1
|
Kacem I, Sghaier I, Ben Rhouma H, Ratti A, Ticozzi N, Silani V, Gouider-Khouja N, Gouider R. Association of Amyotrophic Lateral Sclerosis and Dopa-responsive dystonia in a Tunisian patient. Parkinsonism Relat Disord 2024; 130:107171. [PMID: 39531950 DOI: 10.1016/j.parkreldis.2024.107171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/24/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Dopa-responsive dystonia (DRD) is an autosomal dominant disease with parkinsonian and dystonic symptoms caused by GCH1 gene pathogenic variants affecting dopamine synthesis. The present case report is the first to link DRD with childhood-onset with ALS, suggesting that complex inheritance patterns in the North African population may contribute to multiple disorders.
Collapse
Affiliation(s)
- Imen Kacem
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, 1007, Tunis, Tunisia; Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia
| | - Ikram Sghaier
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia; Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia
| | - Hanene Ben Rhouma
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, 1007, Tunis, Tunisia; LR18SP04, Department of Child and Adolescent Neurology, University of Tunis El Manar, National Institute Mongi Ben Hamida of Neurology, Tunis, Tunisia
| | - Antonia Ratti
- Istituto Auxologico Italiano, IRCCS, Department of Neurology and Laboratory of Neuroscience, Piazzale Brescia 20, 20149, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090, Segrate, Milan, Italy
| | - Nicola Ticozzi
- Istituto Auxologico Italiano, IRCCS, Department of Neurology and Laboratory of Neuroscience, Piazzale Brescia 20, 20149, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Via F. Sforza 35, 20122, Milan, Italy
| | - Vincenzo Silani
- Istituto Auxologico Italiano, IRCCS, Department of Neurology and Laboratory of Neuroscience, Piazzale Brescia 20, 20149, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Via F. Sforza 35, 20122, Milan, Italy
| | - Neziha Gouider-Khouja
- Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, 1007, Tunis, Tunisia; Soukra Medical Center, Tunis, Tunisia
| | - Riadh Gouider
- Clinical Investigation Center (CIC) "Neurosciences and Mental Health", Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia; Faculty of Medicine of Tunis, University of Tunis El Manar, 15, Rue Djebel Lakhdhar, La Rabta, 1007, Tunis, Tunisia; Neurology Department, LR18SP03, Razi University Hospital, 1 rue des orangers Manouba, 2010, Tunis, Tunisia
| |
Collapse
|
2
|
Masahiro Hirai, Suzuki K, Kassai Y, Konishi Y. 3-Nitrotyrosine shortens axons of non-dopaminergic neurons by inhibiting mitochondrial motility. Neurochem Int 2024; 179:105832. [PMID: 39154836 DOI: 10.1016/j.neuint.2024.105832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
3-Nitrotyrosine (3-NT), a byproduct of oxidative and nitrosative stress, is implicated in age-related neurodegenerative disorders. Current literature suggests that free 3-NT becomes integrated into the carboxy-terminal domain of α-tubulin via the tyrosination/detyrosination cycle. Independently of this integration, 3-NT has been associated with the cell death of dopaminergic neurons. Given the critical role of tyrosination/detyrosination in governing axonal morphology and function, the substitution of tyrosine with 3-NT in this process may potentially disrupt axonal homeostasis, although this aspect remains underexplored. In this study, we examined the impact of 3-NT on the axons of cerebellar granule neurons, which is used as a model for non-dopaminergic neurons. Our observations revealed axonal shortening, which correlated with the incorporation of 3-NT into α-tubulin. Importantly, this axonal effect was observed prior to the onset of cellular death. Furthermore, 3-NT was found to diminish mitochondrial motility within the axon, leading to a subsequent reduction in mitochondrial membrane potential. The suppression of syntaphilin, a protein responsible for anchoring mitochondria to microtubules, restored the mitochondrial motility and axonal elongation that were inhibited by 3-NT. These findings underscore the inhibitory role of 3-NT in axonal elongation by impeding mitochondrial movement, suggesting its potential involvement in axonal dysfunction within non-dopaminergic neurons.
Collapse
Affiliation(s)
- Masahiro Hirai
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Kohei Suzuki
- Department of Industrial Innovation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Yusuke Kassai
- Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui, Japan
| | - Yoshiyuki Konishi
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, University of Fukui, Fukui, Japan; Department of Industrial Innovation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan; Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui, Japan; Department of Applied Chemistry and Biotechnology, Faculty of Engineering, University of Fukui, Fukui, Japan; Life Science Innovation Center, University of Fukui, Fukui, Japan.
| |
Collapse
|
3
|
Zheng C, Liu R, Chen J, Li S, Ling Y, Zhang Z. Development of a selective electrochemical microsensor based on molecularly imprinted polydopamine/ZIF-67/laser-induced graphene for point-of-care determination of 3-nitrotyrosine. Biosens Bioelectron 2024; 255:116246. [PMID: 38537430 DOI: 10.1016/j.bios.2024.116246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/15/2024]
Abstract
3-nitrotyrosine (3-NT) is a biomarker closely associated with the early diagnosis of oxidative stress-related disorders. The development of an accurate, cost-effective, point-of-care 3-NT sensor holds significant importance for self-monitoring and clinical treatment. In this study, a selective, sensitive, and portable molecularly imprinted electrochemical sensor was developed. ZIF-67 with strong adsorption capacity was facilely modified on an electrochemically active laser-induced graphene (LIG) substrate (formed ZIF-67/LIG). Subsequently, biocompatible dopamine was chosen as the functional monomer, and interference-free ʟ-tyrosine was used as the dummy template to create molecularly imprinted polydopamine (MIPDA) on the ZIF-67/LIG, endowing the sensor with selectivity. The morphologies, electrochemical properties, and detection performance of the sensor were comprehensively investigated using scanning electron microscopy, cyclic voltammetry, electrochemical impedance spectroscopy, and differential pulse voltammetry. To achieve the best performance, several parameters were optimized, including the number of polymerization cycles (15), elution time (60 min), incubation time (7 min), and pH of the buffer solution (6). The turnaround time for this sensor is 10 min. Benefiting from the alliance of MIPDA, ZIF-67, and LIG, the sensor exhibited excellent sensitivity with a detection limit of 6.71 nM, and distinguished selectivity against 11 interfering substances. To enable convenient clinical diagnosis, a customized electrochemical microsensor with MIPDA/ZIF-67/LIG was designed, showcasing excellent reliability and convenience in detecting biological samples without pretreatment. The proposed microsensor will not only facilitate clinical diagnosis and improve patient care, but also provide inspiration for the development of other portable and accurate electrochemical biosensors.
Collapse
Affiliation(s)
- Chibin Zheng
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Ruwei Liu
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Jianyue Chen
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China; Institute of New Functional Materials Co., Ltd, Guangxi Institute of Industrial Technology, Nanning, 530200, PR China
| | - Shilin Li
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yunhan Ling
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China.
| | - Zhengjun Zhang
- Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
4
|
Chidambaram SB, Anand N, Varma SR, Ramamurthy S, Vichitra C, Sharma A, Mahalakshmi AM, Essa MM. Superoxide dismutase and neurological disorders. IBRO Neurosci Rep 2024; 16:373-394. [PMID: 39007083 PMCID: PMC11240301 DOI: 10.1016/j.ibneur.2023.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/21/2023] [Indexed: 07/16/2024] Open
Abstract
Superoxide dismutase (SOD) is a common antioxidant enzyme found majorly in living cells. The main physiological role of SOD is detoxification and maintain the redox balance, acts as a first line of defence against Reactive nitrogen species (RNS), Reactive oxygen species (ROS), and other such potentially hazardous molecules. SOD catalyses the conversion of superoxide anion free radicals (O 2 -.) into molecular oxygen (O 2) and hydrogen peroxide (H 2O 2) in the cells. Superoxide dismutases (SODs) are expressed in neurons and glial cells throughout the CNS both intracellularly and extracellularly. Endogenous oxidative stress (OS) linked with enlarged production of reactive oxygen metabolites (ROMs), inflammation, deregulation of redox balance, mitochondrial dysfunction and bioenergetic crisis are found to be prerequisite for neuronal loss in neurological diseases. Clinical and genetic studies indicate a direct correlation between mutations in SOD gene and neurodegenerative diseases, like Amyotrophic Lateral Sclerosis (ALS), Huntington's disease (HD), Parkinson's Disease (PD) and Alzheimer's Disease (AD). Therefore, inhibitors of OS are considered as an optimistic approach to prevent neuronal loss. SOD mimetics like Metalloporphyrin Mn (II)-cyclic polyamines, Nitroxides and Mn (III)- Salen complexes are designed and used as therapeutic extensively in the treatment of neurological disorders. SODs and SOD mimetics are promising future therapeutics in the field of various diseases with OS-mediated pathology.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department of Pharmacology, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Antigua, Antigua and Barbuda
| | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Ajman University, 346 Ajman, the United Arab Emirates
- Center of Medical and Bio-allied Health Sciences Research, Ajman University, 346 Ajman, the United Arab Emirates
| | - Srinivasan Ramamurthy
- College of Pharmacy & Health Sciences, University of Science and Technology of Fujairah, 2202 Fujairah, the United Arab Emirates
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ambika Sharma
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
- Ageing and Dementia Research Group, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
5
|
Choi DH, Kang SK, Lee KE, Jung J, Kim EJ, Kim WH, Kwon YG, Kim KP, Jo I, Park YS, Park SI. Nitrosylation of β2-Tubulin Promotes Microtubule Disassembly and Differentiated Cardiomyocyte Beating in Ischemic Mice. Tissue Eng Regen Med 2023; 20:921-937. [PMID: 37679590 PMCID: PMC10519925 DOI: 10.1007/s13770-023-00582-5] [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: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Beating cardiomyocyte regeneration therapies have revealed as alternative therapeutics for heart transplantation. Nonetheless, the importance of nitric oxide (NO) in cardiomyocyte regeneration has been widely suggested, little has been reported concerning endogenous NO during cardiomyocyte differentiation. METHODS Here, we used P19CL6 cells and a Myocardiac infarction (MI) model to confirm NO-induced protein modification and its role in cardiac beating. Two tyrosine (Tyr) residues of β2-tubulin (Y106 and Y340) underwent nitrosylation (Tyr-NO) by endogenously generated NO during cardiomyocyte differentiation from pre-cardiomyocyte-like P19CL6 cells. RESULTS Tyr-NO-β2-tubulin mediated the interaction with Stathmin, which promotes microtubule disassembly, and was prominently observed in spontaneously beating cell clusters and mouse embryonic heart (E11.5d). In myocardial infarction mice, Tyr-NO-β2-tubulin in transplanted cells was closely related with cardiac troponin-T expression with their functional recovery, reduced infarct size and thickened left ventricular wall. CONCLUSION This is the first discovery of a new target molecule of NO, β2-tubulin, that can promote normal cardiac beating and cardiomyocyte regeneration. Taken together, we suggest therapeutic potential of Tyr-NO-β2-tubulin, for ischemic cardiomyocyte, which can reduce unexpected side effect of stem cell transplantation, arrhythmogenesis.
