1
|
Jiang A, You L, Handley RR, Hawkins V, Reid SJ, Jacobsen JC, Patassini S, Rudiger SR, Mclaughlan CJ, Kelly JM, Verma PJ, Bawden CS, Gusella JF, MacDonald ME, Waldvogel HJ, Faull RLM, Lehnert K, Snell RG. Single nuclei RNA-seq reveals a medium spiny neuron glutamate excitotoxicity signature prior to the onset of neuronal death in an ovine Huntington's disease model. Hum Mol Genet 2024; 33:1524-1539. [PMID: 38776957 PMCID: PMC11336116 DOI: 10.1093/hmg/ddae087] [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: 03/07/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Huntington's disease (HD) is a neurodegenerative genetic disorder caused by an expansion in the CAG repeat tract of the huntingtin (HTT) gene resulting in behavioural, cognitive, and motor defects. Current knowledge of disease pathogenesis remains incomplete, and no disease course-modifying interventions are in clinical use. We have previously reported the development and characterisation of the OVT73 transgenic sheep model of HD. The 73 polyglutamine repeat is somatically stable and therefore likely captures a prodromal phase of the disease with an absence of motor symptomatology even at 5-years of age and no detectable striatal cell loss. To better understand the disease-initiating events we have undertaken a single nuclei transcriptome study of the striatum of an extensively studied cohort of 5-year-old OVT73 HD sheep and age matched wild-type controls. We have identified transcriptional upregulation of genes encoding N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors in medium spiny neurons, the cell type preferentially lost early in HD. Further, we observed an upregulation of astrocytic glutamate uptake transporters and medium spiny neuron GABAA receptors, which may maintain glutamate homeostasis. Taken together, these observations support the glutamate excitotoxicity hypothesis as an early neurodegeneration cascade-initiating process but the threshold of toxicity may be regulated by several protective mechanisms. Addressing this biochemical defect early may prevent neuronal loss and avoid the more complex secondary consequences precipitated by cell death.
Collapse
Affiliation(s)
- Andrew Jiang
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Linya You
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Fudan University, 131 Dong'an Road, Shanghai 200032, China
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, 130 Dong'an Road, Shanghai 200032, China
| | - Renee R Handley
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Victoria Hawkins
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Suzanne J Reid
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Jessie C Jacobsen
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Stefano Patassini
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Skye R Rudiger
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Clive J Mclaughlan
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Jennifer M Kelly
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - Paul J Verma
- Aquatic and Livestock Sciences, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - C Simon Bawden
- Molecular Biology and Reproductive Technology Laboratories, South Australian Research and Development Institute, 129 Holland Road, Adelaide, SA 5350, Australia
| | - James F Gusella
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, United States
- Department of Genetics, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, United States
| | - Marcy E MacDonald
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, United States
- Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, United States
| | - Henry J Waldvogel
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging, Centre for Brain Research, Faculty of Medical and Health Science, The University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Klaus Lehnert
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| | - Russell G Snell
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, 3 Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
2
|
Saidia AR, François F, Casas F, Mechaly I, Venteo S, Veechi JT, Ruel J, Puel JL, Wang J. Oxidative Stress Plays an Important Role in Glutamatergic Excitotoxicity-Induced Cochlear Synaptopathy: Implication for Therapeutic Molecules Screening. Antioxidants (Basel) 2024; 13:149. [PMID: 38397748 PMCID: PMC10886292 DOI: 10.3390/antiox13020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024] Open
Abstract
The disruption of the synaptic connection between the sensory inner hair cells (IHCs) and the auditory nerve fiber terminals of the type I spiral ganglion neurons (SGN) has been observed early in several auditory pathologies (e.g., noise-induced or ototoxic drug-induced or age-related hearing loss). It has been suggested that glutamate excitotoxicity may be an inciting element in the degenerative cascade observed in these pathological cochlear conditions. Moreover, oxidative damage induced by free hydroxyl radicals and nitric oxide may dramatically enhance cochlear damage induced by glutamate excitotoxicity. To investigate the underlying molecular mechanisms involved in cochlear excitotoxicity, we examined the molecular basis responsible for kainic acid (KA, a full agonist of AMPA/KA-preferring glutamate receptors)-induced IHC synapse loss and degeneration of the terminals of the type I spiral ganglion afferent neurons using a cochlear explant culture from P3 mouse pups. Our results demonstrated that disruption of the synaptic connection between IHCs and SGNs induced increased levels of oxidative stress, as well as altered both mitochondrial function and neurotrophin signaling pathways. Additionally, the application of exogenous antioxidants and neurotrophins (NT3, BDNF, and small molecule TrkB agonists) clearly increases synaptogenesis. These results suggest that understanding the molecular pathways involved in cochlear excitotoxicity is of crucial importance for the future clinical trials of drug interventions for auditory synaptopathies.
Collapse
Affiliation(s)
- Anissa Rym Saidia
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Florence François
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France;
| | - Ilana Mechaly
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Stéphanie Venteo
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Joseph T. Veechi
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Jérôme Ruel
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université-INSERM, 1263-INRAE 1260, 13385 Marseille, France;
| | - Jean-Luc Puel
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| |
Collapse
|
3
|
Namdeo P, Gidwani B, Tiwari S, Jain V, Joshi V, Shukla SS, Pandey RK, Vyas A. Therapeutic potential and novel formulations of ursolic acid and its derivatives: an updated review. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:4275-4292. [PMID: 36597140 DOI: 10.1002/jsfa.12423] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/15/2022] [Accepted: 01/03/2023] [Indexed: 06/06/2023]
Abstract
Plants produce biologically active metabolites that have been utilised to cure a variety of severe and persistent illnesses. There is a possibility that understanding how these bioactive molecules work would allow researchers to come up with better treatments for diseases including malignancy, cardiac disease and neurological disorders. A triterpene called ursolic acid (UA) is a pentacyclic prevalent triterpenoid found in fruits, leaves, herbs and blooms. The biological and chemical aspects of UA, as well as their presence, plant sources and biosynthesis, and traditional and newer technologies of extraction, are discussed in this review. Because of its biological function in the creation of new therapeutic techniques, UA is a feasible option for the evolution and medical management of a wide range of medical conditions, including cancer and other life threatening diseases. Despite this, the substance's poor solubility in aquatic environments makes it unsuitable for medicinal purposes. This hurdle was resolved in many different ways. The inclusion of UA into various pharmaceutical delivery approaches was found to be quite effective in this respect. This review also describes the properties of UA and its pharmacokinetics, as well as therapeutic applications of UA for cancer, inflammatory and cardiovascular diseases, in addition to its anti-diabetic, immunomodulatory, hepatoprotective and anti-microbial properties. Some of the recent findings related to novel nano-sized carriers as a delivery system for UA and the patents related to the applications of UA and its various derivatives are covered in this review. The analytical study of UA, oleanolic acid and other phytoconstituents by UV, HPLC, high-performance thin-layer chromatography and gas chromatography is also discussed. In the future, UA could be explored in vivo using various animal models and, in addition, the regulatory status regarding UA needs to be explored. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Priya Namdeo
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, India
| | | | - Sakshi Tiwari
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, India
| | - Vishal Jain
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, India
| | - Veenu Joshi
- Center for Basic Science and Research, Pt. Ravishankar Shukla University, Raipur, India
| | | | | | - Amber Vyas
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, India
| |
Collapse
|
4
|
Caramelo B, Monteiro-Alfredo T, Martins J, Sereno J, Castelhano J, Manadas B, Castelo-Branco M, Matafome P. Functional imaging and neurochemistry identify in vivo neuroprotection mechanisms counteracting excitotoxicity and neurovascular changes in the hippocampus and visual cortex of obese and type 2 diabetic animal models. J Neurochem 2023; 165:892-906. [PMID: 37026518 DOI: 10.1111/jnc.15825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023]
Abstract
Functional MRI (fMRI) with 1 H-MRS was combined on the hippocampus and visual cortex of animal models of obesity (high-fat diet, HFD) and type 2 diabetes (T2D) to identify the involved mechanisms and temporal evolution of neurometabolic changes in these disorders that could serve as potentially reliable clinical biomarkers. HFD rats presented elevated levels of N-acetylaspartylglutamate (NAAG) (p = 0.0365 vs. standard diet, SD) and glutathione (GSH) (p = 0.0494 vs. SD) in the hippocampus. NAAG and GSH levels in this structure proved to be correlated (r = 0.4652, p = 0.0336). This mechanism was not observed in diabetic rats. Combining MRS and fMRI-evaluated blood-oxygen-level-dependent (BOLD) response, elevated taurine (p = 0.0326 vs. HFD) and GABA type A receptor (GABAA R) (p = 0.0211 vs. SD and p = 0.0153 vs. HFD) were observed in the visual cortex of only diabetic rats, counteracting the elevated BOLD response and suggesting an adaptative mechanism against hyperexcitability observed in the primary visual cortex (V1) (p = 0.0226 vs. SD). BOLD amplitude was correlated with the glutamate levels (r = 0.4491; p = 0.0316). Therefore, here we found evidence for several biological dichotomies regarding excitotoxicity and neuroprotection in different brain regions, identifying putative markers of their different susceptibility and response to the metabolic and vascular insults of obesity and diabetes.
Collapse
Affiliation(s)
- Beatriz Caramelo
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
| | - Tamaeh Monteiro-Alfredo
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
| | - João Martins
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - José Sereno
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Coimbra Chemistry Centre (CQC), Chemistry Department, University of Coimbra, Coimbra, Portugal
| | - João Castelhano
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| |
Collapse
|
5
|
Hameed MQ, Hodgson N, Lee HHC, Pascual-Leone A, MacMullin PC, Jannati A, Dhamne SC, Hensch TK, Rotenberg A. N-acetylcysteine treatment mitigates loss of cortical parvalbumin-positive interneuron and perineuronal net integrity resulting from persistent oxidative stress in a rat TBI model. Cereb Cortex 2023; 33:4070-4084. [PMID: 36130098 PMCID: PMC10068300 DOI: 10.1093/cercor/bhac327] [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: 05/26/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) increases cerebral reactive oxygen species production, which leads to continuing secondary neuronal injury after the initial insult. Cortical parvalbumin-positive interneurons (PVIs; neurons responsible for maintaining cortical inhibitory tone) are particularly vulnerable to oxidative stress and are thus disproportionately affected by TBI. Systemic N-acetylcysteine (NAC) treatment may restore cerebral glutathione equilibrium, thus preventing post-traumatic cortical PVI loss. We therefore tested whether weeks-long post-traumatic NAC treatment mitigates cortical oxidative stress, and whether such treatment preserves PVI counts and related markers of PVI integrity and prevents pathologic electroencephalographic (EEG) changes, 3 and 6 weeks after fluid percussion injury in rats. We find that moderate TBI results in persistent oxidative stress for at least 6 weeks after injury and leads to the loss of PVIs and the perineuronal net (PNN) that surrounds them as well as of per-cell parvalbumin expression. Prolonged post-TBI NAC treatment normalizes the cortical redox state, mitigates PVI and PNN loss, and - in surviving PVIs - increases per-cell parvalbumin expression. NAC treatment also preserves normal spectral EEG measures after TBI. We cautiously conclude that weeks-long NAC treatment after TBI may be a practical and well-tolerated treatment strategy to preserve cortical inhibitory tone post-TBI.
