51
|
Choi Y, Shin T. Alendronate Enhances Functional Recovery after Spinal Cord Injury. Exp Neurobiol 2022; 31:54-64. [PMID: 35256544 PMCID: PMC8907254 DOI: 10.5607/en21030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury is a destructive disease characterized by motor/sensory dysfunction and severe inflammation. Alendronate is an anti-inflammatory molecule and may therefore be of benefit in the treatment of the inflammation associated with spinal cord injury. This study aimed to evaluate whether alendronate attenuates motor/sensory dysfunction and the inflammatory response in a thoracic spinal cord clip injury model. Alendronate was intraperitoneally administered at 1 mg/kg/day or 5 mg/kg/day from day (D) 0 to 28 post-injury (PI). The histopathological evaluation showed an alleviation of the inflammatory response, including the infiltration of inflammatory cells, and a decrease in gliosis. Alendronate also led to reductions in the levels of inflammation-related molecules, including mitogen-activated protein kinase, p53, pro-inflammatory cytokines, and pro-inflammatory mediators. Neuro-behavioral assessments, including the Basso, Beattie, and Bresnahan scale for locomotor function, the von Frey filament test, the hot plate test, and the cold stimulation test for sensory function, and the horizontal ladder test for sensorimotor function improved significantly in the alendronate-treated group at D28PI. Taken together, these results suggest that alendronate treatment can inhibit the inflammatory response in spinal cord injury thus improving functional responses.
Collapse
Affiliation(s)
- Yuna Choi
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
52
|
Probing Cell Redox State and Glutathione-Modulating Factors Using a Monochlorobimane-Based Microplate Assay. Antioxidants (Basel) 2022; 11:antiox11020391. [PMID: 35204274 PMCID: PMC8869332 DOI: 10.3390/antiox11020391] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 12/10/2022] Open
Abstract
Thiol compounds including predominantly glutathione (GSH) are key components of redox homeostasis, which are involved in the protection and regulation of mammalian cells. The assessment of cell redox status by means of in situ analysis of GSH in living cells is often preferable over established assays in cell lysates due to fluctuations of the GSH pool. For this purpose, we propose a microplate assay with monochlorobimane (MCB) as an available fluorescent probe for GSH, although poorly detected in the microplate format. In addition to the new procedure for improved MCB-assisted GSH detection in plate-grown cells and its verification with GSH modulators, this study provides a useful methodology for the evaluation of cell redox status probed through relative GSH content and responsiveness to both supplemented thiols and variation in oxygen pressure. The roles of extracellular interactions of thiols and natural variability of cellular glutathione on the assay performance were emphasized and discussed. The results are of broad interest in cell biology research and should be particularly useful for the characterization of pathological cells with decreased GSH status and increased oxidative status as well as redox-modulating factors.
Collapse
|
53
|
Iskusnykh IY, Zakharova AA, Pathak D. Glutathione in Brain Disorders and Aging. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27010324. [PMID: 35011559 PMCID: PMC8746815 DOI: 10.3390/molecules27010324] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/06/2023]
Abstract
Glutathione is a remarkably functional molecule with diverse features, which include being an antioxidant, a regulator of DNA synthesis and repair, a protector of thiol groups in proteins, a stabilizer of cell membranes, and a detoxifier of xenobiotics. Glutathione exists in two states—oxidized and reduced. Under normal physiological conditions of cellular homeostasis, glutathione remains primarily in its reduced form. However, many metabolic pathways involve oxidization of glutathione, resulting in an imbalance in cellular homeostasis. Impairment of glutathione function in the brain is linked to loss of neurons during the aging process or as the result of neurological diseases such as Huntington’s disease, Parkinson’s disease, stroke, and Alzheimer’s disease. The exact mechanisms through which glutathione regulates brain metabolism are not well understood. In this review, we will highlight the common signaling cascades that regulate glutathione in neurons and glia, its functions as a neuronal regulator in homeostasis and metabolism, and finally a mechanistic recapitulation of glutathione signaling. Together, these will put glutathione’s role in normal aging and neurological disorders development into perspective.
Collapse
Affiliation(s)
- Igor Y. Iskusnykh
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: authors: (I.Y.I.); (D.P.)
| | - Anastasia A. Zakharova
- Department of Medical Biochemistry, Faculty of Biomedicine, Pirogov Russian National Research Medical University, Ostrovitianov St. 1, 117997 Moscow, Russia;
| | - Dhruba Pathak
- Department of Psychology, Temple University, Philadelphia, PA 19122, USA
- Correspondence: authors: (I.Y.I.); (D.P.)
| |
Collapse
|
54
|
Chavda V, Singh K, Patel V, Mishra M, Mishra AK. Neuronal Glial Crosstalk: Specific and Shared Mechanisms in Alzheimer’s Disease. Brain Sci 2022; 12:brainsci12010075. [PMID: 35053818 PMCID: PMC8773743 DOI: 10.3390/brainsci12010075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The human brain maintains billions of neurons functional across the lifespan of the individual. The glial, supportive cells of the brain are indispensable to neuron elasticity. They undergo various states (active, reactive, macrophage, primed, resting) and carefully impose either quick repair or the cleaning of injured neurons to avoid damage extension. Identifying the failure of these interactions involving the relation of the input of glial cells to the inception and/or progression of chronic neurodegenerative diseases (ND) is crucial in identifying therapeutic options, given the well-built neuro-immune module of these diseases. In the present review, we scrutinize different interactions and important factors including direct cell–cell contact, intervention by the CD200 system, various receptors present on their surfaces, CXC3RI and TREM2, and chemokines and cytokines with special reference to Alzheimer’s disease (AD). The present review of the available literature will elucidate the contribution of microglia and astrocytes to the pathophysiology of AD, thus evidencing glial cells as obligatory transducers of pathology and superlative targets for interference.
Collapse
Affiliation(s)
- Vishal Chavda
- Division of Anesthesia, Dreamzz IVF Center and Women’s Care Hospital, Ahmedabad 382350, Gujarat, India;
| | - Kavita Singh
- Centre for Translational Research, Jiwaji University, Gwalior 474011, Madhya Pradesh, India;
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Meerambika Mishra
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (M.M.); (A.K.M.)
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Korea
- Correspondence: (M.M.); (A.K.M.)
| |
Collapse
|
55
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
56
|
Morales-Rosales SL, Santín-Márquez R, Posadas-Rodriguez P, Rincon-Heredia R, Montiel T, Librado-Osorio R, Luna-López A, Rivero-Segura NA, Torres C, Cano-Martínez A, Silva-Palacios A, Cortés-Hernández P, Morán J, Massieu L, Konigsberg M. Senescence in Primary Rat Astrocytes Induces Loss of the Mitochondrial Membrane Potential and Alters Mitochondrial Dynamics in Cortical Neurons. Front Aging Neurosci 2021; 13:766306. [PMID: 34924995 PMCID: PMC8672143 DOI: 10.3389/fnagi.2021.766306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/27/2021] [Indexed: 01/10/2023] Open
Abstract
The decline in brain function during aging is one of the most critical health problems nowadays. Although senescent astrocytes have been found in old-age brains and neurodegenerative diseases, their impact on the function of other cerebral cell types is unknown. The aim of this study was to evaluate the effect of senescent astrocytes on the mitochondrial function of a neuron. In order to evaluate neuronal susceptibility to a long and constant senescence-associated secretory phenotype (SASP) exposure, we developed a model by using cellular cocultures in transwell plates. Rat primary cortical astrocytes were seeded in transwell inserts and induced to premature senescence with hydrogen peroxide [stress-induced premature senescence (SIPS)]. Independently, primary rat cortical neurons were seeded at the bottom of transwells. After neuronal 6 days in vitro (DIV), the inserts with SIPS-astrocytes were placed in the chamber and cocultured with neurons for 6 more days. The neuronal viability, the redox state [reduced glutathione/oxidized glutathione (GSH/GSSG)], the mitochondrial morphology, and the proteins and membrane potential were determined. Our results showed that the neuronal mitochondria functionality was altered after being cocultured with senescent astrocytes. In vivo, we found that old animals had diminished mitochondrial oxidative phosphorylation (OXPHOS) proteins, redox state, and senescence markers as compared to young rats, suggesting effects of the senescent astrocytes similar to the ones we observed in vitro. Overall, these results indicate that the microenvironment generated by senescent astrocytes can affect neuronal mitochondria and physiology.
Collapse
Affiliation(s)
- Sandra Lizbeth Morales-Rosales
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Roberto Santín-Márquez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Pedro Posadas-Rodriguez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Ruth Rincon-Heredia
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Teresa Montiel
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Raúl Librado-Osorio
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | - Armando Luna-López
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | - Claudio Torres
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Paulina Cortés-Hernández
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Atlixco, Mexico
| | - Julio Morán
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| |
Collapse
|
57
|
Verkhratsky A, Parpura V, Li B, Scuderi C. Astrocytes: The Housekeepers and Guardians of the CNS. ADVANCES IN NEUROBIOLOGY 2021; 26:21-53. [PMID: 34888829 DOI: 10.1007/978-3-030-77375-5_2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Astroglia are a diverse group of cells in the central nervous system. They are of the ectodermal, neuroepithelial origin and vary in morphology and function, yet, they can be collectively defined as cells having principle function to maintain homeostasis of the central nervous system at all levels of organisation, including homeostasis of ions, pH and neurotransmitters; supplying neurones with metabolic substrates; supporting oligodendrocytes and axons; regulating synaptogenesis, neurogenesis, and formation and maintenance of the blood-brain barrier; contributing to operation of the glymphatic system; and regulation of systemic homeostasis being central chemosensors for oxygen, CO2 and Na+. Their basic physiological features show a lack of electrical excitability (inapt to produce action potentials), but display instead a rather active excitability based on variations in cytosolic concentrations of Ca2+ and Na+. It is expression of neurotransmitter receptors, pumps and transporters at their plasmalemma, along with transports on the endoplasmic reticulum and mitochondria that exquisitely regulate the cytosolic levels of these ions, the fluctuation of which underlies most, if not all, astroglial homeostatic functions.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| |
Collapse
|
58
|
Ferah Okkay I, Okkay U, Gundogdu OL, Bayram C, Mendil AS, Ertugrul MS, Hacimuftuoglu A. Syringic acid protects against thioacetamide-induced hepatic encephalopathy: Behavioral, biochemical, and molecular evidence. Neurosci Lett 2021; 769:136385. [PMID: 34871743 DOI: 10.1016/j.neulet.2021.136385] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
The objective of this study was to elucidate the effects of syringic acid on thioacetamide-induced hepatic encephalopathy which is a complex serious syndrome with neuropsychiatric abnormalities related to acute liver dysfunctions like cirrhosis. Rats were treated with syringic acid (50 and 100 mg/kg, p.o.) for 14 days in treatment groups. Hepatic encephalopathy was induced by three doses of (200 mg/kg i.p.) thioacetamide injection. Syringic acid effectively alleviated thioacetamide-induced hepatic injury via reduction in ammonia, AST, ALT, ALP, LDH and decrease in oxidative stress (decreased MDA, ROS and increased SOD and GSH). Syringic acid also attenuated inflammatory injury by suppressing TNF-α, IL-1β, and NF-κB and increasing IL-10. The caspase-3 expression was also down-regulated in both liver and brain tissues. Immunohistochemical results confirmed the recovery with syringic acid by downregulation of iNOS, 8-OHdG and GFAP expression. Syringic acid decreased the deteriorating effects of thioacetamide as seen by reduced ammonia concentration and also preserved astrocyte and hepatocyte structure. The behavioral test results from elevated plus maze test, similar to the open-field locomotor test results, confirmed that syringic acid can reverse behavioral impairments. In conclusion, syringic acid exerted hepatoprotective and neuroprotective effects against hepatic encephalopathy by mitigating hepatotoxicity biomarkers, exerting antioxidant, anti-inflammatory effects in addition to suppressing hyperammonemia.
