1
|
Tian J, An M, Zhao X, Wang Y, Hasan M. Advances in Fluorescent Sensing Carbon Dots: An Account of Food Analysis. ACS OMEGA 2023; 8:9031-9039. [PMID: 36936334 PMCID: PMC10018703 DOI: 10.1021/acsomega.2c07986] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Illuminating the use of nanomaterials, carbon quantum dots (CQDs) have transfigured the food safety arena because of their bright luminescence, optical properties, low toxicity, and enhanced biocompatibility. Therefore, fluorescent resonance energy transfer, photoinduced electron transfer, and an internal filtering effect mechanism allow precise detection of food additives, heavy metal ions, pathogenic bacteria, veterinary drug residues, and food nutrients. In this review, we describe the primal mechanism of CQD-based fluorescence sensors for food safety inspection. This is an abridged description of the nanodesign and future perspectives of more advanced CQD-based sensors for food safety analysis.
Collapse
Affiliation(s)
- Jixiang Tian
- Institute
of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Minmei An
- Taian
Traditional Chinese Medicine Hospital, Taian 271000, China
| | - Xiaoang Zhao
- Institute
of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yun Wang
- Institute
of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Murtaza Hasan
- Faculty
of Biological and Chemical Sciences, Department of Biotechnology, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- School
of Chemistry and Chemical Engineering, Zhongkai
University of Agriculture and Engineering, Guangzhou 510225, China
| |
Collapse
|
2
|
Buck SA, De Miranda BR, Logan RW, Fish KN, Greenamyre JT, Freyberg Z. VGLUT2 Is a Determinant of Dopamine Neuron Resilience in a Rotenone Model of Dopamine Neurodegeneration. J Neurosci 2021; 41:4937-4947. [PMID: 33893220 PMCID: PMC8260163 DOI: 10.1523/jneurosci.2770-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is characterized by progressive dopamine (DA) neuron loss in the SNc. In contrast, DA neurons in the VTA are relatively protected from neurodegeneration, but the underlying mechanisms for this resilience remain poorly understood. Recent work suggests that expression of the vesicular glutamate transporter 2 (VGLUT2) selectively impacts midbrain DA neuron vulnerability. We investigated whether altered DA neuron VGLUT2 expression determines neuronal resilience in rats exposed to rotenone, a mitochondrial complex I inhibitor and toxicant model of PD. We discovered that VTA/SNc DA neurons that expressed VGLUT2 are more resilient to rotenone-induced DA neurodegeneration. Surprisingly, the density of neurons with detectable VGLUT2 expression in the VTA and SNc increases in response to rotenone. Furthermore, dopaminergic terminals within the NAc, where the majority of VGLUT2-expressing DA neurons project, exhibit greater resilience compared with DA terminals in the caudate/putamen. More broadly, VGLUT2-expressing terminals are protected throughout the striatum from rotenone-induced degeneration. Together, our data demonstrate that a distinct subpopulation of VGLUT2-expressing DA neurons are relatively protected from rotenone neurotoxicity. Rotenone-induced upregulation of the glutamatergic machinery in VTA and SNc neurons and their projections may be part of a broader neuroprotective mechanism. These findings offer a putative new target for neuronal resilience that can be manipulated to prevent toxicant-induced DA neurodegeneration in PD.SIGNIFICANCE STATEMENT Environmental exposures to pesticides contribute significantly to pathologic processes that culminate in Parkinson's disease (PD). The pesticide rotenone has been used to generate a PD model that replicates key features of the illness, including dopamine neurodegeneration. To date, longstanding questions remain: are there dopamine neuron subpopulations resilient to rotenone; and if so, what are the molecular determinants of this resilience? Here we show that the subpopulation of midbrain dopaminergic neurons that express the vesicular glutamate transporter 2 (VGLUT2) are more resilient to rotenone-induced neurodegeneration. Rotenone also upregulates VGLUT2 more broadly in the midbrain, suggesting that VGLUT2 expression generally confers increased resilience to rotenone. VGLUT2 may therefore be a new target for boosting neuronal resilience to prevent toxicant-induced DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Silas A Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - Briana R De Miranda
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, 02118
- Center for Systems Neurogenetics of Addiction, The Jackson Laboratory, Bar Harbor, Maine, 04609
| | - Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| | - J Timothy Greenamyre
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213
| |
Collapse
|
3
|
A novel 3D porous graphene foam prepared by chemical vapor deposition using nickel nanoparticles: Electrochemical determination of levodopa in the presence of uric acid. Microchem J 2019. [DOI: 10.1016/j.microc.2019.02.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
4
|
Selvakumar GP, Iyer SS, Kempuraj D, Raju M, Thangavel R, Saeed D, Ahmed ME, Zahoor H, Raikwar SP, Zaheer S, Zaheer A. Glia Maturation Factor Dependent Inhibition of Mitochondrial PGC-1α Triggers Oxidative Stress-Mediated Apoptosis in N27 Rat Dopaminergic Neuronal Cells. Mol Neurobiol 2018; 55:7132-7152. [PMID: 29383690 PMCID: PMC6066475 DOI: 10.1007/s12035-018-0882-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/08/2018] [Indexed: 02/08/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease affecting over five million individuals worldwide. The exact molecular events underlying PD pathogenesis are still not clearly known. Glia maturation factor (GMF), a neuroinflammatory protein in the brain plays an important role in the pathogenesis of PD. Mitochondrial dysfunctions and oxidative stress trigger apoptosis leading to dopaminergic neuronal degeneration in PD. Peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α or PPARGC-α) acts as a transcriptional co-regulator of mitochondrial biogenesis and energy metabolism by controlling oxidative phosphorylation, antioxidant activity, and autophagy. In this study, we found that incubation of immortalized rat dopaminergic (N27) neurons with GMF influences the expression of peroxisome PGC-1α and increases oxidative stress, mitochondrial dysfunction, and apoptotic cell death. We show that incubation with GMF reduces the expression of PGC-1α with concomitant decreases in the mitochondrial complexes. Besides, there is increased oxidative stress and depolarization of mitochondrial membrane potential (MMP) in these cells. Further, GMF reduces tyrosine hydroxylase (TH) expression and shifts Bax/Bcl-2 expression resulting in release of cytochrome-c and increased activations of effector caspase expressions. Transmission electron microscopy analyses revealed alteration in the mitochondrial architecture. Our results show that GMF acts as an important upstream regulator of PGC-1α in promoting dopaminergic neuronal death through its effect on oxidative stress-mediated apoptosis. Our current data suggest that GMF is a critical risk factor for PD and suggest that it could be explored as a potential therapeutic target to inhibit PD progression.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Murugesan Raju
- Department of Ophthalmology, University of Missouri, Columbia, MO, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Daniyal Saeed
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Harris Zahoor
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.
