C, Jiang P, Xue H, Peterson SE, Tran HT, McCann AE, Parast MM, Li S, Pleasure DE, Laurent LC, Loring JF, Liu Y, Deng W. Role of astroglia in Down's syndrome revealed by patient-derived human-induced pluripotent stem cells. Nat Commun 2014; 5:4430. [PMID: 25034944 PMCID: PMC4109022 DOI: 10.1038/ncomms5430] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 06/17/2014] [Indexed: 12/19/2022] Open
Abstract
Down's syndrome (DS), caused by trisomy of human chromosome 21, is the most common genetic cause of intellectual disability. Here we use induced pluripotent stem cells (iPSCs) derived from DS patients to identify a role for astrocytes in DS pathogenesis. DS astroglia exhibit higher levels of reactive oxygen species and lower levels of synaptogenic molecules. Astrocyte-conditioned medium collected from DS astroglia causes toxicity to neurons, and fails to promote neuronal ion channel maturation and synapse formation. Transplantation studies show that DS astroglia do not promote neurogenesis of endogenous neural stem cells in vivo. We also observed abnormal gene expression profiles from DS astroglia. Finally, we show that the FDA-approved antibiotic drug, minocycline, partially corrects the pathological phenotypes of DS astroglia by specifically modulating the expression of S100B, GFAP, inducible nitric oxide synthase, and thrombospondins 1 and 2 in DS astroglia. Our studies shed light on the pathogenesis and possible treatment of DS by targeting astrocytes with a clinically available drug.
Collapse
Affiliation(s)
- Chen Chen
- 1] Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California 95817, USA [3] Department of Neurology, Institute of Neurology, Tianjin General Hospital, Tianjin Medical University, Tianjin 300070, China [4]
| | - Peng Jiang
- 1] Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California 95817, USA [3]
| | - Haipeng Xue
- 1] Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA [2] Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA [3] Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92037, USA [4] Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Suzanne E Peterson
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ha T Tran
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Anna E McCann
- 1] Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA [2] Present address: Department of Biology, University of Washington, Seattle, Washington 98195, USA
| | - Mana M Parast
- Department of Pathology, University of California, San Diego, La Jolla, California 92093, USA
| | - Shenglan Li
- 1] Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA [2] Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - David E Pleasure
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California 95817, USA
| | - Louise C Laurent
- 1] Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92037, USA [2] Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jeanne F Loring
- 1] Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92037, USA [2] Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ying Liu
- 1] Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA [2] Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA [3] Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92037, USA [4] Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Wenbin Deng
- 1] Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California 95817, USA
| |
Collapse
|
30
|
Villarreal A, Seoane R, González Torres A, Rosciszewski G, Angelo MF, Rossi A, Barker PA, Ramos AJ. S100B protein activates a RAGE-dependent autocrine loop in astrocytes: implications for its role in the propagation of reactive gliosis. J Neurochem 2014; 131:190-205. [DOI: 10.1111/jnc.12790] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/09/2014] [Accepted: 06/10/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Alejandro Villarreal
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Rocío Seoane
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Agustina González Torres
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Gerardo Rosciszewski
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Maria Florencia Angelo
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Alicia Rossi
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Philip A. Barker
- Montreal Neurological Institute; Center for Neuronal Survival; McGill University; Montreal Québec Canada
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular; Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis”; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
31
|
Inflammatory Response in Patients under Coronary Artery Bypass Grafting Surgery and Clinical Implications: A Review of the Relevance of Dexmedetomidine Use. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/905238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the fact that coronary artery bypass grafting surgery (CABG) with cardiopulmonary bypass (CPB) prolongs life and reduces symptoms in patients with severe coronary artery diseases, these benefits are accompanied by increased risks. Morbidity associated with cardiopulmonary bypass can be attributed to the generalized inflammatory response induced by blood-xenosurfaces interactions during extracorporeal circulation and the ischemia/reperfusion implications, including exacerbated inflammatory response resembling the systemic inflammatory response syndrome (SIRS). The use of specific anesthetic agents with anti-inflammatory activity can modulate the deleterious inflammatory response. Consequently, anti-inflammatory anesthetics may accelerate postoperative recovery and better outcomes than classical anesthetics. It is known that the stress response to surgery can be attenuated by sympatholytic effects caused by activation of central (α-)2-adrenergic receptor, leading to reductions in blood pressure and heart rate, and more recently, that they can have anti-inflammatory properties. This paper discusses the clinical significance of the dexmedetomidine use, a selective (α-)2-adrenergic agonist, as a coadjuvant in general anesthesia. Actually, dexmedetomidine use is not in anesthetic routine, but this drug can be considered a particularly promising agent in perioperative multiple organ protection.