Collapse
Affiliation(s)
- Irmak Ferah Okkay
- Department of Pharmacology, Faculty of Pharmacy, Ataturk University, Turkey.
| | - Ufuk Okkay
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Turkey
| | - Omer Lutfi Gundogdu
- Department of Neurology, Faculty of Medicine, Recep Tayyip Erdogan University, Turkey
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Turkey
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | | | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Turkey
| |
Collapse
|
59
|
Savio LEB, Leite-Aguiar R, Alves VS, Coutinho-Silva R, Wyse ATS. Purinergic signaling in the modulation of redox biology. Redox Biol 2021; 47:102137. [PMID: 34563872 PMCID: PMC8479832 DOI: 10.1016/j.redox.2021.102137] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
Purinergic signaling is a cell communication pathway mediated by extracellular nucleotides and nucleosides. Tri- and diphosphonucleotides are released in physiological and pathological circumstances activating purinergic type 2 receptors (P2 receptors): P2X ion channels and P2Y G protein-coupled receptors. The activation of these receptors triggers the production of reactive oxygen and nitrogen species and alters antioxidant defenses, modulating the redox biology of cells. The activation of P2 receptors is controlled by ecto-enzymes named ectonucleotidases, E-NTPDase1/CD39 and ecto-5'-nucleotidase/CD73) being the most relevant. The first enzyme hydrolyzes adenosine triphosphate (ATP) and adenosine diphosphate (ADP) into adenosine monophosphate (AMP), and the second catalyzes the hydrolysis of AMP to adenosine. The activity of these enzymes is diminished by oxidative stress. Adenosine actives P1 G-coupled receptors that, in general, promote the maintenance of redox hemostasis by decreasing reactive oxygen species (ROS) production and increase antioxidant enzymes. Intracellular purine metabolism can also contribute to ROS generation via xanthine oxidase activity, which converts hypoxanthine into xanthine, and finally, uric acid. In this review, we describe the mechanisms of redox biology modulated by purinergic signaling and how this signaling may be affected by disturbances in the redox homeostasis of cells.
Collapse
Affiliation(s)
- Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Raíssa Leite-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinícius Santos Alves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
60
|
Fan Q, Gao Y, Mazur F, Chandrawati R. Nanoparticle-based colorimetric sensors to detect neurodegenerative disease biomarkers. Biomater Sci 2021; 9:6983-7007. [PMID: 34528639 DOI: 10.1039/d1bm01226f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurodegenerative disorders (NDDs) are progressive, incurable health conditions that primarily affect brain cells, and result in loss of brain mass and impaired function. Current sensing technologies for NDD detection are limited by high cost, long sample preparation, and/or require skilled personnel. To overcome these limitations, optical sensors, specifically colorimetric sensors, have garnered increasing attention towards the development of a cost-effective, simple, and rapid alternative approach. In this review, we evaluate colorimetric sensing strategies of NDD biomarkers (e.g. proteins, neurotransmitters, bio-thiols, and sulfide), address the limitations and challenges of optical sensor technologies, and provide our outlook on the future of this field.
Collapse
Affiliation(s)
- Qingqing Fan
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Yuan Gao
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Federico Mazur
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| |
Collapse
|
61
|
Stetska VO, Dovbynchuk TV, Makedon YS, Dziubenko NV. The effect of water-soluble pristine C60 fullerene on 6-OHDA-induced Parkinson’s disease in rats. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Oxidative stress is thought to be one of the mechanisms that leads to the dysfunction and degeneration of dopaminergic neurons in Parkinson’s disease pathogenesis and presumed to be underway during the prodromal phase. Therefore, therapy, which is effective against pre-motor symptoms, might be effective in preventing or delaying the development and progression of Parkinson’s disease. The aim of our study was to investigate the therapeutic efficiency of pristine C60 fullerene aqueous solution (C60FAS) during Parkinson’s disease in rats. The unilateral dopamine deficiency was induced in male Wistar rats (220–250 g) by stereotaxic microinjection of neurotoxin 6-hydroxydopamine (6-OHDA, 12 μg). C60FAS was injected to rats intraperitoneally daily for 10 days (0.65 mg/kg per day). The percentage of destroyed dopaminergic neurons was determined by the apomorphine test and by IHC staining of tyrosine hydroxylase-positive neurons in substantia nigra. We evaluated the rat body weight, the water and food intake, Open Field behavioural test, the level of biochemical antioxidant system, the activity of peritoneal macrophages. Levels of spontaneous and carbachol-stimulated colon motility were estimated by ballonographic method in vivo. C60FAS showed a positive tendency to increase the number of tyrosine hydroxylase-positive cells in the midbrain, which was associated with more profound improvement in apomorphine-rotation behaviour and slight relief of the anxiety level in Open Field test. Furthermore, C60FAS treatment increased the index of stimulated distal colon motor activity while it did not have a significant effect on water content in feces and total gastrointestinal transit time. C60FAS treatment did not affect water intake behaviour or body weight changes while it induced an increase of glutathione level and decrease activity of glutathione peroxidase in the brain as well as an increase in activity of peritoneal macrophages in 6-OHDA-Parkinson’s disease rats. These findings confirmed the potential therapeutic effectiveness of water-soluble pristine C60 fullerene in Parkinson’s disease pathogenesis, though there is ground for caution because of its systemic mild toxic effect.
Collapse
|
62
|
Faverzani JL, Steinmetz A, Deon M, Marchetti DP, Guerreiro G, Sitta A, de Moura Coelho D, Lopes FF, Nascimento LVM, Steffens L, Henn JG, Ferro MB, Brito VB, Wajner M, Moura DJ, Vargas CR. L-carnitine protects DNA oxidative damage induced by phenylalanine and its keto acid derivatives in neural cells: a possible pathomechanism and adjuvant therapy for brain injury in phenylketonuria. Metab Brain Dis 2021; 36:1957-1968. [PMID: 34216350 DOI: 10.1007/s11011-021-00780-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 06/07/2021] [Indexed: 11/24/2022]
Abstract
Although phenylalanine (Phe) is known to be neurotoxic in phenylketonuria (PKU), its exact pathogenetic mechanisms of brain damage are still poorly known. Furthermore, much less is known about the role of the Phe derivatives phenylacetic (PAA), phenyllactic (PLA) and phenylpyruvic (PPA) acids that also accumulate in this this disorder on PKU neuropathology. Previous in vitro and in vivo studies have shown that Phe elicits oxidative stress in brain of rodents and that this deleterious process also occurs in peripheral tissues of phenylketonuric patients. In the present study, we investigated whether Phe and its derivatives PAA, PLA and PPA separately or in combination could induce reactive oxygen species (ROS) formation and provoke DNA damage in C6 glial cells. We also tested the role of L-carnitine (L-car), which has been recently considered an antioxidant agent and easily cross the blood brain barrier on the alterations of C6 redox status provoked by Phe and its metabolites. We first observed that cell viability was not changed by Phe and its metabolites. Furthermore, Phe, PAA, PLA and PPA, at concentrations found in plasma of PKU patients, provoked marked DNA damage in the glial cells separately and when combined. Of note, these effects were totally prevented (Phe, PAA and PPA) or attenuated (PLA) by L-car pre-treatment. In addition, a potent ROS formation also induced by Phe and PAA, whereas only moderate increases of ROS were caused by PPA and PLA. Pre-treatment with L-car also prevented Phe- and PAA-induced ROS generation, but not that provoked by PLA and PPA. Thus, our data show that Phe and its major metabolites accumulated in PKU provoke extensive DNA damage in glial cells probably by ROS formation and that L-car may potentially represent an adjuvant therapeutic agent in PKU treatment.
Collapse
Affiliation(s)
- Jéssica Lamberty Faverzani
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Aline Steinmetz
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Marion Deon
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Desirèe Padilha Marchetti
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilian Guerreiro
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - Franciele Fatima Lopes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - Luiza Steffens
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Jeferson Gustavo Henn
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Matheus Bernardes Ferro
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Verônica Bidinotto Brito
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
- Departamento de Fisioterapia, Faculdades Integradas de Taquara (FACCAT), Taquara, Brazil
| | - Moacir Wajner
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Dinara Jaqueline Moura
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
63
|
Mehta K, Kaur B, Pandey KK, Dhar P, Kaler S. Resveratrol protects against inorganic arsenic-induced oxidative damage and cytoarchitectural alterations in female mouse hippocampus. Acta Histochem 2021; 123:151792. [PMID: 34634674 DOI: 10.1016/j.acthis.2021.151792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023]
Abstract
Prolonged inorganic arsenic (iAs) exposure is widely associated with brain damage particularly in the hippocampus via oxidative and apoptotic pathways. Resveratrol (RES) has gained considerable attention because of its benefits to human health. However, its neuroprotective potential against iAs-induced toxicity in CA1 region of hippocampus remains unexplored. Therefore, we investigated the neuroprotective efficacy of RES against arsenic trioxide (As2O3)-induced adverse effects on neuronal morphology, apoptotic markers and oxidative stress parameters in mouse CA1 region (hippocampus). Adult female Swiss albino mice of reproductive maturity were orally exposed to either As2O3 (2 and 4 mg/kg bw) alone or in combination with RES (40 mg/kg bw) for a period of 45 days. After animal sacrifice on day 46, the perfusion fixed brain samples were used for the observation of neuronal morphology and studying the morphometric features. While the freshly dissected hippocampi were processed for biochemical estimation of oxidative stress markers and western blotting of apoptosis-associated proteins. Chronic iAs exposure led to significant decrease in Stratum Pyramidale layer thickness along with reduction in cell density and area of Pyramidal neurons in contrast to the controls. Biochemical analysis showed reduced hippocampal GSH content but no change in total nitrite (NO) levels following iAs exposure. Western blotting showed apparent changes in the expression levels of Bax and Bcl-2 proteins following iAs exposure, however the change was statistically insignificant. Contrastingly, iAs +RES co-treatment exhibited substantial reversal in morphological and biochemical observations. Together, these findings provide preliminary evidence of neuroprotective role of RES on structural and biochemical alterations pertaining to mouse hippocampus following chronic iAs exposure.
Collapse
Affiliation(s)
- K Mehta
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - B Kaur
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - K K Pandey
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - P Dhar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - S Kaler
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
64
|
Asari MA, Sirajudeen K, Mohd Yusof NA, Mohd Amin MSI. DHA-rich fish oil and Tualang honey reduce chronic stress-induced oxidative damage in the brain of rat model. J Tradit Complement Med 2021; 12:361-366. [PMID: 35747355 PMCID: PMC9209864 DOI: 10.1016/j.jtcme.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 07/28/2021] [Accepted: 10/01/2021] [Indexed: 12/03/2022] Open
Abstract
Background Exposure to chronic stress induces oxidative damage which alters the dynamic balance between antioxidant and pro-oxidant activities in the brain. Tualang honey (TH) is a Malaysian wild multifloral honey which has been shown to contain high amount antioxidants. DHA-rich fish oil is a form of omega-3 fatty acids found in fish which also possesses some antioxidant activity. This study aimed to evaluate anti-stress activity of DHA-rich fish oil, TH and their combination on several parameters of oxidative stress in chronic stress rat model. Methods Fifty male Sprague Dawley rats were divided into (i) control, (ii) stress-exposed, (iii) stress-exposed and treated with TH (1 g/kg body weight twice daily), (iv) stress-exposed and treated with DHA-rich fish oil (450 mg/kg body weight twice daily), and (v) stress-exposed and treated with a combination of TH and DHA-rich fish oil. The chronic stress regimen consisted of a combination of restraint stress and a swim stress test for 28 days. Results DHA-rich fish oil and TH significantly (p < 0.05) supressed stress-induced elevation of serum corticosterone and lipid peroxidation, and caused a significant increase in total antioxidant capacity. For glutathione status, only TH significantly reduced stress-induced elevation of oxidised glutathione (GSSG) and normalised GSH/GSSG ratio. Conclusion: Both DHA-rich fish oil and TH have protective effects against brain oxidative stress but consuming these substances together does not seem to provide an additional benefit compared to consuming them separately. Investigated the effect of honey, DHA and their combination in single experimental setting. Tualang honey and DHA-rich fish oil can attenuate brain oxidative stress in chronic stress rat model. Combination of Tualang honey and DHA -rich fish oil is not superior than consuming these substances separately.