Collapse
Affiliation(s)
- Mustafa Q Hameed
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Nathaniel Hodgson
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Henry H C Lee
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Andres Pascual-Leone
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Paul C MacMullin
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Ali Jannati
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Sameer C Dhamne
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Takao K Hensch
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Department of Molecular & Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138, United States
| | - Alexander Rotenberg
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
6
|
Briyal S, Ranjan AK, Gulati A. Oxidative stress: A target to treat Alzheimer's disease and stroke. Neurochem Int 2023; 165:105509. [PMID: 36907516 DOI: 10.1016/j.neuint.2023.105509] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/01/2023] [Accepted: 03/05/2023] [Indexed: 03/13/2023]
Abstract
Oxidative stress has been established as a well-known pathological condition in several neurovascular diseases. It starts with increased production of highly oxidizing free-radicals (e.g. reactive oxygen species; ROS and reactive nitrogen species; RNS) and becomes too high for the endogenous antioxidant system to neutralize them, which results in a significantly disturbed balance between free-radicals and antioxidants levels and causes cellular damage. A number of studies have evidently shown that oxidative stress plays a critical role in activating multiple cell signaling pathways implicated in both progression as well as initiation of neurological diseases. Therefore, oxidative stress continues to remain a key therapeutic target for neurological diseases. This review discusses the mechanisms involved in reactive oxygen species (ROS) generation in the brain, oxidative stress, and pathogenesis of neurological disorders such as stroke and Alzheimer's disease (AD) and the scope of antioxidant therapies for these disorders.
Collapse
Affiliation(s)
- Seema Briyal
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA.
| | - Amaresh K Ranjan
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA
| | - Anil Gulati
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA; Pharmazz Inc. Research and Development, Willowbrook, IL, USA
| |
Collapse
|
7
|
Jia R, Yuan X, Zhang X, Song P, Han S, Wang S, Li Y, Zhang S, Zhao X, Zhang Y, Cheng J, Song X. Oxidative stress impairs cognitive function by affecting hippocampal fimbria volume in drug-naïve, first-episode schizophrenia. Front Neurosci 2023; 17:1153439. [PMID: 37139526 PMCID: PMC10149877 DOI: 10.3389/fnins.2023.1153439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Objective The aim of the present study was to explore influencing factors of cognitive impairments and their interrelationships in drug-naïve, first-episode schizophrenia (SCZ). Methods Patients with drug naïve, first episode SCZ and healthy controls (HCs) were enrolled. Cognitive function was assessed by the MATRICS Consensus Cognitive Battery (MCCB). Serum levels of oxidative stress indices, including folate, superoxide dismutase (SOD), uric acid (UA) and homocysteine (Hcy), were determined after an overnight fast. Hippocampal subfield volumes were measured using FreeSurfer. Mediation models were conducted using the SPSS PROCESS v3.4 macro. A false discovery rate (FDR) correction was applied for multiple comparisons. Results Sixty-seven patients with SCZ and 65 HCs were enrolled in our study. The patient group had significantly lower serum levels of folate and SOD and higher serum levels of HCY compared with the HCs (all p < 0.05). The patient group had a significantly smaller volume of the whole hippocampus than the HC group (p < 0.05). We also found significant volume differences between the two groups in the following subfields: CA1, molecular layer, GC-ML-DG and fimbria (all p < 0.05, uncorrected). The partial correlation analysis controlling for age and sex showed that the fimbria volume in the patient group was significantly positively associated with NAB scores (r = 0.382, pFDR = 0.024); serum levels of SOD in the patient group showed a significantly positive correlation with fimbria volume (r = 0.360, pFDR = 0.036). Mediation analyses controlling for age and sex showed that the serum levels of SOD in patients with SCZ had significant indirect effects on the NAB scores which were mediated by the fimbria volume [indirect effect = 0.0565, 95% CI from the bootstrap test excluding zero (0.0066 to 0.0891)]. Conclusion Oxidative stress, a reduction in hippocampal subfield volumes and cognitive impairments occur in early SCZ. Oxidative stress impairs cognitive function by affecting hippocampal subfield volumes.
Collapse
Affiliation(s)
- Rufei Jia
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xiaoyun Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Peilun Song
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuying Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yajun Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Siwei Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xinyi Zhao
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Jingliang Cheng, ;10
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China
- Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
- *Correspondence: Xueqin Song,
| |
Collapse
|
8
|
Tucci P, Lattanzi R, Severini C, Saso L. Nrf2 Pathway in Huntington's Disease (HD): What Is Its Role? Int J Mol Sci 2022; 23:ijms232315272. [PMID: 36499596 PMCID: PMC9739588 DOI: 10.3390/ijms232315272] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease that occurs worldwide. Despite some progress in understanding the onset of HD, drugs that block or delay symptoms are still not available. In recent years, many treatments have been proposed; among them, nuclear transcriptional factor-2 (Nrf2) enhancer compounds have been proposed as potential therapeutic agents to treat HD. Nrf2 triggers an endogenous antioxidant pathway activated in different neurodegenerative disorders. Probably, the stimulation of Nrf2 during either the early phase or before HD symptoms' onset, could slow or prevent striatum degeneration. In this review, we present the scientific literature supporting the role of Nrf2 in HD and the potential prophylactic and therapeutic role of this compound.
Collapse
Affiliation(s)
- Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cinzia Severini
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Viale del Policlinico 155, 00161 Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
Ramya EM, Kumar GP, Chandrasekhar Y, Anilakumar KR. Adaptogenic potential of ginsenosides against domoic acid-induced toxicity by regulating neuronal stress and kinate receptors: Ex vivo and in silico studies. J Food Biochem 2022; 46:e14089. [PMID: 35102569 DOI: 10.1111/jfbc.14089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022]
Abstract
This study is focused on potential effects of ginsenosides from Panax ginseng (PG) against amnesic shell fish poison, that is, domoic acid-induced excitotoxicity. Mice received PG at two different dosages by oral feeding for a period of 28 days (50 and 100 mg kg-1 bwt.-1 ). Domoic acid was injected to the mice to induce excitotoxicity (DA; 3 mg kg-1 bwt.-1 ) and piracetam-injected animals (PIR; 100 mg kg-1 bwt.-1 ) were treated as positive control. DA-induced cognitive impairment was reverted by PG supplementation, which was observed in Morris water maze and novel object task. Moreover, PG supplementation restored levels of GABA and antioxidant enzymes. Our results further elucidated ameliorative effects of PG supplementation on DA-induced changes in the expression of synaptic plasticity (BDNF), inflammation (NFkB), and apoptotic (Bcl2, Bax, and Caspase 3) markers. Hence, this study elucidates potential nootropic effects of ginsenosides from P. ginseng extract against DA-induced neuronal impairments via, modulation of behavioral and biochemical mechanisms involved in excitotoxicity, oxidative stress, neuro-inflammation, and apoptosis. PRACTICAL APPLICATIONS: This study highlights potential effects of ginsenosides from Panax ginseng against amnesic shell fish poison, that is, domoic acid-induced excitotoxicity for the first time. This study confirms that ginsenosides have the beneficial effects against amelioration of DA-induced toxicity. This study elucidates the potential nootropic effects of P. ginseng extract against DA-induced neuronal impairments via, modulation of synaptic plasticity markers and oxido-inflammatory responses leading to apoptosis. This study will be helpful in offering various mechanisms involved in pharmacological applications of P. ginseng in the management of DA-induced excitotoxicity.
Collapse
Affiliation(s)
| | - Garlapati Phani Kumar
- Food Quality Assurance Division, Defense Food Research Laboratory (DRDO), Mysore, India
| | | | | |
Collapse
|
10
|
Brandley ET, Kirkland AE, Baron M, Baraniuk JN, Holton KF. The Effect of the Low Glutamate Diet on the Reduction of Psychiatric Symptoms in Veterans With Gulf War Illness: A Pilot Randomized-Controlled Trial. Front Psychiatry 2022; 13:926688. [PMID: 35795023 PMCID: PMC9251130 DOI: 10.3389/fpsyt.2022.926688] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The objective of this pilot study was to examine the effects of the low glutamate diet on anxiety, post-traumatic stress disorder (PTSD), and depression in veterans with Gulf War Illness (GWI). The low glutamate diet removes dietary excitotoxins and increases consumption of micronutrients which are protective against glutamatergic excitotoxicity. This study was registered at ClinicalTrials.gov (NCT#03342482). Forty veterans with GWI completed psychiatric questionnaires at baseline and after 1-month following the low glutamate diet. Participants were then randomized into a double-blind, placebo-controlled crossover challenge with monosodium glutamate (MSG; a dietary excitotoxin) vs. placebo over three consecutive days per week, with assessments on day three. Data were analyzed across the full sample and with participants categorized by baseline symptom severity. Pre-post-dietary intervention change scores were analyzed with Wilcoxon signed-rank tests and paired sample t-tests across the full sample, and changes across symptom severity categories were analyzed using ANOVA. Crossover challenge results were analyzed with linear mixed modeling accounting for challenge material (MSG v. placebo), sequence (MSG/placebo v. placebo/MSG), period (challenge week 1 v. week 2), pre-diet baseline symptom severity category (minimal/mild, moderate, or severe), and the challenge material*symptom severity category interaction. A random effect of ID (sequence) was also included. All three measures showed significant improvement after 1 month on the diet, with significant differences between baseline severity categories. Individuals with severe psychological symptoms at baseline showed the most improvement after 1 month on the diet, while those with minimal/mild symptoms showed little to no change. Modeling results from the challenge period demonstrated a significant worsening of anxiety from MSG in only the most severe group, with no significant effects of MSG challenge on depression nor PTSD symptoms. These results suggest that the low glutamate diet may be an effective treatment for depression, anxiety, and PTSD, but that either (a) glutamate is only a direct cause of symptoms in anxiety, or (b) underlying nutrient intake may prevent negative psychiatric effects from glutamate exposure. Future, larger scale clinical trials are needed to confirm these findings and to further explore the potential influence of increased micronutrient intake on the improvements observed across anxiety, PTSD, and depression.
