1
|
Hellwig M, Diel P, Eisenbrand G, Grune T, Guth S, Henle T, Humpf HU, Joost HG, Marko D, Raupbach J, Roth A, Vieths S, Mally A. Dietary glycation compounds - implications for human health. Crit Rev Toxicol 2024; 54:485-617. [PMID: 39150724 DOI: 10.1080/10408444.2024.2362985] [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: 03/26/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 08/17/2024]
Abstract
The term "glycation compounds" comprises a wide range of structurally diverse compounds that are formed endogenously and in food via the Maillard reaction, a chemical reaction between reducing sugars and amino acids. Glycation compounds produced endogenously are considered to contribute to a range of diseases. This has led to the hypothesis that glycation compounds present in food may also cause adverse effects and thus pose a nutritional risk to human health. In this work, the Senate Commission on Food Safety (SKLM) of the German Research Foundation (DFG) summarized data on formation, occurrence, exposure and toxicity of glycation compounds (Part A) and systematically assessed potential associations between dietary intake of defined glycation compounds and disease, including allergy, diabetes, cardiovascular and renal disease, gut/gastrotoxicity, brain/cognitive impairment and cancer (Part B). A systematic search in Pubmed (Medline), Scopus and Web of Science using a combination of keywords defining individual glycation compounds and relevant disease patterns linked to the subject area of food, nutrition and diet retrieved 253 original publications relevant to the research question. Of these, only 192 were found to comply with previously defined quality criteria and were thus considered suitable to assess potential health risks of dietary glycation compounds. For each adverse health effect considered in this assessment, however, only limited numbers of human, animal and in vitro studies were identified. While studies in humans were often limited due to small cohort size, short study duration, and confounders, experimental studies in animals that allow for controlled exposure to individual glycation compounds provided some evidence for impaired glucose tolerance, insulin resistance, cardiovascular effects and renal injury in response to oral exposure to dicarbonyl compounds, albeit at dose levels by far exceeding estimated human exposures. The overall database was generally inconsistent or inconclusive. Based on this systematic review, the SKLM concludes that there is at present no convincing evidence for a causal association between dietary intake of glycation compounds and adverse health effects.
Collapse
Affiliation(s)
- Michael Hellwig
- Chair of Special Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | | | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Sabine Guth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | - Thomas Henle
- Chair of Food Chemistry, TU Dresden, Dresden, Germany
| | | | - Hans-Georg Joost
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Jana Raupbach
- Institute of Food Chemistry, Technische Universität Braunschweig, Braunschweig, Germany
| | - Angelika Roth
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Dortmund, Germany
| | | | - Angela Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Najjar JA, Calvert JW. Effects of protein glycation and protective mechanisms against glycative stress. Curr Opin Pharmacol 2024; 76:102464. [PMID: 38796877 PMCID: PMC11229435 DOI: 10.1016/j.coph.2024.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/15/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Glycation is a posttranslational modification of proteins that contributes to the vast array of biological information that can be conveyed via a singular proteome. Understanding the role of advanced glycation end-products (AGEs) in human health and pathophysiology can be difficult, as the physiological effects of AGEs have been associated with multiple biological processes and disease state development, including acute myocardial ischemia-reperfusion injury, heart failure, and atherosclerosis, as well as tumor cell migration. The critical role of the glyoxalase system in the detoxification of methylglyoxal and other AGEs has been well established. Recently, evidence has emerged that DJ-1 displays antiglycative activity and may contribute to another mechanism of protection against protein glycation outside of the glyoxalase system. Identification of potential substrates of DJ-1 and determination of the pathways in which DJ-1 operates, is needed to fully understand the role of this protein in modulating biological homeostasis and the development of disease.
Collapse
Affiliation(s)
- Jade A Najjar
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, USA
| | - John W Calvert
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, USA.
| |
Collapse
|
3
|
Yang Z, Zhang W, Lu H, Cai S. Methylglyoxal in the Brain: From Glycolytic Metabolite to Signalling Molecule. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227905. [PMID: 36432007 PMCID: PMC9696358 DOI: 10.3390/molecules27227905] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
Advances in molecular biology technology have piqued tremendous interest in glycometabolism and bioenergetics in homeostasis and neural development linked to ageing and age-related diseases. Methylglyoxal (MGO) is a by-product of glycolysis, and it can covalently modify proteins, nucleic acids, and lipids, leading to cell growth inhibition and, eventually, cell death. MGO can alter intracellular calcium homeostasis, which is a major cell-permeant precursor to advanced glycation end-products (AGEs). As side-products or signalling molecules, MGO is involved in several pathologies, including neurodevelopmental disorders, ageing, and neurodegenerative diseases. In this review, we demonstrate that MGO (the metabolic side-product of glycolysis), the GLO system, and their analogous relationship with behavioural phenotypes, epigenetics, ageing, pain, and CNS degeneration. Furthermore, we summarise several therapeutic approaches that target MGO and the glyoxalase (GLO) system in neurodegenerative diseases.
Collapse
Affiliation(s)
- Zeyong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Huashan Rd. 1961, Shanghai 200030, China
- Correspondence: (Z.Y.); (S.C.)
| | - Wangping Zhang
- Department of Anesthesiology, Women and Children’s Hospital of Jiaxing University, No. 2468 Zhonghuan East Road, Jiaxing 314000, China
| | - Han Lu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shu Cai
- School of Nursing, Guangdong Pharmaceutical University, No. 283 Jianghai Avenue, Haizhu District, Guangzhou 510310, China
- Correspondence: (Z.Y.); (S.C.)
| |
Collapse
|
4
|
Toriumi K, Miyashita M, Suzuki K, Tabata K, Horiuchi Y, Ishida H, Itokawa M, Arai M. Role of glyoxalase 1 in methylglyoxal detoxification-the broad player of psychiatric disorders. Redox Biol 2021; 49:102222. [PMID: 34953453 PMCID: PMC8718652 DOI: 10.1016/j.redox.2021.102222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022] Open
Abstract
Methylglyoxal (MG) is a highly reactive α-ketoaldehyde formed endogenously as a byproduct of the glycolytic pathway. To remove MG, various detoxification systems work together in vivo, including the glyoxalase system, which enzymatically degrades MG using glyoxalase 1 (GLO1) and GLO2. Recently, numerous reports have shown that GLO1 expression and MG accumulation in the brain are involved in the pathogenesis of psychiatric disorders, such as anxiety disorder, depression, autism, and schizophrenia. Furthermore, it has been reported that GLO1 inhibitors may be promising drugs for the treatment of psychiatric disorders. In this review, we discuss the recent findings of the effects of altered GLO1 function on mental behavior, especially focusing on results obtained from animal models.