Collapse
|
65
|
Golubiani G, Lagani V, Solomonia R, Müller M. Metabolomic Fingerprint of Mecp2-Deficient Mouse Cortex: Evidence for a Pronounced Multi-Facetted Metabolic Component in Rett Syndrome. Cells 2021; 10:cells10092494. [PMID: 34572143 PMCID: PMC8472238 DOI: 10.3390/cells10092494] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/10/2023] Open
Abstract
Using unsupervised metabolomics, we defined the complex metabolic conditions in the cortex of a mouse model of Rett syndrome (RTT). RTT, which represents a cause of mental and cognitive disabilities in females, results in profound cognitive impairment with autistic features, motor disabilities, seizures, gastrointestinal problems, and cardiorespiratory irregularities. Typical RTT originates from mutations in the X-chromosomal methyl-CpG-binding-protein-2 (Mecp2) gene, which encodes a transcriptional modulator. It then causes a deregulation of several target genes and metabolic alterations in the nervous system and peripheral organs. We identified 101 significantly deregulated metabolites in the Mecp2-deficient cortex of adult male mice; 68 were increased and 33 were decreased compared to wildtypes. Pathway analysis identified 31 mostly upregulated metabolic pathways, in particular carbohydrate and amino acid metabolism, key metabolic mitochondrial/extramitochondrial pathways, and lipid metabolism. In contrast, neurotransmitter-signaling is dampened. This metabolic fingerprint of the Mecp2-deficient cortex of severely symptomatic mice provides further mechanistic insights into the complex RTT pathogenesis. The deregulated pathways that were identified—in particular the markedly affected amino acid and carbohydrate metabolism—confirm a complex and multifaceted metabolic component in RTT, which in turn signifies putative therapeutic targets. Furthermore, the deregulated key metabolites provide a choice of potential biomarkers for a more detailed rating of disease severity and disease progression.
Collapse
Affiliation(s)
- Gocha Golubiani
- Institut für Neuro- und Sinnesphysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, D-37130 Göttingen, Germany;
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Vincenzo Lagani
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Revaz Solomonia
- Institute of Chemical Biology, Ilia State University, 0162 Tbilisi, Georgia; (V.L.); (R.S.)
| | - Michael Müller
- Institut für Neuro- und Sinnesphysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, D-37130 Göttingen, Germany;
- Correspondence: ; Tel.: +49-551-39-22933
| |
Collapse
|
66
|
Bottino F, Lucignani M, Napolitano A, Dellepiane F, Visconti E, Rossi Espagnet MC, Pasquini L. In Vivo Brain GSH: MRS Methods and Clinical Applications. Antioxidants (Basel) 2021; 10:antiox10091407. [PMID: 34573039 PMCID: PMC8468877 DOI: 10.3390/antiox10091407] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/22/2021] [Accepted: 08/30/2021] [Indexed: 01/31/2023] Open
Abstract
Glutathione (GSH) is an important antioxidant implicated in several physiological functions, including the oxidation−reduction reaction balance and brain antioxidant defense against endogenous and exogenous toxic agents. Altered brain GSH levels may reflect inflammatory processes associated with several neurologic disorders. An accurate and reliable estimation of cerebral GSH concentrations could give a clear and thorough understanding of its metabolism within the brain, thus providing a valuable benchmark for clinical applications. In this context, we aimed to provide an overview of the different magnetic resonance spectroscopy (MRS) technologies introduced for in vivo human brain GSH quantification both in healthy control (HC) volunteers and in subjects affected by different neurological disorders (e.g., brain tumors, and psychiatric and degenerative disorders). Additionally, we aimed to provide an exhaustive list of normal GSH concentrations within different brain areas. The definition of standard reference values for different brain areas could lead to a better interpretation of the altered GSH levels recorded in subjects with neurological disorders, with insights into the possible role of GSH as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Francesca Bottino
- Medical Physics Department, Bambino Gesù Children’s Hospital IRCCS, 00165 Rome, Italy; (F.B.); (M.L.)
| | - Martina Lucignani
- Medical Physics Department, Bambino Gesù Children’s Hospital IRCCS, 00165 Rome, Italy; (F.B.); (M.L.)
| | - Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children’s Hospital IRCCS, 00165 Rome, Italy; (F.B.); (M.L.)
- Correspondence: ; Tel.: +39-333-3214614
| | - Francesco Dellepiane
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, 00189 Rome, Italy; (F.D.); (M.C.R.E.); (L.P.)
| | - Emiliano Visconti
- Neuroradiology Unit, Surgery and Trauma Department, Maurizio Bufalini Hospital, 47521 Cesena, Italy;
| | - Maria Camilla Rossi Espagnet
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, 00189 Rome, Italy; (F.D.); (M.C.R.E.); (L.P.)
- Neuroradiology Unit, Bambino Gesù Children’s Hospital IRCCS, 00165 Rome, Italy
| | - Luca Pasquini
- Neuroradiology Unit, NESMOS Department, Sant’Andrea Hospital, La Sapienza University, 00189 Rome, Italy; (F.D.); (M.C.R.E.); (L.P.)
- Neuroradiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
67
|
Ozawa H, Miyazawa T, Miyazawa T. Effects of Dietary Food Components on Cognitive Functions in Older Adults. Nutrients 2021; 13:2804. [PMID: 34444965 PMCID: PMC8398286 DOI: 10.3390/nu13082804] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/21/2022] Open
Abstract
Population aging has recently been an important issue as the number of elderly people is growing worldwide every year, and the extension of social security costs is financially costly. The increase in the number of elderly people with cognitive decline is a serious problem related to the aging of populations. Therefore, it is necessary to consider not only physical care but also cognitive patterns in the future care of older adults. Since food contains a variety of bioactive substances, dietary patterns may help improve age-related cognitive decline. However, the relationship between cognitive function and individual food components remains ambiguous as no clear efficacy or mechanism has been confirmed. Against this background, this review summarizes previous reports on the biological process of cognitive decline in the elderly and the relationship between individual compounds in foods and cognitive function, as well as the role of individual components of food in cognitive function, in the following order: lipids, carotenoids, vitamins, phenolic compounds, amino acids, peptides, and proteins. Based on the research presented in this review, a proper diet that preserves cognitive function has the potential to improve age-related cognitive decline, Alzheimer's disease, and Parkinson's disease. Hopefully, this review will help to trigger the development of new foods and technologies that improve aging and cognitive functions and extend the healthy life span.
Collapse
Affiliation(s)
| | | | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai 980-8579, Japan; (H.O.); (T.M.)
| |
Collapse
|
68
|
Asanuma M, Miyazaki I. Glutathione and Related Molecules in Parkinsonism. Int J Mol Sci 2021; 22:ijms22168689. [PMID: 34445395 PMCID: PMC8395390 DOI: 10.3390/ijms22168689] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis, and release in/from surrounding astrocytes. Transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), a detoxifying master transcription factor, is expressed mainly in astrocytes and activates the gene expression of various phase II drug-metabolizing enzymes or antioxidants including GSH-related molecules and metallothionein by binding to the antioxidant response element (ARE) of these genes. Accumulating evidence has shown the involvement of dysfunction of antioxidative molecules including GSH and its related molecules in the pathogenesis of Parkinson’s disease (PD) or parkinsonian models. Furthermore, we found several agents targeting GSH synthesis in the astrocytes that protect nigrostriatal dopaminergic neuronal loss in PD models. In this article, the neuroprotective effects of supplementation and enhancement of GSH and its related molecules in PD pathology are reviewed, along with introducing new experimental findings, especially targeting of the xCT-GSH synthetic system and Nrf2–ARE pathway in astrocytes.
Collapse
|
69
|
Nutraceutical Strategy to Counteract Eye Neurodegeneration and Oxidative Stress in Drosophila melanogaster Fed with High-Sugar Diet. Antioxidants (Basel) 2021; 10:antiox10081197. [PMID: 34439445 PMCID: PMC8388935 DOI: 10.3390/antiox10081197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022] Open
Abstract
Aberrant production of reactive oxygen species (ROS) is a common feature of damaged retinal neurons in diabetic retinopathy, and antioxidants may exert both preventive and therapeutic action. To evaluate the beneficial and antioxidant properties of food supplementation with Lisosan G, a powder of bran and germ of grain (Triticum aestivum) obtained by fermentation with selected lactobacillus and natural yeast strains, we used an in vivo model of hyperglycemia-induced retinal damage, the fruit fly Drosophila melanogaster fed with high-sucrose diet. Lisosan G positively affected the visual system of hyperglycemic flies at structural/functional level, decreased apoptosis, and reactivated protective autophagy at the retina internal network. Also, in high sucrose-fed Drosophila, Lisosan G reduced the levels of brain ROS and retina peroxynitrite. The analysis of oxidative stress-related metabolites suggested 7,8-dihydrofolate, uric acid, dihydroorotate, γ-L-glutamyl-L-cysteine, allantoin, cysteinyl-glycine, and quinolate as key mediators of Lisosan G-induced inhibition of neuronal ROS, along with the upregulation of glutathione system. Of note, Lisosan G may impact oxidative stress and the ensuing retinal cell death, also independently from autophagy, although the autophagy-ROS cross-talk is critical. This study demonstrated that the continuous supplementation with the alimentary integrator Lisosan G exerts a robust and multifaceted antioxidant effect on retinal neurons, thus providing efficacious neuroprotection of hyperglycemic eye.
Collapse
|
70
|
Wang C, Gong Y, Deng F, Ding E, Tang J, Codling G, Challis JK, Green D, Wang J, Chen Q, Xie Y, Su S, Yang Z, Raine J, Jones PD, Tang S, Giesy JP. Remodeling of Arctic char (Salvelinus alpinus) lipidome under a stimulated scenario of Arctic warming. GLOBAL CHANGE BIOLOGY 2021; 27:3282-3298. [PMID: 33837644 DOI: 10.1111/gcb.15638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/14/2021] [Indexed: 06/12/2023]
Abstract
Arctic warming associated with global climate change poses a significant threat to populations of wildlife in the Arctic. Since lipids play a vital role in adaptation of organisms to variations in temperature, high-resolution mass-spectrometry-based lipidomics can provide insights into adaptive responses of organisms to a warmer environment in the Arctic and help to illustrate potential novel roles of lipids in the process of thermal adaption. In this study, we studied an ecologically and economically important species-Arctic char (Salvelinus alpinus)-with a detailed multi-tissue analysis of the lipidome in response to chronic shifts in temperature using a validated lipidomics workflow. In addition, dynamic alterations in the hepatic lipidome during the time course of shifts in temperature were also characterized. Our results showed that early life stages of Arctic char were more susceptible to variations in temperature. One-year-old Arctic char responded to chronic increases in temperature with coordinated regulation of lipids, including headgroup-specific remodeling of acyl chains in glycerophospholipids (GP) and extensive alterations in composition of lipids in membranes, such as less lyso-GPs, and more ether-GPs and sphingomyelin. Glycerolipids (e.g., triacylglycerol, TG) also participated in adaptive responses of the lipidome of Arctic char. Eight-week-old Arctic char exhibited rapid adaptive alterations of the hepatic lipidome to stepwise decreases in temperature while showing blunted responses to gradual increases in temperature, implying an inability to adapt rapidly to warmer environments. Three common phosphatidylethanolamines (PEs) (PE 36:6|PE 16:1_20:5, PE 38:7|PE 16:1_22:6, and PE 40:7|PE 18:1_22:6) were finally identified as candidate lipid biomarkers for temperature shifts via machine learning approach. Overall, this work provides additional information to a better understanding of underlying regulatory mechanisms of the lipidome of Arctic organisms in the face of near-future warming.