Collapse
Affiliation(s)
- Elizabeth T Brandley
- Department of Health Studies, American University, Washington, DC, United States
| | - Anna E Kirkland
- Medical University of South Carolina, Charleston, SC, United States
| | - Michael Baron
- Department of Mathematics and Statistics, American University, Washington, DC, United States
| | - James N Baraniuk
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Kathleen F Holton
- Department of Health Studies, American University, Washington, DC, United States.,Department of Neuroscience, American University, Washington, DC, United States.,Center for Neuroscience and Behavior, American University, Washington, DC, United States
| |
Collapse
|
11
|
Alam M, Ali S, Ahmed S, Elasbali AM, Adnan M, Islam A, Hassan MI, Yadav DK. Therapeutic Potential of Ursolic Acid in Cancer and Diabetic Neuropathy Diseases. Int J Mol Sci 2021; 22:12162. [PMID: 34830043 PMCID: PMC8621142 DOI: 10.3390/ijms222212162] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
Ursolic acid (UA) is a pentacyclic triterpenoid frequently found in medicinal herbs and plants, having numerous pharmacological effects. UA and its analogs treat multiple diseases, including cancer, diabetic neuropathy, and inflammatory diseases. UA inhibits cancer proliferation, metastasis, angiogenesis, and induced cell death, scavenging free radicals and triggering numerous anti- and pro-apoptotic proteins. The biochemistry of UA has been examined broadly based on the literature, with alterations frequently having been prepared on positions C-3 (hydroxyl), C12-C13 (double bonds), and C-28 (carboxylic acid), leading to several UA derivatives with increased potency, bioavailability and water solubility. UA could be used as a protective agent to counter neural dysfunction via anti-oxidant and anti-inflammatory effects. It is a potential therapeutic drug implicated in the treatment of cancer and diabetic complications diseases provide novel machinery to the anti-inflammatory properties of UA. The pharmacological efficiency of UA is exhibited by the therapeutic theory of one-drug → several targets → one/multiple diseases. Hence, UA shows promising therapeutic potential for cancer and diabetic neuropathy diseases. This review aims to discuss mechanistic insights into promising beneficial effects of UA. We further explained the pharmacological aspects, clinical trials, and potential limitations of UA for the management of cancer and diabetic neuropathy diseases.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (A.I.); (M.I.H.)
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (A.I.); (M.I.H.)
| | - Sarfraz Ahmed
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India;
| | - Abdelbaset Mohamed Elasbali
- Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail P.O. Box 2440, Saudi Arabia;
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (A.I.); (M.I.H.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (A.I.); (M.I.H.)
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon 21924, Korea
| |
Collapse
|
12
|
Holton KF. Micronutrients May Be a Unique Weapon Against the Neurotoxic Triad of Excitotoxicity, Oxidative Stress and Neuroinflammation: A Perspective. Front Neurosci 2021; 15:726457. [PMID: 34630015 PMCID: PMC8492967 DOI: 10.3389/fnins.2021.726457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Excitotoxicity has been implicated in many neurological disorders and is a leading cause of oxidative stress and neuroinflammation in the nervous system. Most of the research to date has focused on each of these conditions individually; however, excitotoxicity, oxidative stress, and neuroinflammation have the ability to influence one another in a self-sustaining manner, thus functioning as a "neurotoxic triad." This perspective article re-introduces the concept of the neurotoxic triad and reviews how specific dietary micronutrients have been shown to protect against not only oxidative stress, but also excitotoxicity and neuroinflammation. Future dietary interventions for neurological disorders could focus on the effects on all three aspects of the neurotoxic triad.
Collapse
Affiliation(s)
- Kathleen F Holton
- Nutritional Neuroscience Lab, Department of Health Studies, Center for Neuroscience and Behavior, American University, Washington, DC, United States
| |
Collapse
|
13
|
Hiraoka H, Nomura R, Takasugi N, Akai R, Iwawaki T, Kumagai Y, Fujimura M, Uehara T. Spatiotemporal analysis of the UPR transition induced by methylmercury in the mouse brain. Arch Toxicol 2021; 95:1241-1250. [PMID: 33454823 DOI: 10.1007/s00204-021-02982-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/06/2021] [Indexed: 01/11/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures a specific area of the brain. MeHg-mediated neurotoxicity is believed to be caused by oxidative stress and endoplasmic reticulum (ER) stress but the mechanism by which those stresses lead to neuronal loss is unclear. Here, by utilizing the ER stress-activated indicator (ERAI) system, we investigated the signaling alterations in the unfolded protein response (UPR) prior to neuronal apoptosis in the mouse brain. In ERAI transgenic mice exposed to MeHg (25 mg/kg, S.C.), the ERAI signal, which indicates activation of the cytoprotective pathway of the UPR, was detected in the brain. Interestingly, detailed ex vivo analysis showed that the ERAI signal was localized predominantly in neurons. Time course analysis of MeHg exposure (30 ppm in drinking water) showed that whereas the ERAI signal was gradually attenuated at the late phase after increasing at the early phase, activation of the apoptotic pathway of the UPR was enhanced in proportion to the exposure time. These results suggest that MeHg induces not only ER stress but also neuronal cell death via a UPR shift. UPR modulation could be a therapeutic target for treating neuropathy caused by electrophiles similar to MeHg.
Collapse
Affiliation(s)
- Hideki Hiraoka
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Ryosuke Nomura
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Nobumasa Takasugi
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan
| | - Ryoko Akai
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Masatake Fujimura
- Department of Basic Medical Science, National Institute for Minamata Disease, Kumamoto, 867-0008, Japan
| | - Takashi Uehara
- Department of Medicinal Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8530, Japan.
| |
Collapse
|
14
|
Dionysopoulou S, Wikström P, Walum E, Thermos K. Effect of NADPH oxidase inhibitors in an experimental retinal model of excitotoxicity. Exp Eye Res 2020; 200:108232. [DOI: 10.1016/j.exer.2020.108232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023]
|
15
|
Kozin SV, Kravtsov AA, Zlischeva EI, Shurygina LV, Malyshko VV, Moiseev AV, Elkina AA, Baryshev MG. The Influence of a Deuterium Depleted Drinking Diet on the Functional State of the Central Nervous System of Animals in Hypoxia. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920060093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
16
|
Neurotoxicity in Gulf War Illness and the potential role of glutamate. Neurotoxicology 2020; 80:60-70. [DOI: 10.1016/j.neuro.2020.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
|
17
|
Tang KS. The potential role of nanoyttria in alleviating oxidative stress biomarkers: Implications for Alzheimer's disease therapy. Life Sci 2020; 259:118287. [PMID: 32814066 DOI: 10.1016/j.lfs.2020.118287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease that requires immediate attention. Oxidative stress that leads to the generation of reactive oxygen species is a contributing factor to the disease progression by promoting synthesis and deposition of amyloid-β, the main hallmark protein in AD. It has been previously demonstrated that nanoyttria possesses antioxidant properties and can alleviate cellular oxidative injury in various toxicity and disease models. This review proposed that nanoyttria could be used for the treatment of AD. In this paper, the evidence on the antioxidant potential of nanoyttria is presented and its prospects on AD therapy are discussed.
Collapse
Affiliation(s)
- Kim San Tang
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia; Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
18
|
Armada-Moreira A, Gomes JI, Pina CC, Savchak OK, Gonçalves-Ribeiro J, Rei N, Pinto S, Morais TP, Martins RS, Ribeiro FF, Sebastião AM, Crunelli V, Vaz SH. Going the Extra (Synaptic) Mile: Excitotoxicity as the Road Toward Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:90. [PMID: 32390802 PMCID: PMC7194075 DOI: 10.3389/fncel.2020.00090] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
Excitotoxicity is a phenomenon that describes the toxic actions of excitatory neurotransmitters, primarily glutamate, where the exacerbated or prolonged activation of glutamate receptors starts a cascade of neurotoxicity that ultimately leads to the loss of neuronal function and cell death. In this process, the shift between normal physiological function and excitotoxicity is largely controlled by astrocytes since they can control the levels of glutamate on the synaptic cleft. This control is achieved through glutamate clearance from the synaptic cleft and its underlying recycling through the glutamate-glutamine cycle. The molecular mechanism that triggers excitotoxicity involves alterations in glutamate and calcium metabolism, dysfunction of glutamate transporters, and malfunction of glutamate receptors, particularly N-methyl-D-aspartic acid receptors (NMDAR). On the other hand, excitotoxicity can be regarded as a consequence of other cellular phenomena, such as mitochondrial dysfunction, physical neuronal damage, and oxidative stress. Regardless, it is known that the excessive activation of NMDAR results in the sustained influx of calcium into neurons and leads to several deleterious consequences, including mitochondrial dysfunction, reactive oxygen species (ROS) overproduction, impairment of calcium buffering, the release of pro-apoptotic factors, among others, that inevitably contribute to neuronal loss. A large body of evidence implicates NMDAR-mediated excitotoxicity as a central mechanism in the pathogenesis of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and epilepsy. In this review article, we explore different causes and consequences of excitotoxicity, discuss the involvement of NMDAR-mediated excitotoxicity and its downstream effects on several neurodegenerative disorders, and identify possible strategies to study new aspects of these diseases that may lead to the discovery of new therapeutic approaches. With the understanding that excitotoxicity is a common denominator in neurodegenerative diseases and other disorders, a new perspective on therapy can be considered, where the targets are not specific symptoms, but the underlying cellular phenomena of the disease.
Collapse
Affiliation(s)
- Adam Armada-Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Joana I. Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Carolina Campos Pina
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Oksana K. Savchak
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Sara Pinto
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Tatiana P. Morais
- Neuroscience Division, School of Bioscience, Cardiff University, Cardiff, United Kingdom
| | - Robertta Silva Martins
- Laboratório de Neurofarmacologia, Instituto Biomédico, Universidade Federal Fluminense, Niterói, Brazil
| | - Filipa F. Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Vincenzo Crunelli
- Neuroscience Division, School of Bioscience, Cardiff University, Cardiff, United Kingdom
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Sandra H. Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
19
|
Deletraz A, Zéamari K, Hua K, Combes M, Villamena FA, Tuccio B, Callizot N, Durand G. Substituted α-Phenyl and α-Naphthlyl- N- tert-butyl Nitrones: Synthesis, Spin-Trapping, and Neuroprotection Evaluation. J Org Chem 2020; 85:6073-6085. [PMID: 32267700 DOI: 10.1021/acs.joc.0c00563] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
New derivatives of α-phenyl-N-tert-butyl nitrone (PBN) bearing a hydroxyl, an acetate, or an acetamide substituent on the N-tert-butyl moiety and para-substituted phenyl or naphthlyl moieties were synthesized. Their ability to trap hydroxymethyl radical was evaluated by electron paramagnetic resonance spectroscopy. The presence of two electron-withdrawing substituents on both sides of the nitronyl function improves the spin-trapping properties, with 4-HOOC-PBN-CH2OAc and 4-HOOC-PBN-CH2NHAc being ∼4× more reactive than PBN. The electrochemical properties of the derivatives were further investigated by cyclic voltammetry and showed that the redox potentials of the nitrones are largely influenced by the nature of the substituents both on the aromatic ring and on the N-tert-butyl function. The acetamide derivatives PBN-CH2NHAc, 4-AcNHCH2-PBN-CH2NHAc, and 4-MeO-PBN-CH2NHAc were the easiest to oxidize. A computational approach was used to rationalize the effect of functionalization on the free energies of nitrone reactivity with hydroxymethyl radical as well as on the electron affinity and ionization potential. Finally, the neuroprotection of the derivatives was evaluated in an in vitro model of cellular injury on cortical neurons. Five derivatives showed good protection at very low concentrations (0.1-10 μM), with PBN-CH2NHAc and 4-HOOC-PBN being the two most promising agents.