- Department of Neurology and The Center for Translational Neuroscience, M741A Medical Science Building, School of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, USA.
| |
Collapse
|
5
|
Mythri RB, Raghunath NR, Narwade SC, Pandareesh MDR, Sabitha KR, Aiyaz M, Chand B, Sule M, Ghosh K, Kumar S, Shankarappa B, Soundararajan S, Alladi PA, Purushottam M, Gayathri N, Deobagkar DD, Laxmi TR, Srinivas Bharath MM. Manganese- and 1-methyl-4-phenylpyridinium-induced neurotoxicity display differences in morphological, electrophysiological and genome-wide alterations: implications for idiopathic Parkinson's disease. J Neurochem 2017; 143:334-358. [DOI: 10.1111/jnc.14147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Rajeswara Babu Mythri
- Department of Neurochemistry; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
- Neurotoxicology Laboratory-Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Narayana Reddy Raghunath
- Department of Neurochemistry; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
- Neurotoxicology Laboratory-Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | | | - Mirazkar Dasharatha Rao Pandareesh
- Department of Neurochemistry; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
- Neurotoxicology Laboratory-Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Kollarkandi Rajesh Sabitha
- Department of Neurophysiology; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Mohamad Aiyaz
- Genotypic Technology Pvt. Ltd; Bangalore Karnataka India
| | - Bipin Chand
- Genotypic Technology Pvt. Ltd; Bangalore Karnataka India
| | - Manas Sule
- InterpretOmics; Shezan Lavelle; Bangalore Karnataka India
| | - Krittika Ghosh
- InterpretOmics; Shezan Lavelle; Bangalore Karnataka India
| | - Senthil Kumar
- InterpretOmics; Shezan Lavelle; Bangalore Karnataka India
| | - Bhagyalakshmi Shankarappa
- Molecular Genetics Laboratory - Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Soundarya Soundararajan
- Molecular Genetics Laboratory - Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Phalguni Anand Alladi
- Department of Neurophysiology; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Meera Purushottam
- Molecular Genetics Laboratory - Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Narayanappa Gayathri
- Department of Neuropathology; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | | | - Thenkanidiyoor Rao Laxmi
- Department of Neurophysiology; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| | - Muchukunte Mukunda Srinivas Bharath
- Department of Neurochemistry; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
- Neurotoxicology Laboratory-Neurobiology Research Center; National Institute of Mental Health and Neurosciences (NIMHANS); Bangalore Karnataka India
| |
Collapse
|
6
|
Aguirre-Vidal Y, Monroy-Noyola A, Anaya-Ramos L, Arteaga-Silva M, Mendez-Armenta M, Ostoa-Saloma P, Díaz-Zaragoza M, Morales-Montor J, Ríos C, Montes S. β-Estradiol-3-benzoate confers neuroprotection in Parkinson MPP + rat model through inhibition of lipid peroxidation. Steroids 2017; 126:7-14. [PMID: 28827046 DOI: 10.1016/j.steroids.2017.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/27/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022]
Abstract
Estradiol (E2), in addition to its known hormone function, is a neuroactive steroid that has shown neuroprotective profile in several models of neurological diseases. The present study explores the antioxidant effect of β-estradiol-3-benzoate (EB) on the neurotoxicity elicited by MPP+ in rat striatum. Male Wistar rats, that were gonadectomized 30days prior to EB, were given 100µgEB per rat every 48h for 11days and animals were infused with MPP+ via intrastriatal at day six after beginning EB treatment. EB treatment completely prevented the fall in dopamine caused by MPP+, such result was related with decreased lipid peroxidation, a marker of oxidative stress; diminished number of ipsilateral-to-lesion turns and increased signal of the dopamine-synthesizing enzyme Tyrosin Hydroxylase in substantia nigra. The protection elicited by EB was not related to Mn or Cu-Zn superoxide dismutase enzymatic activities or glutathione modulation since none of these parameters were influenced by EB at the times assayed. Whereas, increased expression of PON2 as a result of EB treatment was observed, this phenomenon could be one of the mechanism by which the steroid conferred protection to dopaminergic cells against MPP+ injury.
Collapse
Affiliation(s)
- Yoshajandith Aguirre-Vidal
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Antonio Monroy-Noyola
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Laura Anaya-Ramos
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Marcela Arteaga-Silva
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, Mexico
| | - Marisela Mendez-Armenta
- Laboratorio de Neuropatologia experimental, Instituto Nacional de Neurología y Neurocirugía, Dr. Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 Ciudad de México, Mexico
| | - Mariana Díaz-Zaragoza
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 Ciudad de México, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70228, 04510 Ciudad de México, Mexico
| | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Dr. Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Sergio Montes
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Dr. Manuel Velasco Suárez, Ciudad de México, Mexico.