Collapse
Affiliation(s)
- Chao Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yufeng Gong
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Fuchang Deng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Enmin Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jie Tang
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
- School of Resources and Environment, Anhui Agricultural University, Hefei, Anhui, China
| | - Garry Codling
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
- Research Centre for Contaminants in the Environment, Masaryk University, Brno, Czech Republic
| | | | - Derek Green
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jing Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Qiliang Chen
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Yuwei Xie
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Shu Su
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zilin Yang
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jason Raine
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Paul D Jones
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - John P Giesy
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Environmental Sciences, Baylor University, Waco, TX, USA
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| |
Collapse
|
71
|
Glutathione in the Nervous System as a Potential Therapeutic Target to Control the Development and Progression of Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2021; 10:antiox10071011. [PMID: 34201812 PMCID: PMC8300718 DOI: 10.3390/antiox10071011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/25/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare neurological disorder that affects the motor neurons responsible for regulating muscle movement. However, the molecular pathogenic mechanisms of ALS remain poorly understood. A deficiency in the antioxidant tripeptide glutathione (GSH) in the nervous system appears to be involved in several neurodegenerative diseases characterized by the loss of neuronal cells. Impaired antioxidant defense systems, and the accumulation of oxidative damage due to increased dysfunction in GSH homeostasis are known to be involved in the development and progression of ALS. Aberrant GSH metabolism and redox status following oxidative damage are also associated with various cellular organelles, including the mitochondria and nucleus, and are crucial factors in neuronal toxicity induced by ALS. In this review, we provide an overview of the implications of imbalanced GSH homeostasis and its molecular characteristics in various experimental models of ALS.
Collapse
|
72
|
Kolar D, Kleteckova L, Brozka H, Vales K. Mini-review: Brain energy metabolism and its role in animal models of depression, bipolar disorder, schizophrenia and autism. Neurosci Lett 2021; 760:136003. [PMID: 34098028 DOI: 10.1016/j.neulet.2021.136003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/13/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Mitochondria are cellular organelles essential for energy metabolism and antioxidant defense. Mitochondrial impairment is implicated in many psychiatric disorders, including depression, bipolar disorder, schizophrenia, and autism. To characterize and eventually find effective treatments of bioenergetic impairment in psychiatric disease, researchers find animal models indispensable. The present review focuses on brain energetics in several environmental, genetic, drug-induced, and surgery-induced animal models of depression, bipolar disorder, schizophrenia, and autism. Most reported deficits included decreased activity in the electron transport chain, increased oxidative damage, decreased antioxidant defense, decreased ATP levels, and decreased mitochondrial potential. Models of depression, bipolar disorder, schizophrenia, and autism shared many bioenergetic deficits. This is in concordance with the absence of a disease-specific brain energy phenotype in human patients. Unfortunately, due to the absence of null results in examined literature, indicative of reporting bias, we refrain from making generalized conclusions. Present review can be a valuable tool for comparing current findings, generating more targeted hypotheses, and selecting fitting models for further preclinical research.
Collapse
Affiliation(s)
- David Kolar
- National Institute of Mental Health, Klecany, Czech Republic.
| | | | - Hana Brozka
- Institute of Physiology, Academy of Sciences, Prague, Czech Republic.
| | - Karel Vales
- National Institute of Mental Health, Klecany, Czech Republic.
| |
Collapse
|
73
|
Vitalakumar D, Sharma A, Flora SJS. Ferroptosis: A potential therapeutic target for neurodegenerative diseases. J Biochem Mol Toxicol 2021; 35:e22830. [PMID: 34047408 DOI: 10.1002/jbt.22830] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/25/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022]
Abstract
Ferroptosis is a newly identified regulated form of cell death, which is thought to play a major role in neurodegenerative diseases. In this review, we discuss recent studies elucidating the molecular mechanisms involved in the regulation and execution of ferroptotic cell death and also its role in the brain. Ferroptosis is regulated mainly via iron homeostasis, glutathione metabolism, and lipid peroxidation. Ferroptotic cell death and pro-ferroptotic factors are correlated with the etiopathogenesis of Parkinson's disease (PD) and Alzheimer's disease (AD). Ferroptosis and etiological factors act synergistically in PD and AD pathogenesis. Furthermore, several preclinical and clinical studies targeting ferroptosis in PD and AD have also shown positive results. Evidence of ferroptosis in the brain thus gives new insights into understanding neurodegenerative diseases. Ferroptosis studies in the brain are still in their infancy, but the existing pieces of evidence suggest a strong correlation between ferroptotic cell death and neurodegenerative diseases. Thus, ferroptosis might be a promising target for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- D Vitalakumar
- Department of Biotechnology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Ankita Sharma
- Department of Biotechnology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Swaran J S Flora
- Department of Biotechnology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| |
Collapse
|
74
|
Oyefeso FA, Muotri AR, Wilson CG, Pecaut MJ. Brain organoids: A promising model to assess oxidative stress-induced central nervous system damage. Dev Neurobiol 2021; 81:653-670. [PMID: 33942547 DOI: 10.1002/dneu.22828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) is one of the most significant propagators of systemic damage with implications for widespread pathologies such as vascular disease, accelerated aging, degenerative disease, inflammation, and traumatic injury. OS can be induced by numerous factors such as environmental conditions, lifestyle choices, disease states, and genetic susceptibility. It is tied to the accumulation of free radicals, mitochondrial dysfunction, and insufficient antioxidant protection, which leads to cell aging and tissue degeneration over time. Unregulated systemic increase in reactive species, which contain harmful free radicals, can lead to diverse tissue-specific OS responses and disease. Studies of OS in the brain, for example, have demonstrated how this state contributes to neurodegeneration and altered neural plasticity. As the worldwide life expectancy has increased over the last few decades, the prevalence of OS-related diseases resulting from age-associated progressive tissue degeneration. Unfortunately, vital translational research studies designed to identify and target disease biomarkers in human patients have been impeded by many factors (e.g., limited access to human brain tissue for research purposes and poor translation of experimental models). In recent years, stem cell-derived three-dimensional tissue cultures known as "brain organoids" have taken the spotlight as a novel model for studying central nervous system (CNS) diseases. In this review, we discuss the potential of brain organoids to model the responses of human neural cells to OS, noting current and prospective limitations. Overall, brain organoids show promise as an innovative translational model to study CNS susceptibility to OS and elucidate the pathophysiology of the aging brain.
Collapse
Affiliation(s)
- Foluwasomi A Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Alysson R Muotri
- Department of Pediatrics/Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD, Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Michael J Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
75
|
Raj Rai S, Bhattacharyya C, Sarkar A, Chakraborty S, Sircar E, Dutta S, Sengupta R. Glutathione: Role in Oxidative/Nitrosative Stress, Antioxidant Defense, and Treatments. ChemistrySelect 2021. [DOI: 10.1002/slct.202100773] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sristi Raj Rai
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| | | | - Anwita Sarkar
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| | - Surupa Chakraborty
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| | - Esha Sircar
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| | - Sreejita Dutta
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Amity University Kolkata 700135, W.B. India
| |
Collapse
|
76
|
Mishra P, Mittal AK, Rajput SK, Sinha JK. Cognition and memory impairment attenuation via reduction of oxidative stress in acute and chronic mice models of epilepsy using antiepileptogenic Nux vomica. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113509. [PMID: 33141053 DOI: 10.1016/j.jep.2020.113509] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/03/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
UNLABELLED Ethnopharmacological relevance Processed Nux vomica seed extracts and homeopathic medicinal preparations (HMPs) are widely used in traditional Indian and Chinese medicine for respiratory, digestive, neurological and behavioral disorders. Antioxidant property of Nux vomica is well known and recent investigation has highlighted the anticonvulsant potential of its homeopathic formulation. AIM OF THE STUDY To explore the anticonvulsant and antiepileptogenic potential of Nux vomica HMPs (6CH, 12CH and 30CH potency) in pentylenetetrazole (PTZ) induced acute and chronic experimental seizure models in mice and investigate their effects on cognition, memory, motor activity and oxidative stress markers in kindled animals. MATERIALS AND METHODS Acute seizures were induced in the animals through 70 mg/kg (i.p.) administration of PTZ followed by the evaluation of latency and duration of Generalized tonic-clonic seizures (GTCS). Subconvulsive PTZ doses (35 mg/kg, i.p.) induced kindling in 29 days, which was followed by assessment of cognition, memory and motor impairment through validated behavioral techniques. The status of oxidative stress was estimated through measurement of MDA, GSH and SOD. RESULTS HMPs delayed the latency and reduced the duration of GTCS in acute model signifying possible regulation of GABAergic neurotransmission. Kindling was significantly hindered by the HMPs that justified the ameliorated cognition, memory and motor activity impairment. The HMPs attenuated lipid peroxidation by reducing MDA level and strengthened the antioxidant mechanism by enhancing the GSH and SOD levels in the kindled animals. CONCLUSIONS Nux vomica HMPs showed anticonvulsant and antiepileptogenic potency in acute and chronic models of epilepsy. The test drugs attenuated behavioral impairment and reduced the oxidative stress against PTZ induced kindling owing to which they can be further explored for their cellular and molecular mechanism(s).
Collapse
Affiliation(s)
- Priya Mishra
- Amity Institute of Neuropsychology and Neurosciences (AINN), Amity University, Noida, Uttar Pradesh, 201303, India
| | - Amit Kumar Mittal
- Amity Institute of Indian System of Medicine (AIISM), Amity University, Noida, Uttar Pradesh, 201303, India
| | - Satyendra Kumar Rajput
- Amity Institute of Indian System of Medicine (AIISM), Amity University, Noida, Uttar Pradesh, 201303, India; Department of Pharmaceutical Sciences, Gurukul Kangri (deemed to be University), Haridwar, Uttrakhand, 249404, India.
| | - Jitendra Kumar Sinha
- Amity Institute of Neuropsychology and Neurosciences (AINN), Amity University, Noida, Uttar Pradesh, 201303, India.
| |
Collapse
|
77
|
Azar J, Yousef MH, El-Fawal HAN, Abdelnaser A. Mercury and Alzheimer's disease: a look at the links and evidence. Metab Brain Dis 2021; 36:361-374. [PMID: 33411216 DOI: 10.1007/s11011-020-00649-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/20/2020] [Indexed: 12/01/2022]
Abstract
This review paper investigates a specific environmental-disease interaction between mercury exposure and Alzheimer's disease hallmarks. Alzheimer's disease is a neurodegenerative disorder affecting predominantly the memory of the affected individual. It prevails mostly in the elderly, rendering many factors as possible causative agents, which potentially contribute to the disease pathogenicity cumulatively. Alzheimer's disease affects nearly 50 million people worldwide and is considered one the most devastating diseases not only for the patient, but also for their families and caregivers. Mercury is a common environmental toxin, found in the atmosphere mostly due to human activity, such as coal burning for heating and cooking. Natural release of mercury into the atmosphere occurs by volcanic eruptions, in the form of vapor, or weathering rocks. The most toxic form of mercury to humans is methylmercury, to which humans are exposed to by ingestion of fish. Methylmercury was found to exert its toxic effects on different parts of the human body, with predominance on the brain. There is no safe concentration for mercury in the atmosphere, even trace amounts can elicit harm to humans in the long term. Mercury's effect on Alzheimer's disease hallmarks formation, extracellular senile plaques and intracellular neurofibrillary tangles, has been widely studied. This review demonstrates the involvement of mercury, in its different forms, in the pathway of amyloid beta deposition and tau tangles formation. It aims to understand the link between mercury exposure and Alzheimer's disease so that, in the future, prevention strategies can be applied to halt the progression of this disease.