Collapse
|
32
|
Qin B, Panickar KS, Anderson RA. Cinnamon polyphenols regulate S100β, sirtuins, and neuroactive proteins in rat C6 glioma cells. Nutrition 2013; 30:210-7. [PMID: 24239092 DOI: 10.1016/j.nut.2013.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/26/2013] [Accepted: 07/02/2013] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Increasing evidence suggests that cinnamon has many health benefits when used in herbal medicine and as a dietary ingredient. The aim of this study was to investigate the effects of an aqueous extract of cinnamon, high in type A polyphenols, on molecular targets in rat C6 glioma cells that underlie their protective effects. METHODS C6 rat glioma cells were seeded in 35-mm culture dishes or six-well plates, then were incubated with cinnamon polyphenols at doses of 10 and 20 μg/mL for 24 h. The targeting protein expression, secretion, and phosphorylation were evaluated by immunoprecitation/immunoblotting and immunofluorescence imaging. RESULTS Cinnamon polyphenols significantly enhanced secretion of S100β, a Ca(2+)-binding protein, and increased intracellular S100β expression after 24 h of incubation, in rat C6 glioma cells. Cinnamon polyphenols also enhanced protein levels of sirtuin 1, 2, and 3, deacetylases important in cell survival, and the tumor suppressor protein, p53, and inhibited the inflammatory factors, tumor necrosis factor alpha, and phospho-p65, a subunit of nuclear factor-κβ. Cinnamon polyphenols also up-regulated levels of phospho-p38, extracellular signal-regulated protein and mitogen-activated protein and kinase-activated protein kinases that may be important for prosurvival functions. CONCLUSION Our results indicate that the effects of cinnamon polyphenols on upregulating prosurvival proteins, activating mitogen-activated protein kinase pathways, and decreasing proinflammatory cytokines may contribute to their neuroprotective effects.
Collapse
Affiliation(s)
- Bolin Qin
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, USA; Integrity Nutraceuticals International, Spring Hill, TN, USA.
| | - Kiran S Panickar
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, USA; Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard A Anderson
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, USA
| |
Collapse
|
33
|
Abstract
The S100 protein family consists of 24 members functionally distributed into three main subgroups: those that only exert intracellular regulatory effects, those with intracellular and extracellular functions and those which mainly exert extracellular regulatory effects. S100 proteins are only expressed in vertebrates and show cell-specific expression patterns. In some instances, a particular S100 protein can be induced in pathological circumstances in a cell type that does not express it in normal physiological conditions. Within cells, S100 proteins are involved in aspects of regulation of proliferation, differentiation, apoptosis, Ca2+ homeostasis, energy metabolism, inflammation and migration/invasion through interactions with a variety of target proteins including enzymes, cytoskeletal subunits, receptors, transcription factors and nucleic acids. Some S100 proteins are secreted or released and regulate cell functions in an autocrine and paracrine manner via activation of surface receptors (e.g. the receptor for advanced glycation end-products and toll-like receptor 4), G-protein-coupled receptors, scavenger receptors, or heparan sulfate proteoglycans and N-glycans. Extracellular S100A4 and S100B also interact with epidermal growth factor and basic fibroblast growth factor, respectively, thereby enhancing the activity of the corresponding receptors. Thus, extracellular S100 proteins exert regulatory activities on monocytes/macrophages/microglia, neutrophils, lymphocytes, mast cells, articular chondrocytes, endothelial and vascular smooth muscle cells, neurons, astrocytes, Schwann cells, epithelial cells, myoblasts and cardiomyocytes, thereby participating in innate and adaptive immune responses, cell migration and chemotaxis, tissue development and repair, and leukocyte and tumor cell invasion.