Collapse
Affiliation(s)
- Kazuya Toriumi
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Mitsuhiro Miyashita
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan; Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital, Setagaya-ku, Tokyo, 156-0057, Japan; Department of Psychiatry, Takatsuki Hospital, Hachioji, Tokyo, 192-0005, Japan
| | - Kazuhiro Suzuki
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan; Department of Psychiatry, Graduate School of Medicine, Shinshu University, Nagano, 390-8621, Japan
| | - Koichi Tabata
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan; Department of Psychiatry and Behavioral Science, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Yasue Horiuchi
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Hiroaki Ishida
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Masanari Itokawa
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan; Department of Psychiatry, Tokyo Metropolitan Matsuzawa Hospital, Setagaya-ku, Tokyo, 156-0057, Japan
| | - Makoto Arai
- Schizophrenia Research Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan.
| |
Collapse
|
5
|
Malard E, Valable S, Bernaudin M, Pérès E, Chatre L. The Reactive Species Interactome in the Brain. Antioxid Redox Signal 2021; 35:1176-1206. [PMID: 34498917 DOI: 10.1089/ars.2020.8238] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: Redox pioneer Helmut Sies attempted to explain reactive species' challenges faced by organelles, cells, tissues, and organs via three complementary definitions: (i) oxidative stress, that is, the disturbance in the prooxidant-antioxidant defense balance in favor of the prooxidants; (ii) oxidative eustress, the low physiological exposure to prooxidants; and (iii) oxidative distress, the supraphysiological exposure to prooxidants. Recent Advances: Identification, concentration, and interactions are the most important elements to improve our understanding of reactive species in physiology and pathology. In this context, the reactive species interactome (RSI) is a new multilevel redox regulatory system that identifies reactive species families, reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species, and it integrates their interactions with their downstream biological targets. Critical Issues: We propose a united view to fully combine reactive species identification, oxidative eustress and distress, and the RSI system. In this view, we also propose including the forgotten reactive carbonyl species, an increasingly rediscovered reactive species family related to the other reactive families, and key enzymes within the RSI. We focus on brain physiology and pathology to demonstrate why this united view should be considered. Future Directions: More studies are needed for an improved understanding of the contributions of reactive species through their identification, concentration, and interactions, including in the brain. Appreciating the RSI in its entirety should unveil new molecular players and mechanisms in physiology and pathology in the brain and elsewhere.
Collapse
Affiliation(s)
- Elise Malard
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Elodie Pérès
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Laurent Chatre
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| |
Collapse
|
6
|
Barkley-Levenson AM, Lee A, Palmer AA. Genetic and Pharmacological Manipulations of Glyoxalase 1 Mediate Ethanol Withdrawal Seizure Susceptibility in Mice. Brain Sci 2021; 11:127. [PMID: 33478138 PMCID: PMC7835754 DOI: 10.3390/brainsci11010127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/03/2022] Open
Abstract
Central nervous system (CNS) hyperexcitability is a clinically significant feature of acute ethanol withdrawal. There is evidence for a genetic contribution to withdrawal severity, but specific genetic risk factors have not been identified. The gene glyoxalase 1 (Glo1) has been previously implicated in ethanol consumption in mice, and GLO1 inhibition can attenuate drinking in mice and rats. Here, we investigated whether genetic and pharmacological manipulations of GLO1 activity can also mediate ethanol withdrawal seizure severity in mice. Mice from two transgenic lines overexpressing Glo1 on different genetic backgrounds (C57BL/6J (B6) and FVB/NJ (FVB)) were tested for handling-induced convulsions (HICs) as a measure of acute ethanol withdrawal. Following an injection of 4 g/kg alcohol, both B6 and FVB mice overexpressing Glo1 showed increases in HICs compared to wild-type littermates, though only the FVB line showed a statistically significant difference. We also administered daily ethanol injections (2 g/kg + 9 mg/kg 4-methylpyrazole) to wild-type B6 mice for 10 days and tested them for HICs on the 10th day following treatment with either a vehicle or a GLO1 inhibitor (S-bromobenzylglutathione cyclopentyl diester (pBBG)). Treatment with pBBG reduced HICs, although this effect was only statistically significant following two 10-day cycles of ethanol exposure and withdrawal. These results provide converging genetic and pharmacological evidence that GLO1 can mediate ethanol withdrawal seizure susceptibility.
Collapse
Affiliation(s)
- Amanda M. Barkley-Levenson
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (A.L.); (A.A.P.)
| | - Amy Lee
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (A.L.); (A.A.P.)
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA; (A.L.); (A.A.P.)
- Institute for Genomic Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Wang K, Li N, Xu M, Huang M, Huang F. Glyoxalase 1 Inhibitor Alleviates Autism-like Phenotype in a Prenatal Valproic Acid-Induced Mouse Model. ACS Chem Neurosci 2020; 11:3786-3792. [PMID: 33166134 DOI: 10.1021/acschemneuro.0c00482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is a severe neurological and developmental disorder that impairs a person's ability to socialize and communicate and affects behavior. The number of patients diagnosed with ASD has risen rapidly. However, the pathophysiology of ASD is poorly understood, and drugs for ASD treatment are strikingly limited. This study aims to evaluate the roles of glyoxalase 1 (GLO1)-methylglyoxal (MG)-γ-aminobutyric acid (GABA) signaling in ASD using a valproic acid (VPA)-induced animal model of autism. The GLO1 levels were analyzed by RT-qPCR and Western blot assay, and MG levels were measured with a Methylglyoxal Assay Kit. The open-field and sniff duration tests were used to assess the interest and anxiety of VPA mice. The three-chamber, marble-burying, and tail-flick tests were applied to determine the sociability, repetitive behavior, and nociceptive threshold of VPA mice. Our results demonstrated that increased GLO1 and decreased MG were observed in VPA mice. Administration of S-p-bromobenzylglutathione cyclopentyl diester (BrBzGCp2), a GLO1 inhibitor, was beneficial for alleviating anxiety, reducing repetitive behavior, and improving the impaired sociability and nociceptive threshold of VPA mice. BrBzGCp2 treatment may be developed as a promising therapeutic strategy for patients with ASD.
Collapse
Affiliation(s)
- Kui Wang
- Psychiatric Ward, Qingdao Mental Health Center, Qingdao University, No 299 Nanjing Road, Qingdao, 266034 Shandong, China
| | - Na Li
- Psychiatric Ward, Qingdao Mental Health Center, Qingdao University, No 299 Nanjing Road, Qingdao, 266034 Shandong, China
| | - Min Xu
- Psychiatric Ward, Qingdao Mental Health Center, Qingdao University, No 299 Nanjing Road, Qingdao, 266034 Shandong, China
| | - Meng Huang
- Department of Laboratory Medicine, Lao-shan Disease Area, the Affiliated Hospital of Qingdao University, Qingdao, 266000 Shandong, China
| | - Fei Huang
- Psychiatric Ward, Qingdao Mental Health Center, Qingdao University, No 299 Nanjing Road, Qingdao, 266034 Shandong, China
| |
Collapse
|
8
|
Ruan QT, Yazdani N, Reed ER, Beierle JA, Peterson LP, Luttik KP, Szumlinski KK, Johnson WE, Ash PEA, Wolozin B, Bryant CD. 5' UTR variants in the quantitative trait gene Hnrnph1 support reduced 5' UTR usage and hnRNP H protein as a molecular mechanism underlying reduced methamphetamine sensitivity. FASEB J 2020; 34:9223-9244. [PMID: 32401417 DOI: 10.1096/fj.202000092r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/17/2020] [Accepted: 04/24/2020] [Indexed: 12/20/2022]
Abstract
We previously identified a 210 kb region on chromosome 11 (50.37-50.58 Mb, mm10) containing two protein-coding genes (Hnrnph1, Rufy1) that was necessary for reduced methamphetamine-induced locomotor activity in C57BL/6J congenic mice harboring DBA/2J polymorphisms. Gene editing of a small deletion in the first coding exon supported Hnrnph1 as a quantitative trait gene. We have since shown that Hnrnph1 mutants also exhibit reduced methamphetamine-induced reward, reinforcement, and dopamine release. However, the quantitative trait variants (QTVs) that modulate Hnrnph1 function at the molecular level are not known. Nine single nucleotide polymorphisms and seven indels distinguish C57BL/6J from DBA/2J within Hnrnph1, including four variants within the 5' untranslated region (UTR). Here, we show that a 114 kb introgressed region containing Hnrnph1 and Rufy1 was sufficient to cause a decrease in MA-induced locomotor activity. Gene-level transcriptome analysis of striatal tissue from 114 kb congenics vs Hnrnph1 mutants identified a nearly perfect correlation of fold-change in expression for those differentially expressed genes that were common to both mouse lines, indicating functionally similar effects on the transcriptome and behavior. Exon-level analysis (including noncoding exons) revealed decreased 5' UTR usage of Hnrnph1 and immunoblot analysis identified a corresponding decrease in hnRNP H protein in 114 kb congenic mice. Molecular cloning of the Hnrnph1 5' UTR containing all four variants (but none of them individually) upstream of a reporter induced a decrease in reporter signal in both HEK293 and N2a cells, thus, identifying a set of QTVs underlying molecular regulation of Hnrnph1.