Collapse
Affiliation(s)
- Jihan Azar
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt
| | - Mohamed H Yousef
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo, Cairo, Egypt
| | - Hassan A N El-Fawal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt
| | - Anwar Abdelnaser
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box: 74, New Cairo, Egypt.
| |
Collapse
|
78
|
Dahabiyeh LA, Mahmoud NN, Al-Natour MA, Safo L, Kim DH, Khalil EA, Abu-Dahab R. Phospholipid-Gold Nanorods Induce Energy Crisis in MCF-7 Cells: Cytotoxicity Evaluation Using LC-MS-Based Metabolomics Approach. Biomolecules 2021; 11:364. [PMID: 33673519 PMCID: PMC7997200 DOI: 10.3390/biom11030364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/19/2022] Open
Abstract
Phospholipid-modified gold nanorods (phospholipid-GNRs) have demonstrated drastic cytotoxicity towards MCF-7 breast cancer cells compared to polyethylene glycol-coated GNRs (PEG-GNRs). In this study, the mechanism of cytotoxicity of phospholipid-GNRs towards MCF-7 cells was investigated using mass spectrometry-based global metabolic profiling and compared to PEGylated counterparts. The results showed that when compared to PEG-GNRs, phospholipid-GNRs induced significant and more pronounced impact on the metabolic profile of MCF-7 cells. Phospholipid-GNRs significantly decreased the levels of metabolic intermediates and end-products associated with cellular energy metabolisms resulting in dysfunction in TCA cycle, a reduction in glycolytic activity, and imbalance of the redox state. Additionally, phospholipid-GNRs disrupted several metabolism pathways essential for the normal growth and proliferation of cancer cells including impairment in purine, pyrimidine, and glutathione metabolisms accompanied by lower amino acid pools. On the other hand, the effects of PEG-GNRs were limited to alteration of glycolysis and pyrimidine metabolism. The current work shed light on the importance of metabolomics as a valuable analytical approach to explore the molecular effects of GNRs with different surface chemistry on cancer cell and highlights metabolic targets that might serve as promising treatment strategy in cancer.
Collapse
Affiliation(s)
- Lina A. Dahabiyeh
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Nouf N. Mahmoud
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Mohammad A. Al-Natour
- Department of Pharmaceutics and Pharmaceutical Technology, The Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan;
| | - Laudina Safo
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK; (L.S.); (D.-H.K.)
| | - Dong-Hyun Kim
- Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK; (L.S.); (D.-H.K.)
| | - Enam A. Khalil
- Department of Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Rana Abu-Dahab
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| |
Collapse
|
79
|
Du Y, Wei J, Yang X, Dou Y, Zhao L, Qi X, Yu X, Guo W, Wang Q, Deng W, Li M, Lin D, Li T, Ma X. Plasma metabolites were associated with spatial working memory in major depressive disorder. Medicine (Baltimore) 2021; 100:e24581. [PMID: 33663067 PMCID: PMC7909221 DOI: 10.1097/md.0000000000024581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/14/2021] [Indexed: 02/05/2023] Open
Abstract
Major depressive disorder (MDD) is a common disease with both affective and cognitive disorders. Alterations in metabolic systems of MDD patients have been reported, but the underlying mechanisms still remains unclear. We sought to identify abnormal metabolites in MDD by metabolomics and to explore the association between differential metabolites and neurocognitive dysfunction.Plasma samples from 53 MDD patients and 83 sex-, gender-, BMI-matched healthy controls (HCs) were collected. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) system was then used to detect metabolites in those samples. Two different algorithms were applied to identify differential metabolites in 2 groups. Of the 136 participants, 35 MDD patients and 48 HCs had completed spatial working memory test. Spearman rank correlation coefficient was applied to explore the relationship between differential metabolites and working memory in these 2 groups.The top 5 metabolites which were found in sparse partial least squares-discriminant analysis (sPLS-DA) model and random forest (RF) model were the same, and significant difference was found in 3 metabolites between MDD and HCs, namely, gamma-glutamyl leucine, leucine-enkephalin, and valeric acid. In addition, MDD patients had higher scores in spatial working memory (SWM) between errors and total errors than HCs. Valeric acid was positively correlated with working memory in MDD group.Gamma-glutamyl leucine, leucine-enkephalin, and valeric acid were preliminarily proven to be decreased in MDD patients. In addition, MDD patients performed worse in working memory than HCs. Dysfunction in working memory of MDD individuals was associated with valeric acid.
Collapse
Affiliation(s)
- Yue Du
- Psychiatric Laboratory and Mental Health Center
| | - Jinxue Wei
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xiao Yang
- Psychiatric Laboratory and Mental Health Center
| | - Yikai Dou
- Psychiatric Laboratory and Mental Health Center
| | - Liansheng Zhao
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xueyu Qi
- Psychiatric Laboratory and Mental Health Center
| | - Xueli Yu
- Psychiatric Laboratory and Mental Health Center
| | - Wanjun Guo
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Qiang Wang
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Wei Deng
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Minli Li
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Dongtao Lin
- College of Foreign Languages and Cultures, Sichuan University, PR China
| | - Tao Li
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu
| |
Collapse
|
80
|
Yeasts as Complementary Model Systems for the Study of the Pathological Repercussions of Enhanced Synphilin-1 Glycation and Oxidation. Int J Mol Sci 2021; 22:ijms22041677. [PMID: 33562355 PMCID: PMC7915245 DOI: 10.3390/ijms22041677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 01/22/2023] Open
Abstract
Synphilin-1 has previously been identified as an interaction partner of α-Synuclein (αSyn), a primary constituent of neurodegenerative disease-linked Lewy bodies. In this study, the repercussions of a disrupted glyoxalase system and aldose reductase function on Synphilin-1 inclusion formation characteristics and cell growth were investigated. To this end, either fluorescent dsRed-tagged or non-tagged human SNCAIP, which encodes the Synphilin-1 protein, was expressed in Saccharomyces cerevisiae and Schizosaccharomyces pombe yeast strains devoid of enzymes Glo1, Glo2, and Gre3. Presented data shows that lack of Glo2 and Gre3 activity in S. cerevisiae increases the formation of large Synphilin-1 inclusions. This correlates with enhanced oxidative stress levels and an inhibitory effect on exponential growth, which is most likely caused by deregulation of autophagic degradation capacity, due to excessive Synphilin-1 aggresome build-up. These findings illustrate the detrimental impact of increased oxidation and glycation on Synphilin-1 inclusion formation. Similarly, polar-localised inclusions were observed in wild-type S. pombe cells and strains deleted for either glo1+ or glo2+. Contrary to S. cerevisiae, however, no growth defects were observed upon expression of SNCAIP. Altogether, our findings show the relevance of yeasts, especially S. cerevisiae, as complementary models to unravel mechanisms contributing to Synphilin-1 pathology in the context of neurodegenerative diseases.
Collapse
|
81
|
Iravanpour F, Dargahi L, Rezaei M, Haghani M, Heidari R, Valian N, Ahmadiani A. Intranasal insulin improves mitochondrial function and attenuates motor deficits in a rat 6-OHDA model of Parkinson's disease. CNS Neurosci Ther 2021; 27:308-319. [PMID: 33497031 PMCID: PMC7871791 DOI: 10.1111/cns.13609] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Aims Experimental and clinical evidences demonstrate that common dysregulated pathways are involved in Parkinson’s disease (PD) and type 2 diabetes. Recently, insulin treatment through intranasal (IN) approach has gained attention in PD, although the underlying mechanism of its potential therapeutic effects is still unclear. In this study, we investigated the effects of insulin treatment in a rat model of PD with emphasis on mitochondrial function indices in striatum. Methods Rats were treated with a daily low dose (4IU/day) of IN insulin, starting 72 h after 6‐OHDA‐induced lesion and continued for 14 days. Motor performance, dopaminergic cell survival, mitochondrial dehydrogenases activity, mitochondrial swelling, mitochondria permeability transition pore (mPTP), mitochondrial membrane potential (Δψm), reactive oxygen species (ROS) formation, and glutathione (GSH) content in mitochondria, mitochondrial adenosine triphosphate (ATP), and the gene expression of PGC‐1α, TFAM, Drp‐1, GFAP, and Iba‐1 were assessed. Results Intranasal insulin significantly reduces 6‐OHDA‐induced motor dysfunction and dopaminergic cell death. In parallel, it improves mitochondrial function indices and modulates mitochondria biogenesis and fission as well as activation of astrocytes and microglia. Conclusion Considering the prominent role of mitochondrial dysfunction in PD pathology, IN insulin as a disease‐modifying therapy for PD should be considered for extensive research.
Collapse
Affiliation(s)
- Farideh Iravanpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rezaei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Haghani
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
82
|
Kim SW, Kim Y, Kim SE, An JY. Ferroptosis-Related Genes in Neurodevelopment and Central Nervous System. BIOLOGY 2021; 10:35. [PMID: 33419148 PMCID: PMC7825574 DOI: 10.3390/biology10010035] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Ferroptosis, first introduced as a new form of regulated cell death induced by erastin, is accompanied by the accumulation of iron and lipid peroxides, thus it can be inhibited either by iron chelators or by lipophilic antioxidants. In the past decade, multiple studies have introduced the potential importance of ferroptosis in many human diseases, including cancer and neurodegenerative diseases. In this review, we will discuss the genetic association of ferroptosis with neurological disorders and development of the central nervous system.
Collapse
Affiliation(s)
- Soo-Whee Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Sung Eun Kim
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Joon-Yong An
- Department of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (S.-W.K.); (Y.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
83
|
Asanuma M, Miyazaki I. [Anti-oxidants in astrocytes as target of neuroprotection for Parkinson's disease]. Nihon Yakurigaku Zasshi 2021; 156:14-20. [PMID: 33390474 DOI: 10.1254/fpj.20071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Recently, it has been reported that dysfunction of astrocytes is involved vulnerability of neuronal cells in several neurological disorders. Glutathione (GSH) is the most abundant intrinsic antioxidant in the central nervous system, and its substrate cysteine is readily becomes the oxidized dimeric cystine. Since neurons lack a cystine transport system, neuronal GSH synthesis depends on cystine uptake via the cystine/glutamate exchange transporter (xCT), GSH synthesis and release in/from surrounding astrocytes. The expression and release of the zinc-binding protein metallothionein (MT) in astrocytes, which is a strong antioxidant, is induced and exerts neuroprotective in the case of dopaminergic neuronal damage. In addition, the transcription factor Nrf2 induces expression of MT-1 and GSH related molecules. We previously revealed that several antiepileptic drugs, serotonin 5-HT1A receptor agonists, plant-derived chemicals (phytochemicals) increased xCT expression, Nrf2 activation, GSH or MT expression and release in/from astrocytes, and exerted a neuroprotective effect against dopaminergic neurodegeneration in Parkinson's disease model. Our serial studies on neuroprotection via antioxidant defense mechanism of astrocytes have found three target molecular systems of astrocytes for neuroprotection: (1) xCT-GSH synthetic system, (2) Nrf2 system and (3) 5-HT1A receptor-Nrf2-MT system, 5-HT1A-S100β system. In this article, possible neuroprotective strategy for Parkinson's disease has been reviewed targeting antioxidative molecules in astrocytes.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentist and Pharmaceutical Sciences
| |
Collapse
|
84
|
Physiological and antioxidant responses of Euryale ferox salisb seedlings to microcystins. Toxicon 2020; 190:50-57. [PMID: 33338447 DOI: 10.1016/j.toxicon.2020.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/23/2020] [Accepted: 12/13/2020] [Indexed: 11/22/2022]
Abstract
Lake Taihu is the third largest freshwater lake located in eastern China. In recent years, it has experienced extensive cyanobacterial (Microcystis spp.) blooms that produce toxic microcystins (MCs), which may have acute and chronic hepatotoxic effects in animals and humans. Although the impact of MCs on both terrestrial and aquatic plants is well documented, the effects and underlying mechanisms of the harmful toxin MC-LR on Euryale ferox Salisb seedlings have rarely been reported. Thus, herein, the antioxidant response mechanisms and the biosynthesis of secondary metabolites during the exposure of E. ferox Salisb seedlings to varying MC-LR concentrations (0.05, 0.2, 1, and 5 μg/L) were thoroughly investigated after exposure periods (7, 14, 21 d). Our study revealed that the seedling growth was inhibited with increasing MC-LR exposure concentration that significantly induced at 1 μg/L and reached a maximum level at 5 μg/L, whereas the activity of the antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), and peroxidase (POD) in the seedling cells increased gradually with increasing MC-LR concentration and longer exposure time. The maximum malondialdehyde (MDA) content was 4.3-fold higher than that of the control group under an MC-LR concentration of 5.0 μg/L after 7 days of exposure treatment. The study of the seedling detoxification mechanism revealed that the content of total glutathione (tGSH) and reduced glutathione (GSH), as well as the activities of GSH sparse transferase (GST) and glutathione reductase (GR), increased to varying degrees and reached a maximum level at 1 μg/L. Therefore, the exposure to MC-LR can promote the accumulation of secondary metabolites and increase the activities of secondary metabolic enzymes in the seedlings. Further investigation of these antioxidative mechanisms will provide additional information for the identification and development of bio-indicators to evaluate the environmental impact of MCs on aquatic ecosystems.