Collapse
Affiliation(s)
- Da Hyeon Choi
- Department of Biological Sciences and Biotechnology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Seong Ki Kang
- Division of Intractable Diseases, Center for Biomedical Sciences, Korea National Institute of Health (KNIH), Cheongju, Republic of Korea
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin, Republic of Korea
| | - Kyeong Eun Lee
- Department of Biological Sciences and Biotechnology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Jongsun Jung
- AI Drug Platform Center, Syntekabio, Daejeon, Republic of Korea
| | - Eun Ju Kim
- Department of Applied Chemistry, Kyung Hee University, Yongin, Republic of Korea
| | - Won-Ho Kim
- Division of Cardiovascular and Rare Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Kyung Hee University, Yongin, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, College of Ewha Womans University, Seoul, Republic of Korea
- Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Shin Park
- Department of Biological Sciences and Biotechnology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea.
| | - Sang Ick Park
- Division of Intractable Diseases, Center for Biomedical Sciences, Korea National Institute of Health (KNIH), Cheongju, Republic of Korea.
| |
Collapse
|
6
|
Steinert JR, Amal H. The contribution of an imbalanced redox signalling to neurological and neurodegenerative conditions. Free Radic Biol Med 2023; 194:71-83. [PMID: 36435368 DOI: 10.1016/j.freeradbiomed.2022.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Nitric oxide and other redox active molecules such as oxygen free radicals provide essential signalling in diverse neuronal functions, but their excess production and insufficient scavenging induces cytotoxic redox stress which is associated with numerous neurodegenerative and neurological conditions. A further component of redox signalling is mediated by a homeostatic regulation of divalent metal ions, the imbalance of which contributes to neuronal dysfunction. Additional antioxidant molecules such as glutathione and enzymes such as super oxide dismutase are involved in maintaining a physiological redox status within neurons. When cellular processes are perturbed and generation of free radicals overwhelms the antioxidants capacity of the neurons, a resulting redox damage leads to neuronal dysfunction and cell death. Cellular sources for production of redox-active molecules may include NADPH oxidases, mitochondria, cytochrome P450 and nitric oxide (NO)-generating enzymes, such as endothelial, neuronal and inducible NO synthases. Several neurodegenerative and developmental neurological conditions are associated with an imbalanced redox state as a result of neuroinflammatory processes leading to nitrosative and oxidative stress. Ongoing research aims at understanding the causes and consequences of such imbalanced redox homeostasis and its role in neuronal dysfunction.
Collapse
Affiliation(s)
- Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
7
|
Kurogi K, Rasool MI, Alherz FA, El Daibani AA, Bairam AF, Abunnaja MS, Yasuda S, Wilson LJ, Hui Y, Liu MC. SULT genetic polymorphisms: physiological, pharmacological and clinical implications. Expert Opin Drug Metab Toxicol 2021; 17:767-784. [PMID: 34107842 DOI: 10.1080/17425255.2021.1940952] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Cytosolic sulfotransferases (SULTs)-mediated sulfation is critically involved in the metabolism of key endogenous compounds, such as catecholamines and thyroid/steroid hormones, as well as a variety of drugs and other xenobiotics. Studies performed in the past three decades have yielded a good understanding about the enzymology of the SULTs and their structural biology, phylogenetic relationships, tissue/organ-specific/developmental expression, as well as the regulation of the SULT gene expression. An emerging area is related to the functional impact of the SULT genetic polymorphisms. AREAS COVERED The current review aims to summarize our current knowledge about the above-mentioned aspects of the SULT research. An emphasis is on the information concerning the effects of the polymorphisms of the SULT genes on the functional activity of the SULT allozymes and the associated physiological, pharmacological, and clinical implications. EXPERT OPINION Elucidation of how SULT SNPs may influence the drug-sulfating activity of SULT allozymes will help understand the differential drug metabolism and eventually aid in formulating personalized drug regimens. Moreover, the information concerning the differential sulfating activities of SULT allozymes toward endogenous compounds may allow for the development of strategies for mitigating anomalies in the metabolism of these endogenous compounds in individuals with certain SULT genotypes.
Collapse
Affiliation(s)
- Katsuhisa Kurogi
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Mohammed I Rasool
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Pharmacology, College of Pharmacy, University of Karbala, Karbala, Iraq
| | - Fatemah A Alherz
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amal A El Daibani
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA
| | - Ahsan F Bairam
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Pharmacology, College of Pharmacy, University of Kufa, Najaf, Iraq
| | - Maryam S Abunnaja
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA
| | - Shin Yasuda
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Bioscience, School of Agriculture, Tokai University, Kumamoto City, Kumamoto 862-8652, Japan
| | - Lauren J Wilson
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA
| | - Ying Hui
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA.,Department of Obstetrics and Gynecology, Beijing Hospital, Beijing, China
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614 USA
| |
Collapse
|
8
|
Lu CC, Nyam TTE, Kuo JR, Lee YL, Chio CC, Wang CC. The neuroprotective effects of AMN082 on neuronal apoptosis in rats after traumatic brain injury. BMC Neurosci 2021; 22:44. [PMID: 34171999 PMCID: PMC8228939 DOI: 10.1186/s12868-021-00649-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate whether AMN082 exerts its neuroprotective effect by attenuating glutamate receptor-associated neuronal apoptosis and improving functional outcomes after traumatic brain injury (TBI). METHODS Anesthetized male Sprague-Dawley rats were divided into the sham-operated, TBI + vehicle, and TBI + AMN082 groups. AMN082 (10 mg/kg) was intraperitoneally injected 0, 24, or 48 h after TBI. In the 120 min after TBI, heart rate, mean arterial pressure, intracranial pressure (ICP), and cerebral perfusion pressure (CPP) were continuously measured. Motor function, the infarct volume, neuronal nitrosative stress-associated apoptosis, and N-methyl-D-aspartate receptor 2A (NR2A) and NR2B expression in the pericontusional cortex were measured on the 3rd day after TBI. RESULTS The results showed that the AMN082-treated group had a lower ICP and higher CPP after TBI. TBI-induced motor deficits, the increase in infarct volume, neuronal apoptosis, and 3-nitrotyrosine and inducible nitric oxide synthase expression in the pericontusional cortex were significantly improved by AMN082 therapy. Simultaneously, AMN082 increased NR2A and NR2B expression in neuronal cells. CONCLUSIONS We concluded that intraperitoneal injection of AMN082 for 3 days may ameliorate TBI by attenuating glutamate receptor-associated nitrosative stress and neuronal apoptosis in the pericontusional cortex. We suggest that AMN082 administration in the acute stage may be a promising strategy for TBI.
Collapse
Affiliation(s)
- Chung-Che Lu
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Tee-Tau Eric Nyam
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Jinn-Rung Kuo
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yao-Lin Lee
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
| | - Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, 901 Chung Hwa Road, Yung Kang City, Tainan, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
- Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| |
Collapse
|
9
|
Wang F, Yuan Q, Chen F, Pang J, Pan C, Xu F, Chen Y. Fundamental Mechanisms of the Cell Death Caused by Nitrosative Stress. Front Cell Dev Biol 2021; 9:742483. [PMID: 34616744 PMCID: PMC8488117 DOI: 10.3389/fcell.2021.742483] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Nitrosative stress, as an important oxygen metabolism disorder, has been shown to be closely associated with cardiovascular diseases, such as myocardial ischemia/reperfusion injury, aortic aneurysm, heart failure, hypertension, and atherosclerosis. Nitrosative stress refers to the joint biochemical reactions of nitric oxide (NO) and superoxide (O2 -) when an oxygen metabolism disorder occurs in the body. The peroxynitrite anion (ONOO-) produced during this process can nitrate several biomolecules, such as proteins, lipids, and DNA, to generate 3-nitrotyrosine (3-NT), which further induces cell death. Among these, protein tyrosine nitration and polyunsaturated fatty acid nitration are the most studied types to date. Accordingly, an in-depth study of the relationship between nitrosative stress and cell death has important practical significance for revealing the pathogenesis and strategies for prevention and treatment of various diseases, particularly cardiovascular diseases. Here, we review the latest research progress on the mechanisms of nitrosative stress-mediated cell death, primarily involving several regulated cell death processes, including apoptosis, autophagy, ferroptosis, pyroptosis, NETosis, and parthanatos, highlighting nitrosative stress as a unique mechanism in cardiovascular diseases.
Collapse
Affiliation(s)
- Fulin Wang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Qiuhuan Yuan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Fengying Chen
- Emergency Department, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Fengying Chen,
| | - Jiaojiao Pang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Chang Pan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Feng Xu,
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, China
- Chest Pain Center, Qilu Hospital, Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital, Shandong University, Jinan, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
- Yuguo Chen,
| |
Collapse
|
10
|
Amal H, Barak B, Bhat V, Gong G, Joughin BA, Wang X, Wishnok JS, Feng G, Tannenbaum SR. Shank3 mutation in a mouse model of autism leads to changes in the S-nitroso-proteome and affects key proteins involved in vesicle release and synaptic function. Mol Psychiatry 2020; 25:1835-1848. [PMID: 29988084 PMCID: PMC6614015 DOI: 10.1038/s41380-018-0113-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/25/2022]
Abstract
Mutation in the SHANK3 human gene leads to different neuropsychiatric diseases including Autism Spectrum Disorder (ASD), intellectual disabilities and Phelan-McDermid syndrome. Shank3 disruption in mice leads to dysfunction of synaptic transmission, behavior, and development. Protein S-nitrosylation, the nitric oxide (NO•)-mediated posttranslational modification (PTM) of cysteine thiols (SNO), modulates the activity of proteins that regulate key signaling pathways. We tested the hypothesis that Shank3 mutation would generate downstream effects on PTM of critical proteins that lead to modification of synaptic functions. SNO-proteins in two ASD-related brain regions, cortex and striatum of young and adult InsG3680(+/+) mice (a human mutation-based Shank3 mouse model), were identified by an innovative mass spectrometric method, SNOTRAP. We found changes of the SNO-proteome in the mutant compared to WT in both ages. Pathway analysis showed enrichment of processes affected in ASD. SNO-Calcineurin in mutant led to a significant increase of phosphorylated Synapsin1 and CREB, which affect synaptic vesicle mobilization and gene transcription, respectively. A significant increase of 3-nitrotyrosine was found in the cortical regions of the adult mutant, signaling both oxidative and nitrosative stress. Neuronal NO• Synthase (nNOS) was examined for levels and localization in neurons and no significant difference was found in WT vs. mutant. S-nitrosoglutathione concentrations were higher in mutant mice compared to WT. This is the first study on NO•-related molecular changes and SNO-signaling in the brain of an ASD mouse model that allows the characterization and identification of key proteins, cellular pathways, and neurobiological mechanisms that might be affected in ASD.