Collapse
Affiliation(s)
- Qiu T Ruan
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.,Biomolecular Pharmacology Training Program, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Transformative Training Program in Addiction Science, Boston University School of Medicine, Boston, MA, USA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.,Biomolecular Pharmacology Training Program, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Transformative Training Program in Addiction Science, Boston University School of Medicine, Boston, MA, USA
| | - Eric R Reed
- Ph.D. Program in Bioinformatics, Boston University, Boston, MA, USA
| | - Jacob A Beierle
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.,Biomolecular Pharmacology Training Program, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Transformative Training Program in Addiction Science, Boston University School of Medicine, Boston, MA, USA
| | - Lucy P Peterson
- Biomolecular Pharmacology Training Program, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Kimberly P Luttik
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, Molecular, Cellular and Developmental Biology, Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - William E Johnson
- Department of Medicine, Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Peter E A Ash
- Laboratory of Neurodegeneration, Department of Pharmacology and Experimental Therapeutics and Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Benjamin Wolozin
- Laboratory of Neurodegeneration, Department of Pharmacology and Experimental Therapeutics and Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.,Biomolecular Pharmacology Training Program, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Transformative Training Program in Addiction Science, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Nirwan N, Siraj F, Vohora D. Inverted-U response of lacosamide on pilocarpine-induced status epilepticus and oxidative stress in C57BL/6 mice is independent of hippocampal collapsin response mediator protein-2. Epilepsy Res 2018; 145:93-101. [PMID: 29935443 DOI: 10.1016/j.eplepsyres.2018.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/09/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Currently, lacosamide (LCM) is not approved for use in status epilepticus (SE) but several shreds of evidence are available to support its use. The present study was, therefore, undertaken to evaluate the effect of LCM on pilocarpine (PILO) induced SE and neurodegeneration in C57BL/6 mice and to ascertain the involvement of CRMP-2 in mediating above effect. METHODS Pilocarpine-induced SE model was developed to explore the effect of LCM 20, 40 and 80 mg/kg in mice. We assessed the seizure severity, seizure latency, spontaneous alternation behavior (SAB) and motor coordination by behavioral observation. Histopathological evaluation and measurement of the levels of CRMP-2, reduced glutathione (GSH) and malondialdehyde (MDA) were carried out in mice hippocampus. RESULTS LCM exhibited a biphasic effect i.e., protection against SE at 20 mg/kg and 40 mg/kg dose whilst aggravated seizure-like behavior and mortality at 80 mg/kg. Further, it increased percentage alternation (i.e., restored spatial memory) in SAB and elevated motor impairment with increasing dose. Histologically, LCM 20 mg/kg and 40 mg/kg (but not 80 mg/kg) reduced neurodegeneration. LCM 20 mg/kg and 40 mg/kg reversed the elevated MDA and GSH levels while 80 mg/kg showed a tendency to increase oxidative stress. In contrast, LCM (at all doses) reversed the pilocarpine-induced elevation of collapsin response mediator protein-2 (CRMP-2). CONCLUSION LCM protected against pilocarpine-induced SE, associated neurodegeneration and improved pilocarpine-associated impairment of spatial memory. The study reveals that CRMP-2 may not be mediating the inverted-U-response of LCM at least in pilocarpine model. Therefore, the anti-oxidant effect of LCM (and not its ability to modulate CRMP-2) was anticipated as the mechanism underlying neuroprotection.
Collapse
Affiliation(s)
- Nikita Nirwan
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Fouzia Siraj
- National Institute of Pathology (ICMR), Safdarjang Hospital Campus, New Delhi 110029, India
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
10
|
Barkley-Levenson AM, Lagarda FA, Palmer AA. Glyoxalase 1 (GLO1) Inhibition or Genetic Overexpression Does Not Alter Ethanol's Locomotor Effects: Implications for GLO1 as a Therapeutic Target in Alcohol Use Disorders. Alcohol Clin Exp Res 2018. [PMID: 29532486 DOI: 10.1111/acer.13623] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Glyoxalase 1 (GLO1) is an enzyme that metabolizes methylglyoxal (MG), which is a competitive partial agonist at GABAA receptors. Inhibition of GLO1 increases concentrations of MG in the brain and decreases binge-like ethanol (EtOH) drinking. This study assessed whether inhibition of GLO1, or genetic overexpression of Glo1, would also alter the locomotor effects of EtOH, which might explain reduced EtOH consumption following GLO1 inhibition. We used the prototypical GABAA receptor agonist muscimol as a positive control. METHODS Male C57BL/6J mice were pretreated with either the GLO1 inhibitor S-bromobenzylglutathione cyclopentyl diester (pBBG; 7.5 mg/kg; Experiment 1) or muscimol (0.75 mg/kg; Experiment 2), or their corresponding vehicle. We then determined whether locomotor response to a range of EtOH doses (0, 0.5, 1.0, 1.5, 2.0, and 2.5) was altered by either pBBG or muscimol pretreatment. We also examined the locomotor response to a range of EtOH doses in FVB/NJ wild-type and transgenic Glo1 overexpressing mice (Experiment 3). Anxiety-like behavior (time spent in the center of the open field) was assessed in all 3 experiments. RESULTS The EtOH dose-response curve was not altered by pretreatment with pBBG or by transgenic overexpression of Glo1. In contrast, muscimol blunted locomotor stimulation at low EtOH doses and potentiated locomotor sedation at higher EtOH doses. No drug or genotype differences were seen in anxiety-like behavior after EtOH treatment. CONCLUSIONS The dose of pBBG used in this study is within the effective range shown previously to reduce EtOH drinking. Glo1 overexpression has been previously shown to increase EtOH drinking. However, neither manipulation altered the dose-response curve for EtOH's locomotor effects, whereas muscimol appeared to enhance the locomotor sedative effects of EtOH. The present data demonstrate that reduced EtOH drinking caused by GLO1 inhibition is not due to potentiation of EtOH's stimulant or depressant effects.
Collapse
Affiliation(s)
| | - Frances A Lagarda
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, California.,Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
11
|
Tao H, Zhou X, Zhao B, Li K. Conflicting Effects of Methylglyoxal and Potential Significance of miRNAs for Seizure Treatment. Front Mol Neurosci 2018; 11:70. [PMID: 29556176 PMCID: PMC5845011 DOI: 10.3389/fnmol.2018.00070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/19/2018] [Indexed: 01/09/2023] Open
Abstract
According to an update from the World Health Organization, approximately 50 million people worldwide suffer from epilepsy, and nearly one-third of these individuals are resistant to the currently available antiepileptic drugs, which has resulted in an insistent pursuit of novel strategies for seizure treatment. Recently, methylglyoxal (MG) was demonstrated to serve as a partial agonist of the gamma-aminobutyric acid type A (GABAA) receptor and to play an inhibitory role in epileptic activities. However, MG is also a substrate in the generation of advanced glycation end products (AGEs) that function by activating the receptor of AGEs (RAGE). The AGE/RAGE axis is responsible for the transduction of inflammatory cascades and appears to be an adverse pathway in epilepsy. This study systematically reviewed the significance of GABAergic MG, glyoxalase I (GLO1; responsible for enzymatic catalysis of MG cleavage) and downstream RAGE signaling in epilepsy. This work also discussed the potential of miRNAs that target multiple mRNAs and introduced a preliminary scheme for screening and validating miRNA candidates with the goal of reconciling the conflicting effects of MG for the future development of seizure treatments.