Collapse
|
85
|
Miyazaki I, Asanuma M. Neuron-Astrocyte Interactions in Parkinson's Disease. Cells 2020; 9:cells9122623. [PMID: 33297340 PMCID: PMC7762285 DOI: 10.3390/cells9122623] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. PD patients exhibit motor symptoms such as akinesia/bradykinesia, tremor, rigidity, and postural instability due to a loss of nigrostriatal dopaminergic neurons. Although the pathogenesis in sporadic PD remains unknown, there is a consensus on the involvement of non-neuronal cells in the progression of PD pathology. Astrocytes are the most numerous glial cells in the central nervous system. Normally, astrocytes protect neurons by releasing neurotrophic factors, producing antioxidants, and disposing of neuronal waste products. However, in pathological situations, astrocytes are known to produce inflammatory cytokines. In addition, various studies have reported that astrocyte dysfunction also leads to neurodegeneration in PD. In this article, we summarize the interaction of astrocytes and dopaminergic neurons, review the pathogenic role of astrocytes in PD, and discuss therapeutic strategies for the prevention of dopaminergic neurodegeneration. This review highlights neuron-astrocyte interaction as a target for the development of disease-modifying drugs for PD in the future.
Collapse
|
86
|
Wang Q, Xie C, Xi S, Qian F, Peng X, Huang J, Tang F. Radioprotective Effect of Flavonoids on Ionizing Radiation-Induced Brain Damage. Molecules 2020; 25:5719. [PMID: 33287417 PMCID: PMC7730479 DOI: 10.3390/molecules25235719] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 01/27/2023] Open
Abstract
Patients receiving brain radiotherapy may suffer acute or chronic side effects. Ionizing radiation induces the production of intracellular reactive oxygen species and pro-inflammatory cytokines in the central nervous system, leading to brain damage. Complementary Chinese herbal medicine therapy may reduce radiotherapy-induced side effects. Flavonoids are a class of natural products which can be extracted from Chinese herbal medicine and have been shown to have neuroprotective and radioprotective properties. Flavonoids are effective antioxidants and can also inhibit regulatory enzymes or transcription factors important for controlling inflammatory mediators, affect oxidative stress through interaction with DNA and enhance genomic stability. In this paper, radiation-induced brain damage and the relevant molecular mechanism were summarized. The radio-neuro-protective effect of flavonoids, i.e., antioxidant, anti-inflammatory and maintaining genomic stability, were then reviewed. We concluded that flavonoids treatment may be a promising complementary therapy to prevent radiotherapy-induced brain pathophysiological changes and cognitive impairment.
Collapse
Affiliation(s)
- Qinqi Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.W.); (C.X.); (S.X.)
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Chenghao Xie
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.W.); (C.X.); (S.X.)
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Shijun Xi
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.W.); (C.X.); (S.X.)
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Xiaochun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.W.); (C.X.); (S.X.)
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Jiangrong Huang
- Department of Integrative Medicine, School of Health Sciences, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 1 CREATE Way #04-01, CREATE Tower, Singapore 138602, Singapore
| |
Collapse
|
87
|
Bjørklund G, Tinkov AA, Hosnedlová B, Kizek R, Ajsuvakova OP, Chirumbolo S, Skalnaya MG, Peana M, Dadar M, El-Ansary A, Qasem H, Adams JB, Aaseth J, Skalny AV. The role of glutathione redox imbalance in autism spectrum disorder: A review. Free Radic Biol Med 2020; 160:149-162. [PMID: 32745763 DOI: 10.1016/j.freeradbiomed.2020.07.017] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022]
Abstract
The role of glutathione in autism spectrum disorder (ASD) is emerging as a major topic, due to its role in the maintenance of the intracellular redox balance. Several studies have implicated glutathione redox imbalance as a leading factor in ASD, and both ASD and many other neurodevelopmental disorders involve low levels of reduced glutathione (GSH), high levels of oxidized glutathione (GSSG), and abnormalities in the expressions of glutathione-related enzymes in the blood or brain. Glutathione metabolism, through its impact on redox environment or redox-independent mechanisms, interferes with multiple mechanisms involved in ASD pathogenesis. Glutathione-mediated regulation of glutamate receptors [e.g., N-methyl-d-aspartate (NMDA) receptor], as well as the role of glutamate as a substrate for glutathione synthesis, may be involved in the regulation of glutamate excitotoxicity. However, the interaction between glutathione and glutamate in the pathogenesis of brain diseases may vary from synergism to antagonism. Modulation of glutathione is also associated with regulation of redox-sensitive transcription factors nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) and downstream signaling (proinflammatory cytokines and inducible enzymes), thus providing a significant impact on neuroinflammation. Mitochondrial dysfunction, as well as neuronal apoptosis, may also provide a significant link between glutathione metabolism and ASD. Furthermore, it has been recently highlighted that glutathione can affect and modulate DNA methylation and epigenetics. Review analysis including research studies meeting the required criteria for analysis showed statistically significant differences between the plasma GSH and GSSG levels as well as GSH:GSSG ratio in autistic patients compared with healthy individuals (P = 0.0145, P = 0.0150 and P = 0.0202, respectively). Therefore, the existing data provide a strong background on the role of the glutathione system in ASD pathogenesis. Future research is necessary to investigate the role of glutathione redox signaling in ASD, which could potentially also lead to promising therapeutics.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo I Rana, Norway.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Božena Hosnedlová
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
| | - Rene Kizek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic; CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic; Faculty of Pharmacy with Division of Laboratory Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Olga P Ajsuvakova
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| | - Margarita G Skalnaya
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| | | | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Afaf El-Ansary
- Medicinal Chemistry Department, King Saud University, Riyadh, Saudi Arabia; Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Qasem
- Autism Research and Treatment Center, Riyadh, Saudi Arabia; CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia
| | - James B Adams
- School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, USA
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Federal Research Centre of Biological Systems, Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
| |
Collapse
|
88
|
Seyedan AA, Dezfoulian O, Alirezaei M. Satureja khuzistanica Jamzad essential oil prevents doxorubicin-induced apoptosis via extrinsic and intrinsic mitochondrial pathways. Res Pharm Sci 2020; 15:481-490. [PMID: 33628290 PMCID: PMC7879789 DOI: 10.4103/1735-5362.297851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 05/10/2020] [Accepted: 10/06/2020] [Indexed: 11/06/2022] Open
Abstract
Background and purpose: In addition to hepato-cardiotoxicity, doxorubicin (DOX) also induces nephrotoxicity which is considered as the limiting factor for this drug in cancer therapy. The effect of carvacrol, the main active ingredient of Satureja khuzistanica Jamzad essential oil (SKEO), in the amelioration of DOX- induced cardiotoxicity is well established. The aim of the present study was to evaluate the possible protective effects of SKEO against DOX-induced nephrotoxicity. Experimental approach: SKEO was intraperitoneally administered at 50, 100, and 200 mg/kg to male Wistar rats for 12 consecutive days. Five groups of animals including negative control (saline), vehicle (Tween® 20), SKEO50, DOX (at 8th day of treatment), and SKEO50 + DOX were assessed. Findings/Results: Creatinine, urea concentrations, and caspase-3 activity significantly elevated in the serum of DOX treated group in contrast to other groups after injection of a single dose of DOX (20 mg/kg i.p.), however, SKEO reduced glutathione peroxidase and caspase-3 activity against other groups while SKEO + DOX was also significantly reduced caspase-3 activity against DOX group. Other biochemical markers changes were not significant. Immunohistochemical assessment unveiled that SKEO + DOX improved the activity of Bcl-2 family proteins (Bax and Bcl-2) and caspase-8 protein to the advantage of cell survival in both intrinsic mitochondrial and extrinsic pathway down streamed to the terminal caspase-3 apoptotic molecule., Conclusion and implications: It was concluded that SKEO could have influential effects against apoptosis induced by DOX, but not improperly ameliorate oxidative stress.
Collapse
Affiliation(s)
- Ali Al Seyedan
- Department of Pathobiology, School of Veterinary Medicine, Lorestan University, Khorramabad, I.R. Iran
| | - Omid Dezfoulian
- Department of Pathobiology, School of Veterinary Medicine, Lorestan University, Khorramabad, I.R. Iran
| | - Masoud Alirezaei
- Department of Biochemistry, School of Veterinary Medicine, Lorestan University, Khorramabad, I.R. Iran
| |
Collapse
|
89
|
Alkan Uçkun A, Barım Öz Ö. Acute exposure to the fungicide penconazole affects some biochemical parameters in the crayfish (Astacus leptodactylus Eschscholtz, 1823). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:35626-35637. [PMID: 32601870 DOI: 10.1007/s11356-020-09595-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/04/2020] [Indexed: 06/11/2023]
Abstract
Penconazole is one of the most widely used fungicides all over the world, and since it spreads to large environments, its toxic effects on non-target organisms are of great concern. The toxic effects of penconazole on crayfish (Astacus leptodactylus), which is a bioindicator in freshwater ecosystems and consumed economically, are not known. Therefore, in this study, the purpose was to contribute to the literature on the potential harmful effects of penconazole on a non-target species, Astacus leptodactylus. For this aim, the acute toxicity (96 h) of penconazole was examined. The 96-h LC50 value of penconazole was detected as 18.7 mg L-1. Four concentrations of penconazole (18.7 mg L-1, 9.35 mg L-1, 4.68 mg L-1, 2.34 mg L-1) were applied to crayfish for 96 h. The results showed that penconazole had destructive effects on esterase mechanisms by inhibiting acetylcholinesterase (AChE) and carboxylesterase (CaE) activities. Significant increases were observed in all antioxidant parameters (superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), glutathione S-transferase (GST), reduced glutathione (GSH), malondialdehyde (MDA)) in all doses except the lowest concentration (2.34 mg L-1). All adenosine triphosphatase (ATPase) activities (Na+/K+-ATPase, Mg2+-ATPase, Ca2+-ATPase, total ATPase) had significant dose-related inhibition in both gill and muscle tissues. In summary, our findings show that acute penconazole administration to crayfish causes significant toxic effects on esterase, antioxidative parameters, and metabolic enzymes.