Collapse
Affiliation(s)
- Anaïs Deletraz
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM & Avignon Université, Equipe Chimie Bioorganique et Systèmes Amphiphiles, 301 rue Baruch de Spinoza, BP 21239, Avignon 84916 Cedex 9, France
| | - Kamal Zéamari
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM & Avignon Université, Equipe Chimie Bioorganique et Systèmes Amphiphiles, 301 rue Baruch de Spinoza, BP 21239, Avignon 84916 Cedex 9, France
| | - Kangyu Hua
- The Ohio State University, Department of Biological Chemistry and Pharmacology, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Maud Combes
- Neuro-Sys, 410 Chemin Départemental 60, 13120 Gardanne, France
| | - Frederick A Villamena
- The Ohio State University, Department of Biological Chemistry and Pharmacology, 473 West 12th Avenue, Columbus, Ohio 43210, United States
| | - Béatrice Tuccio
- Aix-Marseille Université, CNRS, ICR UMR 7273, Avenue Escadrille Normandie Niemen, 13397 Marseille Cedex 20, France
| | - Noelle Callizot
- Neuro-Sys, 410 Chemin Départemental 60, 13120 Gardanne, France
| | - Grégory Durand
- Institut des Biomolécules Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM & Avignon Université, Equipe Chimie Bioorganique et Systèmes Amphiphiles, 301 rue Baruch de Spinoza, BP 21239, Avignon 84916 Cedex 9, France
| |
Collapse
|
20
|
Hameed MQ, Hsieh TH, Morales-Quezada L, Lee HHC, Damar U, MacMullin PC, Hensch TK, Rotenberg A. Ceftriaxone Treatment Preserves Cortical Inhibitory Interneuron Function via Transient Salvage of GLT-1 in a Rat Traumatic Brain Injury Model. Cereb Cortex 2019; 29:4506-4518. [PMID: 30590449 PMCID: PMC7150617 DOI: 10.1093/cercor/bhy328] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) results in a decrease in glutamate transporter-1 (GLT-1) expression, the major mechanism for glutamate removal from synapses. Coupled with an increase in glutamate release from dead and dying neurons, this causes an increase in extracellular glutamate. The ensuing glutamate excitotoxicity disproportionately damages vulnerable GABAergic parvalbumin-positive inhibitory interneurons, resulting in a progressively worsening cortical excitatory:inhibitory imbalance due to a loss of GABAergic inhibitory tone, as evidenced by chronic post-traumatic symptoms such as epilepsy, and supported by neuropathologic findings. This loss of intracortical inhibition can be measured and followed noninvasively using long-interval paired-pulse transcranial magnetic stimulation with mechanomyography (LI-ppTMS-MMG). Ceftriaxone, a β-lactam antibiotic, is a potent stimulator of the expression of rodent GLT-1 and would presumably decrease excitotoxic damage to GABAergic interneurons. It may thus be a viable antiepileptogenic intervention. Using a rat fluid percussion injury TBI model, we utilized LI-ppTMS-MMG, quantitative PCR, and immunohistochemistry to test whether ceftriaxone treatment preserves intracortical inhibition and cortical parvalbumin-positive inhibitory interneuron function after TBI in rat motor cortex. We show that neocortical GLT-1 gene and protein expression are significantly reduced 1 week after TBI, and this transient loss is mitigated by ceftriaxone. Importantly, whereas intracortical inhibition declines progressively after TBI, 1 week of post-TBI ceftriaxone treatment attenuates the loss of inhibition compared to saline-treated controls. This finding is accompanied by significantly higher parvalbumin gene and protein expression in ceftriaxone-treated injured rats. Our results highlight prospects for ceftriaxone as an intervention after TBI to prevent cortical inhibitory interneuron dysfunction, partly by preserving GLT-1 expression.
Collapse
Affiliation(s)
- Mustafa Q Hameed
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tsung-Hsun Hsieh
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Physical Therapy & Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Leon Morales-Quezada
- Spaulding Neuromodulation Center, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Henry H C Lee
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ugur Damar
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul C MacMullin
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Takao K Hensch
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular & Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Alexander Rotenberg
- Department of Neurology, Division of Epilepsy and Clinical Neurophysiology, Neuromodulation Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Kuklin V, Akhatov N, Kondratiev T, Konkayev A, Baigenzhin A, Konkayeva M, Karibekov T, Barlow N, Tveita T, Dahl V. The influences of morphine or ketamine pre-treatment on hemodynamic, acid-base status, biochemical markers of brain damage and early survival in rats after asphyxial cardiac arrest. BMC Anesthesiol 2019; 19:214. [PMID: 31747898 PMCID: PMC6868711 DOI: 10.1186/s12871-019-0884-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 10/31/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND In different models of hypoxia, blockade of opioid or N-methyl-D-aspartate (NMDA) receptors shows cardio- and neuroprotective effects with a consequent increase in animal survival. The aim of the study was to investigate effects of pre-treatment with Morphine or Ketamine on hemodynamic, acid-base status, early survival, and biochemical markers of brain damage in a rat model of asphyxial cardiac arrest (ACA). METHODS Under anaesthesia with Thiopental Sodium 60 mg/kg, i.p., Wistar rats (n = 42) were tracheostomized and catheters were inserted in a femoral vein and artery. After randomization, the rats were pre-treated with: Morphine 5 mg/kg i.v. (n = 14); Ketamine 40 mg/kg i.v. (n = 14); or equal volume of i.v. NaCl 0.9% as a Control (n = 14). ACA was induced by corking of the tracheal tube for 8 min, and defined as a mean arterial pressure (MAP) < 20 mmHg. Resuscitation was started at 5 min after cardiac arrest (CA). Invasive MAP was recorded during experiments. Arterial pH and blood gases were sampled at baseline (BL) and 10 min after CA. At the end of experiments, all surviving rats were euthanised, brain and blood samples for measurement of Neuron Specific Enolase (NSE), s100 calcium binding protein B (s100B) and Caspase-3 (CS-3) were retrieved. RESULTS At BL no differences between groups were found in hemodynamic or acid-base status. After 3 min of asphyxia, all animals had cardiac arrest (CA). Return of spontaneous circulation (MAP > 60 mmHg) was achieved in all animals within 3 min after CA. At the end of the experiment, the Ketamine pre-treated group had increased survival (13 of 14; 93%) compared to the Control (7 of 14; 50%) and Morphine (10 of 14; 72%) groups (p = 0.035). Biochemical analysis of plasma concentration of NSE and s100B as well as an analysis of CS-3 levels in the brain tissue did not reveal any differences between the study groups. CONCLUSION In rats after ACA, pre-treatment with Morphine or Ketamine did not have any significant influence on hemodynamic and biochemical markers of brain damage. However, significantly better pH level and increased early survival were found in the Ketamine pre-treated group.
Collapse
Affiliation(s)
- Vladimir Kuklin
- Department of Anaesthesiology and Intensive Care Medicine, Akershus university hospital, Sykehusveien, 25, 1478, Lørenskog, Norway.
| | - Nurlan Akhatov
- Department of Anaesthesiology and Intensive Care Medicine, Astana Medical University, Nur-Sultan, Kazakhstan.,Department of Anaesthesiology and Intensive Care Medicine, National Scientific Medical Center, Nur-Sultan, Kazakhstan
| | - Timofei Kondratiev
- Anaesthesia and Critical Care Research Group, Department of Clinical Medicine, UiT - The Arctic University of Norway, 9037, Tromsø, Norway
| | - Aidos Konkayev
- Department of Anaesthesiology and Intensive Care Medicine, Astana Medical University, Nur-Sultan, Kazakhstan
| | - Abai Baigenzhin
- Department of Anaesthesiology and Intensive Care Medicine, National Scientific Medical Center, Nur-Sultan, Kazakhstan
| | - Maiya Konkayeva
- Department of Anaesthesiology and Intensive Care Medicine, Astana Medical University, Nur-Sultan, Kazakhstan
| | - Temirlan Karibekov
- Department of Anaesthesiology and Intensive Care Medicine, National Scientific Medical Center, Nur-Sultan, Kazakhstan
| | - Nicholas Barlow
- Department of Anaesthesiology and Intensive Care Medicine, Akershus university hospital, Sykehusveien, 25, 1478, Lørenskog, Norway
| | - Torkjel Tveita
- Division of Surgical Medicine and Intensive Care, University Hospital of Northern Norway, 9038, Tromsø, Norway
| | - Vegard Dahl
- Department of Anaesthesiology and Intensive Care Medicine, Akershus university hospital, Sykehusveien, 25, 1478, Lørenskog, Norway.,Department of Anaesthesiology and Intensive Care Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Tobore TO. Towards a comprehensive etiopathogenetic and pathophysiological theory of multiple sclerosis. Int J Neurosci 2019; 130:279-300. [PMID: 31588832 DOI: 10.1080/00207454.2019.1677648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Multiple sclerosis (MS) is a neurodegenerative disease caused by dysfunction of the immune system that affects the central nervous system (CNS). It is characterized by demyelination, chronic inflammation, neuronal and oligodendrocyte loss and reactive astrogliosis. It can result in physical disability and acute neurological and cognitive problems. Despite the gains in knowledge of immunology, cell biology, and genetics in the last five decades, the ultimate etiology or specific elements that trigger MS remain unknown. The objective of this review is to propose a theoretical basis for MS etiopathogenesis.Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(multiple sclerosis* OR EAE) AND (pathophysiology* OR etiopathogenesis)". The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and June 30, 2019. The search was filtered down to 362 articles which were included in this review.Results: A framework to better understand the etiopathogenesis and pathophysiology of MS can be derived from four essential factors; mitochondria dysfunction (MtD) & oxidative stress (OS), vitamin D (VD), sex hormones and thyroid hormones. These factors play a direct role in MS etiopathogenesis and have a modulatory effect on many other factors involved in the disease.Conclusions: For better MS prevention and treatment outcomes, efforts should be geared towards treating thyroid problems, sex hormone alterations, VD deficiency, sleep problems and melatonin alterations. MS patients should be encouraged to engage in activities that boost total antioxidant capacity (TAC) including diet and regular exercise and discouraged from activities that promote OS including smoking and alcohol consumption.
Collapse
|
23
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Sala G, Arosio A, Conti E, Beretta S, Lunetta C, Riva N, Ferrarese C, Tremolizzo L. Riluzole Selective Antioxidant Effects in Cell Models Expressing Amyotrophic Lateral Sclerosis Endophenotypes. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:438-442. [PMID: 31352711 PMCID: PMC6705104 DOI: 10.9758/cpn.2019.17.3.438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/07/2018] [Accepted: 08/12/2018] [Indexed: 11/18/2022]
Abstract
Objective Until recently, riluzole was the only drug licensed for amyotrophic lateral sclerosis (ALS). In spite of its efficacy, the mechanism of action remains elusive, and both blocking of glutamate release and antioxidant properties have been postulated. Here we characterized human SH-SY5Y neuroblastoma cell lines, taking advantage of their insensitivity to excitotoxic insults, in order to selectively assess the presence of a direct antioxidant effect of riluzole. Methods SH-SY5Y cells, either parental or overexpressing the G93A SOD1 mutation, were exposed for 24 hours to the selected stimuli. Results Riluzole (1–10 μM) was able to counteract the effects of H2O2 exposure (200 μM/24 hr), limiting both cell death and whole-cell reactive oxygen species (ROS) increase. The same experiments were repeated using SH-SY5Y cells carrying the familial ALS-related G93A-SOD1 mutation and constitutively expressing two-fold increased whole-cell ROS levels with respect to wild-type cells: riluzole was ineffective in this paradigm. Analogously, riluzole was ineffective in preventing cell death induced by exposing SH-SY5Y cells to 3-morpholino-sydnonimine (SIN-1, 1.5 mM/24 hr), a reactive nitrogen species (RNS) donor. Conclusion Our data support a direct antioxidant action of riluzole. Furthermore, the lack of efficacy of riluzole observed in the SOD1 cell model mirrors the lack of efficacy already demonstrated in cognate mouse models of ALS, plausibly reflecting differences in the underlying pathogenic mechanisms. Finally, riluzole inefficacy against nitrosative stress might support the idea that a combined therapeutic intervention may result more effective in ALS patients, as in the case of co-administration of edaravone, a drug known to reduce RNS.