| |
Collapse
|
7
|
Patel M, McElroy PB. Mitochondrial Dysfunction in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders where oxidative stress and mitochondrial dysfunction have been implicated as etiological factors. Mitochondria are the major producers of reactive oxygen species (ROS) that can have damaging effects to cellular macromolecules leading to neurodegeneration. The most compelling evidence for the role of mitochondria in the pathogenesis of PD has been derived from toxicant-induced models of parkinsonism. Over the years, epidemiological studies have suggested a link between exposure to environmental toxins such as pesticides and the risk of developing PD. Data from human and experimental studies involving the use of chemical agents like paraquat, diquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone and maneb have provided valuable insight into the underlying mitochondrial mechanisms contributing to PD and associated neurodegeneration. In this review, we have discussed the role of mitochondrial ROS and dysfunction in the pathogenesis of PD with a special focus on environmental agent-induced parkinsonism. We have described the various mitochondrial mechanisms by which such chemicals exert neurotoxicity, highlighting some landmark epidemiological and experimental studies that support the role of mitochondrial ROS and oxidative stress in contributing to these effects. Finally, we have discussed the significance of these studies in understanding the mechanistic underpinnings of PD-related dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| | - Pallavi Bhuyan McElroy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| |
Collapse
|
8
|
McElroy PB, Sri Hari A, Day BJ, Patel M. Post-translational Activation of Glutamate Cysteine Ligase with Dimercaprol: A NOVEL MECHANISM OF INHIBITING NEUROINFLAMMATION IN VITRO. J Biol Chem 2017; 292:5532-5545. [PMID: 28202547 DOI: 10.1074/jbc.m116.723700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 01/25/2017] [Indexed: 11/06/2022] Open
Abstract
Neuroinflammation and oxidative stress are hallmarks of various neurological diseases. However, whether and how the redox processes control neuroinflammation is incompletely understood. We hypothesized that increasing cellular glutathione (GSH) levels would inhibit neuroinflammation. A series of thiol compounds were identified to elevate cellular GSH levels by a novel approach (i.e. post-translational activation of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH biosynthesis). These small thiol-containing compounds were examined for their ability to increase intracellular GSH levels in a murine microglial cell line (BV2), of which dimercaprol (2,3-dimercapto-1-propanol (DMP)) was found to be the most effective compound. DMP increased GCL activity and decreased LPS-induced production of pro-inflammatory cytokines and inducible nitric-oxide synthase induction in BV2 cells in a concentration-dependent manner. The ability of DMP to elevate GSH levels and attenuate LPS-induced pro-inflammatory cytokine production was inhibited by buthionine sulfoximine, an inhibitor of GCL. DMP increased the expression of GCL holoenzyme without altering the expression of its subunits or Nrf2 target proteins (NQO1 and HO-1), suggesting a post-translational mechanism. DMP attenuated LPS-induced MAPK activation in BV2 cells, suggesting the MAPK pathway as the signaling mechanism underlying the effect of DMP. Finally, the ability of DMP to increase GSH via GCL activation was observed in mixed cerebrocortical cultures and N27 dopaminergic cells. Together, the data demonstrate a novel mechanism of GSH elevation by post-translational activation of GCL. Post-translational activation of GCL offers a novel targeted approach to control inflammation in chronic neuronal disorders associated with impaired adaptive responses.
Collapse
Affiliation(s)
- Pallavi B McElroy
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Ashwini Sri Hari
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| | - Brian J Day
- the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Manisha Patel
- From the Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045 and
| |
Collapse
|
9
|
Cunha MP, Pazini FL, Lieberknecht V, Budni J, Oliveira Á, Rosa JM, Mancini G, Mazzardo L, Colla AR, Leite MC, Santos ARS, Martins DF, de Bem AF, Gonçalves CAS, Farina M, Rodrigues ALS. MPP +-Lesioned Mice: an Experimental Model of Motor, Emotional, Memory/Learning, and Striatal Neurochemical Dysfunctions. Mol Neurobiol 2016; 54:6356-6377. [PMID: 27722926 DOI: 10.1007/s12035-016-0147-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
Abstract
The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces motor and nonmotor dysfunctions resembling Parkinson's disease (PD); however, studies investigating the effects of 1-methyl-4-phenylpyridinium (MPP+), an active oxidative product of MPTP, are scarce. This study investigated the behavioral and striatal neurochemical changes (related to oxidative damage, glial markers, and neurotrophic factors) 24 h after intracerebroventricular administration of MPP+ (1.8-18 μg/mouse) in C57BL6 mice. MPP+ administration at high dose (18 μg/mouse) altered motor parameters, since it increased the latency to leave the first quadrant and reduced crossing, rearing, and grooming responses in the open-field test and decreased rotarod latency time. MPP+ administration at low dose (1.8 μg/mouse) caused specific nonmotor dysfunctions as it produced a depressive-like effect in the forced swim test and tail suspension test, loss of motivational and self-care behavior in the splash test, anxiety-like effect in the elevated plus maze test, and short-term memory deficit in the step-down inhibitory avoidance task, without altering ambulation. MPP+ at doses of 1.8-18 μg/mouse increased tyrosine hydroxylase (TH) immunocontent and at 18 μg/mouse increased α-synuclein and decreased parkin immunocontent. The astrocytic calcium-binding protein S100B and glial fibrillary acidic protein (GFAP)/S100B ratio was decreased following MPP+ administration (18 μg/mouse). At this highest dose, MPP+ increased the ionized calcium-binding adapter molecule 1 (Iba-1) immunocontent, suggesting microglial activation. Also, MPP+ at a dose of 18 μg/mouse increased thiobarbituric acid reactive substances (TBARS) and glutathione (GSH) levels and increased glutathione peroxidase (GPx) and hemeoxygenase-1 (HO-1) immunocontent, suggesting a significant role for oxidative stress in the MPP+-induced striatal damage. MPP+ (18 μg/mouse) also increased striatal fibroblast growth factor 2 (FGF-2) and brain-derived neurotrophic factor (BDNF) levels. Moreover, MPP+ decreased tropomyosin receptor kinase B (TrkB) immunocontent. Finally, MPP+ (1.8-18 μg/mouse) increased serum corticosterone levels and did not alter acetylcholinesterase (AChE) activity in the striatum but increased it in cerebral cortex and hippocampus. Collectively, these results indicate that MPP+ administration at low doses may be used as a model of emotional and memory/learning behavioral deficit related to PD and that MPP+ administration at high dose could be useful for analysis of striatal dysfunctions associated with motor deficits in PD.