Collapse
Affiliation(s)
- Aysel Alkan Uçkun
- Department of Environmental Engineering, Faculty of Engineering, Adıyaman University, Altınşehir neighborhood, Ataturk Boulevard, No. 1, Central Campus, 02040, Adıyaman, Turkey.
| | - Özden Barım Öz
- Department of Physiology, Faculty of Aquaculture, Fırat University, Elazığ, Turkey
| |
Collapse
|
90
|
Farghali M, Ruga S, Morsanuto V, Uberti F. Can Brain Health Be Supported by Vitamin D-Based Supplements? A Critical Review. Brain Sci 2020; 10:brainsci10090660. [PMID: 32972010 PMCID: PMC7563709 DOI: 10.3390/brainsci10090660] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
This review presents recent knowledge on the neuroprotective effects of vitamin D and their usefulness as oral supplementation when combined with other molecules, such as curcumin. A critical look at the effectiveness of vitamin D in this field is also provided. Vitamin D plays a crucial role in neuroprotection and in the cognitive decline associated with aging, where vitamin D’s levels are related to the levels of several neurotrophic factors. An important role of vitamin D has also been observed in the mechanism of neuroinflammation, which is the basis of several aging conditions, including cognitive decline and neurodegeration; furthermore, the neuroprotective effect of vitamin D in the cognitive decline of aging has recently been reported. For this reason, many food supplements created for humans contain vitamin D alone or combined with other molecules with antioxidant properties. However, recent studies also explored negative consequences of the use at a high dosage of vitamin D. Vitamin D in tissues or brain cells can also modulate calbindin-D28K, parvalbumin, and calretinin, and is involved in immune function, thanks also to the combination with curcumin. Curcumin acts as a free radical scavenger and antioxidant, inhibiting lipid peroxidation and oxidative DNA damage. In particular, curcumin is a potent immune-regulatory agent and its administration has been reported to attenuate cognitive impairments. These effects could be exploited in the future to control the mechanisms that lead to the brain decay typical of neurodegenerative diseases.
Collapse
|
91
|
Yoshioka Y, Negoro R, Kadoi H, Motegi T, Shibagaki F, Yamamuro A, Ishimaru Y, Maeda S. Noradrenaline protects neurons against H 2 O 2 -induced death by increasing the supply of glutathione from astrocytes via β 3 -adrenoceptor stimulation. J Neurosci Res 2020; 99:621-637. [PMID: 32954502 DOI: 10.1002/jnr.24733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 11/06/2022]
Abstract
Oxidative stress has been implicated in a variety of neurodegenerative disorders, such as Alzheimer's and Parkinson's disease. Astrocytes play a significant role in maintaining survival of neurons by supplying antioxidants such as glutathione (GSH) to neurons. Recently, we found that noradrenaline increased the intracellular GSH concentration in astrocytes via β3 -adrenoceptor stimulation. These observations suggest that noradrenaline protects neurons from oxidative stress-induced death by increasing the supply of GSH from astrocytes to neurons via the stimulation of β3 -adrenoceptor in astrocytes. In the present study, we examined the protective effect of noradrenaline against H2 O2 -induced neurotoxicity using two different mixed cultures: the mixed culture of human astrocytoma U-251 MG cells and human neuroblastoma SH-SY5Y cells, and the mouse primary cerebrum mixed culture of neurons and astrocytes. H2 O2 -induced neuronal cell death was significantly attenuated by pretreatment with noradrenaline in both mixed cultures but not in single culture of SH-SY5Y cells or in mouse cerebrum neuron-rich culture. The neuroprotective effect of noradrenaline was inhibited by SR59230A, a selective β3 -adrenoceptor antagonist, and CL316243, a selective β3 -adrenoceptor agonist, mimicked the neuroprotective effect of noradrenaline. DL-buthionine-[S,R]-sulfoximine, a GSH synthesis inhibitor, negated the neuroprotective effect of noradrenaline in both mixed cultures. MK571, which inhibits the export of GSH from astrocytes mediated by multidrug resistance-associated protein 1, also prevented the neuroprotective effect of noradrenaline. These results suggest that noradrenaline protects neurons against H2 O2 -induced death by increasing the supply of GSH from astrocytes via β3 -adrenoceptor stimulation.
Collapse
Affiliation(s)
- Yasuhiro Yoshioka
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Ryosuke Negoro
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Hisatsugu Kadoi
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Toshiki Motegi
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Fumiya Shibagaki
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Akiko Yamamuro
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Yuki Ishimaru
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Sadaaki Maeda
- Laboratory of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| |
Collapse
|
92
|
Dalangin R, Kim A, Campbell RE. The Role of Amino Acids in Neurotransmission and Fluorescent Tools for Their Detection. Int J Mol Sci 2020; 21:E6197. [PMID: 32867295 PMCID: PMC7503967 DOI: 10.3390/ijms21176197] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Neurotransmission between neurons, which can occur over the span of a few milliseconds, relies on the controlled release of small molecule neurotransmitters, many of which are amino acids. Fluorescence imaging provides the necessary speed to follow these events and has emerged as a powerful technique for investigating neurotransmission. In this review, we highlight some of the roles of the 20 canonical amino acids, GABA and β-alanine in neurotransmission. We also discuss available fluorescence-based probes for amino acids that have been shown to be compatible for live cell imaging, namely those based on synthetic dyes, nanostructures (quantum dots and nanotubes), and genetically encoded components. We aim to provide tool developers with information that may guide future engineering efforts and tool users with information regarding existing indicators to facilitate studies of amino acid dynamics.
Collapse
Affiliation(s)
- Rochelin Dalangin
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.D.); (A.K.)
| | - Anna Kim
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.D.); (A.K.)
| | - Robert E. Campbell
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; (R.D.); (A.K.)
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo City, Tokyo 113-0033, Japan
| |
Collapse
|
93
|
Michalska P, León R. When It Comes to an End: Oxidative Stress Crosstalk with Protein Aggregation and Neuroinflammation Induce Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9080740. [PMID: 32806679 PMCID: PMC7463521 DOI: 10.3390/antiox9080740] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are characterized by a progressive loss of neurons in the brain or spinal cord that leads to a loss of function of the affected areas. The lack of effective treatments and the ever-increasing life expectancy is raising the number of individuals affected, having a tremendous social and economic impact. The brain is particularly vulnerable to oxidative damage given the high energy demand, low levels of antioxidant defenses, and high levels of metal ions. Driven by age-related changes, neurodegeneration is characterized by increased oxidative stress leading to irreversible neuronal damage, followed by cell death. Nevertheless, neurodegenerative diseases are known as complex pathologies where several mechanisms drive neuronal death. Herein we discuss the interplay among oxidative stress, proteinopathy, and neuroinflammation at the early stages of neurodegenerative diseases. Finally, we discuss the use of the Nrf2-ARE pathway as a potential therapeutic strategy based on these molecular mechanisms to develop transformative medicines.
Collapse
Affiliation(s)
- Patrycja Michalska
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Correspondence: (P.M.); (R.L.); Tel.: +34-91-497-27-66 (P.M. & R.L.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28006 Madrid, Spain
- Correspondence: (P.M.); (R.L.); Tel.: +34-91-497-27-66 (P.M. & R.L.)
| |
Collapse
|
94
|
Mahoney-Sánchez L, Bouchaoui H, Ayton S, Devos D, Duce JA, Devedjian JC. Ferroptosis and its potential role in the physiopathology of Parkinson's Disease. Prog Neurobiol 2020; 196:101890. [PMID: 32726602 DOI: 10.1016/j.pneurobio.2020.101890] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/15/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's Disease (PD) is a common and progressive neurodegenerative disorder characterised by motor impairments as well as non-motor symptoms. While dopamine-based therapies are effective in fighting the symptoms in the early stages of the disease, a lack of neuroprotective drugs means that the disease continues to progress. Along with the traditionally recognised pathological hallmarks of dopaminergic neuronal death and intracellular α-synuclein (α-syn) depositions, iron accumulation, elevated oxidative stress and lipid peroxidation damage are further conspicuous features of PD pathophysiology. However, the underlying mechanisms linking these pathological hallmarks with neurodegeneration still remain unclear. Ferroptosis, a regulated iron dependent cell death pathway involving a lethal accumulation of lipid peroxides, shares several features with PD pathophysiology. Interestingly, α-syn has been functionally linked with the metabolism of both iron and lipid, suggesting a possible interplay between dysregulated α-syn and other PD pathological hallmarks related to ferroptosis. This review will address the importance for understanding these disease mechanisms that could be targeted therapeutically. Anti-ferroptosis molecules are neuroprotective in PD animal models and the anti-ferroptotic iron chelator, deferiprone, slowed disease progression and improved motor function in two independent clinical trials for PD. An ongoing larger multi-centre phase 2 clinical trial will confirm the therapeutic potential of deferiprone and the relevance of ferroptosis in PD. This review addresses the known pathological features of PD in relation to the ferroptosis pathway with therapeutic implications of targeting this cell death pathway.
Collapse
Affiliation(s)
- Laura Mahoney-Sánchez
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1172, University Hospital Centre, LICEND COEN Centre, LilNCog - Lille Neuroscience & Cognition, 59000, France
| | - Hind Bouchaoui
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1172, University Hospital Centre, LICEND COEN Centre, LilNCog - Lille Neuroscience & Cognition, 59000, France
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria, 3052, Australia
| | - David Devos
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1172, University Hospital Centre, LICEND COEN Centre, LilNCog - Lille Neuroscience & Cognition, 59000, France.
| | - James A Duce
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria, 3052, Australia; ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0AH, United Kingdom.
| | - Jean-Christophe Devedjian
- Department of Medical Pharmacology, Lille University, INSERM UMRS_1172, University Hospital Centre, LICEND COEN Centre, LilNCog - Lille Neuroscience & Cognition, 59000, France; Université du Littoral Côte d'Opale-1, place de l'Yser, BP 72033, 59375, Dunkerque Cedex, France
| |
Collapse
|
95
|
Rutkoski CF, Macagnan N, Folador A, Skovronski VJ, do Amaral AMB, Leitemperger J, Costa MD, Hartmann PA, Müller C, Loro VL, Hartmann MT. Morphological and biochemical traits and mortality in Physalaemus gracilis (Anura: Leptodactylidae) tadpoles exposed to the insecticide chlorpyrifos. CHEMOSPHERE 2020; 250:126162. [PMID: 32092566 DOI: 10.1016/j.chemosphere.2020.126162] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 06/10/2023]
Abstract
Organophosphate insecticides such as chlorpyrifos are commonly detected in surface waters around the world, where they are highly toxic to many organisms. The frog Physalaemus gracilis uses water sources located in open fields as reproductive sites, where it is exposed to insecticides. The study aimed to evaluate the lethal and sublethal effect of a commercial chlorpyrifos formulation on P. gracilis tadpoles (Anura: Leptodactylidae). In acute toxicity tests, five chlorpyrifos concentrations between 750 and 2,000 μg L-1 were tested. Chronic toxicity, swimming activity, morphological and enzymatic changes, as well as levels of non-protein thiols (NPSH), carbonyl proteins and lipid peroxidation were evaluated at five insecticide concentrations between 11 and 500 μg L-1. The highest mortality rate of P. gracilis tadpoles occurred at 24 and 48 h, with an LC50 of 893.59 μg L-1. At all chlorpyrifos concentrations, tadpoles displayed reduced mobility and spasms. Morphological anomalies were observed in the mouth and intestine, especially at the highest concentrations used. Acetylcholinesterase activity decreased at 250 and 500 μg L-1, catalase activity increased at all concentrations, and superoxide dismutase and glutathione S-transferase increased from 90 μg L-1 to 30 μg L-1, respectively. We also observed increases in NPSH levels at chlorpyrifos concentration starting at 30 μg L-1 and increases in carbonyl proteins from 90 μg L-1 of pesticide. Taken together, these data suggest that the insecticide chlorpyrifos presents acute and chronic risks for P. gracilis, causing neurotoxic effects and oxidative damage, culminating in high risk for this species.