Collapse
Affiliation(s)
- R Donato
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy.
| | | | | | | | | | | | | |
Collapse
|
34
|
Gliopreventive effects of guanosine against glucose deprivation in vitro. Purinergic Signal 2013; 9:643-54. [PMID: 23846842 DOI: 10.1007/s11302-013-9377-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022] Open
Abstract
Guanosine, a guanine-based purine, is recognized as an extracellular signaling molecule that is released from astrocytes and confers neuroprotective effects in several in vivo and in vitro studies. Astrocytes regulate glucose metabolism, glutamate transport, and defense mechanism against oxidative stress. C6 astroglial cells are widely used as an astrocyte-like cell line to study the astrocytic function and signaling pathways. Our previous studies showed that guanosine modulates the glutamate uptake activity, thus avoiding glutamatergic excitotoxicity and protecting neural cells. The goal of this study was to determine the gliopreventive effects of guanosine against glucose deprivation in vitro in cultured C6 cells. Glucose deprivation induced cytotoxicity, an increase in reactive oxygen and nitrogen species (ROS/RNS) levels and lipid peroxidation as well as affected the metabolism of glutamate, which may impair important astrocytic functions. Guanosine prevented glucose deprivation-induced toxicity in C6 cells by modulating oxidative and nitrosative stress and glial responses, such as the glutamate uptake, the glutamine synthetase activity, and the glutathione levels. Glucose deprivation decreased the level of EAAC1, the main glutamate transporter present in C6 cells. Guanosine also prevented this effect, most likely through PKC, PI3K, p38 MAPK, and ERK signaling pathways. Taken together, these results show that guanosine may represent an important mechanism for protection of glial cells against glucose deprivation. Additionally, this study contributes to a more thorough understanding of the glial- and redox-related protective properties of guanosine in astroglial cells.
Collapse
|
35
|
de Souza DF, Wartchow K, Hansen F, Lunardi P, Guerra MC, Nardin P, Gonçalves CA. Interleukin-6-induced S100B secretion is inhibited by haloperidol and risperidone. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:14-22. [PMID: 23246638 DOI: 10.1016/j.pnpbp.2012.12.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/21/2012] [Accepted: 12/04/2012] [Indexed: 01/03/2023]
Abstract
Although inflammation may be a physiological defense process, imbalanced neuroinflammation has been associated with the pathophysiology of brain disorders, including major depression and schizophrenia. Activated glia releases a variety of pro-inflammatory cytokines that contribute to neuronal dysfunction. Elevated levels of S100B, a glia derived protein, have been observed in the serum and CSF of schizophrenic patients suggesting a glial role in the disease. We evaluated whether S100B secretion (in C6 glioma cells and hippocampal slices in Wistar rats) could be directly modulated by the main inflammatory cytokines (IL-1β, TNF-α, IL-6 and IL-8) altered in schizophrenia, as well as the possible involvement of mitogen-activated protein kinase (MAPK) pathways in these responses. We also investigated the effects of typical and atypical antipsychotic drugs on glial cytokine-induced S100B release. Our results suggest that S100B secretion is increased by pro-inflammatory cytokines via MAPK and that oxidative stress may be a component of this modulation. These results reinforce the idea that the S100B protein is involved in the inflammatory response observed in many brain diseases, including schizophrenia. Moreover the antipsychotics, haloperidol and risperidone, were able to inhibit the secretion of S100B following IL-6 stimulation in C6 glioma cells.