Collapse
Affiliation(s)
- Mauricio P Cunha
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| | - Francis L Pazini
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Vicente Lieberknecht
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Josiane Budni
- Laboratory of Neurosciences, National Institute for Translational Medicine, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Ágatha Oliveira
- Department of Biochemistry, Institute of Chemistry, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Júlia M Rosa
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Gianni Mancini
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Leidiane Mazzardo
- Department of Morphological Sciences, Center of Biological Science, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - André R Colla
- Centro Universitário Municipal de São José, São José, SC, Brazil
| | - Marina C Leite
- Department of Biochemistry, Institute of Basic Health Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Adair R S Santos
- Department of Physiological Sciences, Center of Biological Science, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Daniel F Martins
- Graduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Pedra Branca, Palhoça, SC, 88137-270, Brazil
| | - Andreza F de Bem
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Carlos Alberto S Gonçalves
- Department of Biochemistry, Institute of Basic Health Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| |
Collapse
|
10
|
Analysis of glutathione levels in the brain tissue samples from HIV-1-positive individuals and subject with Alzheimer's disease and its implication in the pathophysiology of the disease process. BBA CLINICAL 2016; 6:38-44. [PMID: 27335804 PMCID: PMC4908271 DOI: 10.1016/j.bbacli.2016.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 02/07/2023]
Abstract
HIV-1 positive individuals are at high risk for susceptibility to both pulmonary tuberculosis (TB) and extra-pulmonary TB, including TB meningitis (TBM) which is an extreme form of TB. The goals of this study are to determine the mechanisms responsible for compromised levels of glutathione (GSH) in the brain tissue samples derived from HIV-1-infected individuals and individuals with Alzheimer's disease (AD), investigate the possible underlying mechanisms responsible for GSH deficiency in these pathological conditions, and establish a link between GSH levels and pathophysiology of the disease processes. We demonstrated in the autopsied human brain tissues that the levels of total and reduced forms of GSH were significantly compromised in HIV-1 infected individuals compared to in healthy subjects and individuals with AD. Brain tissue samples derived from HIV-1-positive individuals had substantially higher levels of free radicals than that derived from healthy and AD individuals. Enzymes that are responsible for the de novo synthesis of GSH such as γ-glutamate cysteine-ligase catalytic subunit (GCLC-rate limiting step enzyme) and glutathione synthetase (GSS-enzyme involved in the second step reaction) were significantly decreased in the brain tissue samples derived from HIV-1-positive individuals with low CD4 + T-cells (< 200 cells/mm3) compared to healthy and AD individuals. Levels of glutathione reductase (GSR) were also decreased in the brain tissue samples derived from HIV-1 infected individuals. Overall, our findings demonstrate causes for GSH deficiency in the brain tissue from HIV-1 infected individuals explaining the possible reasons for increased susceptibility to the most severe form of extra-pulmonary TB, TBM. Total and reduced forms of GSH were significantly compromised in the brain tissues derived from HIV-1 infected individuals. Brain tissue samples derived from HIV-1-positive individuals had substantially higher levels of free radicals. GSH de novo synthesis enzymes were significantly decreased in HIV-1-positive individuals with low CD4 + T-cells. Levels of GSR were also decreased in the brain tissue samples derived from HIV-1 infected individuals. Overall, our findings demonstrate causes for GSH deficiency in the brain tissue from HIV-1 infected individuals.
Collapse
|
11
|
Vinpocetine attenuates MPTP-induced motor deficit and biochemical abnormalities in Wistar rats. Neuroscience 2014; 286:393-403. [PMID: 25514048 DOI: 10.1016/j.neuroscience.2014.12.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/26/2014] [Accepted: 12/07/2014] [Indexed: 12/28/2022]
Abstract
Up-regulation in phosphodiesterase 1 (PDE1) expression and decreased levels of cyclic nucleotides (cAMP and cGMP) have been reported in patients and experimental animal models of Parkinson's disease (PD). Phosphodiesterase (PDE) inhibitors have been reported to be beneficial in cognitive and motor deficit states. The present study is designed to investigate the effect of vinpocetine, a PDE1 inhibitor in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced experimental PD-like symptoms in rats. To produce stable motor deficit, MPTP was repeatedly administered intranigrally (bilaterally) at an interval of 1 week (days 1, 7 and 14). Following development of stable motor deficit, which was observed after the third infusion of MPTP (day 14) in rats, the animals were treated with vinpocetine (5-, 10- and 20-mg/kg, i.p.) from days 15 to 28. Movement abnormalities were assessed by a battery of behavioral tests. Moreover, levels of malondialdehyde, nitrite and reduced glutathione were measured in striatal brain homogenate to confirm the role of oxidative and nitrosative stress in PD. Repeated intranigral administration of MPTP produced stable motor deficits, reduced the cyclic nucleotides and dopamine levels and caused elevation in oxidative-nitrosative stress markers. Chronic administration of vinpocetine (for 14 days) significantly and dose dependently attenuated movement disabilities and oxidative-nitrosative stress in MPTP-treated rats. Moreover, vinpocetine treatment enhances cyclic nucleotide levels and restores the dopamine level in MPTP-treated rats. The observed results of the present study are indicative of the therapeutic potential of vinpocetine in PD.