Collapse
Affiliation(s)
- Haitham Amal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Boaz Barak
- McGovern Institute for Brain Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | | | - Guanyu Gong
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Brian A. Joughin
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA,Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xin Wang
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - John S. Wishnok
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Steven R. Tannenbaum
- Department of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA,Department of Chemistry, Massachusetts Institute of
Technology, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Low Doses of Arsenic in a Mouse Model of Human Exposure and in Neuronal Culture Lead to S-Nitrosylation of Synaptic Proteins and Apoptosis via Nitric Oxide. Int J Mol Sci 2020; 21:ijms21113948. [PMID: 32486366 PMCID: PMC7312481 DOI: 10.3390/ijms21113948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 11/17/2022] Open
Abstract
Background: Accumulating public health and epidemiological literature support the hypothesis that arsenic in drinking water or food affects the brain adversely. Methods: Experiments on the consequences of nitric oxide (NO) formation in neuronal cell culture and mouse brain were conducted to probe the mechanistic pathways of nitrosative damage following arsenic exposure. Results: After exposure of mouse embryonic neuronal cells to low doses of sodium arsenite (SA), we found that Ca2+ was released leading to the formation of large amounts of NO and apoptosis. Inhibition of NO synthase prevented neuronal apoptosis. Further, SA led to concerted S-nitrosylation of proteins significantly associated with synaptic vesicle recycling and acetyl-CoA homeostasis. Our findings show that low-dose chronic exposure (0.1–1 ppm) to SA in the drinking water of mice led to S-nitrosylation of proteomic cysteines. Subsequent removal of arsenic from the drinking water reversed the biochemical alterations. Conclusions: This work develops a mechanistic understanding of the role of NO in arsenic-mediated toxicity in the brain, incorporating Ca2+ release and S-nitrosylation as important modifiers of neuronal protein function.
Collapse
|
12
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
Wang CC, Wee HY, Hu CY, Chio CC, Kuo JR. The Effects of Memantine on Glutamic Receptor-Associated Nitrosative Stress in a Traumatic Brain Injury Rat Model. World Neurosurg 2018; 112:e719-e731. [PMID: 29382619 DOI: 10.1016/j.wneu.2018.01.140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND The main aim of this study is to elucidate whether the neuroprotective effect of memantine, a noncompetitive N-methyl-d-aspartate receptor 2B (NR2B) antagonist, affects neuronal nitrosative stress, apoptosis, and NR2B expression and improves functional outcomes. METHODS Immediately after the onset of fluid percussion traumatic brain injury (TBI), anesthetized male Sprague-Dawley rats were divided into sham-operated, TBI + vehicle, and TBI + memantine groups. TBI rats were treated with a memantine intraperitoneal injection dose of 20 mg/kg intraperitoneally and then 1 mg/kg every 12 hours intraperitoneally for 6 doses. The motor function, proprioception, infarction volume, and neuronal apoptosis were then measured. Immunofluorescence was used to evaluate astrogliosis, microgliosis, nitrosative stress, and NR2A and NR2B expression in cortical cells. All the parameters were assessed 72 hours after TBI. RESULTS Compared with the sham-operated controls, the TBI-induced motor and proprioception deficits, and increased infraction volume after TBI were significantly attenuated by memantine therapy. The TBI-induced neuronal apoptosis, astrogliosis, and microgliosis, the numbers of neuronal NO synthase and 3-nitro-l-tyrosine expression in neurons, and inducible NO synthase expression in microglia and astrocyte cells in the ischemic cortex after TBI were significantly improved by memantine therapy. Simultaneously, without affecting the NR2A expression in neuronal cells, the NR2B expression significantly decreased after memantine therapy, as evaluated by an immunofluorescence stain. CONCLUSIONS Intraperitoneal injection of memantine in the acute stage may ameliorate TBI in rats by affecting NR2B expression and decreasing neuronal apoptosis and nitrosative stress in the injured cortex. These effects might represent 1 mechanism by which functional recovery occurred.
Collapse
Affiliation(s)
- Che-Chuan Wang
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hsiao-Yue Wee
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chiao-Ya Hu
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chung-Ching Chio
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jinn-Rung Kuo
- Department of Neurosurgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
14
|
|
15
|
Veyrat-Durebex C, Corcia P, Piver E, Devos D, Dangoumau A, Gouel F, Vourc'h P, Emond P, Laumonnier F, Nadal-Desbarats L, Gordon PH, Andres CR, Blasco H. Disruption of TCA Cycle and Glutamate Metabolism Identified by Metabolomics in an In Vitro Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2015; 53:6910-6924. [PMID: 26666663 DOI: 10.1007/s12035-015-9567-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022]
Abstract
This study aims to develop a cellular metabolomics model that reproduces the pathophysiological conditions found in amyotrophic lateral sclerosis in order to improve knowledge of disease physiology. We used a co-culture model combining the motor neuron-like cell line NSC-34 and the astrocyte clone C8-D1A, with each over-expressing wild-type or G93C mutant human SOD1, to examine amyotrophic lateral sclerosis (ALS) physiology. We focused on the effects of mutant human SOD1 as well as oxidative stress induced by menadione on intracellular metabolism using a metabolomics approach through gas chromatography coupled with mass spectrometry (GC-MS) analysis. Preliminary non-supervised analysis by Principal Component Analysis (PCA) revealed that cell type, genetic environment, and time of culture influenced the metabolomics profiles. Supervised analysis using orthogonal partial least squares discriminant analysis (OPLS-DA) on data from intracellular metabolomics profiles of SOD1G93C co-cultures produced metabolites involved in glutamate metabolism and the tricarboxylic acid cycle (TCA) cycle. This study revealed the feasibility of using a metabolomics approach in a cellular model of ALS. We identified potential disruption of the TCA cycle and glutamate metabolism under oxidative stress, which is consistent with prior research in the disease. Analysis of metabolic alterations in an in vitro model is a novel approach to investigation of disease physiology.
Collapse
Affiliation(s)
- Charlotte Veyrat-Durebex
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France. .,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France.
| | - Philippe Corcia
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Service de Neurologie, 37044, Tours, France
| | | | - David Devos
- Département de Pharmacologie médicale, INSERM U1171, Université Lille Nord de France, CHRU de Lille, Lille, France
| | - Audrey Dangoumau
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France
| | - Flore Gouel
- Département de Pharmacologie médicale, INSERM U1171, Université Lille Nord de France, CHRU de Lille, Lille, France
| | - Patrick Vourc'h
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| | - Patrick Emond
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,PPF-ASB, Université François Rabelais de Tours, Tours, France
| | - Frédéric Laumonnier
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France
| | - Lydie Nadal-Desbarats
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,PPF-ASB, Université François Rabelais de Tours, Tours, France
| | | | - Christian R Andres
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| | - Hélène Blasco
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| |
Collapse
|
16
|
Yuste JE, Tarragon E, Campuzano CM, Ros-Bernal F. Implications of glial nitric oxide in neurodegenerative diseases. Front Cell Neurosci 2015; 9:322. [PMID: 26347610 PMCID: PMC4538301 DOI: 10.3389/fncel.2015.00322] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/03/2015] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) is a pleiotropic janus-faced molecule synthesized by nitric oxide synthases (NOS) which plays a critical role in a number of physiological and pathological processes in humans. The physiological roles of NO depend on its local concentrations, as well as its availability and the nature of downstream target molecules. Its double-edged sword action has been linked to neurodegenerative disorders. Excessive NO production, as the evoked by inflammatory signals, has been identified as one of the major causative reasons for the pathogenesis of several neurodegenerative diseases. Moreover, excessive NO synthesis under neuroinflammation leads to the formation of reactive nitrogen species and neuronal cell death. There is an intimate relation between microglial activation, NO and neuroinflammation in the human brain. The role of NO in neuroinflammation has been defined in animal models where this neurotransmitter can modulate the inflammatory process acting on key regulatory pathways, such as those associated with excitotoxicity processes induced by glutamate accumulation and microglial activation. Activated glia express inducible NOS and produce NO that triggers calcium mobilization from the endoplasmic reticulum, activating the release of vesicular glutamate from astroglial cells resulting in neuronal death. This change in microglia potentially contributes to the increased age-associated susceptibility and neurodegeneration. In the current review, information is provided about the role of NO, glial activation and age-related processes in the central nervous system (CNS) that may be helpful in the isolation of new therapeutic targets for aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jose Enrique Yuste
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| | - Ernesto Tarragon
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain ; Département des Sciences Biomédicales et Précliniques/Biochimie et Physiologie du Système Nerveux, Centre de Recherche du Cyclotron, Université de Liège Liège, Belgium
| | - Carmen María Campuzano
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| | - Francisco Ros-Bernal
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| |
Collapse
|
17
|
Chen HJC, Spiers JG, Sernia C, Lavidis NA. Response of the nitrergic system to activation of the neuroendocrine stress axis. Front Neurosci 2015; 9:3. [PMID: 25653586 PMCID: PMC4300918 DOI: 10.3389/fnins.2015.00003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/05/2015] [Indexed: 12/19/2022] Open
Abstract
Exposure to stressful stimuli causes activation of the hypothalamic-pituitary-adrenal axis which rapidly releases high concentrations of glucocorticoid stress hormones, resulting in increased cellular metabolism and spontaneous oxygen and nitrogen radical formation. High concentrations of nitrogen radicals, including nitric oxide, cause damage to cellular proteins in addition to inhibiting components of the mitochondrial transport chain, leading to cellular energy deficiency. During stress exposure, pharmacological inhibition of nitric oxide production reduces indicators of anxiety- and depressive-like behavior in animal models. Therefore, the purpose of this review is to present an overview of the current literature on stress-evoked changes in the nitrergic system, particularly within neural tissue.