Collapse
Affiliation(s)
- Daniela Fraga de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, 90035-003, Porto Alegre, Brazil
| | | | | | | | | | | | | |
Collapse
|
36
|
Quincozes-Santos A, Bobermin LD, Latini A, Wajner M, Souza DO, Gonçalves CA, Gottfried C. Resveratrol protects C6 astrocyte cell line against hydrogen peroxide-induced oxidative stress through heme oxygenase 1. PLoS One 2013; 8:e64372. [PMID: 23691207 PMCID: PMC3654976 DOI: 10.1371/journal.pone.0064372] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/11/2013] [Indexed: 01/21/2023] Open
Abstract
Resveratrol, a polyphenol presents in grapes and wine, displays antioxidant and anti-inflammatory properties and cytoprotective effect in brain pathologies associated to oxidative stress and neurodegeneration. In previous work, we demonstrated that resveratrol exerts neuroglial modulation, improving glial functions, mainly related to glutamate metabolism. Astrocytes are a major class of glial cells and regulate neurotransmitter systems, synaptic processing, energy metabolism and defense against oxidative stress. This study sought to determine the protective effect of resveratrol against hydrogen peroxide (H2O2)-induced cytotoxicity in C6 astrocyte cell line, an astrocytic lineage, on neurochemical parameters and their cellular and biochemical mechanisms. H2O2 exposure increased oxidative-nitrosative stress, iNOS expression, cytokine proinflammatory release (TNFα levels) and mitochondrial membrane potential dysfunction and decreased antioxidant defenses, such as SOD, CAT and creatine kinase activity. Resveratrol strongly prevented C6 cells from H2O2-induced toxicity by modulating glial, oxidative and inflammatory responses. Resveratrol per se increased heme oxygenase 1 (HO1) expression and extracellular GSH content. In addition, HO1 signaling pathway is involved in the protective effect of resveratrol against H2O2-induced oxidative damage in astroglial cells. Taken together, these results show that resveratrol represents an important mechanism for protection of glial cells against oxidative stress.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | | | | | | | | | | | | |
Collapse
|
37
|
Cuesta S, Proietto R, García GB. Astrogliosis and HSP 70 activation in neonate rats' brain exposed to sodium metavanadate through lactation. Neurotoxicol Teratol 2013; 37:57-62. [PMID: 23557781 DOI: 10.1016/j.ntt.2013.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 03/07/2013] [Accepted: 03/18/2013] [Indexed: 12/16/2022]
Abstract
The effect of sodium metavanadate (NaVO3) exposure on lipid oxidative damage in the CNS of suckling rats was studied. Using histological markers of cellular injury, we also studied the morphological alterations of neurons and astroglial cells in different regions of neonate rats CNS after NaVO3 exposure. Dams of treated litters were intraperitoneally injected with 3mgNaVO3/kgbody weight/day during 12days starting on post-natal day (PND) 10. On the 21st PND, four pups of each litter were sacrificed by decapitation and six brain areas were removed for lipid peroxidation assay by the thiobarbituric acid (TBA) reaction, the other four were transcardially perfused-fixed and their brains were removed and cut with a cryostat. Brain sections were processed for: NADPHd histochemistry and anti-HSP70, anti-GFAP and anti-S100 immunohistochemistry. The relative optical density of the NADPHd stained layers and of S100 (+) astrocytes and the GFAP (+) astrocyte surface area in Cer and Hc were measured. Although MDA levels, S100 immunostaining and NADPHd activity didn't show differences between experimental and control groups, both astrogliosis and HSP70 activation were detected in Cer, while only the former was detected in Hc of V-exposed pups.