Collapse
|
12
|
Chorley B, Ward W, Simmons SO, Vallanat B, Veronesi B. The cellular and genomic response of rat dopaminergic neurons (N27) to coated nanosilver. Neurotoxicology 2014; 45:12-21. [PMID: 25194297 DOI: 10.1016/j.neuro.2014.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/21/2014] [Accepted: 08/25/2014] [Indexed: 01/27/2023]
Abstract
This study examined if nanosilver (nanoAg) of different sizes and coatings were differentially toxic to oxidative stress-sensitive neurons. N27 rat dopaminergic neurons were exposed (0.5-5 ppm) to a set of nanoAg of different sizes (10nm, 75 nm) and coatings (PVP, citrate) and their physicochemical, cellular and genomic response measured. Both coatings retained their manufactured sizes in culture media, however, the zeta potentials of both sizes of PVP-coated nanoAg were significantly less electronegative than those of their citrate-coated counterparts. Markers of oxidative stress, measured at 0.5-5 ppm exposure concentrations, indicated that caspase 3/7 activity and glutathione levels were significantly increased by both sizes of PVP-coated nanoAg and by the 75 nm citrate-coated nanoAg. Both sizes of PVP-coated nanoAg also increased intra-neuronal nitrite levels and activated ARE/NRF2, a reporter gene for the oxidative stress-protection pathway. Global gene expression on N27 neurons, exposed to 0.5 ppm for 8h, indicated a dominant effect by PVP-coated nanoAg over citrate. The 75 nm PVP-coated material altered 196 genes that were loosely associated with mitochondrial dysfunction. In contrast, the 10nm PVP-coated nanoAg altered 82 genes that were strongly associated with NRF2 oxidative stress pathways. Less that 20% of the affected genes were shared by both sizes of PVP-coated nanoAg. These cellular and genomic findings suggest that PVP-coated nanoAg is more bioactive than citrate-coated nanoAg. Although both sizes of PVP-coated nanoAg altered the genomic expression of N27 neurons along oxidative stress pathways, exposure to the 75 nm nanoAg favored pathways associated with mitochondrial dysfunction, whereas the 10nm PVP-coated nanoAg affected NRF2 neuronal protective pathways.
Collapse
Affiliation(s)
- Brian Chorley
- National Health and Environmental Effects Research Laboratory (Integrated Systems Toxicology Division), United States
| | - William Ward
- Research Genomics Core, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Steven O Simmons
- National Health and Environmental Effects Research Laboratory (Integrated Systems Toxicology Division), United States
| | - Beena Vallanat
- Research Genomics Core, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Bellina Veronesi
- National Health and Environmental Effects Research Laboratory (Integrated Systems Toxicology Division), United States.
| |
Collapse
|
13
|
Transcriptional and metabolic adaptation of human neurons to the mitochondrial toxicant MPP(+). Cell Death Dis 2014; 5:e1222. [PMID: 24810058 PMCID: PMC4047858 DOI: 10.1038/cddis.2014.166] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 12/20/2022]
Abstract
Assessment of the network of toxicity pathways by Omics technologies and bioinformatic data processing paves the road toward a new toxicology for the twenty-first century. Especially, the upstream network of responses, taking place in toxicant-treated cells before a point of no return is reached, is still little explored. We studied the effects of the model neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) by a combined metabolomics (mass spectrometry) and transcriptomics (microarrays and deep sequencing) approach to provide unbiased data on earliest cellular adaptations to stress. Neural precursor cells (LUHMES) were differentiated to homogeneous cultures of fully postmitotic human dopaminergic neurons, and then exposed to the mitochondrial respiratory chain inhibitor MPP+ (5 μM). At 18–24 h after treatment, intracellular ATP and mitochondrial integrity were still close to control levels, but pronounced transcriptome and metabolome changes were seen. Data on altered glucose flux, depletion of phosphocreatine and oxidative stress (e.g., methionine sulfoxide formation) confirmed the validity of the approach. New findings were related to nuclear paraspeckle depletion, as well as an early activation of branches of the transsulfuration pathway to increase glutathione. Bioinformatic analysis of our data identified the transcription factor ATF-4 as an upstream regulator of early responses. Findings on this signaling pathway and on adaptive increases of glutathione production were confirmed biochemically. Metabolic and transcriptional profiling contributed complementary information on multiple primary and secondary changes that contribute to the cellular response to MPP+. Thus, combined ‘Omics' analysis is a new unbiased approach to unravel earliest metabolic changes, whose balance decides on the final cell fate.
Collapse
|
14
|
Neuroprotective effects of a variety of pomegranate juice extracts against MPTP-induced cytotoxicity and oxidative stress in human primary neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:685909. [PMID: 24223235 PMCID: PMC3816068 DOI: 10.1155/2013/685909] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/02/2013] [Accepted: 08/02/2013] [Indexed: 02/05/2023]
Abstract
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is an environmental toxin which selectively induces oxidative damage and mitochondrial and proteasomal dysfunctions to dopaminergic neurons in the substantia nigra leading to Parkinsonian syndrome in animal models and humans. MPTP is one of the most widely used in vitro models to investigate the pathophysiology of Parkinson's disease (PD) and, screen for novel therapeutic compounds that can slow down or ameliorate this progressive degenerative disease. We investigated the therapeutic effect of pomegranate juice extracts (PJE), Helow, Malasi, Qusum, and Hamadh against MPTP-induced neurotoxicity in primary human neurons by examining extracellular LDH activity, intracellular NAD+ and ATP levels, and endogenous antioxidant levels including lipid peroxidation products, catalase, superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities, and reduced glutathione (GSH) levels. MPTP induced a reduction in SOD and GPx activities and intracellular NAD+, ATP, and GSH levels parallel to an increase in extracellular LDH and CAT activities, although lipid peroxidation was not altered. We report that helow and malasi can ameliorate MPTP-induced neurotoxicity by attenuating the observed changes in redox function to a greater extent than qusum and hamedh. Selected PJE varieties may exhibit properties which may be of therapeutic value to slow down age-related degeneration and neurodegeneration in particular.