Collapse
Affiliation(s)
| | - Jereme G Spiers
- School of Biomedical Sciences, The University of Queensland Brisbane, QLD, Australia
| | - Conrad Sernia
- School of Biomedical Sciences, The University of Queensland Brisbane, QLD, Australia
| | - Nickolas A Lavidis
- School of Biomedical Sciences, The University of Queensland Brisbane, QLD, Australia
| |
Collapse
|
18
|
Sheng H, Chaparro RE, Sasaki T, Izutsu M, Pearlstein RD, Tovmasyan A, Warner DS. Metalloporphyrins as therapeutic catalytic oxidoreductants in central nervous system disorders. Antioxid Redox Signal 2014; 20:2437-64. [PMID: 23706004 DOI: 10.1089/ars.2013.5413] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Metalloporphyrins, characterized by a redox-active transitional metal (Mn or Fe) coordinated to a cyclic porphyrin core ligand, mitigate oxidative/nitrosative stress in biological systems. Side-chain substitutions tune redox properties of metalloporphyrins to act as potent superoxide dismutase mimics, peroxynitrite decomposition catalysts, and redox regulators of transcription factor function. With oxidative/nitrosative stress central to pathogenesis of CNS injury, metalloporphyrins offer unique pharmacologic activity to improve the course of disease. RECENT ADVANCES Metalloporphyrins are efficacious in models of amyotrophic lateral sclerosis, Alzheimer's disease, epilepsy, neuropathic pain, opioid tolerance, Parkinson's disease, spinal cord injury, and stroke and have proved to be useful tools in defining roles of superoxide, nitric oxide, and peroxynitrite in disease progression. The most substantive recent advance has been the synthesis of lipophilic metalloporphyrins offering improved blood-brain barrier penetration to allow intravenous, subcutaneous, or oral treatment. CRITICAL ISSUES Insufficient preclinical data have accumulated to enable clinical development of metalloporphyrins for any single indication. An improved definition of mechanisms of action will facilitate preclinical modeling to define and validate optimal dosing strategies to enable appropriate clinical trial design. Due to previous failures of "antioxidants" in clinical trials, with most having markedly less biologic activity and bioavailability than current-generation metalloporphyrins, a stigma against antioxidants has discouraged the development of metalloporphyrins as CNS therapeutics, despite the consistent definition of efficacy in a wide array of CNS disorders. FUTURE DIRECTIONS Further definition of the metalloporphyrin mechanism of action, side-by-side comparison with "failed" antioxidants, and intense effort to optimize therapeutic dosing strategies are required to inform and encourage clinical trial design.
Collapse
Affiliation(s)
- Huaxin Sheng
- 1 Department of Anesthesiology, Duke University Medical Center (DUMC) , Durham, North Carolina
| | | | | | | | | | | | | |
Collapse
|
19
|
Iqbal J, Li W, Hasan M, Liu K, Awan U, Saeed Y, Zhang Y, Muhammad Khan A, Shah A, Qing H, Deng Y. Differential expression of specific cellular defense proteins in rat hypothalamus under simulated microgravity induced conditions: comparative proteomics. Proteomics 2014; 14:1424-33. [PMID: 24648329 DOI: 10.1002/pmic.201400019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 02/16/2014] [Accepted: 03/10/2014] [Indexed: 12/14/2022]
Abstract
Microgravity severely halts the structural and functional cerebral capacity of astronauts especially affecting their brains due to the stress produced by cephalic fluid shift. We employed a rat tail suspension model to substantiate simulated microgravity (SM) in brain. In this study, comparative mass spectrometry was applied in order to demonstrate the differential expression of 17 specific cellular defense proteins. Gamma-enolase, peptidyl-prolyl cis-trans isomerase A, glial fibrillary acidic protein, heat shock protein HSP 90-alpha, 10 kDa heat shock protein, mitochondrial, heat shock cognate 71 kDa protein, superoxide dismutase 1 and dihydropyrimidinase-related protein 2 were found to be upregulated by HPLC/ESI-TOF. Furthermore, five differentially expressed proteins including 60 kDa heat shock protein, mitochondrial, heat shock protein HSP 90-beta, peroxiredoxin-2, stress-induced-phosphoprotein, and UCHL-1 were found to be upregulated by HPLC/ESI-Q-TOF MS. In addition, downregulated proteins include cytochrome C, superoxide dismutase 2, somatic, and excitatory amino acid transporter 1 and protein DJ-1. Validity of MS results was successfully performed by Western blot analysis of DJ-1 protein. This study will not only help to understand the neurochemical responses produced under microgravity but also will give future direction to cure the proteomic losses and their after effects in astronauts.
Collapse
Affiliation(s)
- Javed Iqbal
- School of Life Sciences, Beijing Institute of Technology, Beijing, P. R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
D’Amico E, Factor-Litvak P, Santella RM, Mitsumoto H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic Biol Med 2013; 65:509-527. [PMID: 23797033 PMCID: PMC3859834 DOI: 10.1016/j.freeradbiomed.2013.06.029] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/14/2013] [Accepted: 06/14/2013] [Indexed: 12/12/2022]
Abstract
Sporadic amyotrophic lateral sclerosis (ALS) is one of the most devastating neurological diseases; most patients die within 3 to 4 years after symptom onset. Oxidative stress is a disturbance in the pro-oxidative/antioxidative balance favoring the pro-oxidative state. Autopsy and laboratory studies in ALS indicate that oxidative stress plays a major role in motor neuron degeneration and astrocyte dysfunction. Oxidative stress biomarkers in cerebrospinal fluid, plasma, and urine are elevated, suggesting that abnormal oxidative stress is generated outside of the central nervous system. Our review indicates that agricultural chemicals, heavy metals, military service, professional sports, excessive physical exertion, chronic head trauma, and certain foods might be modestly associated with ALS risk, with a stronger association between risk and smoking. At the cellular level, these factors are all involved in generating oxidative stress. Experimental studies indicate that a combination of insults that induce modest oxidative stress can exert additive deleterious effects on motor neurons, suggesting that multiple exposures in real-world environments are important. As the disease progresses, nutritional deficiency, cachexia, psychological stress, and impending respiratory failure may further increase oxidative stress. Moreover, accumulating evidence suggests that ALS is possibly a systemic disease. Laboratory, pathologic, and epidemiologic evidence clearly supports the hypothesis that oxidative stress is central in the pathogenic process, particularly in genetically susceptive individuals. If we are to improve ALS treatment, well-designed biochemical and genetic epidemiological studies, combined with a multidisciplinary research approach, are needed and will provide knowledge crucial to our understanding of ALS etiology, pathophysiology, and prognosis.
Collapse
Affiliation(s)
- Emanuele D’Amico
- Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032, ;
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032,
| | - Regina M. Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032,
| | - Hiroshi Mitsumoto
- Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032
| |
Collapse
|
21
|
Advances in cellular models to explore the pathophysiology of amyotrophic lateral sclerosis. Mol Neurobiol 2013; 49:966-83. [PMID: 24198229 DOI: 10.1007/s12035-013-8573-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/15/2013] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), the most common adult-onset motor neuron disorder, is fatal for most patients less than 3 years from when the first symptoms appear. The aetiologies for sporadic and most familial forms of ALS are unknown, but genetic factors are increasingly recognized as causal in a subset of patients. Studies of disease physiology suggest roles for oxidative stress, glutamate-mediated excitotoxicity or protein aggregation; how these pathways interact in the complex pathophysiology of ALS awaits elucidation. Cellular models are being used to examine disease mechanisms. Recent advances include the availability of expanded cell types, from neuronal or glial cell culture to motoneuron-astrocyte co-culture genetically or environmentally modified. Cell culture experiments confirmed the central role of glial cells in ALS. The recent adaptation of induced pluripotent stem cells (iPSC) for ALS modeling could allow a broader perspective and is expected to generate new hypotheses, related particularly to mechanisms underlying genetic factors. Cellular models have provided meaningful advances in the understanding of ALS, but, to date, complete characterization of in vitro models is only partially described. Consensus on methodological approaches, strategies for validation and techniques that allow rapid adaptation to new genetic or environmental influences is needed. In this article, we review the principal cellular models being employed in ALS and highlight their contribution to the understanding of disease mechanisms. We conclude with recommendations on means to enhance the robustness and generalizability of the different concepts for experimental ALS.
Collapse
|
22
|
Abstract
Oxidative stress is a widely recognized cause of cell death associated with neurodegeneration, inflammation, and aging. Tyrosine nitration in these conditions has been reported extensively, but whether tyrosine nitration is a marker or plays a role in the cell-death processes was unknown. Here, we show that nitration of a single tyrosine residue on a small proportion of 90-kDa heat-shock protein (Hsp90), is sufficient to induce motor neuron death by the P2X7 receptor-dependent activation of the Fas pathway. Nitrotyrosine at position 33 or 56 stimulates a toxic gain of function that turns Hsp90 into a toxic protein. Using an antibody that recognizes the nitrated Hsp90, we found immunoreactivity in motor neurons of patients with amyotrophic lateral sclerosis, in an animal model of amyotrophic lateral sclerosis, and after experimental spinal cord injury. Our findings reveal that cell death can be triggered by nitration of a single protein and highlight nitrated Hsp90 as a potential target for the development of effective therapies for a large number of pathologies.
Collapse
|
23
|
Blume YB, Krasylenko YA, Demchuk OM, Yemets AI. Tubulin tyrosine nitration regulates microtubule organization in plant cells. FRONTIERS IN PLANT SCIENCE 2013; 4:530. [PMID: 24421781 PMCID: PMC3872735 DOI: 10.3389/fpls.2013.00530] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 12/10/2013] [Indexed: 05/21/2023]
Abstract
During last years, selective tyrosine nitration of plant proteins gains importance as well-recognized pathway of direct nitric oxide (NO) signal transduction. Plant microtubules are one of the intracellular signaling targets for NO, however, the molecular mechanisms of NO signal transduction with the involvement of cytoskeletal proteins remain to be elucidated. Since biochemical evidence of plant α-tubulin tyrosine nitration has been obtained recently, potential role of this posttranslational modification in regulation of microtubules organization in plant cell is estimated in current paper. It was shown that 3-nitrotyrosine (3-NO2-Tyr) induced partially reversible Arabidopsis primary root growth inhibition, alterations of root hairs morphology and organization of microtubules in root cells. It was also revealed that 3-NO2-Tyr intensively decorates such highly dynamic microtubular arrays as preprophase bands, mitotic spindles and phragmoplasts of Nicotiana tabacum Bright Yellow-2 (BY-2) cells under physiological conditions. Moreover, 3D models of the mitotic kinesin-8 complexes with the tail of detyrosinated, tyrosinated and tyrosine nitrated α-tubulin (on C-terminal Tyr 450 residue) from Arabidopsis were reconstructed in silico to investigate the potential influence of tubulin nitrotyrosination on the molecular dynamics of α-tubulin and kinesin-8 interaction. Generally, presented data suggest that plant α-tubulin tyrosine nitration can be considered as its common posttranslational modification, the direct mechanism of NO signal transduction with the participation of microtubules under physiological conditions and one of the hallmarks of the increased microtubule dynamics.