Collapse
Affiliation(s)
- Santiago Cuesta
- Morphology Department, School of Biochemical and Pharmaceutical Sciences, National University of Rosario, Suipacha 531, Rosario, Santa Fe, Argentina.
| | | | | |
Collapse
|
38
|
Chen L, Chen L, Lv Y, Cui Z, Bei G, Qin G, Zhou J, Ge T. Tetrandrine ameliorates cognitive impairment via inhibiting astrocyte-derived S100B activation in a rat model of chronic cerebral hypoperfusion. Neurol Res 2013; 35:614-21. [PMID: 23561481 DOI: 10.1179/1743132813y.0000000175] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES To investigate the effects of tetrandrine (Tet) on cognitive impairment induced by chronic cerebral hypoperfusion and its potential anti-inflammatory mechanism by modulating the expression of S100B, interleukin-1 beta (IL-1 beta), tumor necrosis factor-alpha (TNF-alpha), and inducible nitric oxide synthase (iNOS). METHODS Chronic cerebral hypoperfusion was induced by ligation of the bilateral common carotid arteries for 8 weeks. Rats were treated with Tet (10 mg/kg or 30 mg/kg) intraperitoneally every 3 days for 4 weeks. Cognitive function of rats was evaluated by the Morris water maze. Hematoxylin eosin (H & E) and Nissl staining were used to observe neuronal damage in the hippocampal CA1 region. Immunofluorescence, quantitative real-time polymerase chain reaction (QT-PCR), and western blot were performed to measure S100B, IL-1 beta, TNF-alpha, and iNOS levels in the CA1 region of chronic cerebral hypoperfusion rats. RESULTS The Tet-treated group significantly decreased the escape latency of chronic cerebral hypoperfusion rats in finding the hidden platform (P <0.05). Compared with the 2-VO (two-vessel occlusion) group, more neurons with regular morphology and/or Nissl bodies in the hippocampus were observed in the Tet-treated group, suggesting attenuated neuronal damage and degeneration. Additionally, S100B, IL-1 beta, TNF-alpha, and iNOS levels were significantly (P <0.05) decreased in the CA1 region of the chronic cerebral hypoperfusion affected rats treated with Tet. CONCLUSION Our results found that Tet could improve cognitive impairment in the chronic cerebral hypoperfusion rats. Tetrandrine may be a novel and promising candidate for future treatment and/or prevention of chronic cerebral hypoperfusion via inhibiting S100B activation and decreasing the expression of IL-1 beta, TNF-alpha, and iNOS in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Lianlian Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Cross-Talk Between Neurons and Astrocytes in Response to Bilirubin: Early Beneficial Effects. Neurochem Res 2013; 38:644-59. [DOI: 10.1007/s11064-012-0963-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 12/31/2022]
|
40
|
Lipoic acid increases glutamate uptake, glutamine synthetase activity and glutathione content in C6 astrocyte cell line. Int J Dev Neurosci 2012; 31:165-70. [DOI: 10.1016/j.ijdevneu.2012.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 12/19/2012] [Accepted: 12/21/2012] [Indexed: 12/23/2022] Open
|
41
|
Bobermin LD, Quincozes-Santos A, Guerra MC, Leite MC, Souza DO, Gonçalves CA, Gottfried C. Resveratrol prevents ammonia toxicity in astroglial cells. PLoS One 2012; 7:e52164. [PMID: 23284918 PMCID: PMC3528750 DOI: 10.1371/journal.pone.0052164] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/15/2012] [Indexed: 02/07/2023] Open
Abstract
Ammonia is implicated as a neurotoxin in brain metabolic disorders associated with hyperammonemia. Acute ammonia toxicity can be mediated by an excitotoxic mechanism, oxidative stress and nitric oxide (NO) production. Astrocytes interact with neurons, providing metabolic support and protecting against oxidative stress and excitotoxicity. Astrocytes also convert excess ammonia and glutamate into glutamine via glutamine synthetase (GS). Resveratrol, a polyphenol found in grapes and red wines, exhibits antioxidant and anti-inflammatory properties and modulates glial functions, such as glutamate metabolism. We investigated the effect of resveratrol on the production of reactive oxygen species (ROS), GS activity, S100B secretion, TNF-α, IL-1β and IL-6 levels in astroglial cells exposed to ammonia. Ammonia induced oxidative stress, decreased GS activity and increased cytokines release, probably by a mechanism dependent on protein kinase A (PKA) and extracellular signal-regulated kinase (ERK) pathways. Resveratrol prevented ammonia toxicity by modulating oxidative stress, glial and inflammatory responses. The ERK and nuclear factor-κB (NF-κB) are involved in the protective effect of resveratrol on cytokines proinflammatory release. In contrast, other antioxidants (e.g., ascorbic acid and trolox) were not effective against hyperammonemia. Thus, resveratrol could be used to protect against ammonia-induced neurotoxicity.