Collapse
Affiliation(s)
- Hua Tao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xu Zhou
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Keshen Li
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Stroke Center, Neurology & Neurosurgery Division, Clinical Medicine Research Institute & the First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
12
|
McMurray KMJ, Ramaker MJ, Barkley-Levenson AM, Sidhu PS, Elkin P, Reddy MK, Guthrie ML, Cook JM, Rawal VH, Arnold LA, Dulawa SC, Palmer AA. Identification of a novel, fast-acting GABAergic antidepressant. Mol Psychiatry 2018; 23:384-391. [PMID: 28322281 PMCID: PMC5608625 DOI: 10.1038/mp.2017.14] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 12/07/2016] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
Current pharmacotherapies for depression exhibit slow onset, side effects and limited efficacy. Therefore, identification of novel fast-onset antidepressants is desirable. GLO1 is a ubiquitous cellular enzyme responsible for the detoxification of the glycolytic byproduct methylglyoxal (MG). We have previously shown that MG is a competitive partial agonist at GABA-A receptors. We examined the effects of genetic and pharmacological inhibition of GLO1 in two antidepressant assay models: the tail suspension test (TST) and the forced swim test (FST). We also examined the effects of GLO1 inhibition in three models of antidepressant onset: the chronic FST (cFST), chronic mild stress (CMS) paradigm and olfactory bulbectomy (OBX). Genetic knockdown of Glo1 or pharmacological inhibition using two structurally distinct GLO1 inhibitors (S-bromobenzylglutathione cyclopentyl diester (pBBG) or methyl-gerfelin (MeGFN)) reduced immobility in the TST and acute FST. Both GLO1 inhibitors also reduced immobility in the cFST after 5 days of treatment. In contrast, the serotonin reuptake inhibitor fluoxetine (FLX) reduced immobility after 14, but not 5 days of treatment. Furthermore, 5 days of treatment with either GLO1 inhibitor blocked the depression-like effects induced by CMS on the FST and coat state, and attenuated OBX-induced locomotor hyperactivity. Finally, 5 days of treatment with a GLO1 inhibitor (pBBG), but not FLX, induced molecular markers of the antidepressant response including brain-derived neurotrophic factor (BDNF) induction and increased phosphorylated cyclic-AMP response-binding protein (pCREB) to CREB ratio in the hippocampus and medial prefrontal cortex (mPFC). Our findings indicate that GLO1 inhibitors may provide a novel and fast-acting pharmacotherapy for depression.
Collapse
Affiliation(s)
- Katherine M. J. McMurray
- Committee on Neurobiology, University of Chicago, Chicago IL 60637, USA,Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Marcia J. Ramaker
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA
| | - Amanda M. Barkley-Levenson
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Preetpal S. Sidhu
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Pavel Elkin
- Department of Chemistry, University of Chicago, Chicago IL 60637, USA
| | - M. Kashi Reddy
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Margaret L. Guthrie
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - James M. Cook
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Viresh H. Rawal
- Department of Chemistry, University of Chicago, Chicago IL 60637, USA
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Stephanie C. Dulawa
- Committee on Neurobiology, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Abraham A. Palmer
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA,Institute for Genome Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA,Corresponding Author: Abraham A. Palmer, Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093,
| |
Collapse
|
13
|
Antognelli C, Trapani E, Delle Monache S, Perrelli A, Daga M, Pizzimenti S, Barrera G, Cassoni P, Angelucci A, Trabalzini L, Talesa VN, Goitre L, Retta SF. KRIT1 loss-of-function induces a chronic Nrf2-mediated adaptive homeostasis that sensitizes cells to oxidative stress: Implication for Cerebral Cavernous Malformation disease. Free Radic Biol Med 2018; 115:202-218. [PMID: 29170092 PMCID: PMC5806631 DOI: 10.1016/j.freeradbiomed.2017.11.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 10/18/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
KRIT1 (CCM1) is a disease gene responsible for Cerebral Cavernous Malformations (CCM), a major cerebrovascular disease of proven genetic origin affecting 0.3-0.5% of the population. Previously, we demonstrated that KRIT1 loss-of-function is associated with altered redox homeostasis and abnormal activation of the redox-sensitive transcription factor c-Jun, which collectively result in pro-oxidative, pro-inflammatory and pro-angiogenic effects, suggesting a novel pathogenic mechanism for CCM disease and raising the possibility that KRIT1 loss-of-function exerts pleiotropic effects on multiple redox-sensitive mechanisms. To address this possibility, we investigated major redox-sensitive pathways and enzymatic systems that play critical roles in fundamental cytoprotective mechanisms of adaptive responses to oxidative stress, including the master Nrf2 antioxidant defense pathway and its downstream target Glyoxalase 1 (Glo1), a pivotal stress-responsive defense enzyme involved in cellular protection against glycative and oxidative stress through the metabolism of methylglyoxal (MG). This is a potent post-translational protein modifier that may either contribute to increased oxidative molecular damage and cellular susceptibility to apoptosis, or enhance the activity of major apoptosis-protective proteins, including heat shock proteins (Hsps), promoting cell survival. Experimental outcomes showed that KRIT1 loss-of-function induces a redox-sensitive sustained upregulation of Nrf2 and Glo1, and a drop in intracellular levels of MG-modified Hsp70 and Hsp27 proteins, leading to a chronic adaptive redox homeostasis that counteracts intrinsic oxidative stress but increases susceptibility to oxidative DNA damage and apoptosis, sensitizing cells to further oxidative challenges. While supporting and extending the pleiotropic functions of KRIT1, these findings shed new light on the mechanistic relationship between KRIT1 loss-of-function and enhanced cell predisposition to oxidative damage, thus providing valuable new insights into CCM pathogenesis and novel options for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
| | - Eliana Trapani
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Science, University of L'Aquila, Italy
| | - Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Torino, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Science, University of L'Aquila, Italy
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy
| | | | - Luca Goitre
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy.
| |
Collapse
|
14
|
López-Díez R, Shen X, Daffu G, Khursheed M, Hu J, Song F, Rosario R, Xu Y, Li Q, Xi X, Zou YS, Li H, Schmidt AM, Yan SF. Ager Deletion Enhances Ischemic Muscle Inflammation, Angiogenesis, and Blood Flow Recovery in Diabetic Mice. Arterioscler Thromb Vasc Biol 2017. [PMID: 28642238 DOI: 10.1161/atvbaha.117.309714] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Diabetic subjects are at higher risk of ischemic peripheral vascular disease. We tested the hypothesis that advanced glycation end products (AGEs) and their receptor (RAGE) block angiogenesis and blood flow recovery after hindlimb ischemia induced by femoral artery ligation through modulation of immune/inflammatory mechanisms. APPROACH AND RESULTS Wild-type mice rendered diabetic with streptozotocin and subjected to unilateral femoral artery ligation displayed increased accumulation and expression of AGEs and RAGE in ischemic muscle. In diabetic wild-type mice, femoral artery ligation attenuated angiogenesis and impaired blood flow recovery, in parallel with reduced macrophage content in ischemic muscle and suppression of early inflammatory gene expression, including Ccl2 (chemokine [C-C motif] ligand-2) and Egr1 (early growth response gene-1) versus nondiabetic mice. Deletion of Ager (gene encoding RAGE) or transgenic expression of Glo1 (reduces AGEs) restored adaptive inflammation, angiogenesis, and blood flow recovery in diabetic mice. In diabetes mellitus, deletion of Ager increased circulating Ly6Chi monocytes and augmented macrophage infiltration into ischemic muscle tissue after femoral artery ligation. In vitro, macrophages grown in high glucose display inflammation that is skewed to expression of tissue damage versus tissue repair gene expression. Further, macrophages grown in high versus low glucose demonstrate blunted macrophage-endothelial cell interactions. In both settings, these adverse effects of high glucose were reversed by Ager deletion in macrophages. CONCLUSIONS These findings indicate that RAGE attenuates adaptive inflammation in hindlimb ischemia; underscore microenvironment-specific functions for RAGE in inflammation in tissue repair versus damage; and illustrate that AGE/RAGE antagonism may fill a critical gap in diabetic peripheral vascular disease.