Collapse
Affiliation(s)
- Yaroslav B. Blume
- *Correspondence: Yaroslav B. Blume, Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics, National Academy of Sciences of Ukraine, Osipovskogo str., 2, Kyiv 04123, Ukraine e-mail:
| | | | | | | |
Collapse
|
24
|
Slominski A, Zmijewski MA, Pawelek J. L-tyrosine and L-dihydroxyphenylalanine as hormone-like regulators of melanocyte functions. Pigment Cell Melanoma Res 2012; 25:14-27. [PMID: 21834848 PMCID: PMC3242935 DOI: 10.1111/j.1755-148x.2011.00898.x] [Citation(s) in RCA: 346] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is evidence that L-tyrosine and L-dihydroxyphenylalanine (L-DOPA), besides serving as substrates and intermediates of melanogenesis, are also bioregulatory agents acting not only as inducers and positive regulators of melanogenesis but also as regulators of other cellular functions. These can be mediated through action on specific receptors or through non-receptor-mediated mechanisms. The substrate induced (L-tyrosine and/or L-DOPA) melanogenic pathway would autoregulate itself as well as regulate the melanocyte functions through the activity of its structural or regulatory proteins and through intermediates of melanogenesis and melanin itself. Dissection of regulatory and autoregulatory elements of this process may elucidate how substrate-induced autoregulatory pathways have evolved from prokaryotic or simple eukaryotic organisms to complex systems in vertebrates. This could substantiate an older theory proposing that receptors for amino acid-derived hormones arose from the receptors for those amino acids, and that nuclear receptors evolved from primitive intracellular receptors binding nutritional factors or metabolic intermediates.
Collapse
Affiliation(s)
- Andrzej Slominski
- Department of Pathology and Laboratory Medicine, University of Tennessee, Memphis, TN, USA.
| | | | | |
Collapse
|
25
|
Yemets AI, Krasylenko YA, Lytvyn DI, Sheremet YA, Blume YB. Nitric oxide signalling via cytoskeleton in plants. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2011; 181:545-54. [PMID: 21893251 DOI: 10.1016/j.plantsci.2011.04.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 04/25/2011] [Accepted: 04/27/2011] [Indexed: 05/19/2023]
Abstract
Nitric oxide (NO) in plant cell mediates processes of growth and development starting from seed germination to pollination, as well as biotic and abiotic stress tolerance. However, proper understanding of the molecular mechanisms of NO signalling in plants has just begun to emerge. Accumulated evidence suggests that in eukaryotic cells NO regulates functions of proteins by their post-translational modifications, namely tyrosine nitration and S-nitrosylation. Among the candidates for NO-downstream effectors are cytoskeletal proteins because of their involvement in many processes regulated by NO. This review discusses new insights in plant NO signalling focused mainly on the involvement of cytoskeleton components into NO-cascades. Herein, examples of NO-related post-translational modifications of cytoskeletal proteins, and also indirect NO impact, are discussed. Special attention is paid to plant α-tubulin tyrosine nitration as an emerging topic in plant NO research.
Collapse
Affiliation(s)
- Alla I Yemets
- Department of Genomics and Molecular Biotechnology, Institute of Food Biotechnology and Genomics, National Academy of Sciences of Ukraine, Osipovskogo Str., 2a, Kyiv 04123, Ukraine.
| | | | | | | | | |
Collapse
|
26
|
Soon CPW, Donnelly PS, Turner BJ, Hung LW, Crouch PJ, Sherratt NA, Tan JL, Lim NKH, Lam L, Bica L, Lim S, Hickey JL, Morizzi J, Powell A, Finkelstein DI, Culvenor JG, Masters CL, Duce J, White AR, Barnham KJ, Li QX. Diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuII(atsm)) protects against peroxynitrite-induced nitrosative damage and prolongs survival in amyotrophic lateral sclerosis mouse model. J Biol Chem 2011; 286:44035-44044. [PMID: 22033929 DOI: 10.1074/jbc.m111.274407] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive paralyzing disease characterized by tissue oxidative damage and motor neuron degeneration. This study investigated the in vivo effect of diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuII(atsm)), which is an orally bioavailable, blood-brain barrier-permeable complex. In vitro the compound inhibits the action of peroxynitrite on Cu,Zn-superoxide dismutase (SOD1) and subsequent nitration of cellular proteins. Oral treatment of transgenic SOD1G93A mice with CuII(atsm) at presymptomatic and symptomatic ages was performed. The mice were examined for improvement in lifespan and motor function, as well as histological and biochemical changes to key disease markers. Systemic treatment of SOD1G93A mice significantly delayed onset of paralysis and prolonged lifespan, even when administered to symptomatic animals. Consistent with the properties of this compound, treated mice had reduced protein nitration and carbonylation, as well as increased antioxidant activity in spinal cord. Treatment also significantly preserved motor neurons and attenuated astrocyte and microglial activation in mice. Furthermore, CuII(atsm) prevented the accumulation of abnormally phosphorylated and fragmented TAR DNA-binding protein-43 (TDP-43) in spinal cord, a protein pivotal to the development of ALS. CuII(atsm) therefore represents a potential new class of neuroprotective agents targeting multiple major disease pathways of motor neurons with therapeutic potential for ALS.
Collapse
Affiliation(s)
- Cynthia P W Soon
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Paul S Donnelly
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Bradley J Turner
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010; Florey Neuroscience Institutes, The University of Melbourne, Parkville, Victoria 3010
| | - Lin W Hung
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Peter J Crouch
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010
| | - Nicki A Sherratt
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Jiang-Li Tan
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010
| | - Nastasia K-H Lim
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010
| | - Linh Lam
- Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Laura Bica
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010
| | - SinChun Lim
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010
| | - James L Hickey
- School of Chemistry, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Julia Morizzi
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Andrew Powell
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - David I Finkelstein
- Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Janetta G Culvenor
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010
| | - Colin L Masters
- Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010
| | - James Duce
- Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010
| | - Anthony R White
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010
| | - Kevin J Barnham
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010.
| | - Qiao-Xin Li
- Department of Pathology, The University of Melbourne, Parkville, Victoria 3010; Mental Health Research Institute, The University of Melbourne, Parkville, Victoria 3010; Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010.
| |
Collapse
|
27
|
Yasuda S, Yasuda T, Liu MY, Shetty S, Idell S, Boggaram V, Suiko M, Sakakibara Y, Fu J, Liu MC. Sulfation of chlorotyrosine and nitrotyrosine by human lung endothelial and epithelial cells: Role of the human SULT1A3. Toxicol Appl Pharmacol 2011; 251:104-9. [DOI: 10.1016/j.taap.2010.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/07/2010] [Accepted: 12/09/2010] [Indexed: 10/18/2022]
|
28
|
Attenuation of brain nitrostative and oxidative damage by brain cooling during experimental traumatic brain injury. J Biomed Biotechnol 2011; 2011:145214. [PMID: 21318143 PMCID: PMC3034961 DOI: 10.1155/2011/145214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 12/16/2010] [Accepted: 01/04/2011] [Indexed: 11/18/2022] Open
Abstract
The aim of the present study was to ascertain whether brain cooling causes attenuation of traumatic brain injury by reducing brain nitrostative and oxidative damage. Brain cooling was accomplished by infusion of 5 mL of 4°C saline over 5 minutes via the external jugular vein. Immediately after the onset of traumatic brain injury, rats were randomized into two groups and given 37°C or 4°C normal saline. Another group of rats were used as sham operated controls. Behavioral and biochemical assessments were conducted on 72 hours after brain injury or sham operation. As compared to those of the sham-operated controls, the 37°C saline-treated brain injured animals displayed motor deficits, higher cerebral contusion volume and incidence, higher oxidative damage (e.g., lower values of cerebral superoxide dismutase, catalase, glutathione peroxidase and glutathione reductase, but higher values of cerebral malondialdehyde), and higher nitrostative damage (e.g., higher values of neuronal nitric oxide synthase and 3-nitrotyrosine). All the motor deficits and brain nitrostative and oxidative damage were significantly reduced by retrograde perfusion of 4°C saline via the jugular vein. Our data suggest that brain cooling may improve the outcomes of traumatic brain injury in rats by reducing brain nitrostative and oxidative damage.
Collapse
|
29
|
Abstract
Cellular damage occurring under oxidative conditions has been ascribed mainly to the formation of peroxynitrite (ONOOH/ONOO(-)) that originates from the reaction of NO(*) with O(2) (*-). The detrimental effects of peroxynitrite are exacerbated by the reaction with CO(2) that leads to ONOOC(O)O(-), which further decays to the strong oxidant radicals NO(2) (*) and CO(3) (*-). The reaction with CO(2), however, may redirect peroxynitrite specificity. An excessive formation of peroxynitrite represents an important mechanism contributing to the DNA damage, the inactivation of metabolic enzymes, ionic pumps, and structural proteins, and the disruption of cell membranes. Because of its ability to oxidize biomolecules, peroxynitrite is implicated in an increasing list of diseases, including neurodegenerative and cardiovascular disorders, inflammation, pain, autoimmunity, cancer, and aging. However, peroxynitrite displays also protective activities: (i) at high concentrations, it shows anti-viral, anti-microbial, and anti-parasitic actions; and (ii) at low concentrations, it stimulates protective mechanisms in the cardiovascular, nervous, and respiratory systems. The detrimental effects of peroxynitrite and related reactive species are impaired by (pseudo-) enzymatic systems, mainly represented by heme-proteins (e.g., hemoglobin and myoglobin). Here, we report biochemical aspects of peroxynitrite actions being at the root of its biomedical effects.