Collapse
Affiliation(s)
- Larissa Daniele Bobermin
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - André Quincozes-Santos
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail:
| | - Maria Cristina Guerra
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marina Concli Leite
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diogo Onofre Souza
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos-Alberto Gonçalves
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carmem Gottfried
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
42
|
Chen X, Guan T, Li C, Shang H, Cui L, Li XM, Kong J. SOD1 aggregation in astrocytes following ischemia/reperfusion injury: a role of NO-mediated S-nitrosylation of protein disulfide isomerase (PDI). J Neuroinflammation 2012; 9:237. [PMID: 23061969 PMCID: PMC3526400 DOI: 10.1186/1742-2094-9-237] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/03/2012] [Indexed: 11/10/2022] Open
Abstract
Background Ubiquitinated-protein aggregates are implicated in cerebral ischemia/reperfusion injury. The very presence of these ubiquitinated-protein aggregates is abnormal and seems to be disease-related. However, it is not clear what leads to aggregate formation and whether the aggregations represent a reaction to aggregate-mediated neurodegeneration. Methods To study the nitrosative stress-induced protein aggregation in cerebral ischemia/reperfusion injury, we used primary astrocyte cultures as a cell model, and systematically examined their iNOS expression and consequent NO generation following oxygen glucose deprivation and reperfusion. The expression of protein disulfide isomerase (PDI) and copper-zinc superoxide dismutase (SOD1) were also examined, and the biochemical interaction between PDI and SOD1 was determined by immunoprecipitation. In addition, the levels of S-nitrosylated PDI in cultured astrocytes after oxygen glucose deprivation and reperfusion treatment were measured using the biotin-switch assay. The formation of ubiquitinated-protein aggregates was detected by immunoblot and immunofluorescence staining. Results Our data showed that the up-regulation of iNOS expression after oxygen glucose deprivation and reperfusion treatment led to excessive NO generation. Up-regulation of PDI and SOD1 was also identified in cultured astrocytes following oxygen glucose deprivation and reperfusion, and these two proteins were found to bind to each other. Furthermore, the increased nitrosative stress due to ischemia/reperfusion injury was highly associated with NO-induced S-nitrosylation of PDI, and this S-nitrosylation of PDI was correlated with the formation of ubiquitinated-protein aggregates; the levels of S-nitrosylated PDI increased in parallel with the formation of aggregates. When NO generation was pharmacologically inhibited by iNOS specific inhibitor 1400W, S-nitrosylation of PDI was significantly blocked. In addition, the formation of ubiquitinated-protein aggregates in cultured astrocytes following oxygen glucose deprivation and reperfusion was also suppressed by 1400W. Interestingly, these aggregates were colocalized with SOD1, which was found to co-immunoprecipitate with PDI. Conclusions NO-mediated S-nitrosylation of PDI may be involved in the formation of the SOD1-linked ubiquitinated-protein aggregates in cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Xueping Chen
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Hirata Y, Furuta K, Suzuki M, Oh-hashi K, Ueno Y, Kiuchi K. Neuroprotective cyclopentenone prostaglandins up-regulate neurotrophic factors in C6 glioma cells. Brain Res 2012; 1482:91-100. [PMID: 22982731 DOI: 10.1016/j.brainres.2012.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/05/2012] [Accepted: 09/06/2012] [Indexed: 12/30/2022]
Abstract
In a previous study, we developed newly synthesized arylthio derivatives of cyclopentenone prostaglandins (GIF-0642, GIF-0643, GIF-0644, GIF-0745 and GIF-0747), which are neuroprotective against both manganese toxicity in PC12 cells and glutamate toxicity in HT22 cells. In the present study, we showed that these compounds and their lead compound, NEPP11, are potent inducers of glial cell line-derived neurotrophic factor (GDNF) expression in C6 glioma cells and primary astrocytes. These neuroprotective cyclopentenone prostaglandins also induced the gene expression of nerve growth factor and, to a lesser extent, brain-derived neurotrophic factor. The induction of GDNF mRNA was transcription-dependent, and the overexpression of dominant-negative