Collapse
Affiliation(s)
- Raquel López-Díez
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Xiaoping Shen
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Gurdip Daffu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Md Khursheed
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Jiyuan Hu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Fei Song
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Rosa Rosario
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Yunlu Xu
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Qing Li
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Xiangmei Xi
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Yu Shan Zou
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Huilin Li
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Ann Marie Schmidt
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York
| | - Shi Fang Yan
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine (R.L.D., X.S., G.D., M.K., F.S., R.R., Y.X., Q.L., X.X., Y.S.Z., A.M.S., S.F.Y.), Department of Population Health (J.H., H.L.), and Department of Environmental Science (H.L.), New York University School of Medicine, New York.
| |
Collapse
|
15
|
Jang S, Kwon DM, Kwon K, Park C. Generation and characterization of mouse knockout for glyoxalase 1. Biochem Biophys Res Commun 2017. [PMID: 28623132 DOI: 10.1016/j.bbrc.2017.06.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glyoxalase 1 (Glo1) is the first enzyme involved in glutathione-dependent detoxification of methylglyoxal, eventually generating d-lactate by the second enzyme glyoxalase 2 (Glo2). An accumulation of intracellular glyoxal and methylglyoxal leads to protein malfunction and mutation via formation of the advanced glycation end products (AGEs). Studies on mouse behavior suggest that methylglyoxal has anxiolytic properties. In this report, we generated and characterized a mouse knockout for Glo1. The knockout mice were viable without a pronounced phenotypic defect. Increased level of AGEs in Glo1 knockout mice was detected by immunoblotting with anti-MGH1 in liver homogenate, but not in brain. Alterations in behavior were observed in open field, light-dark transition, and tail suspension test. Open field data indicate increased exploration for novel environment and entry/stay in center zone in Glo1 knockout mice. In addition, increased light-dark transition and immobility was observed in the knockout mice. These data indicate that Glo1 knockout reduces anxiety-like behavior, but increases depression-like behavior.
Collapse
Affiliation(s)
- Sumi Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291, Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - David Min Kwon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291, Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Kyu Kwon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291, Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Chankyu Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291, Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
16
|
McMurray KMJ, Sidhu PS, Cook JM, Arnold LA, Palmer AA. Genetic and pharmacological manipulation of glyoxalase 1 regulates voluntary ethanol consumption in mice. Addict Biol 2017; 22:381-389. [PMID: 26691867 DOI: 10.1111/adb.12333] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/14/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022]
Abstract
Previous studies have identified an association between the gene glyoxalase 1 (Glo1) and anxiety-like behavior in mice and have shown that the substrate of GLO1, methylglyoxal, is a competitive partial agonist at GABAA receptors. Given the well-established role of GABAA receptors in the behavioral effects of ethanol (EtOH), we investigated the role of Glo1 in voluntary EtOH consumption in mice using the drinking in the dark (DID) paradigm. Transgenic mice overexpressing Glo1 on both FVB/NJ (FVB) or C57BL/6J (B6) backgrounds showed increased voluntary EtOH consumption compared to their wild-type littermates in DID. Furthermore, transgenic Glo1 knockdown mice on a B6 background showed decreased voluntary EtOH consumption in DID. These genetic manipulations of Glo1 had no effect on sucrose, saccharin or water consumption. Finally, we found that a small molecule GLO1 inhibitor (S-bromobenzylglutathione cyclopentyl diester (pBBG; 6.25, 12.5 mg/kg)) reduced EtOH consumption compared to vehicle treated B6 mice without altering saccharin or water consumption. Sucrose consumption was only reduced by the higher (12.5 mg/kg) dose of pBBG. We did not observe differences in the loss of righting reflex (LORR) or EtOH-induced foot slips on the balance beam in response to acute EtOH administration (LORR: 4 g/kg, Balance Beam: 1.25 g/kg) in B6 or FVB mice overexpressing Glo1, nor in B6 mice treated with pBBG. These data are the first to implicate Glo1 in EtOH-related behaviors and suggest that GLO1 inhibitors may have therapeutic potential for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Katherine M. J. McMurray
- Committee on Neurobiology; University of Chicago; Chicago IL 60637 USA
- Department of Human Genetics; University of Chicago; Chicago IL 60637 USA
| | - Preetpal S. Sidhu
- Department of Chemistry and Biochemistry; University of Wisconsin-Milwaukee and the Milwaukee Institute for Drug Discovery; Milwaukee WI USA
| | - James M. Cook
- Department of Chemistry and Biochemistry; University of Wisconsin-Milwaukee and the Milwaukee Institute for Drug Discovery; Milwaukee WI USA
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry; University of Wisconsin-Milwaukee and the Milwaukee Institute for Drug Discovery; Milwaukee WI USA
| | - Abraham A. Palmer
- Department of Human Genetics; University of Chicago; Chicago IL 60637 USA
- Department of Psychiatry and Behavioral Neuroscience; University of Chicago; Chicago IL 60637 USA
- Department of Psychiatry; University of Californian San Diego; La Jolla CA 92093 USA
| |
Collapse
|
17
|
Alomar F, Singh J, Jang H, Rozanzki GJ, Shao CH, Padanilam BJ, Mayhan WG, Bidasee KR. Smooth muscle-generated methylglyoxal impairs endothelial cell-mediated vasodilatation of cerebral microvessels in type 1 diabetic rats. Br J Pharmacol 2016; 173:3307-3326. [PMID: 27611446 PMCID: PMC5738666 DOI: 10.1111/bph.13617] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/26/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelial cell-mediated vasodilatation of cerebral arterioles is impaired in individuals with Type 1 diabetes (T1D). This defect compromises haemodynamics and can lead to hypoxia, microbleeds, inflammation and exaggerated ischaemia-reperfusion injuries. The molecular causes for dysregulation of cerebral microvascular endothelial cells (cECs) in T1D remains poorly defined. This study tests the hypothesis that cECs dysregulation in T1D is triggered by increased generation of the mitochondrial toxin, methylglyoxal, by smooth muscle cells in cerebral arterioles (cSMCs). EXPERIMENTAL APPROACH Endothelial cell-mediated vasodilatation, vascular transcytosis inflammation, hypoxia and ischaemia-reperfusion injury were assessed in brains of male Sprague-Dawley rats with streptozotocin-induced diabetes and compared with those in diabetic rats with increased expression of methylglyoxal-degrading enzyme glyoxalase-I (Glo-I) in cSMCs. KEY RESULTS After 7-8 weeks of T1D, endothelial cell-mediated vasodilatation of cerebral arterioles was impaired. Microvascular leakage, gliosis, macrophage/neutrophil infiltration, NF-κB activity and TNF-α levels were increased, and density of perfused microvessels was reduced. Transient occlusion of a mid-cerebral artery exacerbated ischaemia-reperfusion injury. In cSMCs, Glo-I protein was decreased, and the methylglyoxal-synthesizing enzyme, vascular adhesion protein 1 (VAP-1) and methylglyoxal were increased. Restoring Glo-I protein in cSMCs of diabetic rats to control levels via gene transfer, blunted VAP-1 and methylglyoxal increases, cECs dysfunction, microvascular leakage, inflammation, ischaemia-reperfusion injury and increased microvessel perfusion. CONCLUSIONS AND IMPLICATIONS Methylglyoxal generated by cSMCs induced cECs dysfunction, inflammation, hypoxia and exaggerated ischaemia-reperfusion injury in diabetic rats. Lowering methylglyoxal produced by cSMCs may be a viable therapeutic strategy to preserve cECs function and blunt deleterious downstream consequences in T1D.