Collapse
Affiliation(s)
- Gessica Sala
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| | - Alessandro Arosio
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| | - Elisa Conti
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| | - Simone Beretta
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy.,Neurology Unit, San Gerardo Hospital, Monza, Milano, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicentre (NEMO), Fondazione Serena Onlus, Milano, Italy
| | - Nilo Riva
- San Raffaele Scientific Institute, Milano, Italy
| | - Carlo Ferrarese
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy.,Neurology Unit, San Gerardo Hospital, Monza, Milano, Italy
| | - Lucio Tremolizzo
- Labaratory of Neurobiology, School of Medicine and Surgery and Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy.,Neurology Unit, San Gerardo Hospital, Monza, Milano, Italy
| |
Collapse
|
25
|
Tobore TO. Towards a comprehensive understanding of the contributions of mitochondrial dysfunction and oxidative stress in the pathogenesis and pathophysiology of Huntington's disease. J Neurosci Res 2019; 97:1455-1468. [DOI: 10.1002/jnr.24492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022]
|
26
|
de Oliveira MR, Duarte AR, Chenet AL, de Almeida FJS, Andrade CMB. Carnosic Acid Pretreatment Attenuates Mitochondrial Dysfunction in SH-SY5Y Cells in an Experimental Model of Glutamate-Induced Excitotoxicity. Neurotox Res 2019; 36:551-562. [PMID: 31016690 DOI: 10.1007/s12640-019-00044-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/29/2019] [Accepted: 04/09/2019] [Indexed: 11/29/2022]
Abstract
Mitochondria are the major site of adenosine triphosphate (ATP) production in mammalian cells. Moreover, mitochondria produce most of the reactive oxygen species (ROS) in nucleated cells. Redox and bioenergetic abnormalities have been seen in mitochondria during the onset and progression of neurodegenerative diseases. In that context, excitotoxicity induced by glutamate (GLU) plays an important role in mediating neurotoxicity. Several drugs have been used in the treatment of diseases involving excitotoxicity. Nonetheless, some patients (20-30%) present drug resistance. Thus, it is necessary to find chemicals able to attenuate mitochondrial dysfunction in the case of excitotoxicity. In this work, we treated the human neuroblastoma SH-SY5Y cell line with the diterpene carnosic acid (CA) at 1 μM for 12 h prior to the exposure to GLU for further 24 h. We found that CA prevented the GLU-induced mitochondrion-related redox impairment and bioenergetic decline in SH-SY5Y cells. CA also downregulated the pro-apoptotic stimulus elicited by GLU in this experimental model. CA exerted mitochondrial protection by a mechanism associated with the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), since silencing of this protein with small interfering RNA (siRNA) suppressed the CA-induced protective effects. Future directions include investigating whether CA would be able to modulate mitochondrial function and/or dynamics in in vivo experimental models of excitotoxicity.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil. .,Programa de Pós-Graduação em Química (PPGQ), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil. .,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil.
| | - Adriane Ribeiro Duarte
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil.,Programa de Pós-Graduação em Química (PPGQ), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
| | - Aline Lukasievicz Chenet
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil.,Programa de Pós-Graduação em Química (PPGQ), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
| | - Fhelipe Jolner Souza de Almeida
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil.,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
| | - Claudia Marlise Balbinotti Andrade
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil.,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Mato Grosso (UFMT), Cuiaba, MT, Brazil
| |
Collapse
|
27
|
Hara Y, Crimins JL, Puri R, Wang ACJ, Motley SE, Yuk F, Ramos TM, Janssen WGM, Rapp PR, Morrison JH. Estrogen Alters the Synaptic Distribution of Phospho-GluN2B in the Dorsolateral Prefrontal Cortex While Promoting Working Memory in Aged Rhesus Monkeys. Neuroscience 2019; 394:303-315. [PMID: 30482274 DOI: 10.1016/j.neuroscience.2018.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/06/2023]
Abstract
Age- and menopause-related deficits in working memory can be partially restored with estradiol replacement in women and female nonhuman primates. Working memory is a cognitive function reliant on persistent firing of dorsolateral prefrontal cortex (dlPFC) neurons that requires the activation of GluN2B-containing glutamate NMDA receptors. We tested the hypothesis that the distribution of phospho-Tyr1472-GluN2B (pGluN2B), a predominant form of GluN2B seen at the synapse, is sensitive to aging or estradiol treatment and coupled to working memory performance. First, ovariectomized young and aged rhesus monkeys (Macaca mulatta) received long-term cyclic vehicle (V) or estradiol (E) treatment and were tested on the delayed response (DR) test of working memory. Then, serial section electron microscopic immunocytochemistry was performed to quantitatively assess the subcellular distribution of pGluN2B. While the densities of pGluN2B immunogold particles in dlPFC dendritic spines were not different across age or treatment groups, the percentage of gold particles located within the synaptic compartment was significantly lower in aged-E monkeys compared to young-E and aged-V monkeys. On the other hand, the percentage of pGluN2B gold particles in the spine cytoplasm was decreased with E treatment in young, but increased with E in aged monkeys. In aged monkeys, DR average accuracy inversely correlated with the percentage of synaptic pGluN2B, while it positively correlated with the percentage of cytoplasmic pGluN2B. Together, E replacement may promote cognitive health in aged monkeys, in part, by decreasing the relative representation of synaptic pGluN2B and potentially protecting the dlPFC from calcium toxicity.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Johanna L Crimins
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Rishi Puri
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Athena C J Wang
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Sarah E Motley
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; California National Primate Research Center, Davis, CA 95616, United States
| | - Frank Yuk
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Tiffany M Ramos
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - William G M Janssen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Peter R Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224, United States
| | - John H Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; California National Primate Research Center, Davis, CA 95616, United States; Department of Neurology, School of Medicine, University of California, Davis, CA 95616, United States.
| |
Collapse
|
28
|
Free d-aspartate triggers NMDA receptor-dependent cell death in primary cortical neurons and perturbs JNK activation, Tau phosphorylation, and protein SUMOylation in the cerebral cortex of mice lacking d-aspartate oxidase activity. Exp Neurol 2019; 317:51-65. [PMID: 30822420 DOI: 10.1016/j.expneurol.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/19/2019] [Accepted: 02/24/2019] [Indexed: 11/23/2022]
Abstract
In mammals, free d-aspartate (D-Asp) is abundant in the embryonic brain, while levels remain very low during adulthood as a result of the postnatal expression and activity of the catabolizing enzyme d-aspartate oxidase (DDO). Previous studies have shown that long-lasting exposure to nonphysiological, higher D-Asp concentrations in Ddo knockout (Ddo-/-) mice elicits a precocious decay of synaptic plasticity and cognitive functions, along with a dramatic age-dependent expression of active caspase 3, associated with increased cell death in different brain regions, including hippocampus, prefrontal cortex, and substantia nigra pars compacta. Here, we investigate the yet unclear molecular and cellular events associated with the exposure of abnormally high D-Asp concentrations in cortical primary neurons and in the brain of Ddo-/- mice. For the first time, our in vitro findings document that D-Asp induces in a time-, dose-, and NMDA receptor-dependent manner alterations in JNK and Tau phosphorylation levels, associated with pronounced cell death in primary cortical neurons. Moreover, observations obtained in Ddo-/- animals confirmed that high in vivo levels of D-Asp altered cortical JNK signaling, Tau phosphorylation and enhanced protein SUMOylation, indicating a robust indirect role of DDO activity in regulating these biochemical NMDA receptor-related processes. Finally, no gross modifications in D-Asp concentrations and DDO mRNA expression were detected in the cortex of patients with Alzheimer's disease when compared to age-matched healthy controls.
Collapse
|
29
|
Li HY, Hong X, Huang M, So KF. Voluntary running delays primary degeneration in rat retinas after partial optic nerve transection. Neural Regen Res 2019; 14:728-734. [PMID: 30632515 PMCID: PMC6352605 DOI: 10.4103/1673-5374.247481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Running is believed to be beneficial for human health. Many studies have focused on the neuroprotective effects of voluntary running on animal models. There were both primary and secondary degeneration in neurodegenerative diseases, including glaucoma. However, whether running can delay primary or secondary degeneration or both of them was not clear. Partial optic nerve transection model is a valuable glaucoma model for studying both primary and secondary degeneration because it can separate primary (mainly in the superior retina) from secondary (mainly in the inferior retina) degeneration. Therefore, we compared the survival of retinal ganglion cells between Sprague-Dawley rat runners and non-runners both in the superior and inferior retinas. Excitotoxicity, oxidative stress, and apoptosis are involved in the degeneration of retinal ganglion cells in glaucoma. So we also used western immunoblotting to compare the expression of some proteins involved in apoptosis (phospho-c-Jun N-terminal kinases, p-JNKs), oxidative stress (manganese superoxide dismutase, MnSOD) and excitotoxicity (glutamine synthetase) between runners and non-runners after partial optic nerve transection. Results showed that voluntary running delayed the death of retinal ganglion cells vulnerable to primary degeneration but not those to secondary degeneration. In addition, voluntary running decreased the expression of glutamine synthetase, but not the expression of p-JNKs and MnSOD in the superior retina after partial optic nerve transection. These results illustrated that primary degeneration of retinal ganglion cells might be mainly related with excitotoxicity rather than oxidative stress; and the voluntary running could down-regulate excitotoxicity to delay the primary degeneration of retinal ganglion cells after partial optic nerve transection.