Collapse
Affiliation(s)
- Camila F Rutkoski
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Natani Macagnan
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Alexandre Folador
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Vrandrieli J Skovronski
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Aline M B do Amaral
- Biochemistry and Molecular Biology Department, Federal University of Santa Maria, Camobi Campus, Av Roraima, No 1423, 97105-340, Camobi, RS, Brazil.
| | - Jossiele Leitemperger
- Biochemistry and Molecular Biology Department, Federal University of Santa Maria, Camobi Campus, Av Roraima, No 1423, 97105-340, Camobi, RS, Brazil.
| | - Maiara D Costa
- Biochemistry and Molecular Biology Department, Federal University of Santa Maria, Camobi Campus, Av Roraima, No 1423, 97105-340, Camobi, RS, Brazil.
| | - Paulo A Hartmann
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Caroline Müller
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| | - Vania L Loro
- Biochemistry and Molecular Biology Department, Federal University of Santa Maria, Camobi Campus, Av Roraima, No 1423, 97105-340, Camobi, RS, Brazil.
| | - Marilia T Hartmann
- Ecology and Conservation Laboratory, Federal University of Fronteira Sul, Erechim Campus, RS 135 - Km 72, No 200, 99700-000, Erechim, RS, Brazil.
| |
Collapse
|
96
|
Zhou Y, Chen Q, Wang Y, Wu H, Xu W, Pan Y, Gao S, Dong X, Zhang JH, Shao A. Persistent Neurovascular Unit Dysfunction: Pathophysiological Substrate and Trigger for Late-Onset Neurodegeneration After Traumatic Brain Injury. Front Neurosci 2020; 14:581. [PMID: 32581697 PMCID: PMC7296179 DOI: 10.3389/fnins.2020.00581] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) represents one of the major causes of death worldwide and leads to persisting neurological deficits in many of the survivors. One of the most significant long-term sequelae deriving from TBI is neurodegenerative disease, which is a group of incurable diseases that impose a heavy socio-economic burden. However, mechanisms underlying the increased susceptibility of TBI to neurodegenerative disease remain elusive. The neurovascular unit (NVU) is a functional unit composed of neurons, neuroglia, vascular cells, and the basal lamina matrix. The key role of NVU dysfunction in many central nervous system diseases has been revealed. Studies have proved the presence of prolonged structural and functional abnormalities of the NVU after TBI. Moreover, growing evidence suggests impaired NVU function is also implicated in neurodegenerative diseases. Therefore, we propose the Neurovascular Unit Dysfunction (NVUD) Hypothesis, in which the persistent NVU dysfunction is thought to underlie the development of post-TBI neurodegeneration. We deduce NVUD Hypothesis through relational inference and supporting evidence, and suggest continued NVU abnormalities following TBI serve as the pathophysiological substrate and trigger yielding chronic neuroinflammation, proteinopathies and oxidative stress, consequently leading to the progression of neurodegenerative diseases. The NVUD Hypothesis may provide potential treatment and prevention strategies for TBI and late-onset neurodegenerative diseases.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanbo Pan
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Dong
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
97
|
Nehser M, Dark J, Schweitzer D, Campbell M, Zwicker J, Hitt DM, Little H, Diaz-Correa A, Holley DC, Patel SA, Thompson CM, Bridges RJ. System X c- Antiporter Inhibitors: Azo-Linked Amino-Naphthyl-Sulfonate Analogues of Sulfasalazine. Neurochem Res 2020; 45:1375-1386. [PMID: 31754956 PMCID: PMC10688270 DOI: 10.1007/s11064-019-02901-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 01/18/2023]
Abstract
The cystine/glutamate antiporter system Xc- (SXc-) mediates the exchange of intracellular L-glutamate (L-Glu) with extracellular L-cystine (L-Cys2). Both the import of L-Cys2 and the export of L-Glu take on added significance in CNS cells, especially astrocytes. When the relative activity of SXc- overwhelms the regulatory capacity of the EAATs, the efflux of L-Glu through the antiporter can be significant enough to trigger excitotoxic pathology, as is thought to occur in glioblastoma. This has prompted considerable interest in the pharmacological specificity of SXc- and the development of inhibitors. The present study explores a series of analogues that are structurally related to sulfasalazine, a widely employed inhibitor of SXc-. We identify a number of novel aryl-substituted amino-naphthylsulfonate analogues that inhibit SXc- more potently than sulfasalazine. Interestingly, the inhibitors switch from a competitive to noncompetitive mechanism with increased length and lipophilic substitutions, a structure-activity relationship that was previously observed with aryl-substituted isoxazole. These results suggest that the two classes of inhibitors may interact with some of the same domains on the antiporter protein and that the substrate and inhibitor binding sites may be in close proximity to one another. Molecular modeling is used to explore this possibility.
Collapse
Affiliation(s)
- M Nehser
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - J Dark
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - D Schweitzer
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - M Campbell
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - J Zwicker
- Deciphera Pharmaceuticals, Lawrence, KA, 66044, USA
| | - D M Hitt
- Chemistry Department, Carroll College, Helena, MT, 56925, USA
| | - H Little
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - A Diaz-Correa
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - D C Holley
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - S A Patel
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - C M Thompson
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA
| | - R J Bridges
- Department of Biomedical and Pharmaceutical Science, Center for Structural and Functional Neuroscience, Skaggs School of Pharmacy, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
98
|
Batandier C, Poyot T, Marissal-Arvy N, Couturier K, Canini F, Roussel AM, Hininger-Favier I. Acute emotional stress and high fat/high fructose diet modulate brain oxidative damage through NrF2 and uric acid in rats. Nutr Res 2020; 79:23-34. [PMID: 32610255 DOI: 10.1016/j.nutres.2020.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/30/2020] [Accepted: 05/15/2020] [Indexed: 01/08/2023]
Abstract
Studies focusing on the interaction of dietary and acute emotional stress on oxidative stress in cortex frontal and in brain mitochondria are scarce. Dietary-induced insulin resistance, as observed in Western diets, has been associated with increased oxidative stress causing mitochondrial dysfunction. We hypothesized that acute emotional stress could be an aggravating factor by impacting redox status in cortex and brain mitochondria. Thus, the aim of the present study was to evaluate the combination of an insulin resistance inducing high-fat/high-fructose (HF/HFr) diet and acute emotional stress on brain oxidative stress in rats. We measured several oxidative stress parameters (carbonyls, FRAP, TBARS assays, GSH, GSSG, oxidized DNA, mRNA expression of redox proteins (Nrf2), and uric acid). The HF/HFr diet resulted in increased oxidative stress both in the brain mitochondria and in the frontal cortex and decreased expression of the Nrf2 gene. The emotional stress induced an oxidative response in plasma and in brain mitochondria of the control group. In the HF/HFr group it triggered an increase expression of the redox transcription factor Nrf2 and its downstream antioxidant genes. This suggests an improvement of the redox stress tolerance in response to an enhanced production of reactive oxygen species. Accordingly, a blunted oxidative effect on several markers was observed in plasma and brain of HF/HFr-stressed group. This was confirmed in a parallel study using lipopolysaccharide as a stress model. Beside the Nrf2 increase, the stress induced a stronger UA release in HF/HFr which could take a part in the redox stress.
Collapse
Affiliation(s)
- C Batandier
- Univ. Grenoble Alpes, Inserm, LBFA, 38000 Grenoble, France
| | - T Poyot
- Institut de Recherche Biomédicale des Armées (IRBA), BP73, 91223 Brétigny-sur-Orge, Cedex, France
| | - N Marissal-Arvy
- Bordeaux University, Laboratory of Nutrition, Memory and glucocorticoid, UMR 1286, 33076 Bordeaux Cedex, France; INRA, Laboratory of Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux Cedex, France
| | - K Couturier
- Univ. Grenoble Alpes, Inserm, LBFA, 38000 Grenoble, France
| | - F Canini
- Institut de Recherche Biomédicale des Armées (IRBA), BP73, 91223 Brétigny-sur-Orge, Cedex, France; Ecole du Val de Grâce, 1 place A. Laveran, 75230 Paris, France
| | - A M Roussel
- Univ. Grenoble Alpes, Inserm, LBFA, 38000 Grenoble, France
| | | |
Collapse
|
99
|
Zoufal V, Mairinger S, Krohn M, Wanek T, Filip T, Sauberer M, Stanek J, Kuntner C, Pahnke J, Langer O. Measurement of cerebral ABCC1 transport activity in wild-type and APP/PS1-21 mice with positron emission tomography. J Cereb Blood Flow Metab 2020; 40:954-965. [PMID: 31195936 PMCID: PMC7181082 DOI: 10.1177/0271678x19854541] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/17/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022]
Abstract
Previous data suggest a possible link between multidrug resistance-associated protein 1 (ABCC1) and brain clearance of beta-amyloid (Aβ). We used PET with 6-bromo-7-[11C]methylpurine ([11C]BMP) to measure cerebral ABCC1 transport activity in a beta-amyloidosis mouse model (APP/PS1-21) and in wild-type mice aged 50 and 170 days, without and with pretreatment with the ABCC1 inhibitor MK571. One hundred seventy days-old-animals additionally underwent [11C]PiB PET scans to measure Aβ load. While baseline [11C]BMP PET scans detected no differences in the elimination slope of radioactivity washout from the brain (kelim) between APP/PS1-21 and wild-type mice of both age groups, PET scans after MK571 pretreatment revealed significantly higher kelim values in APP/PS1-21 mice than in wild-type mice aged 170 days, suggesting increased ABCC1 activity. The observed increase in kelim occurred across all investigated brain regions and was independent of the presence of Aβ plaques measured with [11C]PiB. Western blot analysis revealed a trend towards increased whole brain ABCC1 levels in 170 days-old-APP/PS1-21 mice versus wild-type mice and a significant positive correlation between ABCC1 levels and kelim. Our data point to an upregulation of ABCC1 in APP/PS1-21 mice, which may be related to an induction of ABCC1 in astrocytes as a protective mechanism against oxidative stress.
Collapse
Affiliation(s)
- Viktoria Zoufal
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Krohn
- Department of Neuro/Pathology,
University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- University of Lübeck Institute for
Experimental und Clinical Pharmacology and Toxicology Center of Brain, Behavior and
Metabolism (CBBM), Lübeck, Germany
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Claudia Kuntner
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Jens Pahnke
- Department of Neuro/Pathology,
University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- LIED, University of Lübeck,
Germany
- Leibniz-Institute of Plant Biochemistry,
Halle, Germany
- Medical Faculty, Department of
Pharmacology, University of Latvia, Rīga, Latvia
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT
Austrian Institute of Technology GmbH, Seibersdorf, Austria
- Department of Clinical Pharmacology,
Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging und
Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna,
Vienna, Austria
| |
Collapse
|
100
|
Dwivedi D, Megha K, Mishra R, Mandal PK. Glutathione in Brain: Overview of Its Conformations, Functions, Biochemical Characteristics, Quantitation and Potential Therapeutic Role in Brain Disorders. Neurochem Res 2020; 45:1461-1480. [PMID: 32297027 DOI: 10.1007/s11064-020-03030-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/28/2022]
Abstract
Glutathione (GSH) is an important antioxidant found abundantly and synthesized intracellularly in the cytosol in a tightly regulated fashion. It has diverse physiological functions, including protection against reactive oxygen species and nitrogen species, antioxidant defense as well as maintenance of cellular thiol status. The human brain due to the high oxygen consumption is extremely susceptible to the generation of reactive oxygen species. GSH plays a paramount role in brain antioxidant defense, maintaining redox homeostasis. The depletion of brain GSH has also been observed from both autopsies as well as in vivo MRS studies with aging and varied neurological disorders (Alzheimer's disease, Parkinson's disease, etc.). Therefore, GSH enrichment using supplementation is a promising avenue in the therapeutic development for these neurological disorders. This review will enrich the information on the importance of GSH synthesis, metabolism, functions, compartmentation and inter-organ transport, structural conformations and its quantitation via different techniques. The transportation of GSH in the brain via different interventional routes and its potential role in the development of therapeutic strategies for various brain disorders is also addressed. Very recent study found significant improvement of behavioral deficits including cognitive decline, depressive-like behaviors, in APP (NL-G-F/NL-G-FG-) mice due to oral GSH administration. This animal model study put an emergent need to complete GSH supplementation trial in MCI and AD patients for cognitive improvement as proposed earlier.
Collapse
Affiliation(s)
- Divya Dwivedi
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Kanu Megha
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Ritwick Mishra
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Pravat K Mandal
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Manesar, Gurgaon, Haryana, India. .,Florey Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Parkville, Melbourne, Australia.
| |
Collapse
|