Collapse
Affiliation(s)
- Fadhel Alomar
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNEUSA
- Department of PharmacologyUniversity of DammamDammamSaudi Arabia
| | - Jaipaul Singh
- School of Forensic and Applied ScienceUniversity of Central LancashirePrestonUK
| | - Hee‐Seong Jang
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - George J Rozanzki
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
- Nebraska Redox Biology CenterLincolnNEUSA
| | - Chun Hong Shao
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Babu J Padanilam
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - William G Mayhan
- Department of Basic Biomedical Sciences, Sanford School of MedicineUniversity of South DakotaVermillionSDUSA
| | - Keshore R Bidasee
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNEUSA
- Department of Environmental, Agricultural and Occupational HealthUniversity of Nebraska Medical CenterOmahaNEUSA
- Nebraska Redox Biology CenterLincolnNEUSA
| |
Collapse
|
18
|
De Giorgis V, Varesio C, Baldassari C, Piazza E, Olivotto S, Macasaet J, Balottin U, Veggiotti P. Atypical Manifestations in Glut1 Deficiency Syndrome. J Child Neurol 2016; 31:1174-80. [PMID: 27250207 DOI: 10.1177/0883073816650033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 03/27/2016] [Indexed: 11/17/2022]
Abstract
Glucose transporter type 1 deficiency syndrome is a genetically determined, treatable, neurologic disorder that is caused by an insufficient transport of glucose into the brain. It is caused by a mutation in the SCL2A1 gene, which is so far the only known to be associated with this condition. Glucose transporter type 1 deficiency syndrome consists of a wide clinical spectrum that usually presents with cognitive impairment, epilepsy, paroxysmal exercise-induced dyskinesia, acquired microcephaly, hemolytic anemia, gait disturbance, and dyspraxia in different combinations. However, there are other clinical manifestations that we consider equally peculiar but that have so far been poorly described in literature. In this review, supported by a video contribution, we will accurately describe this type of clinical manifestation such as oculogyric crises, weakness, paroxysmal kinesigenic and nonkinesigenic dyskinesia in order to provide an additional instrument for a correct, rapid diagnosis.
Collapse
Affiliation(s)
- V De Giorgis
- Brain and Behaviour Department, University of Pavia, Pavia, Italy
| | - C Varesio
- Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - C Baldassari
- Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - E Piazza
- Brain and Behaviour Department, University of Pavia, Pavia, Italy
| | - S Olivotto
- Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - J Macasaet
- Department of Neurosciences, Makati Medical Center, Manila, Philippines
| | - U Balottin
- Brain and Behaviour Department, University of Pavia, Pavia, Italy Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - P Veggiotti
- Brain and Behaviour Department, University of Pavia, Pavia, Italy Department of Child Neurology and Psychiatry, "C. Mondino" National Neurological Institute, Pavia, Italy
| |
Collapse
|
19
|
Hansen F, Pandolfo P, Galland F, Torres FV, Dutra MF, Batassini C, Guerra MC, Leite MC, Gonçalves CA. Methylglyoxal can mediate behavioral and neurochemical alterations in rat brain. Physiol Behav 2016; 164:93-101. [PMID: 27235733 DOI: 10.1016/j.physbeh.2016.05.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 12/17/2022]
Abstract
Diabetes is associated with loss of cognitive function and increased risk for Alzheimer's disease (AD). Advanced glycation end products (AGEs) are elevated in diabetes and AD and have been suggested to act as mediators of the cognitive decline observed in these pathologies. Methylglyoxal (MG) is an extremely reactive carbonyl compound that propagates glycation reactions and is, therefore, able to generate AGEs. Herein, we evaluated persistent behavioral and biochemical parameters to explore the hypothesis that elevated exogenous MG concentrations, induced by intracerebroventricular (ICV) infusion, lead to cognitive decline in Wistar rats. A high and sustained administration of MG (3μmol/μL; subdivided into 6days) was found to decrease the recognition index of rats, as evaluated by the object-recognition test. However, MG was unable to impair learning-memory processes, as shown by the habituation in the open field (OF) and Y-maze tasks. Moreover, a single high dose of MG induced persistent alterations in anxiety-related behavior, diminishing the anxiety-like parameters evaluated in the OF test. Importantly, MG did not alter locomotion behavior in the different tasks performed. Our biochemical findings support the hypothesis that MG induces persistent alterations in the hippocampus, but not in the cortex, related to glyoxalase 1 activity, AGEs content and glutamate uptake. Glial fibrillary acidic protein and S100B content, as well as S100B secretion (astroglial-related parameters of brain injury), were not altered by ICV MG administration. Taken together, our data suggest that MG interferes directly in brain function and that the time and the levels of exogenous MG determine the different features that can be seen in diabetic patients.
Collapse
Affiliation(s)
- Fernanda Hansen
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil.
| | - Pablo Pandolfo
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, 24020-141 Niterói, RJ, Brazil
| | - Fabiana Galland
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | - Felipe Vasconcelos Torres
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | - Márcio Ferreira Dutra
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Trindade, 88040-970 Florianópolis, SC, Brazil
| | - Cristiane Batassini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | - Maria Cristina Guerra
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, 90035-003 Porto Alegre, RS, Brazil
| |
Collapse
|
20
|
Tao H, Si L, Zhou X, Liu Z, Ma Z, Zhou H, Zhong W, Cui L, Zhang S, Li Y, Ma G, Zhao J, Huang W, Yao L, Xu Z, Zhao B, Li K. Role of glyoxalase I gene polymorphisms in late-onset epilepsy and drug-resistant epilepsy. J Neurol Sci 2016; 363:200-6. [PMID: 27000251 DOI: 10.1016/j.jns.2016.01.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/16/2016] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recent studies indicate that increased expression of glyoxalase I (GLO1) could result in epileptic seizures; thus, this study further explored the association of GLO1 with epilepsy from the perspective of molecular genetics. MATERIAL AND METHODS GLO1 single nucleotide polymorphisms (SNPs; rs1130534, rs4746 and rs1049346) were investigated in cohort I (the initial samples: 249 cases and 289 controls). A replication study designed to confirm the positive findings in cohort I was performed in cohorts II (the additional samples: 130 cases and 191 controls) and I+II. RESULTS In cohorts I, II and I+II, the CC genotype at rs1049346 T>C exerts a protective effect against both late-onset epilepsy (odds ratio [OR]=2.437, p=0.013; OR=2.844, p=0.008; OR=2.645, p=0.000, q=0.003, respectively) and drug-resistant epilepsy (DRE) (OR=2.985, p=0.020; OR=2.943, p=0.014; OR=3.049, p=0.001, q=0.006, respectively). Further analyses in cohort I+II indicate that the presence of the TAC/AAT haplotypes (rs1130534-rs4746-rs1049346) may be used as a marker of predisposition to/protection against DRE (p=0.002, q=0.010; p=0.000, q=0.002, respectively). CONCLUSIONS This study is the first to demonstrate that the GLO1 SNPs are significantly associated with epilepsy. In particular, the rs1049346 T>C SNPs are potentially useful for risk assessment of late-onset epilepsy and DRE.