Collapse
Affiliation(s)
- Hong-Ying Li
- Department of Anatomy, School of Medicine; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Xi Hong
- Department of Anatomy, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Mi Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, Guangdong Province; Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
30
|
Yucel MS, Kayis T. Imidacloprid induced alterations in oxidative stress, biochemical, genotoxic, and immunotoxic biomarkers in non-mammalian model organism Galleria mellonella L. (Lepidoptera: Pyralidae). JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2018; 54:27-34. [PMID: 30426817 DOI: 10.1080/03601234.2018.1530545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Imidacloprid (IMI), a neonicotinoid insecticide, is widely used to control pests in agriculture. We investigated the changes in antioxidant enzyme activities, lipid peroxidation levels, biochemical effects, genotoxic effect, and immunotoxic effect of sublethal doses (0.25, 0.50, 0.75, and 1.00 µg) of IMI at different time periods (24, 48, 72, and 96 h) on a model organism, Galleria mellonella L. The results indicated that there were dose-dependent increases in both antioxidant enzyme activities (SOD and CAT) and MDA levels. Protein content was not affected by IMI at 24th and 48th, whereas it was decreased by the highest dose of IMI (1.00 µg) at 72nd and 96th h. Lipid and carbohydrate contents were reduced with increasing doses of IMI. Micronucleus frequency significantly increased in all IMI doses. All IMI doses caused a significant decrease in THC at 24th, 48th, and 72nd h. Our results can help to illustrate the effects of IMI in target organisms and indirectly may aid to discover potential risk of it on nontarget organisms. Future studies, at molecular levels, will be helpful in understanding the mechanism of action of IMI on these biomarkers.
Collapse
Affiliation(s)
- Mehmet Sait Yucel
- a Institutes of Natural and Applied Sciences, Adiyaman University , Adiyaman , Turkey
| | - Tamer Kayis
- b Faculty of Science and Letters, Department of Biology , Adiyaman University , Adiyaman , Turkey
| |
Collapse
|
31
|
Seo DY, Lee SR, Heo JW, No MH, Rhee BD, Ko KS, Kwak HB, Han J. Ursolic acid in health and disease. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:235-248. [PMID: 29719446 PMCID: PMC5928337 DOI: 10.4196/kjpp.2018.22.3.235] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/22/2022]
Abstract
Ursolic acid (UA) is a natural triterpene compound found in various fruits and vegetables. There is a growing interest in UA because of its beneficial effects, which include anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-carcinogenic effects. It exerts these effects in various tissues and organs: by suppressing nuclear factor-kappa B signaling in cancer cells, improving insulin signaling in adipose tissues, reducing the expression of markers of cardiac damage in the heart, decreasing inflammation and increasing the level of anti-oxidants in the brain, reducing apoptotic signaling and the level of oxidants in the liver, and reducing atrophy and increasing the expression levels of adenosine monophosphate-activated protein kinase and irisin in skeletal muscles. Moreover, UA can be used as an alternative medicine for the treatment and prevention of cancer, obesity/diabetes, cardiovascular disease, brain disease, liver disease, and muscle wasting (sarcopenia). In this review, we have summarized recent data on the beneficial effects and possible uses of UA in health and disease managements.
Collapse
Affiliation(s)
- Dae Yun Seo
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Sung Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.,Department of Convergence Biomedical Science, Inje University, Busan 47392, Korea
| | - Jun-Won Heo
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Mi-Hyun No
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Byoung Doo Rhee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Kyung Soo Ko
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, BK21 Plus Team, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea.,Department of Health Science and Technology, Graduate School, Inje University, Busan 47392, Korea
| |
Collapse
|
32
|
Mitra S, Natarajan R, Ziedonis D, Fan X. Antioxidant and anti-inflammatory nutrient status, supplementation, and mechanisms in patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2017; 78:1-11. [PMID: 28499901 DOI: 10.1016/j.pnpbp.2017.05.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/06/2017] [Accepted: 05/08/2017] [Indexed: 10/19/2022]
Abstract
Over 50 million people around the world suffer from schizophrenia, a severe mental illness characterized by misinterpretation of reality. Although the exact causes of schizophrenia are still unknown, studies have indicated that inflammation and oxidative stress may play an important role in the etiology of the disease. Pro-inflammatory cytokines are crucial for normal central nervous development and proper functioning of neural networks and neurotransmitters. Patients with schizophrenia tend to have abnormal immune activation resulting in elevated pro-inflammatory cytokine levels, ultimately leading to functional brain impairments. Patients with schizophrenia have also been found to suffer from oxidative stress, a result of an imbalance between the production of free radicals and the ability to detoxify their harmful effects. Furthermore, inflammation and oxidative stress are implicated to be related to the severity of psychotic symptoms. Several nutrients are known to have anti-inflammatory and antioxidant functions through various mechanisms in our body. The present review evaluates studies and literature that address the status and supplementation of omega-3 polyunsaturated fatty acids, vitamin D, B vitamins (B6, folate, B12), vitamin E, and carotenoids in different stages of schizophrenia. The possible anti-inflammatory and antioxidant mechanisms of action of each nutrient are discussed.
Collapse
Affiliation(s)
- Sumedha Mitra
- Department of Psychiatry, UMass Memorial Medical Center/University of Massachusetts Medical School, One Biotech, 365 Plantation Street, Worcester, MA 01605, USA
| | - Radhika Natarajan
- Department of Psychiatry, UMass Memorial Medical Center/University of Massachusetts Medical School, One Biotech, 365 Plantation Street, Worcester, MA 01605, USA
| | - Douglas Ziedonis
- Department of Psychiatry, UMass Memorial Medical Center/University of Massachusetts Medical School, One Biotech, 365 Plantation Street, Worcester, MA 01605, USA
| | - Xiaoduo Fan
- Department of Psychiatry, UMass Memorial Medical Center/University of Massachusetts Medical School, One Biotech, 365 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
33
|
Lima C, Pinto S, Napoleão P, Pronto-Laborinho AC, Barros MA, Freitas T, de Carvalho M, Saldanha C. Identification of erythrocyte biomarkers in amyotrophic lateral sclerosis. Clin Hemorheol Microcirc 2017; 63:423-437. [PMID: 27258202 DOI: 10.3233/ch-162066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease of the motor system. It has been hypothesised that red blood cells (RBCs) may be involved in the disease process by the release of damaging molecules. OBJECTIVE The aim of this ex vivo study is to compare RBCs biochemical and hemorheological parameters between ALS patients and healthy donors to identify novel biomarkers of the ALS disease. METHODS We included 82 ALS patients and 40 gender age-matched healthy donors. We performed quantification of erythrocyte aggregation and deformability, nitric oxide (NO) efflux from RBCs, acetylcholinesterase (AChE) enzyme activity and intraerythrocytic concentration of nitrite, nitrate and S-nitrosogluthatione (GSNO). RESULTS Erythrocyte deformability and AChE activity were increased in patients with ALS in comparison to healthy donors. NO efflux from RBCs and concentration of intraerythrocytic nitrite were lower in ALS patients. In patients, we found that for higher NO range of values the respiratory function is worse and that for higher AChE range of values the RBCs nitrite content increase. CONCLUSION The results of the present study indicate that NO efflux from RBCs and RBCs AChE should be further explored as potential biomarkers for ALS.
Collapse
Affiliation(s)
- Catarina Lima
- Instituto de Bioquímica, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Pinto
- Instituto de Fisiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Patrícia Napoleão
- Instituto de Bioquímica, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Catarina Pronto-Laborinho
- Instituto de Fisiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Amparo Barros
- Instituto de Fisiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Teresa Freitas
- Instituto de Bioquímica, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mamede de Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Departamento de Neurociências. Hospital de Santa Maria-CHLN, Lisbon, Portugal
| | - Carlota Saldanha
- Instituto de Bioquímica, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
34
|
Salihu AT, Muthuraju S, Idris Z, Izaini Ghani AR, Abdullah JM. Functional outcome after intracerebral haemorrhage – a review of the potential role of antiapoptotic agents. Rev Neurosci 2016; 27:317-27. [DOI: 10.1515/revneuro-2015-0046] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/21/2015] [Indexed: 11/15/2022]
Abstract
AbstractIntracerebral haemorrhage (ICH) is the second most common form of stroke and is associated with greater mortality and morbidity compared with ischaemic stroke. The current ICH management strategies, which mainly target primary injury mechanisms, have not been shown to improve patient’s functional outcome. Consequently, multimodality treatment approaches that will focus on both primary and secondary pathophysiology have been suggested. During the last decade, a proliferation of experimental studies has demonstrated the role of apoptosis in secondary neuronal loss at the periphery of the clot after ICH. Subsequently, the value of certain antiapoptotic agents in reducing neuronal death and improving functional outcome following ICH was evaluated in animal models. Preliminary evidence from those studies strongly supports the potential role of antiapoptotic agents in reducing neuronal death and improving functional outcome after intracerebral haemorrhage. Expectedly, the ongoing and subsequent clinical trials will substantiate these findings and provide clear information on the most potent and safe antiapoptotic agents, their appropriate dosage, and temporal window of action, thereby making them suitable for the multimodality treatment approach.
Collapse
|
35
|
Woźniak Ł, Skąpska S, Marszałek K. Ursolic Acid--A Pentacyclic Triterpenoid with a Wide Spectrum of Pharmacological Activities. Molecules 2015; 20:20614-41. [PMID: 26610440 PMCID: PMC6332387 DOI: 10.3390/molecules201119721] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/21/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
Ursolic acid (UA) is a natural terpene compound exhibiting many pharmaceutical properties. In this review the current state of knowledge about the health-promoting properties of this widespread, biologically active compound, as well as information about its occurrence and biosynthesis are presented. Particular attention has been paid to the application of ursolic acid as an anti-cancer agent; it is worth noticing that clinical tests suggesting the possibility of practical use of UA have already been conducted. Amongst other pharmacological properties of UA one can mention protective effect on lungs, kidneys, liver and brain, anti-inflammatory properties, anabolic effects on skeletal muscles and the ability to suppress bone density loss leading to osteoporosis. Ursolic acid also exhibits anti-microbial features against numerous strains of bacteria, HIV and HCV viruses and Plasmodium protozoa causing malaria.
Collapse
Affiliation(s)
- Łukasz Woźniak
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| | - Sylwia Skąpska
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| | - Krystian Marszałek
- Department of Fruit and Vegetable Product Technology, Institute of Agricultural and Food Biotechnology, 36 Rakowiecka Street, 02-532 Warsaw, Poland.
| |
Collapse
|
36
|
de Carvalho M, Eisen A, Krieger C, Swash M. Motoneuron firing in amyotrophic lateral sclerosis (ALS). Front Hum Neurosci 2014; 8:719. [PMID: 25294995 PMCID: PMC4170108 DOI: 10.3389/fnhum.2014.00719] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/27/2014] [Indexed: 01/09/2023] Open
Abstract
Amyotrophic lateral sclerosis is an inexorably progressive neurodegenerative disorder involving the classical motor system and the frontal effector brain, causing muscular weakness and atrophy, with variable upper motor neuron signs and often an associated fronto-temporal dementia. The physiological disturbance consequent on the motor system degeneration is beginning to be well understood. In this review we describe aspects of the motor cortical, neuronal, and lower motor neuron dysfunction. We show how studies of the changes in the pattern of motor unit firing help delineate the underlying pathophysiological disturbance as the disease progresses. Such studies are beginning to illuminate the underlying disordered pathophysiological processes in the disease, and are important in designing new approaches to therapy and especially for clinical trials.