Collapse
|
15
|
Garcia-Garcia A, Zavala-Flores L, Rodriguez-Rocha H, Franco R. Thiol-redox signaling, dopaminergic cell death, and Parkinson's disease. Antioxid Redox Signal 2012; 17:1764-84. [PMID: 22369136 PMCID: PMC3474187 DOI: 10.1089/ars.2011.4501] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Parkinson's disease (PD) is characterized by the selective loss of dopaminergic neurons of the substantia nigra pars compacta, which has been widely associated with oxidative stress. However, the mechanisms by which redox signaling regulates cell death progression remain elusive. RECENT ADVANCES Early studies demonstrated that depletion of glutathione (GSH), the most abundant low-molecular-weight thiol and major antioxidant defense in cells, is one of the earliest biochemical events associated with PD, prompting researchers to determine the role of oxidative stress in dopaminergic cell death. Since then, the concept of oxidative stress has evolved into redox signaling, and its complexity is highlighted by the discovery of a variety of thiol-based redox-dependent processes regulating not only oxidative damage, but also the activation of a myriad of signaling/enzymatic mechanisms. CRITICAL ISSUES GSH and GSH-based antioxidant systems are important regulators of neurodegeneration associated with PD. In addition, thiol-based redox systems, such as peroxiredoxins, thioredoxins, metallothioneins, methionine sulfoxide reductases, transcription factors, as well as oxidative modifications in protein thiols (cysteines), including cysteine hydroxylation, glutathionylation, and nitrosylation, have been demonstrated to regulate dopaminergic cell loss. FUTURE DIRECTIONS In this review, we summarize major advances in the understanding of the role of thiol-redox signaling in dopaminergic cell death in experimental PD. Future research is still required to clearly understand how integrated thiol-redox signaling regulates the activation of the cell death machinery, and the knowledge generated should open new avenues for the design of novel therapeutic approaches against PD.
Collapse
Affiliation(s)
- Aracely Garcia-Garcia
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | | | | | | |
Collapse
|
16
|
Abstract
SIGNIFICANCE Glutathione (GSH) depletion is a central signaling event that regulates the activation of cell death pathways. GSH depletion is often taken as a marker of oxidative stress and thus, as a consequence of its antioxidant properties scavenging reactive species of both oxygen and nitrogen (ROS/RNS). RECENT ADVANCES There is increasing evidence demonstrating that GSH loss is an active phenomenon regulating the redox signaling events modulating cell death activation and progression. CRITICAL ISSUES In this work, we review the role of GSH depletion by its efflux, as an important event regulating alterations in the cellular redox balance during cell death independent from oxidative stress and ROS/RNS formation. We discuss the mechanisms involved in GSH efflux during cell death progression and the redox signaling events by which GSH depletion regulates the activation of the cell death machinery. FUTURE DIRECTIONS The evidence summarized here clearly places GSH transport as a central mechanism mediating redox signaling during cell death progression. Future studies should be directed toward identifying the molecular identity of GSH transporters mediating GSH extrusion during cell death, and addressing the lack of sensitive approaches to quantify GSH efflux.
Collapse
Affiliation(s)
- Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | | |
Collapse
|
17
|
Singh M, Murthy V, Ramassamy C. Standardized Extracts of Bacopa monniera Protect Against MPP+- and Paraquat-Induced Toxicity by Modulating Mitochondrial Activities, Proteasomal Functions, and Redox Pathways. Toxicol Sci 2011; 125:219-32. [DOI: 10.1093/toxsci/kfr255] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
18
|
Cantu D, Fulton RE, Drechsel DA, Patel M. Mitochondrial aconitase knockdown attenuates paraquat-induced dopaminergic cell death via decreased cellular metabolism and release of iron and H₂O₂. J Neurochem 2011; 118:79-92. [PMID: 21517855 PMCID: PMC3182850 DOI: 10.1111/j.1471-4159.2011.07290.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondrial oxidative stress is a contributing factor in the etiology of numerous neuronal disorders. However, the precise mechanism(s) by which mitochondrial reactive oxygen species modify cellular targets to induce neurotoxicity remains unknown. In this study, we determined the role of mitochondrial aconitase (m-aconitase) in neurotoxicity by decreasing its expression. Incubation of the rat dopaminergic cell line, N27, with paraquat (PQ(2+) ) resulted in aconitase inactivation, increased hydrogen peroxide (H(2) O(2) ) and increased ferrous iron (Fe(2+) ) at times preceding cell death. To confirm the role of m-aconitase in dopaminergic cell death, we knocked down m-aconitase expression via RNA interference. Incubation of m-aconitase knockdown N27 cells with PQ(2+) resulted in decreased H(2) O(2) production, Fe(2+) accumulation, and cell death compared with cells expressing basal levels of m-aconitase. To determine the metabolic role of m-aconitase in mediating neuroprotection, we conducted a complete bioenergetic profile. m-Aconitase knockdown N27 cells showed a global decrease in metabolism (glycolysis and oxygen consumption rates) which blocked PQ(2+) -induced H(+) leak and respiratory capacity deficiency. These findings suggest that dopaminergic cells are protected from death by decreasing release of H(2) O(2) and Fe(2+) in addition to decreased cellular metabolism.
Collapse
Affiliation(s)
- David Cantu
- Graduate Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
- Department of Neuroscience, Tufts University School of Medicine 136 Harrison Ave., SC201, Boston, MA 02111
| | - Ruth E. Fulton
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Derek A. Drechsel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Manisha Patel
- Graduate Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
19
|
24-Epibrassinolide, a Phytosterol from the Brassinosteroid Family, Protects Dopaminergic Cells against MPP-Induced Oxidative Stress and Apoptosis. J Toxicol 2011; 2011:392859. [PMID: 21776258 PMCID: PMC3135132 DOI: 10.1155/2011/392859] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/07/2011] [Accepted: 03/28/2011] [Indexed: 12/03/2022] Open
Abstract
Oxidative stress and apoptosis are frequently cited to explain neuronal cell damage in various neurodegenerative disorders, such as Parkinson' s disease. Brassinosteroids (BRs) are phytosterols recognized to promote stress tolerance of vegetables via modulation of the antioxidative enzyme cascade. However, their antioxidative effects on mammalian neuronal cells have never been examined so far.
We analyzed the ability of 24-epibrassinolide (24-Epi), a natural BR, to protect neuronal PC12 cells from 1-methyl-4-phenylpyridinium- (MPP+-) induced oxidative stress and consequent apoptosis in dopaminergic neurons. Our results demonstrate that 24-Epi reduces the levels of intracellular reactive oxygen species and modulates superoxide dismutase, catalase, and glutathione peroxidase activities. Finally, we determined that the antioxidative properties of 24-Epi lead to the inhibition of MPP+-induced apoptosis by reducing DNA fragmentation as well as the Bax/Bcl-2 protein ratio and cleaved caspase-3. This is the first time that the potent antioxidant and neuroprotective role of 24-Epi has been shown in a mammalian neuronal cell line.