Collapse
Affiliation(s)
- Hua Tao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Ligang Si
- Department of Pediatrics, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Xu Zhou
- Clinical Research Center, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhou Liu
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhonghua Ma
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Haihong Zhou
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Wangtao Zhong
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Lili Cui
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Shuyan Zhang
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - You Li
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Guoda Ma
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Jianghao Zhao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Wenhui Huang
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Lifen Yao
- Department of Pediatrics, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Zhien Xu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Bin Zhao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| | - Keshen Li
- Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| |
Collapse
|
21
|
Kazdoba TM, Hagerman RJ, Zolkowska D, Rogawski MA, Crawley JN. Evaluation of the neuroactive steroid ganaxolone on social and repetitive behaviors in the BTBR mouse model of autism. Psychopharmacology (Berl) 2016; 233:309-23. [PMID: 26525567 PMCID: PMC4703522 DOI: 10.1007/s00213-015-4115-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 10/06/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Abnormalities in excitatory/inhibitory neurotransmission are hypothesized to contribute to autism spectrum disorder (ASD) etiology. BTBR T (+) Itpr3 (tf) /J (BTBR), an inbred mouse strain, displays social deficits and repetitive self-grooming, offering face validity to ASD diagnostic symptoms. Reduced GABAergic neurotransmission in BTBR suggests that GABAA receptor positive allosteric modulators (PAMs) could improve ASD-relevant BTBR phenotypes. The neuroactive steroid ganaxolone acts as a PAM, displaying anticonvulsant properties in rodent epilepsy models and an anxiolytic-like profile in the elevated plus-maze. OBJECTIVES We evaluated ganaxolone in BTBR and C57BL/6J mice in standardized assays for sociability and repetitive behaviors. Open field and anxiety-related behaviors were tested as internal controls and for comparison with the existing neuroactive steroid literature. RESULTS Ganaxolone improved aspects of social approach and reciprocal social interactions in BTBR, with no effect on repetitive self-grooming, and no detrimental effects in C57BL/6J. Ganaxolone increased overall exploratory activity in BTBR and C57BL/6J in the open field, social approach, and elevated plus-maze, introducing a confound for the interpretation of social improvements. Allopregnanolone and diazepam similarly increased total entries in the elevated plus-maze, indicating that behavioral activation may be a general property of GABAA receptor PAMs in these strains. CONCLUSIONS Ganaxolone shows promise for improving sociability. In addition, ganaxolone, as well as other GABAA receptor PAMs, enhanced overall BTBR activity. The translational implications of specific sociability improvements and nonspecific behavioral activation by ganaxolone in the BTBR model remain to be determined. Future studies to explore whether PAMs provide a novel profile with unique benefits for ASD treatment will be worthwhile.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
| | - Randi J Hagerman
- MIND Institute, Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Dorota Zolkowska
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Michael A Rogawski
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| |
Collapse
|
22
|
Dafre AL, Goldberg J, Wang T, Spiegel DA, Maher P. Methylglyoxal, the foe and friend of glyoxalase and Trx/TrxR systems in HT22 nerve cells. Free Radic Biol Med 2015; 89:8-19. [PMID: 26165190 PMCID: PMC5624793 DOI: 10.1016/j.freeradbiomed.2015.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 06/03/2015] [Accepted: 07/06/2015] [Indexed: 01/28/2023]
Abstract
Methylglyoxal (MGO) is a major glycating agent that reacts with basic residues of proteins and promotes the formation of advanced glycation end products (AGEs) which are believed to play key roles in a number of pathologies, such as diabetes, Alzheimer's disease, and inflammation. Here, we examined the effects of MGO on immortalized mouse hippocampal HT22 nerve cells. The endpoints analyzed were MGO and thiol status, the glyoxalase system, comprising glyoxalase 1 and 2 (GLO1/2), and the cytosolic and mitochondrial Trx/TrxR systems, as well as nuclear Nrf2 and its target genes. We found that nuclear Nrf2 is induced by MGO treatment in HT22 cells, as corroborated by induction of the Nrf2-controlled target genes and proteins glutamate cysteine ligase and heme oxygenase 1. Nrf2 knockdown prevented MGO-dependent induction of glutamate cysteine ligase and heme oxygenase 1. The cystine/glutamate antiporter, system xc(-), which is also controlled by Nrf2, was also induced. The increased cystine import (system xc(-)) activity and GCL expression promoted GSH synthesis, leading to increased levels of GSH. The data indicate that MGO can act as both a foe and a friend of the glyoxalase and the Trx/TrxR systems. At low concentrations of MGO (0.3mM), GLO2 is strongly induced, but at high MGO (0.75 mM) concentrations, GLO1 is inhibited and GLO2 is downregulated. The cytosolic Trx/TrxR system is impaired by MGO, where Trx is downregulated yet TrxR is induced, but strong MGO-dependent glycation may explain the loss in TrxR activity. We propose that Nrf2 can be the unifying element to explain the observed upregulation of GSH, GCL, HO1, TrxR1, Trx2, TrxR2, and system xc(-) system activity.
Collapse
Affiliation(s)
- A L Dafre
- Biochemistry Department, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| | - J Goldberg
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - T Wang
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - D A Spiegel
- Department of Chemistry, Yale University, New Haven, CT 06520, USA
| | - P Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
23
|
Abstract
Many current pharmacological treatments for neuropsychiatric disorders, such as anxiety and depression, are limited by a delayed onset of therapeutic effect, adverse side effects, abuse potential or lack of efficacy in many patients. These off-target effects highlight the need to identify novel mechanisms and targets for treatment. Recently, modulation of Glo1 (glyoxalase I) activity was shown to regulate anxiety-like behaviour and seizure-susceptibility in mice. These effects are likely to be mediated through the regulation of MG (methylglyoxal) by Glo1, as MG acts as a competitive partial agonist at GABA(A) (γ-aminobutyric acid A) receptors. Thus modulation of MG by Glo1 represents a novel target for treatment. In the present article, we evaluate the therapeutic potential of indirectly modulating MG concentrations through Glo1 inhibitors for the treatment of neuropsychiatric disorders.
Collapse
|
24
|
Allaman I, Bélanger M, Magistretti PJ. Methylglyoxal, the dark side of glycolysis. Front Neurosci 2015; 9:23. [PMID: 25709564 PMCID: PMC4321437 DOI: 10.3389/fnins.2015.00023] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/13/2015] [Indexed: 12/05/2022] Open
Abstract
Glucose is the main energy substrate for the brain. There is now extensive evidence indicating that the metabolic profile of neural cells with regard to glucose utilization and glycolysis rate is not homogenous, with a marked propensity for glycolytic glucose processing in astrocytes compared to neurons. Methylglyoxal, a highly reactive dicarbonyl compound, is inevitably formed as a by-product of glycolysis. Methylglyoxal is a major cell-permeant precursor of advanced glycation end-products (AGEs), which are associated with several pathologies including diabetes, aging and neurodegenerative diseases. In normal situations, cells are protected against methylglyoxal toxicity by different mechanisms and in particular the glyoxalase system, which represents the most important pathway for the detoxification of methylglyoxal. While the neurotoxic effects of methylglyoxal and AGEs are well characterized, our understanding the glyoxalase system in the brain is more scattered. Considering the high energy requirements (i.e., glucose) of the brain, one should expect that the cerebral glyoxalase system is adequately fitted to handle methylglyoxal toxicity. This review focuses on our actual knowledge on the cellular aspects of the glyoxalase system in brain cells, in particular with regard to its activity in astrocytes and neurons. A main emerging concept is that these two neural cell types have different and energetically adapted glyoxalase defense mechanisms which may serve as protective mechanism against methylglyoxal-induced cellular damage.
Collapse
Affiliation(s)
- Igor Allaman
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Mireille Bélanger
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Pierre J Magistretti
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology Thuwal, Saudi Arabia ; Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| |
Collapse
|
25
|
Alter AS, Engelstad K, Hinton VJ, Montes J, Pearson TS, Akman CI, De Vivo DC. Long-term clinical course of Glut1 deficiency syndrome. J Child Neurol 2015; 30:160-9. [PMID: 24789115 DOI: 10.1177/0883073814531822] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our objective is to characterize the long-term course of Glut1 deficiency syndrome. Longitudinal outcome measures, including Columbia Neurological Scores, neuropsychological tests, and adaptive behavior reports, were collected for 13 participants with Glut1 deficiency syndrome who had been followed for an average of 14.2 (range = 8.9-23.6) years. A parent questionnaire assessed manifestations throughout development. The 6-Minute Walk Test captured gait disturbances and triggered paroxysmal exertional dyskinesia. All longitudinal outcomes remained stable over time. Epilepsy dominated infancy and improved during childhood. Dystonia emerged during childhood or adolescence. Earlier introduction of the ketogenic diet correlated with better long-term outcomes on some measures. Percent-predicted 6-Minute Walk Test distance correlated significantly with Columbia Neurological Scores. We conclude that Glut1 deficiency syndrome is a chronic condition, dominated by epilepsy in infancy and by movement disorders thereafter. Dietary treatment in the first postnatal months may effect improved outcomes, emphasizing the importance of early diagnosis and treatment.