Collapse
|
30
|
Mir M, Asensio VJ, Tolosa L, Gou-Fabregas M, Soler RM, Lladó J, Olmos G. Tumor necrosis factor alpha and interferon gamma cooperatively induce oxidative stress and motoneuron death in rat spinal cord embryonic explants. Neuroscience 2009; 162:959-71. [PMID: 19477238 DOI: 10.1016/j.neuroscience.2009.05.049] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 05/15/2009] [Accepted: 05/21/2009] [Indexed: 12/13/2022]
Abstract
The accumulation of reactive microglia in the degenerating areas of amyotrophic lateral sclerosis (ALS) tissue is a key cellular event creating a chronic inflammatory environment that results in motoneuron death. We have developed a new culture system that consists in rat spinal cord embryonic explants in which motoneurons migrate outside the explant, growing as a monolayer in the presence of glial cells. The proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interferon gamma (IFN-gamma) have been proposed to be involved in ALS-linked microglial activation. In our explants, the combined exposure to these cytokines resulted in an increased expression of the pro-oxidative enzymes inducible nitric oxide synthase (iNOS), the catalytic subunit of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, gp91(phox) and cyclooxygenase-2 (COX-2), as compared to each cytokine alone. This effect was related to their cooperation in the activation of the transcription factor nuclear factor kappa B (NF-kappaB). TNF-alpha and IFN-gamma also cooperated to promote protein oxidation and nitration, thus increasing the percentage of motoneurons immunoreactive for nitrotyrosine. Apoptotic motoneuron death, measured through annexin V-Cy3 and active caspase-3 immunoreactivities, was also found cooperatively induced by TNF-alpha and IFN-gamma. Interestingly, these cytokines did not affect the viability of purified spinal cord motoneurons in the absence of glial cells. It is proposed that the proinflammatory cytokines TNF-alpha and IFN-gamma have cooperative/complementary roles in inflammation-induced motoneuron death.
Collapse
Affiliation(s)
- M Mir
- Grup de Neurobiologia Cel.lular, Institut Universitari d'Investigacions en Ciències de la Salut/Departament de Biologia, Universitat de les Illes Balears, Cra. de Valldemossa, km 7.5, E-07122 Palma de Mallorca, Illes Balears, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Ischiropoulos H. Protein tyrosine nitration--an update. Arch Biochem Biophys 2008; 484:117-21. [PMID: 19007743 DOI: 10.1016/j.abb.2008.10.034] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 10/24/2008] [Accepted: 10/24/2008] [Indexed: 10/21/2022]
Abstract
Tyrosine nitration is a covalent post-translational protein modification derived from the reaction of proteins with nitrating agents. Tyrosine nitration has been used as a marker of oxidant burden in human diseases. However, it remains unclear whether protein nitration is responsible for alterations in protein function that imparts an increased risk for disease development or unfavorable outcomes. Emerging data implicate tyrosine nitration as a mediator of immune responses suggesting a novel biological function for this protein modification.
Collapse
Affiliation(s)
- Harry Ischiropoulos
- Stokes Research Institute and Departments of Pediatrics and Pharmacology, Children's Hospital of Philadelphia and The University of Pennsylvania, 417 Abramson Research Center, Philadelphia, PA 19104-4318, USA.
| |
Collapse
|
32
|
Morphological features and responses to AMPA receptor-mediated excitotoxicity of mouse motor neurons: comparison in purified, mixed anterior horn or motor neuron/glia cocultures. J Neurosci Methods 2008; 170:85-95. [DOI: 10.1016/j.jneumeth.2007.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 11/30/2007] [Accepted: 12/30/2007] [Indexed: 11/23/2022]
|
33
|
Qi JS, Li YN, Zhang BS, Niu T, Liang JH. Peroxynitrite mediates high glucose-induced osteoblast apoptosis. J Endocrinol Invest 2008; 31:314-20. [PMID: 18475049 DOI: 10.1007/bf03346364] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
High glucose (HG) state is closely related to diabetic complications, and among these one main pathogenesis involves apoptosis of important functional cells. However, the mechanism of osteoblast (OB) apoptosis induced by HG state is not clear. Peroxynitrite (ONOO-), the strongest oxidant, can mediate apoptosis in various kinds of cells, including OB. Therefore, this study was aimed at investigating whether HG state could induce OB apoptosis through ONOO-. Cultured OB from rat calvariae explanted from E21 fetuses were treated with HG solution. The ratio of OB apoptosis and ONOOcontent was assayed by TUNEL, flow cytometry, and immunohistochemistry. The results showed that HG state could induce ONOO- overformation (p<0.01), and excessive OB apoptosis (p<0.001). However, the HG-induced OB apoptosis could be attenuated by peonol, a potent scavenger of ONOO-. In conclusion, HG state might trigger excessive ONOO- formation, mediating OB apoptosis.
Collapse
Affiliation(s)
- J S Qi
- Department of Biochemistry, Hebei Medical University, 050017 Shijiazhuang, PR China.
| | | | | | | | | |
Collapse
|
34
|
Ma TC, Mihm MJ, Bauer JA, Hoyt KR. Bioenergetic and oxidative effects of free 3-nitrotyrosine in culture: selective vulnerability of dopaminergic neurons and increased sensitivity of non-dopaminergic neurons to dopamine oxidation. J Neurochem 2007; 103:131-44. [PMID: 17877636 DOI: 10.1111/j.1471-4159.2007.04735.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Protein bound and free 3-nitrotyrosine (3NT) levels are elevated in neurodegenerative diseases and have been used as evidence for peroxynitrite generation. Intrastriatal injection of free 3NT causes dopaminergic neuron injury and represents a new mouse model of Parkinson's disease (PD). We are investigating the nature of free 3NT neurotoxicity. In primary ventral midbrain cultures, free 3NT damaged dopaminergic neurons, while adjacent non-dopaminergic neurons were unaffected. Combined treatment with free 3NT and subtoxic amounts of dopamine caused extensive death of non-dopaminergic forebrain neurons in culture. Free 3NT alone directly inhibited mitochondrial complex I, decreased ATP, sensitized neurons to mitochondrial depolarization, and increased superoxide production. Subtoxic concentrations of rotenone (instead of free 3NT) caused similar results. Additionally, free 3NT and dopamine combined increased extraneuronal hydrogen peroxide and decreased intraneuronal glutathione levels more than dopamine alone. Oxidative and bioenergetic processes have been proposed to contribute to neurodegeneration in PD. As free 3NT is a compound that is increased in PD, damages dopamine neurons in vivo and in vitro and has detrimental effects on neuronal bioenergetics, it is possible that free 3NT is an endogenous contributing factor to neuronal loss, in addition to being a marker of oxidative and nitrative processes.
Collapse
Affiliation(s)
- Thong C Ma
- Division of Pharmacology, The Ohio State University, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
35
|
Duleu S, Van Der Velden C, Poulletier de Gannes F, Tranchant MC, Geffard M. Circulating antibodies to NO- and ONOO-modified antigens in amyotrophic lateral sclerosis, Alzheimer's disease and multiple sclerosis. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.immbio.2007.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Darwish RS, Amiridze N, Aarabi B. Nitrotyrosine as an Oxidative Stress Marker: Evidence for Involvement in Neurologic Outcome in Human Traumatic Brain Injury. ACTA ACUST UNITED AC 2007; 63:439-42. [PMID: 17693848 DOI: 10.1097/ta.0b013e318069178a] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Oxidative stress has been indicated as a mechanism of secondary neuronal injury in traumatic brain injury (TBI). Nitrotyrosine in the cerebrospinal fluid (CSF) may be an in vivo marker of oxidative nitric oxide damage. We tested the hypothesis that increased levels of nitrotyrosine correlate with poor neurologic outcomes in patients with TBI and attempted to identify the source of increased CSF nitrotyrosine. METHODS This institutional-review-board-approved study included 10 adults with severe closed TBI (Glasgow Coma Scale score <8) and no documented hypoxic brain injury. These patients underwent routine evaluation and, when indicated, placement of an intraventricular catheter. CSF samples (n = 27) were collected 2 to 72 hours after TBI and were also obtained from four healthy individuals. Nitrotyrosine levels were measured, and immunohistochemistry was performed. Neurologic follow-up extended to 1 month after injury. RESULTS Nitrotyrosine was not detected in the control samples but was detected in 13 CSF samples from 7 TBI patients (range, 22.4-97.6 nM/mL). Seven patients had poor outcomes, and, in each, nitrotyrosine was detected. Nitrotyrosine immunoreactivity was detected in neurons and glia and confirmed in brain homogenate. CONCLUSION Oxidative stress contributes to secondary brain injury in patients with TBI. Poor neurologic outcome is associated with increased levels of nitrotyrosine in the CSF. Identifying patients or the stage at which oxidative stress is more active using CSF markers of oxidative injury may help in the development of more targeted treatments.
Collapse
Affiliation(s)
- Ribal S Darwish
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | | | |
Collapse
|
37
|
Ye Y, Quijano C, Robinson KM, Ricart KC, Strayer AL, Sahawneh MA, Shacka JJ, Kirk M, Barnes S, Accavitti-Loper MA, Radi R, Beckman JS, Estévez AG. Prevention of peroxynitrite-induced apoptosis of motor neurons and PC12 cells by tyrosine-containing peptides. J Biol Chem 2007; 282:6324-37. [PMID: 17200124 DOI: 10.1074/jbc.m610800200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Although peroxynitrite stimulates apoptosis in many cell types, whether peroxynitrite acts directly as an oxidant or the induction of apoptosis is because of the radicals derived from peroxynitrite decomposition remains unknown. Before undergoing apoptosis because of trophic factor deprivation, primary motor neuron cultures become immunoreactive for nitrotyrosine. We show here using tyrosine-containing peptides that free radical processes mediated by peroxynitrite decomposition products were required for triggering apoptosis in primary motor neurons and in PC12 cells cultures. The same concentrations of tyrosine-containing peptides required to prevent the nitration and apoptosis of motor neurons induced by trophic factor deprivation and of PC12 cells induced by peroxynitrite also prevented peroxynitrite-mediated nitration of motor neurons, brain homogenates, and PC12 cells. The heat shock protein 90 chaperone was nitrated in both trophic factor-deprived motor neurons and PC12 cells incubated with peroxynitrite. Tyrosine-containing peptides did not affect the induction of PC12 cell death by hydrogen peroxide. Tyrosine-containing peptides should protect by scavenging peroxynitrite-derived radicals and not by direct reactions with peroxynitrite as they neither increase the rate of peroxynitrite decomposition nor decrease the bimolecular peroxynitrite-mediated oxidation of thiols. These results reveal an important role for free radical-mediated nitration of tyrosine residues, in apoptosis induced by endogenously produced and exogenously added peroxynitrite; moreover, tyrosine-containing peptides may offer a novel strategy to neutralize the toxic effects of peroxynitrite.