Collapse
|
20
|
Ghosh A, Chandran K, Kalivendi SV, Joseph J, Antholine WE, Hillard CJ, Kanthasamy A, Kanthasamy A, Kalyanaraman B. Neuroprotection by a mitochondria-targeted drug in a Parkinson's disease model. Free Radic Biol Med 2010; 49:1674-84. [PMID: 20828611 PMCID: PMC4020411 DOI: 10.1016/j.freeradbiomed.2010.08.028] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 08/05/2010] [Accepted: 08/27/2010] [Indexed: 12/21/2022]
Abstract
The objective of this study was to assess the neuroprotective effects of a mitochondria-targeted antioxidant, Mito-Q(10), the coenzyme-Q analog attached to a triphenylphosphonium cation that targets the antioxidant to mitochondria, in experimental models of Parkinson's disease (PD). Primary mesencephalic neuronal cells and cultured dopaminergic cells were treated with 1-methyl-4-phenylpyridinium (MPP(+)), an active metabolite of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and mice were used for testing the efficacy of Mito-Q(10). MPP(+) treatment caused a dose-dependent loss of tyrosine hydroxylase and membrane potential and an increase in caspase-3 activation in dopaminergic cells, which were reversed by Mito-Q(10). MPTP treatment induced a loss of striatal dopamine and its metabolites, inactivation of mitochondrial aconitase in the substantia nigra, and a loss of locomotor activity in mice. Treatment with Mito-Q(10) significantly inhibited both MPP(+)- and MPTP-induced neurotoxicity in cell culture and mouse models. Collectively, these results indicate that mitochondrial targeting of antioxidants is a promising neuroprotective strategy in this preclinical mouse model of PD.
Collapse
Affiliation(s)
- Anamitra Ghosh
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Karunakaran Chandran
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shasi V. Kalivendi
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Joy Joseph
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - William E. Antholine
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cecilia J. Hillard
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Corresponding author. Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Miwaukee, WI 53226, USA. Fax: +1 414 456 6512. (B. Kalyanaraman)
| |
Collapse
|
21
|
Huang JY, Chuang JI. Fibroblast growth factor 9 upregulates heme oxygenase-1 and gamma-glutamylcysteine synthetase expression to protect neurons from 1-methyl-4-phenylpyridinium toxicity. Free Radic Biol Med 2010; 49:1099-108. [PMID: 20615462 DOI: 10.1016/j.freeradbiomed.2010.06.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 05/14/2010] [Accepted: 06/22/2010] [Indexed: 10/19/2022]
Abstract
Oxidative stress and lower levels of trophic factors involved in nigrostriatal dopaminergic neurodegeneration are a hallmark of Parkinson disease. Our previous studies found that fibroblast growth factor 9 (FGF9) prevented 1-methyl-4-phenylpyridinium (MPP(+))-induced nigral dopaminergic neuron death and was involved in the neuroprotection of the antioxidant melatonin. However, the protective mechanisms mediated by FGF9 remain unclear. Herein, we explored whether FGF9 regulated the cellular antioxidant defense protecting dopaminergic neurons against MPP(+) intoxication. We found that FGF9 treatment alone induced a decrease in hydrogen peroxide (H(2)O(2)) level, an increase in glutathione content, and an upregulation of gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase 1 (HO-1) expression in primary cortical neurons but not in astrocytes. Simultaneous treatment with FGF9 and MPP(+) prevented MPP(+)-induced neuron death and H(2)O(2) overproduction but did not affect the FGF9-increased gamma-GCS and HO-1 protein expression. Inhibition of gamma-GCS or HO-1 prevented the inhibitory effect of FGF9 on MPP(+)-induced H(2)O(2) production and death in mesencephalic dopaminergic and cortical neurons. However, in the absence of MPP(+), the FGF9-induced H(2)O(2) reduction was blocked by HO-1 inhibitors, but not by gamma-GCS inhibitors. These results indicate that FGF9 upregulates gamma-GCS and HO-1 expression to protect cortical and dopaminergic neurons from MPP(+)-induced oxidative insult.
Collapse
Affiliation(s)
- Jui-Yen Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | | |
Collapse
|
22
|
Gupta A, Kumar A, Kulkarni SK. Licofelone attenuates MPTP-induced neuronal toxicity: behavioral, biochemical and cellular evidence. Inflammopharmacology 2010; 18:223-32. [PMID: 20697819 DOI: 10.1007/s10787-010-0052-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 07/13/2010] [Indexed: 01/14/2023]
Abstract
Neuroinflammation and oxidative stress play critical role in the pathophysiology of neurodegenerative diseases including Parkinson's disease (PD). Recent reports indicate the beneficial effect of anti-inflammatory drugs in attenuating the progression of PD. Therefore, the present study is aimed to evaluate the possible role of licofelone, a dual COX/LOX-inhibitor against MPTP-induced neurotoxicity in mice. Administration of MPTP (40 mg/kg in divided doses of four injections of 10 mg/kg, i.p. each at 1 h interval) significantly impaired locomotor activity and induced catatonia, oxidative damage (elevated levels of lipid peroxidation, superoxide anion and nitrite, and decreased levels of non-protein thiols) as compared with vehicle-treated animals. Biochemical studies revealed significant alterations in mitochondrial enzyme complex activities (decreased complex-I activity and mitochondrial viability) and increased levels of caspase-3 and NF-κB/p65 as compared to vehicle treated group. Licofelone (2.5, 5 or 10 mg/kg/day, p.o.) treatment for 7 days significantly improved locomotor activity, attenuated the severity of catatonia, oxidative damage and restored mitochondrial enzyme complex activity as compared to MPTP-treated group. Licofelone treatment also attenuated the expression of apoptotic factor (caspase-3) and transcription factor (NF-κB/p65) as compared to MPTP-treated group. The findings of the present study suggest that licofelone (dual inhibitor of COX and LOX) represents a new class of anti-inflammatory agent which may provide a novel therapeutic alternative for the treatment and management of PD.