Nrf2 attenuated the ability of the (arylthio)cyclopentenone prostaglandins to stimulate GDNF gene expression. These results suggest that (arylthio)cyclopentenone prostaglandins increase GDNF gene expression partly via the Keap1/Nrf2 pathway. A growing number of reports demonstrate the importance of increasing the amounts of neurotrophic factors, especially GDNF, in neuropathological states. Although the precise mechanisms by which the GIF compounds inhibit cell death are under investigation, an increase in neurotrophic factors may contribute to the diverse pharmacological properties of (arylthio)cyclopentenone prostaglandins in vivo and will make them potentially valuable in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yoko Hirata
- Department of Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido, Gifu 501 1193, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Brunswick AS, Hwang BY, Appelboom G, Hwang RY, Piazza MA, Connolly ES. Serum biomarkers of spontaneous intracerebral hemorrhage induced secondary brain injury. J Neurol Sci 2012; 321:1-10. [PMID: 22857988 DOI: 10.1016/j.jns.2012.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 06/13/2012] [Accepted: 06/23/2012] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke associated with a high rate of morbidity and mortality. It is now believed that much of this damage occurs in the subacute period following the initial insult via a cascade of complex pathophysiologic pathways that continues to be investigated. Increased levels of certain serum proteins have been identified as biomarkers that may reflect or directly participate in the inflammation, blood brain barrier disruption, endothelial dysfunction, and neuronal and glial toxicity that occur during this secondary period of cerebral injury. Some of these biomarkers have the potential to serve as therapeutic targets or surrogate endpoints for future research or clinical trials. Others may someday augment current clinical techniques in diagnosis, risk-stratification, prognostication, treatment decision and measurement of therapeutic efficacy. While much work remains to be done, biomarkers show significant potential to expand clinical options and improve clinical management, thereby reducing mortality and improving functional outcomes in ICH patients.
Collapse
Affiliation(s)
- Andrew S Brunswick
- Department of Neurological Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
45
|
Oda Y, Tsuruta R, Fujita M, Kaneda K, Kawamura Y, Izumi T, Kasaoka S, Maruyama I, Maekawa T. Prediction of the neurological outcome with intrathecal high mobility group box 1 and S100B in cardiac arrest victims: A pilot study. Resuscitation 2012; 83:1006-12. [DOI: 10.1016/j.resuscitation.2012.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 01/17/2012] [Accepted: 01/23/2012] [Indexed: 11/17/2022]
|
46
|
Liu XP, Zheng HY, Qu M, Zhang Y, Cao FY, Wang Q, Ke D, Liu GP, Wang JZ. Upregulation of astrocytes protein phosphatase-2A stimulates astrocytes migration via inhibiting p38 MAPK in tg2576 mice. Glia 2012; 60:1279-88. [PMID: 22729898 DOI: 10.1002/glia.22347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/13/2012] [Indexed: 12/30/2022]
Abstract
One of the earliest neuropathological changes in Alzheimer disease (AD) is the accumulation of astrocytes at sites of β-amyloid (Aβ) deposits, but the cause of this cellular response is unclear. As the activity of protein phosphatase 2A (PP2A) is significantly decreased in the AD brains, we studied the role of PP2A in astrocytes migration. We observed unexpectedly that PP2A activity associated with glial fibrillary acidic protein, an astrocyte marker, was significantly upregulated in tg2576 mice, demonstrated by an increased enzyme activity, a decreased demethylation at leucine-309 (DM-PP2Ac), and a decreased phosphorylation at tyrosine-307 of PP2A (pY307-PP2Ac). Further studies by using in vitro wound-healing model and transwell assay demonstrated that upregulation of PP2A pharmacologically and genetically could stimulate astrocytes migration. Activation of PP2A promotes actin organization and inhibits p38 mitogen-activated protein kinases (p38 MAPK), while simultaneous activation of p38 MAPK partially abolishes the PP2A-induced astrocytes migration. Our data suggest that activation of astrocytes PP2A in tg2567 mice may stimulate the migration of astrocytes to the amyloid plaques by p38 MAPK inhibition, implying that PP2A deficits observed in AD may cause Aβ accumulation via hindering the astrocytes migration.