Collapse
Affiliation(s)
- Aliza S Alter
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Veronica J Hinton
- Department of Neurology, Columbia University, New York, NY, USA Gertrude Sergievsky Center, Columbia University, New York, NY, USA
| | | | - Toni S Pearson
- Department of Neurology, Columbia University, New York, NY, USA
| | - Cigdem I Akman
- Department of Neurology, Columbia University, New York, NY, USA
| | | |
Collapse
|
26
|
Inhibition by active site directed covalent modification of human glyoxalase I. Bioorg Med Chem 2014; 22:3301-8. [DOI: 10.1016/j.bmc.2014.04.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/18/2014] [Accepted: 04/28/2014] [Indexed: 11/22/2022]
|
27
|
Makinson CD, Tanaka BS, Lamar T, Goldin AL, Escayg A. Role of the hippocampus in Nav1.6 (Scn8a) mediated seizure resistance. Neurobiol Dis 2014; 68:16-25. [PMID: 24704313 DOI: 10.1016/j.nbd.2014.03.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 10/25/2022] Open
Abstract
SCN1A mutations are the main cause of the epilepsy disorders Dravet syndrome (DS) and genetic epilepsy with febrile seizures plus (GEFS+). Mutations that reduce the activity of the mouse Scn8a gene, in contrast, are found to confer seizure resistance and extend the lifespan of mouse models of DS and GEFS+. To investigate the mechanism by which reduced Scn8a expression confers seizure resistance, we induced interictal-like burst discharges in hippocampal slices of heterozygous Scn8a null mice (Scn8a(med/+)) with elevated extracellular potassium. Scn8a(med/+) mutants exhibited reduced epileptiform burst discharge activity after P20, indicating an age-dependent increased threshold for induction of epileptiform discharges. Scn8a deficiency also reduced the occurrence of burst discharges in a GEFS+ mouse model (Scn1a(R1648H/+)). There was no detectable change in the expression levels of Scn1a (Nav1.1) or Scn2a (Nav1.2) in the hippocampus of adult Scn8a(med/+) mutants. To determine whether the increased seizure resistance associated with reduced Scn8a expression was due to alterations that occurred during development, we examined the effect of deleting Scn8a in adult mice. Global Cre-mediated deletion of a heterozygous floxed Scn8a allele in adult mice was found to increase thresholds to chemically and electrically induced seizures. Finally, knockdown of Scn8a gene expression in the adult hippocampus via lentiviral Cre injection resulted in a reduction in the number of EEG-confirmed seizures following the administration of picrotoxin. Our results identify the hippocampus as an important structure in the mediation of Scn8a-dependent seizure protection and suggest that selective targeting of Scn8a activity might be efficacious in patients with epilepsy.
Collapse
Affiliation(s)
| | - Brian S Tanaka
- Departments of Microbiology and Molecular Genetics and Anatomy and Neurobiology, University of California, Irvine, CA 92697
| | - Tyra Lamar
- Department of Human Genetics, Emory University, Atlanta, GA 30322
| | - Alan L Goldin
- Departments of Microbiology and Molecular Genetics and Anatomy and Neurobiology, University of California, Irvine, CA 92697
| | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, GA 30322
| |
Collapse
|
28
|
Hassan W, Silva CEB, Mohammadzai IU, da Rocha JBT, Landeira-Fernandez J. Association of oxidative stress to the genesis of anxiety: implications for possible therapeutic interventions. Curr Neuropharmacol 2014; 12:120-39. [PMID: 24669207 PMCID: PMC3964744 DOI: 10.2174/1570159x11666131120232135] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 06/16/2013] [Accepted: 11/02/2013] [Indexed: 12/27/2022] Open
Abstract
Oxidative stress caused by reactive species, including reactive oxygen species, reactive nitrogen species, and unbound, adventitious metal ions (e.g., iron [Fe] and copper [Cu]), is an underlying cause of various neurodegenerative diseases. These reactive species are an inevitable by-product of cellular respiration or other metabolic processes that may cause the oxidation of lipids, nucleic acids, and proteins. Oxidative stress has recently been implicated in depression and anxiety-related disorders. Furthermore, the manifestation of anxiety in numerous psychiatric disorders, such as generalized anxiety disorder, depressive disorder, panic disorder, phobia, obsessive-compulsive disorder, and posttraumatic stress disorder, highlights the importance of studying the underlying biology of these disorders to gain a better understanding of the disease and to identify common biomarkers for these disorders. Most recently, the expression of glutathione reductase 1 and glyoxalase 1, which are genes involved in antioxidative metabolism, were reported to be correlated with anxiety-related phenotypes. This review focuses on direct and indirect evidence of the potential involvement of oxidative stress in the genesis of anxiety and discusses different opinions that exist in this field. Antioxidant therapeutic strategies are also discussed, highlighting the importance of oxidative stress in the etiology, incidence, progression, and prevention of psychiatric disorders.
Collapse
Affiliation(s)
- Waseem Hassan
- Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Chemical Sciences, University of Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | - Imdad Ullah Mohammadzai
- Institute of Chemical Sciences, University of Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Joao Batista Teixeira da Rocha
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | |
Collapse
|
29
|
Mendes Arent A, de Souza LF, Walz R, Dafre AL. Perspectives on molecular biomarkers of oxidative stress and antioxidant strategies in traumatic brain injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:723060. [PMID: 24689052 PMCID: PMC3943200 DOI: 10.1155/2014/723060] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/04/2013] [Accepted: 12/09/2013] [Indexed: 11/23/2022]
Abstract
Traumatic brain injury (TBI) is frequently associated with abnormal blood-brain barrier function, resulting in the release of factors that can be used as molecular biomarkers of TBI, among them GFAP, UCH-L1, S100B, and NSE. Although many experimental studies have been conducted, clinical consolidation of these biomarkers is still needed to increase the predictive power and reduce the poor outcome of TBI. Interestingly, several of these TBI biomarkers are oxidatively modified to carbonyl groups, indicating that markers of oxidative stress could be of predictive value for the selection of therapeutic strategies. Some drugs such as corticosteroids and progesterone have already been investigated in TBI neuroprotection but failed to demonstrate clinical applicability in advanced phases of the studies. Dietary antioxidants, such as curcumin, resveratrol, and sulforaphane, have been shown to attenuate TBI-induced damage in preclinical studies. These dietary antioxidants can increase antioxidant defenses via transcriptional activation of NRF2 and are also known as carbonyl scavengers, two potential mechanisms for neuroprotection. This paper reviews the relevance of redox biology in TBI, highlighting perspectives for future studies.
Collapse
Affiliation(s)
- André Mendes Arent
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
- Faculty of Medicine, University of South Santa Catarina (Unisul), 88137-270 Palhoça, SC, Brazil
- Neurosurgery Service, São José Regional Hospital (HRSJ-HMG), 88103-901 São José, SC, Brazil
| | - Luiz Felipe de Souza
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
| | - Roger Walz
- Applied Neurosciences Centre (CeNAp) and Department of Medical Clinics, University Hospital, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Alcir Luiz Dafre
- Department of Biochemistry, Federal University of Santa Catarina, Biological Sciences Centre, 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|