Collapse
Affiliation(s)
- Mamede de Carvalho
- Institute of Physiology and Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Department of Neurosciences, Hospital Santa Maria, Faculty of Medicine, University of Lisbon Lisbon, Portugal
| | - Andrew Eisen
- Emeritus Professor of Neurology, University of British Columbia Vancouver, BC, Canada
| | - Charles Krieger
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby BC, Canada ; Department of Medicine (Neurology), University of British Columbia, Vancouver BC, Canada
| | - Michael Swash
- Institute of Physiology and Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Department of Neurosciences, Hospital Santa Maria, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Institute of Neuroscience, Barts and The London School of Medicine, Queen Mary University of London London, UK
| |
Collapse
|
37
|
Kuklin V. Survival rate in patients after sudden cardiac arrest at the university hospital of northern Norway treated with or without opioids: A retrospective evaluation. Saudi J Anaesth 2013; 7:310-4. [PMID: 24015136 PMCID: PMC3757806 DOI: 10.4103/1658-354x.115355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Experimental studies both in vivo and in vitro show significantly increased survival rate in animals and in cortical neurons respectively exposed to acute hypoxia and pre-treated with opioids compared to non-treated counterparts. Thus, the main aim of the study was to examine survival rates in patients after sudden cardiac arrest (SCA) in the hospital who were or were not treated with opioids before and/or during cardiac pulmonary resuscitation (CPR). METHODS The registry SCA database at the University Hospital of Northern Norway (UNN) for the period of January 2006-December 2009 was used to obtain data for the evaluation. Inclusion criteria were observed SCA at UNN for patients with American Society of Anesthesiologists (ASA) 1-3. Exclusion criteria included ASA four to five patients and unobserved SCA. Study patients were divided into two groups: Those not treated with opioids and those treated with opioids not more then 3 h before and/or during CPR. Survival rate 1, 2, 3 and 28 days post CPR were compared for the two groups. RESULTS A total of 117 patients were registered in the SCA database at UNN for the period from January 2006 to December 2009. Sixty seven patients were excluded from the study: 17 patients had an unknown time of SCA dιbut, two patients had only syncope and 48 were ASA four to five patients. A total of 50 ASA one to three patients were included in the study, 33 and 17 patients respectively in the control and opioid-treated groups. The patients who were treated with opioids before or during CPR had a significantly higher 1, 2, 3 and 28 days survival rate as compared to those receiving only conventional CPR. The model was adjusted for duration of CPR (P=0.047) and treatment with adrenaline (P=0.779) in the groups. Adjusted Odds ratio was 0.075 (95% confidence interval (CI): 0.015-0.387). Relative risk of fatal outcome in the opioids group was 0.2944 (95% CI: 0.1549-0.5594). CONCLUSION Significantly higher 1, 2, 3 and 28 days survival rate and reduced duration of CPR were found in the patients additionally treated with opioids compared to ordinary resuscitation. Further prospective, randomized, controlled trials are needed to investigate the effects of early administration of opioids during CPR on survival and brain function in patients with witnessed in-hospital SCA.
Collapse
Affiliation(s)
- Vladimir Kuklin
- Department of Anaesthesiology, Kongsberg Hospital, Kongsberg, Norway
| |
Collapse
|
38
|
Cardoso LM, Fernandes LG, Alves AM, Pedrosa ML, Silva ME, Colombari E, Moraes MFD, Júnior DAC. Cardiopulmonary reflex is attenuated in iron overload conscious rats. Nutr Neurosci 2013; 10:121-8. [DOI: 10.1080/10284150701413964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Chen CC, Wang KY, Shen CKJ. DNA 5-methylcytosine demethylation activities of the mammalian DNA methyltransferases. J Biol Chem 2013; 288:9084-91. [PMID: 23393137 PMCID: PMC3610981 DOI: 10.1074/jbc.m112.445585] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/06/2013] [Indexed: 01/19/2023] Open
Abstract
Methylation at the 5-position of DNA cytosine on the vertebrate genomes is accomplished by the combined catalytic actions of three DNA methyltransferases (DNMTs), the de novo enzymes DNMT3A and DNMT3B and the maintenance enzyme DNMT1. Although several metabolic routes have been suggested for demethylation of the vertebrate DNA, whether active DNA demethylase(s) exist has remained elusive. Surprisingly, we have found that the mammalian DNMTs, and likely the vertebrates DNMTs in general, can also act as Ca(2+) ion- and redox state-dependent active DNA demethylases. This finding suggests new directions for reinvestigation of the structures and functions of these DNMTs, in particular their roles in Ca(2+) ion-dependent biological processes, including the genome-wide/local DNA demethylation during early embryogenesis, cell differentiation, neuronal activity-regulated gene expression, and carcinogenesis.
Collapse
Affiliation(s)
- Chun-Chang Chen
- From the Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei 112 and
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Keh-Yang Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan
| |
Collapse
|
40
|
Ohno A, Yoshino F, Yoshida A, Hori N, Ono Y, Kimoto K, Onozuka M, Lee MCI. Soft-food diet induces oxidative stress in the rat brain. Neurosci Lett 2012; 508:42-6. [DOI: 10.1016/j.neulet.2011.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 11/30/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
|
41
|
Ferretti G, Bacchetti T. Peroxidation of lipoproteins in multiple sclerosis. J Neurol Sci 2011; 311:92-7. [PMID: 21967834 DOI: 10.1016/j.jns.2011.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 01/29/2023]
Abstract
Human plasma low density lipoproteins (LDL) and high density lipoproteins (HDL) are involved in the transport of lipids, modulate membrane lipid composition and regulate signal transduction. HDL-like lipoproteins have been shown also in human cerebrospinal fluid and it has been hypothesized that they could have a role in lipid transport in central nervous system. After synthesis, lipoproteins are susceptible to lipid peroxidation triggered by reactive oxygen species (ROS and RNS) produced by peripheral and brain cells. Aim of the paper has been to review the scientific literature on the role of lipid peroxidation of LDL and HDL in the molecular mechanisms of multiple sclerosis (MS). Several studies have demonstrated a significant increase in lipid peroxidation products in brain, plasma and cerebrospinal fluid of MS patients. The increase of antibodies against ox-LDL in plasma and the presence of ox-LDL in demyelinating plaques in MS brain suggests that the disease is associated with oxidative damage of lipoproteins. The impairment of antioxidant systems or an increase in the production of ROS and RNS could contribute to lipoprotein peroxidation in MS. Oxidized lipoproteins show several alterations of their functions, they are neurotoxic and have pro-inflammatory properties. Therefore lipoprotein lipid peroxidation products could be involved in demyelination and axonal injury in MS.
Collapse
Affiliation(s)
- Gianna Ferretti
- Dipartimento di Scienze Cliniche Sperimentali e Odontostomatologiche, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | | |
Collapse
|
42
|
Monitoring of transglutaminase 2 under different oxidative stress conditions. Amino Acids 2011; 42:1037-43. [PMID: 21805137 DOI: 10.1007/s00726-011-1018-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/05/2011] [Indexed: 10/17/2022]
Abstract
Transglutaminase 2 (TG2) is a multifunctional calcium-dependent enzyme which catalyzes the post-translational protein crosslinking with formation of intra- or inter-molecular epsilon(gamma-glutamyl)lysine bonds or polyamine incorporation. The up-regulation and activation of TG2 have been reported in a variety of physiological events, including cell differentiation, signal transduction, apoptosis, and wound healing, as well as in cell response to stress evoked by different internal and external stimuli. Here we review TG2 role in cell response to redox state imbalance both under physiological and pathological conditions, such as neurodegenerative disorders, inflammation, autoimmune diseases and cataractogenesis, in which oxidative stress plays a pathogenetic role and also accelerates disease progression. The increase in TG activity together with mitochondrial impairment and collapse of antioxidant enzymatic cell defences have been reported to be the prominent biochemical alterations becoming evident prior to neurodegeneration. Moreover, oxidative stress-induced TG2 pathway is involved in autophagy inhibition and aggresome formation, and TG2 has been suggested to function as a link between oxidative stress and inflammation by driving the decision as to whether a protein should undergo SUMO-mediated regulation or proteasomal degradation. Literature data suggest a strong association between oxidative stress and TG2 up-regulation, which in turn may result in cell survival or apoptosis, depending on cell type, kind of stressor, duration of insult, as well as TG2 intracellular localization and activity state. In particular, it may be suggested that TG2 plays a pro-survival role when the alteration of cell redox state homeostasis is not associated with intracellular calcium increase triggering TG2 transamidation activity.
Collapse
|
43
|
Witherick J, Wilkins A, Scolding N, Kemp K. Mechanisms of oxidative damage in multiple sclerosis and a cell therapy approach to treatment. Autoimmune Dis 2010; 2011:164608. [PMID: 21197107 PMCID: PMC3010615 DOI: 10.4061/2011/164608] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/10/2010] [Indexed: 12/14/2022] Open
Abstract
Although significant advances have recently been made in the understanding and treatment of multiple sclerosis, reduction of long-term disability remains a key goal. Evidence suggests that inflammation and oxidative stress within the central nervous system are major causes of ongoing tissue damage in the disease. Invading inflammatory cells, as well as resident central nervous system cells, release a number of reactive oxygen and nitrogen species which cause demyelination and axonal destruction, the pathological hallmarks of multiple sclerosis. Reduction in oxidative damage is an important therapeutic strategy to slow or halt disease processes. Many drugs in clinical practice or currently in trial target this mechanism. Cell-based therapies offer an alternative source of antioxidant capability. Classically thought of as being important for myelin or cell replacement in multiple sclerosis, stem cells may, however, have a more important role as providers of supporting factors or direct attenuators of the disease. In this paper we focus on the antioxidant properties of mesenchymal stem cells and discuss their potential importance as a cell-based therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Jonathan Witherick
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Alastair Wilkins
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Neil Scolding
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| |
Collapse
|
44
|
Yang ZJ, Xie Y, Bosco GM, Chen C, Camporesi EM. Hyperbaric oxygenation alleviates MCAO-induced brain injury and reduces hydroxyl radical formation and glutamate release. Eur J Appl Physiol 2009; 108:513-22. [PMID: 19851780 DOI: 10.1007/s00421-009-1229-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2009] [Indexed: 11/30/2022]
Abstract
The present study examined the effect of hyperbaric oxygen (HBO) on the formation of 2,3-dihydroxybenzoic acid (2,3-DHBA) and 2,5-dihydroxybenzoic acid (2,5-DHBA), the products of salicylate trapping of hydroxyl free radicals, and glutamate release in the striatum during acute ischemia and reperfusion. Non-HBO rats (n = 8) were subjected to 1-h ischemia. Study rats (n = 8) were treated with HBO at 2.8 ATA for 1 h during ischemia. Artificial CSF solution containing 5 mM sodium salicylate was perfused at 1 microl/min. Samples were continuously collected at 15 min intervals and the levels of 2,3-DHBA, 2,5-DHBA, and glutamate were analyzed. The lesion volume was determined by TTC stain. Occlusion of the middle cerebral artery induced a significant increase in the levels of 2,3-DHBA and 2,5-DHBA. A peak of approximately two and fourfold of baseline levels was reached at 45 min and was maintained at elevated levels during reperfusion. The level of glutamate increased approximately two times at 30 min during ischemia, continued to increase, and reached approximately three times baseline level during reperfusion. HBO significantly alleviated brain injury associated with decreased levels of 2,3-DHBA, 2,5-DHBA and glutamate. This study suggests that the decreased glutamate release and the reduced formation of hydroxyl free radicals might contribute to the neuroprotective effect of HBO.