Collapse
Affiliation(s)
- Yaozu Ye
- Burke Medical Research Institute, White Plains, New York 10605, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yasuda S, Idell S, Liu MC. Generation and release of nitrotyrosine O-sulfate by HepG2 human hepatoma cells upon SIN-1 stimulation: identification of SULT1A3 as the enzyme responsible. Biochem J 2007; 401:497-503. [PMID: 17002600 PMCID: PMC1820819 DOI: 10.1042/bj20060536] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In addition to serving as a biomarker of oxidative/nitrative stress, elevated levels of nitrotyrosine have been shown to cause DNA damage or trigger apoptosis. Whether the body is equipped with mechanisms for protecting against the potentially harmful nitrotyrosine remains unknown. The present study was designed to investigate the possibility that sulfation serves as a pathway for the metabolism/regulation of nitrotyrosine. Using metabolic labelling, nitrotyrosine O-[35S]sulfate was found to be produced and released into the medium of HepG2 human hepatoma cells labelled with [35S]sulfate in the presence of nitrotyrosine. To identify the enzyme(s) responsible for nitrotyrosine sulfation, a systematic study of all eleven known human cytosolic SULTs (sulfotransferases) was performed. Of the 11 enzymes tested, only SULT1A3 displayed sulfating activity toward nitrotyrosine. The pH-dependence and kinetic constants of SULT1A3 with nitrotyrosine or dopamine as substrate were determined. To examine whether the sulfation of nitrotyrosine occurs in the context of cellular physiology, HepG2 cells labelled with [35S]sulfate were treated with SIN-1 (morpholinosydnonimine), a peroxynitrite generator. Increments of nitrotyrosine O-[35S]sulfate were detected in the medium of HepG2 cells treated with higher concentrations of SIN-1. To gain insight into the physiological relevance of nitrotyrosine sulfation, a time-course study was performed using [3H]tyrosine-labelled HepG2 cells treated with SIN-1. The findings confirm that the bulk of free [3H]nitrotyrosine inside the cells was present in the unconjugated form. The proportion of sulfated [3H]nitrotyrosine increased dramatically in the medium over time, implying that sulfation may play a significant role in the metabolism of free nitrotyrosine.
Collapse
Affiliation(s)
- Shin Yasuda
- Biomedical Research Center and The Texas Lung Injury Institute, The University of Texas Health Center, 11937 U.S. Highway 271, Tyler, TX 75708, U.S.A
| | - Steven Idell
- Biomedical Research Center and The Texas Lung Injury Institute, The University of Texas Health Center, 11937 U.S. Highway 271, Tyler, TX 75708, U.S.A
| | - Ming-Cheh Liu
- Biomedical Research Center and The Texas Lung Injury Institute, The University of Texas Health Center, 11937 U.S. Highway 271, Tyler, TX 75708, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
39
|
Rakhit R, Chakrabartty A. Structure, folding, and misfolding of Cu,Zn superoxide dismutase in amyotrophic lateral sclerosis. Biochim Biophys Acta Mol Basis Dis 2006; 1762:1025-37. [PMID: 16814528 DOI: 10.1016/j.bbadis.2006.05.004] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 05/15/2006] [Accepted: 05/16/2006] [Indexed: 11/16/2022]
Abstract
Fourteen years after the discovery that mutations in Cu, Zn superoxide dismutase (SOD1) cause a subset of familial amyotrophic lateral sclerosis (fALS), the mechanism by which mutant SOD1 exerts toxicity remains unknown. The two principle hypotheses are (a) oxidative damage stemming from aberrant SOD1 redox chemistry, and (b) misfolding of the mutant protein. Here we review the structure and function of wild-type SOD1, as well as the changes to the structure and function in mutant SOD1. The relative merits of the two hypotheses are compared and a common unifying principle is outlined. Lastly, the potential for therapies targeting SOD1 misfolding is discussed.
Collapse
Affiliation(s)
- Rishi Rakhit
- Department of Biochemistry, University of Toronto, University Health Network, Toronto Medical Discovery Tower, Medical and Related Sciences (MaRS), 101 College Street, Toronto, ON, Canada, M5G 1L7
| | | |
Collapse
|
40
|
Petri S, Kiaei M, Damiano M, Hiller A, Wille E, Manfredi G, Calingasan NY, Szeto HH, Beal MF. Cell-permeable peptide antioxidants as a novel therapeutic approach in a mouse model of amyotrophic lateral sclerosis. J Neurochem 2006; 98:1141-8. [PMID: 16895581 DOI: 10.1111/j.1471-4159.2006.04018.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Reactive oxygen species (ROS) play a major role in the pathogenesis of neurodegenerative diseases. They are important contributors to necrotic and apoptotic cell death. A major proportion of cellular ROS is generated at the inner mitochondrial membrane by the respiratory chain. In the present study, we investigated a novel peptide antioxidant (SS-31) targeted to the inner mitochondrial membrane for its therapeutic effects both in vitro and in vivo in the G93A mouse model of amyotrophic lateral sclerosis (ALS). SS-31 protected against cell death induced by hydrogen peroxide in vitro in neuronal cells stably transfected with either wild-type or mutant Cu/Zn superoxide dismutase (SOD1). Daily intraperitoneal injections of SS-31 (5 mg/kg), starting at 30 days of age, led to a significant improvement in survival and motor performance. In comparison with vehicle-treated G93A mice, SS-31-treated mice showed a decreased cell loss and a decrease in immunostaining for markers of oxidative stress in the lumbar spinal cord. This further enhances the concept that pharmacological modification of oxidative stress is a therapeutic option for the treatment of ALS.
Collapse
Affiliation(s)
- Susanne Petri
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Blanchard-Fillion B, Prou D, Polydoro M, Spielberg D, Tsika E, Wang Z, Hazen SL, Koval M, Przedborski S, Ischiropoulos H. Metabolism of 3-nitrotyrosine induces apoptotic death in dopaminergic cells. J Neurosci 2006; 26:6124-30. [PMID: 16763020 PMCID: PMC6675196 DOI: 10.1523/jneurosci.1038-06.2006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intrastriatal injection of 3-nitrotyrosine, which is a biomarker for nitrating oxidants, provokes dopaminergic neuronal death in rats by unknown mechanisms. Herein, we show that extracellular 3-nitrotyrosine is transported via the l-aromatic amino acid transporter in nondopaminergic NT2 cells, whereas in dopaminergic PC12 cells, it is transported by both the l-aromatic amino acid and the dopamine transporters. In both cell lines, 3-nitrotyrosine is a substrate for tyrosine tubulin ligase, resulting in its incorporation into the C terminus of alpha-tubulin. In NT2 cells, incorporation of 3-nitrotyrosine into alpha-tubulin induces a progressive, reversible reorganization of the microtubule architecture. In PC12 cells, 3-nitrotyrosine decreases intracellular dopamine levels and is metabolized by the concerted action of the aromatic amino acid decarboxylase and monoamine oxidase. Intracellular levels of 133 micromol of 3-nitrotyrosine per mole of tyrosine did not alter NT2 viability but induced PC12 apoptosis. The cell death was reversed by caspases and aromatic amino acid decarboxylase and monoamine oxidase inhibitors. 3-Nitrotyrosine induced loss of tyrosine hydroxylase-positive primary rat neurons, which was also prevented by an aromatic amino acid decarboxylase inhibitor. These findings provide a novel mechanism by which products generated by reactive nitrogen species induce dopaminergic neuron death and thus may contribute to the selective neurodegeneration in Parkinson's disease.
Collapse
|
42
|
Peluffo H, Acarin L, Arís A, González P, Villaverde A, Castellano B, González B. Neuroprotection from NMDA excitotoxic lesion by Cu/Zn superoxide dismutase gene delivery to the postnatal rat brain by a modular protein vector. BMC Neurosci 2006; 7:35. [PMID: 16638118 PMCID: PMC1462999 DOI: 10.1186/1471-2202-7-35] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 04/25/2006] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Superoxide mediated oxidative stress is a key neuropathologic mechanism in acute central nervous system injuries. We have analyzed the neuroprotective efficacy of the transient overexpression of antioxidant enzyme Cu/Zn Superoxide dismutase (SOD) after excitotoxic injury to the immature rat brain by using a recently constructed modular protein vector for non-viral gene delivery termed NLSCt. For this purpose, animals were injected with the NLSCt vector carrying the Cu/Zn SOD or the control GFP transgenes 2 hours after intracortical N-methyl-D-aspartate (NMDA) administration, and daily functional evaluation was performed. Moreover, 3 days after, lesion volume, neuronal degeneration and nitrotyrosine immunoreactivity were evaluated. RESULTS Overexpression of Cu/Zn SOD transgene after NMDA administration showed improved functional outcome and a reduced lesion volume at 3 days post lesion. In secondary degenerative areas, increased neuronal survival as well as decreased numbers of degenerating neurons and nitrotyrosine immunoreactivity was seen. Interestingly, injection of the NLSCt vector carrying the control GFP transgene also displayed a significant neuroprotective effect but less pronounced. CONCLUSION When the appropriate levels of Cu/Zn SOD are expressed transiently after injury using the non-viral modular protein vector NLSCt a neuroprotective effect is seen. Thus recombinant modular protein vectors may be suitable for in vivo gene therapy, and Cu/Zn SOD should be considered as an interesting therapeutic transgene.
Collapse
Affiliation(s)
- Hugo Peluffo
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Laia Acarin
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Anna Arís
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Pau González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Antoni Villaverde
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Bernardo Castellano
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Berta González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| |
Collapse
|
43
|
Guix FX, Uribesalgo I, Coma M, Muñoz FJ. The physiology and pathophysiology of nitric oxide in the brain. Prog Neurobiol 2005; 76:126-52. [PMID: 16115721 DOI: 10.1016/j.pneurobio.2005.06.001] [Citation(s) in RCA: 480] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 06/10/2005] [Accepted: 06/14/2005] [Indexed: 12/11/2022]
Abstract
Nitric oxide (NO) is a molecule with pleiotropic effects in different tissues. NO is synthesized by NO synthases (NOS), a family with four major types: endothelial, neuronal, inducible and mitochondrial. They can be found in almost all the tissues and they can even co-exist in the same tissue. NO is a well-known vasorelaxant agent, but it works as a neurotransmitter when produced by neurons and is also involved in defense functions when it is produced by immune and glial cells. NO is thermodynamically unstable and tends to react with other molecules, resulting in the oxidation, nitrosylation or nitration of proteins, with the concomitant effects on many cellular mechanisms. NO intracellular signaling involves the activation of guanylate cyclase but it also interacts with MAPKs, apoptosis-related proteins, and mitochondrial respiratory chain or anti-proliferative molecules. It also plays a role in post-translational modification of proteins and protein degradation by the proteasome. However, under pathophysiological conditions NO has damaging effects. In disorders involving oxidative stress, such as Alzheimer's disease, stroke and Parkinson's disease, NO increases cell damage through the formation of highly reactive peroxynitrite. The paradox of beneficial and damaging effects of NO will be discussed in this review.