Collapse
Affiliation(s)
- Xiu-Ping Liu
- Pathophysiology Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
High-Glucose and S100B Stimulate Glutamate Uptake in C6 Glioma Cells. Neurochem Res 2012; 37:1399-408. [DOI: 10.1007/s11064-012-0722-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 01/31/2012] [Accepted: 02/07/2012] [Indexed: 10/28/2022]
|
48
|
Reali C, Pillai R, Saba F, Cabras S, Michetti F, Sogos V. S100B modulates growth factors and costimulatory molecules expression in cultured human astrocytes. J Neuroimmunol 2012; 243:95-9. [DOI: 10.1016/j.jneuroim.2011.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 11/24/2011] [Accepted: 11/25/2011] [Indexed: 12/18/2022]
|
49
|
Michetti F, Corvino V, Geloso MC, Lattanzi W, Bernardini C, Serpero L, Gazzolo D. The S100B protein in biological fluids: more than a lifelong biomarker of brain distress. J Neurochem 2012; 120:644-59. [PMID: 22145907 DOI: 10.1111/j.1471-4159.2011.07612.x] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
S100B is a calcium-binding protein concentrated in glial cells, although it has also been detected in definite extra-neural cell types. Its biological role is still debated. When secreted, S100B is believed to have paracrine/autocrine trophic effects at physiological concentrations, but toxic effects at higher concentrations. Elevated S100B levels in biological fluids (CSF, blood, urine, saliva, amniotic fluid) are thus regarded as a biomarker of pathological conditions, including perinatal brain distress, acute brain injury, brain tumors, neuroinflammatory/neurodegenerative disorders, psychiatric disorders. In the majority of these conditions, high S100B levels offer an indicator of cell damage when standard diagnostic procedures are still silent. The key question remains as to whether S100B is merely leaked from injured cells or is released in concomitance with both physiological and pathological conditions, participating at high concentrations in the events leading to cell injury. In this respect, S100B levels in biological fluids have been shown to increase in physiological conditions characterized by stressful physical and mental activity, suggesting that it may be physiologically regulated and raised during conditions of stress, with a putatively active role. This possibility makes this protein a candidate not only for a biomarker but also for a potential therapeutic target.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica Sacro Cuore, Roma, Italy.
| | | | | | | | | | | | | |
Collapse
|
50
|
Yammani RR. S100 proteins in cartilage: role in arthritis. Biochim Biophys Acta Mol Basis Dis 2012; 1822:600-6. [PMID: 22266138 DOI: 10.1016/j.bbadis.2012.01.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 12/20/2011] [Accepted: 01/05/2012] [Indexed: 01/15/2023]
Abstract
S100 proteins are low molecular weight calcium binding proteins expressed in vertebrates. The family constitutes 21 known members that are expressed in several tissues and cell types and play a major role in various cellular functions. Uniquely, members of the S100 family have both intracellular and extracellular functions. Several members of the S100 family (S100A1, S100A2, S100A4, S1008, S100A9, S100A11, and S100B) have been identified in human articular cartilage, and their expression is upregulated in diseased tissue. These S100 proteins elicit a catabolic signaling pathway via receptor for advanced glycation end products (RAGE) in cartilage and may promote progression of arthritis. This review summarizes our current understanding of the role of S100 proteins in cartilage biology and in the development of arthritis.
Collapse
Affiliation(s)
- Raghunatha R Yammani
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157, USA.
| |
Collapse
|