Collapse
Affiliation(s)
- Zhong-jin Yang
- Department of Anesthesiology, Upstate Medical University, Syracuse, NY 13210, USA.
| | | | | | | | | |
Collapse
|
45
|
Gürbay A, Gonthier B, Barret L, Favier A, Hincal F. Cytotoxic effect of ciprofloxacin in primary culture of rat astrocytes and protection by Vitamin E. Toxicology 2007; 229:54-61. [PMID: 17098346 DOI: 10.1016/j.tox.2006.09.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 09/27/2006] [Indexed: 01/07/2023]
Abstract
The aim of this study was to investigate the possible cytotoxic and oxidative stress inducing effects of ciprofloxacin (CPFX) on primary cultures of rat astrocytes. The cultured cells were incubated with various concentrations of CPFX (0.5-300mg/l), and cytotoxicity was determined by neutral red (NR) and MTT assays. Survival profile of cells was biphasic in NR assay: CPFX did not cause any alteration at any concentration for 7h, whereas < or =50mg/l concentrations induced significant cell proliferation in incubation periods of 24, 48, 72, and 96h. However, cell proliferation gradually decreased at higher concentrations, and 200 and 300mg/l of CPFX exposure was found to be significantly (p<0.05) cytotoxic at all time periods. With MTT assay, no alteration was noted for incubation period of 7h, as observed with NR assay. But, cell viability decreased with approximately > or =50mg/l CPFX exposure in all other time periods. Cell proliferation was only seen in 24h of incubation with 0.5 and 5mg/l CPFX. Vitamin E pretreatment of cell cultures were found to be providing complete protection against cytotoxicity of 300mg/l CPFX in 96h incubation when measured with both NR and MTT assays. The SOD pretreatment was partially protective with NR assay, but no protection was noted when measured with MTT. A significant enhancement of lipid peroxidation was observed with the cytotoxic concentration of the drug, but total glutathione content and catalase activity of cells did not change. The data obtained in this study suggest that, in accordance with our previous results with fibroblast cells, CPFX-induced cytotoxicity is related to oxidative stress. And the biphasic effect of CPFX possibly resulted from the complex dose-dependent relationships between reactive oxygen species, cell proliferation, and cell viability.
Collapse
Affiliation(s)
- Aylin Gürbay
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 06100 Ankara, Turkey.
| | | | | | | | | |
Collapse
|
46
|
Komatsu T, Lee MCI, Miyagi A, Shoji H, Yoshino F, Maehata Y, Maetani T, Kawamura Y, Ikeda M, Kubota E. Reactive oxygen species generation in gingival fibroblasts of Down syndrome patients detected by electron spin resonance spectroscopy. Redox Rep 2006; 11:71-7. [PMID: 16686997 DOI: 10.1179/135100006x101039] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Oral manifestations of Down syndrome include high susceptibility to gingival inflammation with early onset and rapidly progressive periodontitis. The influence of reactive oxygen species (ROS) on periodontitis of Down syndrome is unclear. The aim of this study was to characterize ROS formation in Down syndrome-gingival fibroblasts (DS-GF) using electron spin resonance (ESR) spin trapping with 5,5-dimetyl-1-pyrolline-N-oxide (DMPO), and to determine whether ROS generation plays a role in the pathogenesis of periodontitis in Down syndrome patients. We observed formation of the DMPO-OH spin adduct, indicating HO* generation from cultured DS-GF and non-DS-GF. The increased HO* generation in cultured DS-GF was strongly decreased in the presence of the H2O2 scavenger, catalase, or the iron chelator, desferal. This may due to the enzymatic ability of over-expressed CuZn-superoxide dismutase in Down syndrome to catalyze the formation of H2O2 from O2*-, thereby increasing the availability of substrate H2O2 for the iron-dependent generation of HO* via the Fenton reaction, suggesting that HO* generated from DS-GF may be involved in progressive periodontitis of Down syndrome.
Collapse
Affiliation(s)
- Tomoko Komatsu
- Department of Clinical Care Medicine, Division of Dentistry for Special Patients, Kanagawa Dental College, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
McMichael MA, Ruaux CG, Baltzer WI, Kerwin SC, Hosgood GL, Steiner JM, Williams DA. Concentrations of 15F2t isoprostane in urine of dogs with intervertebral disk disease. Am J Vet Res 2006; 67:1226-31. [PMID: 16817747 DOI: 10.2460/ajvr.67.7.1226] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To measure 15F(2t) isoprostane concentrations in the urine of dogs undergoing ovariohysterectomy (OHE) and dogs undergoing surgery because of intervertebral disk disease (IVDD) and to assess relationships between urinary concentrations of 15F(2t) isoprostanes and neurologic score in dogs with IVDD. ANIMALS 11 dogs undergoing OHE and 32 dogs with IVDD undergoing hemilaminectomy. PROCEDURES Paired urine samples were obtained at induction of anesthesia and approximately 1 hour after OHE (controls) and were collected from dogs with IVDD at induction of anesthesia (28 samples) and approximately 1 hour after hemilaminectomy (31 samples); 26 paired urine samples were obtained from dogs with IVDD. Urinary isoprostane concentrations were measured by use of a commercial ELISA, and results were adjusted on the basis of urinary creatinine concentrations. Differences in the mean isoprostane-to-creatinine ratio were analyzed. Neurologic score was determined in dogs with IVDD by use of the modified Frankel scoring system. RESULTS Urinary isoprostane-to-creatinine ratios were significantly higher in dogs with IVDD than in control dogs before and after surgery. There was no significant difference between values before and after surgery for either group. There was a significant correlation of neurologic score and urinary isoprostane-to-creatinine ratio because dogs that had higher neurologic scores (ie, less severely affected) generally had higher isoprostane-to-creatinine ratios. CONCLUSIONS AND CLINICAL RELEVANCE Urinary isoprostane-to-creatinine ratios were higher in dogs with IVDD before and after surgery. Analysis of these data suggests that dogs with IVDD are in a state of oxidative stress and that preemptive treatment with antioxidants warrants further investigation.
Collapse
Affiliation(s)
- Maureen A McMichael
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, Texas A&M University, College Station, 77843-4474, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Miyake S, Sasaguri K, Hori N, Shoji H, Yoshino F, Miyazaki H, Anzai K, Ikota N, Ozawa T, Toyoda M, Sato S, Lee MCI. Biting reduces acute stress-induced oxidative stress in the rat hypothalamus. Redox Rep 2006; 10:19-24. [PMID: 15829107 DOI: 10.1179/135100005x21417] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We investigated the inhibitory effect of para-masticatory activity, namely biting, on restraint stress-induced oxidative stress. A blood brain barrier-permeable nitroxyl spin probe, 3-methoxycarbonyl-2,2,5,5,-tetramethylpyrrolidine-1-oxyl (MC-PROXYL), was administered to rats and L-band electron spin resonance (ESR) and ESR-computerized tomography (ESR-CT) imaging were used to show that the decay rate constant of MC-PROXYL in the hypothalamus of isolated brain after 30 min of restraint stress was more rapid than in unrestrained control rats, suggesting that restraint was associated with oxidative stress. Interestingly, biting during restraint stress caused the decay rate constant of MC-PROXYL in isolated brain to approach that of the control group. These observations suggest that biting suppresses oxidative stress induced by restraint stress, and that the anti-stress effect of masticatory motor activity movements, such as biting, are important for reducing the adverse effects associated with exposure to psychological stressors.
Collapse
Affiliation(s)
- Shinjiro Miyake
- Department of Craniofacial Growth and Development Dentistry, Kanagawa Dental College, Yokosuka, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
van Meeteren ME, Teunissen CE, Dijkstra CD, van Tol EAF. Antioxidants and polyunsaturated fatty acids in multiple sclerosis. Eur J Clin Nutr 2006; 59:1347-61. [PMID: 16118655 DOI: 10.1038/sj.ejcn.1602255] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Oligodendrocyte damage and subsequent axonal demyelination is a hallmark of this disease. Different pathomechanisms, for example, immune-mediated inflammation, oxidative stress and excitotoxicity, are involved in the immunopathology of MS. The risk of developing MS is associated with increased dietary intake of saturated fatty acids. Polyunsaturated fatty acid (PUFA) and antioxidant deficiencies along with decreased cellular antioxidant defence mechanisms have been observed in MS patients. Furthermore, antioxidant and PUFA treatment in experimental allergic encephalomyelitis, an animal model of MS, decreased the clinical signs of disease. Low-molecular-weight antioxidants may support cellular antioxidant defences in various ways, including radical scavenging, interfering with gene transcription, protein expression, enzyme activity and by metal chelation. PUFAs may not only exert immunosuppressive actions through their incorporation in immune cells but also may affect cell function within the CNS. Both dietary antioxidants and PUFAs have the potential to diminish disease symptoms by targeting specific pathomechanisms and supporting recovery in MS.
Collapse
Affiliation(s)
- M E van Meeteren
- Department of Biomedical Research, Numico Research BV, Wageningen, The Netherlands
| | | | | | | |
Collapse
|
50
|
Liu D, Bao F, Prough DS, Dewitt DS. Peroxynitrite Generated at the Level Produced by Spinal Cord Injury Induces Peroxidation of Membrane Phospholipids in Normal Rat Cord: Reduction by a Metalloporphyrin. J Neurotrauma 2005; 22:1123-33. [PMID: 16238488 DOI: 10.1089/neu.2005.22.1123] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The goal of the present study was to determine in vivo whether peroxynitrite, at the concentration and duration produced by SCI, contributes to membrane lipid peroxidation (MLP) after traumatic spinal cord injury (SCI) and the capability of a broad spectrum scavenger of reactive species, Mn (III) tetrakis (4-benzoic acid) porphyrin (MnTBAP), to reduce MLP. This was accomplished by administering a peroxynitrite donor 3-morpholinosydnonimine (SIN-1) into the gray matter of an uninjured rat spinal cord through a microdialysis fiber to generate ONOO at the SCI-elevated levels. The resulting MLP was characterized by measuring the productions of extracellular malondialdehyde and of intracellular 4-hydroxynonenal. We demonstrated that extracellular SIN- 1 administration significantly increased the concentration of malondialdehyde (p < 0.001) and the numbers of hydroxynonenal-positive cells (p < 0.001) as compared to a control group in which ACSF was administered. Simultaneous administration of MnTBAP through a second microdialysis fiber significantly reduced SIN-1-induced malondialdehyde production (p < 0.001) and the numbers of HNE-positive cells (p < 0.001). There was no significant difference between MnTBAP-treated and ACSF-controls (p = 0.3). These results demonstrate in vivo that (1) SCI-produced levels of peroxynitrite sufficient to cause MLP, and therefore that peroxynitrite is an agent of secondary damage after acute SCI; (2) MnTBAP can efficiently reduce SIN-1-induced MLP.
Collapse
Affiliation(s)
- Danxia Liu
- Departments of Neurology, Human Biological Chemistry & Genetics, University of Texas Medical Branch, Galveston, TX 77555-0881, USA.
| | | | | | | |
Collapse
|