Collapse
Affiliation(s)
- Amit Gupta
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Center of Advanced Study (UGC-CAS), Punjab University, Chandigarh, India
| | | | | |
Collapse
|
23
|
Levesque S, Wilson B, Gregoria V, Thorpe LB, Dallas S, Polikov VS, Hong JS, Block ML. Reactive microgliosis: extracellular micro-calpain and microglia-mediated dopaminergic neurotoxicity. ACTA ACUST UNITED AC 2010; 133:808-21. [PMID: 20123724 DOI: 10.1093/brain/awp333] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Microglia, the innate immune cells in the brain, can become chronically activated in response to dopaminergic neuron death, fuelling a self-renewing cycle of microglial activation followed by further neuron damage (reactive microgliosis), which is implicated in the progressive nature of Parkinson's disease. Here, we use an in vitro approach to separate neuron injury factors from the cellular actors of reactive microgliosis and discover molecular signals responsible for chronic and toxic microglial activation. Upon injury with the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium, N27 cells (dopaminergic neuron cell line) released soluble neuron injury factors that activated microglia and were selectively toxic to dopaminergic neurons in mixed mesencephalic neuron-glia cultures through nicotinamide adenine dinucleotide phosphate oxidase. mu-Calpain was identified as a key signal released from damaged neurons, causing selective dopaminergic neuron death through activation of microglial nicotinamide adenine dinucleotide phosphate oxidase and superoxide production. These findings suggest that dopaminergic neurons may be inherently susceptible to the pro-inflammatory effects of neuron damage, i.e. reactive microgliosis, providing much needed insight into the chronic nature of Parkinson's disease.
Collapse
Affiliation(s)
- Shannon Levesque
- Department of Anatomy & Neurobiology, Sanger Hall, Room 9-048, 1101 E. Marshall Street, Virginia Commonwealth University Medical Campus, Box 980709, Richmond, VA 23298-0709, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Martin HL, Teismann P. Glutathione--a review on its role and significance in Parkinson's disease. FASEB J 2009; 23:3263-72. [PMID: 19542204 DOI: 10.1096/fj.08-125443] [Citation(s) in RCA: 218] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting over a million people in the United States alone, and is characterized by rigidity, bradykinesia, resting tremor, and postural instability. Its main neuropathological feature is the loss of dopaminergic neurons of the substantia nigra pars compacta. However, the pathogenesis of this loss is not understood fully. One of the earliest biochemical changes seen in PD is a reduction in the levels of total glutathione, a key cellular antioxidant. Traditionally, it has been thought that this decrease in GSH levels is the consequence of increased oxidative stress, a process heavily implicated in PD pathogenesis. However, emerging evidence suggests that GSH depletion may itself play an active role in PD pathogenesis. This review aims to explore the contribution of GSH depletion to PD pathogenesis.
Collapse
Affiliation(s)
- Heather L Martin
- School of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | | |
Collapse
|
25
|
Jarrett SG, Milder JB, Liang LP, Patel M. The ketogenic diet increases mitochondrial glutathione levels. J Neurochem 2008; 106:1044-51. [DOI: 10.1111/j.1471-4159.2008.05460.x] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
26
|
Ardestani A, Yazdanparast R, Nejad AS. 2-Deoxy-d-ribose-induced oxidative stress causes apoptosis in human monocytic cells: Prevention by pyridoxal-5′-phosphate. Toxicol In Vitro 2008; 22:968-79. [DOI: 10.1016/j.tiv.2008.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2007] [Revised: 01/26/2008] [Accepted: 02/06/2008] [Indexed: 01/09/2023]
|
27
|
Neuroprotective effect of the natural iron chelator, phytic acid in a cell culture model of Parkinson's disease. Toxicology 2007; 245:101-8. [PMID: 18255213 DOI: 10.1016/j.tox.2007.12.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2007] [Revised: 12/14/2007] [Accepted: 12/17/2007] [Indexed: 12/15/2022]
Abstract
Disrupted iron metabolism and excess iron accumulation has been reported in the brains of Parkinson's disease (PD) patients. Because excessive iron can induce oxidative stress subsequently causing degradation of nigral dopaminergic neurons in PD, we determined the protective effect of a naturally occurring iron chelator, phytic acid (IP6), on 1-methyl-4-phenylpyridinium (MPP(+))-induced cell death in immortalized rat mesencephalic/dopaminergic cells. Cell death was induced with MPP(+) in normal and iron-excess conditions and cytotoxicity was measured by thiazolyl blue tetrazolium bromide (MTT assay) and trypan blue staining. Apoptotic cell death was also measured with caspase-3 activity, DNA fragmentation, and Hoechst nuclear staining. Compared to MPP(+) treatment, IP6 (30 micromol/L) increased cell viability by 19% (P<0.05) and decreased cell death by 22% (P<0.05). A threefold increase in caspase-3 activity (P<0.001) and a twofold increase in DNA fragmentation (P<0.05) with MPP(+) treatment was decreased by 55% (P<0.01) and 52% (P<0.05), respectively with IP6. Cell survival was increased by 18% (P<0.05) and 42% (P<0.001) with 30 and 100 micromol/L of IP6, respectively in iron-excess conditions. A 40% and 52% (P<0.001) protection was observed in caspase-3 activity with 30 and 100 micromol/L IP6, respectively in iron-excess condition. Similarly, a 45% reduction (P<0.001) in DNA fragmentation was found with 100 micromol/L IP6. In addition, Hoechst nuclear staining results confirmed the protective effect of IP6 against apoptosis. Similar protection was also observed with the differentiated cells. Collectively, our results demonstrate a significant neuroprotective effect of phytate in a cell culture model of PD.
Collapse
|