Collapse
Affiliation(s)
- F X Guix
- Laboratori de Fisiologia Molecular, Unitat de Senyalització Cellular, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Carrer Dr. Aiguader, 80, Barcelona 08003, Spain
| | | | | | | |
Collapse
|
44
|
Tedeschi G, Cappelletti G, Negri A, Pagliato L, Maggioni MG, Maci R, Ronchi S. Characterization of nitroproteome in neuron-like PC12 cells differentiated with nerve growth factor: Identification of two nitration sites in α-tubulin. Proteomics 2005; 5:2422-32. [PMID: 15887183 DOI: 10.1002/pmic.200401208] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Nitric oxide (NO) is a precursor of reactive nitrating species, peroxynitrite and nitrogen dioxide, which modify proteins to generate oxidized species such as 3-nitrotyrosine that has been used as a hallmark of peroxynitrite-mediated oxidative stress on proteins. In the last few years however, a growing body of evidence indicates that NO also regulates a myriad of physiologic responses by modifying tyrosine residues. Looking for the molecular event triggered by NO in nerve growth factor (NGF)-induced neuronal differentiation, we recently reported that in differentiating PC12 cells, the cytoskeleton becomes the main cellular fraction containing nitrotyrosinated proteins, and alpha-tubulin is the major target. In the present work, we focus on the investigation of the sites of tyrosine nitration in alpha-tubulin purified by two-dimensional gel electrophoresis following anti-alpha-tubulin immunoprecipitation of protein extract from NGF-treated PC12 cells. Using Western blotting and matrix-assisted laser desorption/ionization-time of flight analysis, we show for the first time, both in vivo and in vitro, that nitration can occur on alpha-tubulin at sites other than the C-terminus and we positively identify Tyr 161 and Tyr 357 as two specific amino acids endogenously nitrated.
Collapse
|
45
|
Peluffo H, Acarin L, Faiz M, Castellano B, Gonzalez B. Cu/Zn superoxide dismutase expression in the postnatal rat brain following an excitotoxic injury. J Neuroinflammation 2005; 2:12. [PMID: 15929797 PMCID: PMC1164430 DOI: 10.1186/1742-2094-2-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Accepted: 06/01/2005] [Indexed: 01/11/2023] Open
Abstract
Background In the nervous system, as in other organs, Cu/Zn superoxide dismutase (Cu/Zn SOD) is a key antioxidant enzyme involved in superoxide detoxification in normal cellular metabolism and after cell injury. Although it has been suggested that immature brain has a different susceptibility to oxidative damage than adult brain, the distribution and cell-specific expression of this enzyme in immature brain and after postnatal brain damage has not been documented. Methods In this study, we used immunohistochemistry and western blot to analyze the expression of Cu/Zn SOD in intact immature rat brain and in immature rat brain after an NMDA-induced excitotoxic cortical injury performed at postnatal day 9. Double immunofluorescence labelling was used to identify Cu/Zn SOD-expressing cell populations. Results In intact immature brain, Cu/Zn SOD enzyme was widely expressed at high levels in neurons mainly located in cortical layers II, III and V, in the sub-plate, in the pyriform cortex, in the hippocampus, and in the hypothalamus. Glial fibrillary acidic protein-positive cells only showed Cu/Zn SOD expression in the glia limitans and in scattered cells of the ventricle walls. No expression was detected in interfascicular oligodendroglia, microglia or endothelial cells. Following excitotoxic damage, neuronal Cu/Zn SOD was rapidly downregulated (over 2–4 hours) at the injection site before neurodegeneration signals and TUNEL staining were observed. Later, from 1 day post-lesion onward, an upregulation of Cu/Zn SOD was found due to increased expression in astroglia. A further increase was observed at 3, 5 and 7 days that corresponded to extensive induction of Cu/Zn SOD in highly reactive astrocytes and in the astroglial scar. Conclusion We show here that, in the intact immature brain, the expression of Cu/Zn SOD was mainly found in neurons. When damage occurs, a strong and very rapid downregulation of this enzyme precedes neuronal degeneration, and is followed by an upregulation of Cu/Zn SOD in astroglial cells.
Collapse
Affiliation(s)
- Hugo Peluffo
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Laia Acarin
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Maryam Faiz
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Bernardo Castellano
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| | - Berta Gonzalez
- Unit of Histology, Department Of Cell Biology, Physiology, and Immunology; Autonomous University of Barcelona, 08193, Spain
- Institute of Neuroscience, Autonomous University of Barcelona, 08193, Spain
| |
Collapse
|
46
|
Nowoslawski L, Klocke BJ, Roth KA. Molecular Regulation of Acute Ethanol-Induced Neuron Apoptosis. J Neuropathol Exp Neurol 2005; 64:490-7. [PMID: 15977640 DOI: 10.1093/jnen/64.6.490] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ethanol is a potent neurotoxin particularly for the developing nervous system. Intrauterine exposure to ethanol during the last trimester of human gestation can produce a broad spectrum of neuropathologic consequences. This period of human brain development is roughly equivalent to the first week of rodent postnatal life and acute exposure of neonatal mice to ethanol produces massive neuronal apoptosis throughout the brain. We have previously demonstrated that ethanol-induced neuron apoptosis is critically dependent on expression of Bax, a proapoptotic member of the Bcl-2 family. To further define the molecular pathway regulating ethanol-induced neuron apoptosis, we analyzed the effects of acute ethanol exposure on cerebellar internal granule cell neurons both in vivo and in vitro. Ethanol produced extensive Bax-dependent caspase-3 activation and neuron apoptosis in the cerebellar internal granule cell layer, which was maximal at approximately 6 hours postadministration. This effect was recapitulated in vitro and required new gene transcription, protein translation, Bax expression, and caspase activation. Ethanol-induced neuron death was independent of p53 expression and was unaffected by deficiency in the proapoptotic Bcl-2 family members Bid or Bad. These studies indicate that ethanol activates an intrinsic apoptotic death program in neurons that is likely to contribute to the neuropathologic effects of human fetal alcohol exposure.
Collapse
Affiliation(s)
- Lisa Nowoslawski
- Department of Pathology, Division of Neuropathology University of Alabama at Birmingham, Birmingham, Alabama 35294-0017, USA
| | | | | |
Collapse
|
47
|
Acarin L, Peluffo H, Barbeito L, Castellano B, González B. Astroglial nitration after postnatal excitotoxic damage: correlation with nitric oxide sources, cytoskeletal, apoptotic and antioxidant proteins. J Neurotrauma 2005; 22:189-200. [PMID: 15665612 DOI: 10.1089/neu.2005.22.189] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxygen free radicals and nitric oxide (NO) participate in the pathogenesis of acute central nervous system (CNS) injury by forming peroxynitrite, which promotes oxidative damage and tyrosine nitration. Neuronal nitration is associated with cell death, but little is known of the characteristics and cell fate of nitrated astrocytes. In this study, we have used a postnatal excitotoxic lesion model (intracortical NMDA injection) and our aims were (i) to evaluate the temporal and spatial pattern of astroglial nitration in correlation with the neuropathological process and the sources of NO; and (ii) to establish, if any, the correlation among astrocyte nitration and other events such as expression of cytoskeletal proteins, antioxidant enzymes, and cell death markers to cope with nitration and/or undergo cell death. Our results show that after postnatal excitotoxic damage two distinct waves of nitration were observed in relation to astrocytes. At 24 h post-lesion, early-nitrated astrocytes were found within the neurodegenerating area, coinciding with the time of maximal cell death. These early-nitrated astrocytes are highly ramified protoplasmic cells, showing diffuse glial fibrillary acidic protein (GFAP) content and expressing inducible NOS. At later time-points, when astrogliosis is morphologically evident, nitrated hypertrophied reactive astrocytes are observed in the penumbra and the neurodegenerated area, displaying increased expression of GFAP and vimentin cytoskeletal proteins and of metallothionein I-II and Cu/Zn superoxide dismutase antioxidant proteins. Moreover, despite revealing activated caspase-3, they do not show TUNEL labeling. In summary, we show that nitrated astrocytes in vivo constitute a subpopulation of highly reactive astrocytes which display high resistance towards oxidative stress induced cell death.
Collapse
Affiliation(s)
- Laia Acarin
- Unit of Histology, School of Medicine, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma Barcelona, Bellaterra 08193, Spain.
| | | | | | | | | |
Collapse
|
48
|
Wootz H, Hansson I, Korhonen L, Näpänkangas U, Lindholm D. Caspase-12 cleavage and increased oxidative stress during motoneuron degeneration in transgenic mouse model of ALS. Biochem Biophys Res Commun 2004; 322:281-6. [PMID: 15313203 DOI: 10.1016/j.bbrc.2004.07.118] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2004] [Indexed: 11/16/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by loss of motoneurons in the spinal cord and brain stem. We have characterized motoneuron death in transgenic mice carrying the mutant human copper/zinc superoxide dismutase, as a model for familial ALS. Previous studies have shown the involvement of mitochondria in nerve cell demise in these animals. We report here an early cleavage of caspase-12, residing in the endoplasmic reticulum (ER), in the spinal cord during the course of the disease. Apart from caspase-12, caspase-9, and caspase-3 were activated in the transgenic ALS mice. Staining with an antibody for nitrotyrosine, as a marker for oxidative stress, showed a large increase in the ALS mice. The results indicate that oxidative and ER induced stress causing caspase-12 activation are involved in neuronal death and disease progression in ALS. Caspase-12 and the ER pathway for cell death may constitute potential novel targets for the treatment of ALS.
Collapse
Affiliation(s)
- Hanna Wootz
- Department of Neuroscience, Unit of Neurobiology, Uppsala University, Biomedical Centre, Box 587, S-751 23 Uppsala, Sweden
| | | | | | | | | |
Collapse
|